1
|
Maciel EVS, Habeck T, Meyners C, Lermyte F. Self-packed size-exclusion columns enable versatile high-throughput native, top-down, and ion mobility-mass spectrometry studies on proteins and complexes. Talanta 2025; 291:127868. [PMID: 40056653 DOI: 10.1016/j.talanta.2025.127868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Native MS (nMS) is a key structural biology technique that makes it possible to study intact proteins and their interactions. Unfortunately, non-volatile salts are incompatible with nMS, which demands a laborious desalting procedure. Non-denaturing size-exclusion chromatography (SEC) allows both rapid desalting and separation and has previously been explored for nMS automation. However, SEC at conventional scale requires rather large sample amounts as well as harsh ESI conditions, which can cause protein unfolding. Capillary LC allows softer conditions; however, the few commercially available SEC columns appropriate for this flow rate are prohibitively expensive for many laboratories. Existing protocols for packing buffer exchange columns rely on specialized equipment and/or result in columns that have limited capacity for size-based protein separation. Here, we present self-packed miniaturized SEC columns with different stationary phases and customizable dimensions. The columns, produced via slurry packing with an ordinary LC pump were used across a range of samples in several applications including nMS, top-down MS (TDMS), ligand screening, and ion mobility (IM)-MS. Native separation allowed acquisition of data from samples containing more than one protein. We acquired native TDMS data of 3 proteins in 12 min, with up to 47 % sequence coverage. IM-MS of alpha-synuclein at different charge states was measured in ca. 60 min (including calibrants), with results that match the literature. Finally, we used SEC-nMS to rapidly screen proteolysis-targeting chimera candidates and performed collision-induced unfolding (CIU) of a PROTAC-induced ternary complex. Through this work, we highlight the potential of SEC to support developments in structural MS.
Collapse
Affiliation(s)
- Edvaldo Vasconcelos Soares Maciel
- Clemens Schöpf Institute, Department of Chemistry, Technical University of Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany.
| | - Tanja Habeck
- Clemens Schöpf Institute, Department of Chemistry, Technical University of Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany.
| | - Christian Meyners
- Clemens Schöpf Institute, Department of Chemistry, Technical University of Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany.
| | - Frederik Lermyte
- Clemens Schöpf Institute, Department of Chemistry, Technical University of Darmstadt, Peter-Grünberg-Straße 4, 64287, Darmstadt, Germany.
| |
Collapse
|
2
|
Eisert J, Maciel EVS, Dederer V, Berwanger A, Bailey HJ, Đikić I, Knapp S, Empting M, Mathea S, Jensen H, Lermyte F. Native Taylor/Non-Taylor Dispersion-Mass Spectrometry (TNT-MS) Allows Rapid Protein Desalting and Multiplexed, Label-Free Ligand Screening. SMALL METHODS 2025:e2500658. [PMID: 40331427 DOI: 10.1002/smtd.202500658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Indexed: 05/08/2025]
Abstract
Native mass spectrometry (MS) is an important technique in structural biology and drug discovery, due to its ability to study non-covalent assemblies in the gas phase. Drawbacks include the incompatibility of electrospray ionization (ESI) with non-volatile salts and the risk of protein signal suppression by small molecules. Overcoming these often requires offline buffer exchange and/or parallel sample preparation to other methods, reducing the adoption and throughput of native MS. Here, we exploit the dynamics of analytes flowing through an open tubular capillary to keep molecules with a small hydrodynamic radius (e.g., salts) inside a Taylor dispersion regime while pushing larger species (e.g., proteins) into a non-Taylor regime. As such, larger species elute earlier, and are effectively buffer exchanged within the capillary in seconds. In addition to desalting of proteins injected in biologically relevant buffers we demonstrate separation of unbound small molecules from protein-ligand complexes, enabling multiplexed ligand screening. Finally, we investigated the dependence of the critical flow rate for non-Taylor behavior on protein size, enabling limited size-based separation of proteins. Taylor/non-Taylor dispersion mass spectrometry (TNT-MS) was implemented using an unmodified liquid chromatography - mass spectrometry (LC-MS) system operated without a chromatographic column and coupled to an autosampler, which allowed significant automation.
Collapse
Affiliation(s)
- Jonathan Eisert
- Department of Chemistry, Clemens-Schöpf-Institute of Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, 64287, Darmstadt, Germany
| | - Edvaldo Vasconcelos Soares Maciel
- Department of Chemistry, Clemens-Schöpf-Institute of Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, 64287, Darmstadt, Germany
| | - Verena Dederer
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Aylin Berwanger
- Helmholtz Institut for Pharmaceutical Research Saarland (HIPS)/Helmholtz Center for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Henry J Bailey
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, 60438, Frankfurt am Main, Germany
| | - Ivan Đikić
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, 60438, Frankfurt am Main, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60596, Frankfurt am Main, Germany
| | - Martin Empting
- Helmholtz Institut for Pharmaceutical Research Saarland (HIPS)/Helmholtz Center for Infection Research (HZI), Campus E8.1, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, 66123, Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123, Saarbrücken, Germany
| | - Sebastian Mathea
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
- Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Henrik Jensen
- Fida Biosystems Aps, Generatorvej 6, Soborg, 2860, Denmark
| | - Frederik Lermyte
- Department of Chemistry, Clemens-Schöpf-Institute of Chemistry and Biochemistry, Technical University of Darmstadt, Peter-Grünberg-Strasse 4, 64287, Darmstadt, Germany
| |
Collapse
|
3
|
Gomes FP, Durbin KR, Schauer K, Nwachukwu JC, R Kobylski R, Njeri JW, Seath CP, Saviola AJ, McClatchy DB, Diedrich JK, Garrett PT, Papa AB, Ciolacu I, Kelleher NL, Nettles KW, Yates JR. Native top-down proteomics enables discovery in endocrine-resistant breast cancer. Nat Chem Biol 2025:10.1038/s41589-025-01866-8. [PMID: 40186031 DOI: 10.1038/s41589-025-01866-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 02/24/2025] [Indexed: 04/07/2025]
Abstract
Oligomerization of proteoforms produces functional protein complexes. Characterization of these assemblies within cells is critical to understanding the molecular mechanisms involved in disease and to designing effective drugs. Here we present a native top-down proteomics (nTDP) strategy to identify protein assemblies (≤70 kDa) in breast cancer cells and in cells that overexpress epidermal growth factor receptor (EGFR), which serves as a resistance model of estrogen receptor-alpha (ER)-targeted therapies. This nTDP approach identified ~104 complexoforms from 17 protein complexes, which revealed several molecular features of the breast cancer proteome, including EGFR-induced dissociation of nuclear transport factor 2 (NUTF2) assemblies that modulate ER activity. We found that the K4 and K55 post-translational modification sites discovered with nTDP differentially impact the effects of NUTF2 on the inhibition of the ER signaling pathway. The characterization of endogenous proteoform-proteoform/ligand interactions revealed the molecular diversity of complexoforms and their role in breast cancer growth.
Collapse
Affiliation(s)
| | | | | | - Jerome C Nwachukwu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | - Robin R Kobylski
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Jacqline W Njeri
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA
| | - Ciaran P Seath
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
| | | | | | | | | | - Alexandra B Papa
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- Florida Atlantic University, Jupiter, FL, USA
| | - Ianis Ciolacu
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA
- Florida Atlantic University, Jupiter, FL, USA
| | - Neil L Kelleher
- Proteinaceous, Evanston, IL, USA
- Northwestern University, National Resource for Translational and Developmental Proteomics, Evanston, IL, USA
| | - Kendall W Nettles
- Department of Immunology and Microbiology, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- The Skaggs Graduate School of Chemical and Biological Sciences, The Scripps Research Institute, La Jolla, CA, USA.
| | | |
Collapse
|
4
|
Campuzano IDG, Loo JA. Evolution of Mass Spectrometers for High m/ z Biological Ion Formation, Transmission, Analysis and Detection: A Personal Perspective. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:632-652. [PMID: 40043050 DOI: 10.1021/jasms.4c00348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Mass spectrometry (MS) has become an essential tool in virtually all academic, pharmaceutical, and biopharmaceutical analytical laboratories. The specialized and bespoke area of MS research and application of high m/z ion (>m/z 6000 and high mass, >150 kDa) formation, transmission, analysis, and detection is a relatively new area of focus for MS that has seen dramatic acceleration in interest over the last two decades. Herein we delve into this exciting aspect of MS, discussing how MS instrumentation has been refined and evolved for native-MS analysis. We cover the early groundbreaking experiments showing high m/z ion formation, transmission, and preservation of protein structure in the gas phase. Additionally, we discuss specific instrument optimizations and modifications that have advanced high m/z ion generation, transmission, analysis, and detection, contributing to the research area known as gas-phase structural biology. Native-MS sample introduction methods, emerging technologies, and future perspectives are also examined. Finally, we share personal opinions, observations, and experiences that are new to the community or previously unpublished.
Collapse
Affiliation(s)
- Iain D G Campuzano
- Large Molecule Discovery and Research Data Science, Amgen Research, Amgen, Thousand Oaks, California 91320, United States
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, University of California-Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
5
|
Fischer MS, Rogers HT, Chapman EA, Jin S, Ge Y. Native Top-Down Proteomics of Endogenous Protein Complexes Enabled by Online Two-Dimensional Liquid Chromatography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645965. [PMID: 40236213 PMCID: PMC11996319 DOI: 10.1101/2025.03.28.645965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Protein complexes are essential for virtually all biological processes, yet their structural characterization remains a major challenge due to their heterogeneous, dynamic nature and the complexity of the proteome. Native top-down mass spectrometry (nTDMS) has emerged as a powerful tool for comprehensive structural characterization of purified protein complexes, but its application to endogenous protein complexes in the proteome is challenging and typically requires labor-intensive and time-consuming prefractionation. Here, for the first time, we develop a nondenaturing online two-dimensional liquid chromatography (2D-LC) method for native top-down proteomics (nTDP), enabling high-throughput structural analysis of endogenous protein complexes. The automated, online interfacing of size-exclusion and mixed-bed ion-exchange chromatography achieves high coverage of endogenous protein complexes. We further develop a multistage nTDMS approach that enables comprehensive structural characterization within the chromatographic timescale, capturing intact non-covalent complexes, released subunits/cofactors, and backbone fragments. Our analysis detected 133 native proteoforms and endogenous protein complexes (up to 350 kDa) from human heart tissue in less than two hours. Such technological leaps in high-throughput structural characterization of endogenous protein complexes will advance large-scale nTDP studies in health and disease.
Collapse
|
6
|
Liu X, Roberts DS, Bingman CA, Jin S, Ge Y, Gellman SH. Rational design of a foldon-derived heterotrimer guided by quantitative native mass spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645370. [PMID: 40196687 PMCID: PMC11974886 DOI: 10.1101/2025.03.25.645370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Designing stable hetero-oligomeric protein complexes with defined inter-subunit stoichiometries remains a significant challenge. In this study, we report the design of a highly selective heterotrimeric assembly derived from the well-known foldon homotrimer. We generated an aab heterotrimer by introducing the Q11E modification to destabilize the homotrimer and a compensatory modification, either V14A or V14L, to stabilize the hydrophobic core of the heterotrimer. Native mass spectrometry (MS) was essential for guiding the design process, enabling precise characterization of oligomeric states and their equilibrium distributions. The heterotrimer structure was validated by x-ray crystallography. Our findings highlight the effectiveness of combining rational design with native MS to develop specific hetero-oligomeric assemblies.
Collapse
|
7
|
Zhang Z, Liu AP, Wang H, Schuessler HA. Post-column denaturation-assisted hydrophobic interaction chromatography-mass spectrometry for rapid and in-depth characterization of positional isomers in cysteine-based antibody-drug conjugates. J Pharm Biomed Anal 2025; 255:116635. [PMID: 39700863 DOI: 10.1016/j.jpba.2024.116635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a significant advancement in targeted cancer therapy, offering the potential to selectively deliver cytotoxic drugs to tumor cells while minimizing systemic toxicity. However, the structural complexity of ADCs, particularly those conjugated through cysteine residues, poses significant analytical challenges. Due to the hydrophobicity of ADCs, Hydrophobic interaction chromatography (HIC) is often the method of choice to analyze the drug-to-antibody ratio (DAR). However, it requires high-concentration salts, which are often incompatible with mass spectrometry (MS) analysis. By employing ammonium acetate as an MS-compatible salt and integrating a 4-way liquid junction cross configuration for simultaneous introduction of the makeup flow and splitting the flow right before coupling to a mass spectrometer, we achieve high-quality separation and sensitive mass spectrometric analysis. This innovative setup allows for simultaneous DAR measurement and positional isomer characterization by switching the makeup flow solvent from water to a denaturation solution. Our method offers a streamlined and effective approach to ADC characterization, facilitating the identification of positional isomers without the need for fractionation or multiple chromatographic steps. The versatility and robustness of this HIC-MS method are demonstrated through the analysis of two ADCs, highlighting its potential for broad application in ADC development and quality control.
Collapse
Affiliation(s)
- Zhengqi Zhang
- Analytical Research & Development, Merck & Co., Inc., Rahway, NJ 07065, USA.
| | - Anita P Liu
- Analytical Research & Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Hongxia Wang
- Analytical Research & Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | | |
Collapse
|
8
|
He W, Yu S, Li J, Li S, Chen Z, Zhang J, Liu Y, Zhou M, Yang T, Cheng W, Dai SS. From inflammation to remodelling: A novel BASP1 + monocyte subset as a catalyst for acute aortic dissection. J Adv Res 2025:S2090-1232(25)00144-4. [PMID: 40057028 DOI: 10.1016/j.jare.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/10/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025] Open
Abstract
INTRODUCTION Monocytes comprise heterogeneous cell populations. However, beyond traditionally considered as precursors of tissue macrophages, heterogeneity and detailed effects of monocytes in acute aortic dissection (AAD) are largely unknown. OBJECTIVES To investigate the role of brain soluble acid protein 1 positive (BASP1+) monocyte subset in promoting AAD development as well as the underlying mechanism. METHODS Monocyte/macrophage heterogeneity in both human peripheral blood and aortic tissues were assayed by scRNA-seq. Monocyte trafficking and lineage tracing were detected by immunofluorescence and using BASP1-CreER/Lyz2-DreER-tdT reporter mice with AAD. The effects and underlying mechanism were investigated by laser speckle image, ultrasound imaging, Co-IP, ChIP-sequencing. Conditional knockout of BASP1 on monocyte and BASP1 siRNA were used to observe BASP1+ monocyte subset-targeted AAD intervention. RESULTS "PIP2-SP1-ACTN1/VAV3" and "ITGB1-Rac1-GSN" signalling mediated BASP1+ monocyte subset as the first line immune cells infiltrating aortic tissues in AAD induction and partial of them transformed to BASP1+ macrophages to amplify the inflammation. Meanwhile, BASP1+ monocyte subset induced an inflammatory phenotype vascular smooth muscle cell (VSMC) subset and a ROS-enriched endothelial cell (EC) subset accompanied with promoting the apoptosis of normal VSMC and EC, contributing to vascular remodelling and dampening the myo-endothelial gap junction. Selective deletion of BASP1+ monocyte subset and interference with BASP1 expression in monocytes both inhibited the development of AAD in mice. CONCLUSION Interpretation BASP1+ monocyte subset and its regulatory network provides deep insight into AAD pathogenesis and a novel potential target for early intervention in AAD formation.
Collapse
Affiliation(s)
- Wenhui He
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Sanjiu Yu
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Siyu Li
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Zongtao Chen
- Health Management Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China
| | - Jingyu Zhang
- Department of Neurosurgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yangwuyue Liu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Mi Zhou
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Teng Yang
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China.
| | - Shuang-Shuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
9
|
Refaat H, Ahmad A, Kamel E, Gurvich VJ, Rathore AS, Suryanarayanan R, Nejadnik R. Challenges with effective removal of surfactants from monoclonal antibody formulations. Int J Pharm 2025; 670:125146. [PMID: 39743160 DOI: 10.1016/j.ijpharm.2024.125146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/22/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Buffer exchange is a critical step in biologics development, playing a pivotal role in removing contaminants, adjusting sample conditions, and facilitating compatibility studies. The efficiency of centrifugal concentrators for polysorbate removal was compared to a two-step approach involving a surfactant removal column followed by buffer exchange. Trastuzumab-pkrb from Herzuma® was used. While a 30 kDa centrifugal concentrator was ineffective in polysorbate removal, a 50 kDa concentrator caused partial removal. Surfactant removal column proved more effective in removing polysorbates. Buffer exchange using polysorbate-containing formulation buffer, even with a 50 kDa concentrator, accumulated polysorbate, revealing the need for a different approach in small-scale formulation. Adding polysorbate in a separate step after buffer exchange appeared to be a good strategy to prevent this problem. The two approaches did not reveal any differences in the protein aggregation behavior.
Collapse
Affiliation(s)
- Hesham Refaat
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, the United States of America
| | - Aziz Ahmad
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, the United States of America
| | - Eman Kamel
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, the United States of America
| | - Vadim J Gurvich
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, the United States of America
| | - Anurag S Rathore
- Department of Chemical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Raj Suryanarayanan
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, the United States of America.
| | - Reza Nejadnik
- Department of Pharmaceutical Sciences & Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, the United States of America.
| |
Collapse
|
10
|
Dowling QM, Park YJ, Fries CN, Gerstenmaier NC, Ols S, Yang EC, Wargacki AJ, Dosey A, Hsia Y, Ravichandran R, Walkey CD, Burrell AL, Veesler D, Baker D, King NP. Hierarchical design of pseudosymmetric protein nanocages. Nature 2025; 638:553-561. [PMID: 39695230 PMCID: PMC11821544 DOI: 10.1038/s41586-024-08360-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/06/2024] [Indexed: 12/20/2024]
Abstract
Discrete protein assemblies ranging from hundreds of kilodaltons to hundreds of megadaltons in size are a ubiquitous feature of biological systems and perform highly specialized functions1,2. Despite remarkable recent progress in accurately designing new self-assembling proteins, the size and complexity of these assemblies has been limited by a reliance on strict symmetry3. Here, inspired by the pseudosymmetry observed in bacterial microcompartments and viral capsids, we developed a hierarchical computational method for designing large pseudosymmetric self-assembling protein nanomaterials. We computationally designed pseudosymmetric heterooligomeric components and used them to create discrete, cage-like protein assemblies with icosahedral symmetry containing 240, 540 and 960 subunits. At 49, 71 and 96 nm diameter, these nanocages are the largest bounded computationally designed protein assemblies generated to date. More broadly, by moving beyond strict symmetry, our work substantially broadens the variety of self-assembling protein architectures that are accessible through design.
Collapse
Affiliation(s)
- Quinton M Dowling
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Chelsea N Fries
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Neil C Gerstenmaier
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Sebastian Ols
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Erin C Yang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Adam J Wargacki
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Annie Dosey
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Yang Hsia
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Rashmi Ravichandran
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Carl D Walkey
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Anika L Burrell
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - David Baker
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Neil P King
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| |
Collapse
|
11
|
Lanzillotti MB, Brodbelt JS. Progress in Tandem Mass Spectrometry Data Analysis for Nucleic Acids. MASS SPECTROMETRY REVIEWS 2025. [PMID: 39797409 DOI: 10.1002/mas.21923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025]
Abstract
Mass spectrometry (MS) has become a critical tool in the characterization of covalently modified nucleic acids. Well-developed bottom-up approaches, where nucleic acids are digested with an endonuclease and the resulting oligonucleotides are separated before MS and MS/MS analysis, provide substantial insight into modified nucleotides in biological and synthetic nucleic. Top-down MS presents an alternative approach where the entire nucleic acid molecule is introduced to the mass spectrometer intact and then fragmented by MS/MS. Current top-down MS workflows have incorporated automated, on-line HPLC workflows to enable rapid desalting of nucleic acid samples for facile mass analysis without complication from adduction. Furthermore, optimization of MS/MS parameters utilizing collision, electron, or photon-based activation methods have enabled effective bond cleavage throughout the phosphodiester backbone while limiting secondary fragmentation, allowing characterization of progressively larger (~100 nt) nucleic acids and localization of covalent modifications. Development of software applications to perform automated identification of fragment ions has accelerated the broader adoption of mass spectrometry for analysis of nucleic acids. This review focuses on progress in tandem mass spectrometry for characterization of nucleic acids with particular emphasis on the software tools that have proven critical for advancing the field.
Collapse
|
12
|
Su P, McGee JP, Hollas MAR, Fellers RT, Durbin KR, Greer JB, Early BP, Yip PF, Zabrouskov V, Srzentić K, Senko MW, Compton PD, Kelleher NL, Kafader JO. Standardized workflow for multiplexed charge detection mass spectrometry on orbitrap analyzers. Nat Protoc 2025:10.1038/s41596-024-01091-y. [PMID: 39747675 DOI: 10.1038/s41596-024-01091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/11/2024] [Indexed: 01/04/2025]
Abstract
Individual ion mass spectrometry (I2MS) is the Orbitrap-based extension of the niche mass spectrometry technique known as charge detection mass spectrometry (CDMS). While traditional CDMS analysis is performed on in-house-built instruments such as the electrostatic linear ion trap, I2MS extends CDMS analysis to Orbitrap analyzers, allowing charge detection analysis to be available to the scientific community at large. I2MS simultaneously measures the mass-to-charge ratios (m/z) and charges (z) of hundreds to thousands of individual ions within one acquisition event, creating a spectral output directly into the mass domain without the need for further spectral deconvolution. A mass distribution or 'profile' can be created for any desired sample regardless of composition or heterogeneity. To assist in reducing I2MS analysis to practice, we developed this workflow for data acquisition and subsequent data analysis, which includes (i) protein sample preparation, (ii) attenuation of ion signals to obtain individual ions, (iii) the creation of a charge-calibration curve from standard proteins with known charge states and finally (iv) producing a meaningful mass spectral output from a complex or unknown sample by using the STORIboard software. This protocol is suitable for users with prior experience in mass spectrometry and bioanalytical chemistry. First, the analysis of protein standards in native and denaturing mode is presented, setting the foundation for the analysis of complex mixtures that are intractable via traditional mass spectrometry techniques. Examples of complex mixtures included here demonstrate the relevant analysis of an intact human monoclonal antibody and its intricate glycosylation patterns.
Collapse
Affiliation(s)
- Pei Su
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering and the Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - John P McGee
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering and the Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
- ImmPro, Inc., Evanston, IL, USA
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Michael A R Hollas
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Ryan T Fellers
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kenneth R Durbin
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
- Proteinaceous, Inc., Evanston, IL, USA
| | - Joseph B Greer
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Bryan P Early
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Ping F Yip
- Thermo Fisher Scientific, San Jose, CA, USA
| | | | | | | | - Philip D Compton
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
- Integrated Protein Technologies, Evanston, IL, USA
| | - Neil L Kelleher
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering and the Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Jared O Kafader
- Departments of Molecular Biosciences, Chemistry and Chemical and Biological Engineering and the Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
- Proteomics Center of Excellence, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
13
|
Li Y, Li W, Zheng Y, Wang T, Pu R, Zhang Z. Desalting strategies for native mass spectrometry. Talanta 2025; 281:126824. [PMID: 39250868 DOI: 10.1016/j.talanta.2024.126824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
In native mass spectrometry (MS) salts are indispensable for preserving the native structures of biomolecules, but detrimental to mass sensitivity, resolution, and accuracy. Such a conflict makes desalting in native MS more challenging, distinctive, and sample-dependent than in peptide-centric MS. This review first briefly introduces the charged residue mechanism whereby native-like gaseous protein ions are released from electrospray droplets, revealing a higher degree of salt adduction than denatured proteins. Subsequently, this review summarizes and explores the existing strategies, underlying mechanisms and future perspectives of desalting in native MS. These strategies mainly focus on buffer exchange into volatile salts (offline and online approaches), addition of solution additives (e.g., anion, supercharging reagent, solution phase chelator and amino acid), use of submicron electrospray emitters (down to 60 nm), and other potential approaches (e.g., induced and electrophoretic nanoelectrospray ionization). The strategies of online buffer exchange and using nanoscale electrospray emitters are highlighted. This review would not only be a valuable addition to the field of sample preparation in MS, but would also serve as a beginner's guide to desalting in native MS.
Collapse
Affiliation(s)
- Yun Li
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Weijie Li
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Yajun Zheng
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.
| | - Tong Wang
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Ruijin Pu
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China
| | - Zhiping Zhang
- School of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, 710065, China.
| |
Collapse
|
14
|
Kibler RD, Lee S, Kennedy MA, Wicky BIM, Lai SM, Kostelic MM, Carr A, Li X, Chow CM, Nguyen TK, Carter L, Wysocki VH, Stoddard BL, Baker D. Design of pseudosymmetric protein hetero-oligomers. Nat Commun 2024; 15:10684. [PMID: 39695145 PMCID: PMC11655659 DOI: 10.1038/s41467-024-54913-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
Pseudosymmetric hetero-oligomers with three or more unique subunits with overall structural (but not sequence) symmetry play key roles in biology, and systematic approaches for generating such proteins de novo would provide new routes to controlling cell signaling and designing complex protein materials. However, the de novo design of protein hetero-oligomers with three or more distinct chains with nearly identical structures is a challenging unsolved problem because it requires the accurate design of multiple protein-protein interfaces simultaneously. Here, we describe a divide-and-conquer approach that breaks the multiple-interface design challenge into a set of more tractable symmetric single-interface redesign tasks, followed by structural recombination of the validated homo-oligomers into pseudosymmetric hetero-oligomers. Starting from de novo designed circular homo-oligomers composed of 9 or 24 tandemly repeated units, we redesigned the inter-subunit interfaces to generate 19 new homo-oligomers and structurally recombined them to make 24 new hetero-oligomers, including ABC heterotrimers, A2B2 heterotetramers, and A3B3 and A2B2C2 heterohexamers which assemble with high structural specificity. The symmetric homo-oligomers and pseudosymmetric hetero-oligomers generated for each system have identical or nearly identical backbones, and hence are ideal building blocks for generating and functionalizing larger symmetric and pseudosymmetric assemblies.
Collapse
Affiliation(s)
- Ryan D Kibler
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Sangmin Lee
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Madison A Kennedy
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98006, USA
| | - Basile I M Wicky
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Stella M Lai
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Marius M Kostelic
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Ann Carr
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Cameron M Chow
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Tina K Nguyen
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH, 43210, USA
| | - Barry L Stoddard
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98006, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
15
|
Kim D, Liu W, Viner R, Cherezov V. Native mass spectrometry prescreening of G protein-coupled receptor complexes for cryo-EM structure determination. Structure 2024; 32:2206-2219.e4. [PMID: 39471802 PMCID: PMC11625002 DOI: 10.1016/j.str.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 11/01/2024]
Abstract
G protein-coupled receptors (GPCRs) are essential transmembrane proteins playing key roles in human health and disease. Understanding their atomic-level molecular structure and conformational states is imperative for advancing drug development. Recent breakthroughs in single-particle cryogenic electron microscopy (cryo-EM) have propelled the structural biology of GPCRs into a new era. Nevertheless, the preparation of suitable GPCR samples and their complexes for cryo-EM analysis remains challenging due to their poor stability and highly dynamic nature. Here, we present our online buffer exchange-native MS method combined with Direct Mass Technology (OBE-nMS+DMT) which facilitates high-throughput analysis and guides sample preparation. We applied this method to optimize the GPR119-Gs complex sample prior to cryo-EM analysis, leading to a 3.51 Å resolution structure from only 396 movies collected on a 200 kV Glacios. This study suggests that the OBE-nMS+DMT method emerges as a powerful tool for prescreening sample conditions in cryo-EM studies of GPCRs and other membrane protein complexes.
Collapse
Affiliation(s)
- Donggyun Kim
- Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Weijing Liu
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA 95134, USA
| | - Rosa Viner
- Thermo Fisher Scientific, 355 River Oaks Pkwy, San Jose, CA 95134, USA.
| | - Vadim Cherezov
- Bridge Institute, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
16
|
Habeck T, Brown KA, Des Soye B, Lantz C, Zhou M, Alam N, Hossain MA, Jung W, Keener JE, Volny M, Wilson JW, Ying Y, Agar JN, Danis PO, Ge Y, Kelleher NL, Li H, Loo JA, Marty MT, Paša-Tolić L, Sandoval W, Lermyte F. Top-down mass spectrometry of native proteoforms and their complexes: a community study. Nat Methods 2024; 21:2388-2396. [PMID: 38744918 PMCID: PMC11561160 DOI: 10.1038/s41592-024-02279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
The combination of native electrospray ionization with top-down fragmentation in mass spectrometry (MS) allows simultaneous determination of the stoichiometry of noncovalent complexes and identification of their component proteoforms and cofactors. Although this approach is powerful, both native MS and top-down MS are not yet well standardized, and only a limited number of laboratories regularly carry out this type of research. To address this challenge, the Consortium for Top-Down Proteomics initiated a study to develop and test protocols for native MS combined with top-down fragmentation of proteins and protein complexes across 11 instruments in nine laboratories. Here we report the summary of the outcomes to provide robust benchmarks and a valuable entry point for the scientific community.
Collapse
Affiliation(s)
- Tanja Habeck
- Technische Universität Darmstadt, Darmstadt, Germany
| | - Kyle A Brown
- University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Mowei Zhou
- Pacific Northwest National Laboratory, Richland, WA, USA
- Zhejiang University, Zhejiang, China
| | | | | | | | | | | | - Jesse W Wilson
- Pacific Northwest National Laboratory, Richland, WA, USA
| | - Yujia Ying
- Sun Yat-sen University, Guangzhou, China
| | - Jeffrey N Agar
- Northeastern University, Boston, MA, USA
- Consortium for Top-Down Proteomics, Cambridge, MA, USA
| | - Paul O Danis
- Consortium for Top-Down Proteomics, Cambridge, MA, USA
| | - Ying Ge
- University of Wisconsin-Madison, Madison, WI, USA
- Consortium for Top-Down Proteomics, Cambridge, MA, USA
| | - Neil L Kelleher
- Northwestern University, Evanston, IL, USA
- Consortium for Top-Down Proteomics, Cambridge, MA, USA
| | - Huilin Li
- Sun Yat-sen University, Guangzhou, China
| | - Joseph A Loo
- University of California, Los Angeles, CA, USA
- Consortium for Top-Down Proteomics, Cambridge, MA, USA
| | | | - Ljiljana Paša-Tolić
- Pacific Northwest National Laboratory, Richland, WA, USA
- Consortium for Top-Down Proteomics, Cambridge, MA, USA
| | | | | |
Collapse
|
17
|
Bai X, Xu K, Zhao Z, Qin H, Nam KH, Quan C, Ha NC, Xu Y. Structural and Biochemical Analysis of Butanol Dehydrogenase From Thermotoga maritima. Proteins 2024; 92:1357-1365. [PMID: 39023292 DOI: 10.1002/prot.26731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/10/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
Butanol dehydrogenase (BDH) plays a crucial role in butanol biosynthesis by catalyzing the conversion of butanal to butanol using the coenzyme NAD(P)H. In this study, we observed that BDH from Thermotoga maritima (TmBDH) exhibits dual coenzyme specificity and catalytic activity with NADPH as the coenzyme under highly alkaline conditions. Additionally, a thermal stability analysis on TmBDH demonstrated its excellent activity retention even at elevated temperatures of 80°C. These findings demonstrate the superior thermal stability of TmBDH and suggest that it is a promising candidate for large-scale industrial butanol production. Furthermore, we discovered that TmBDH effectively catalyzes the conversion of aldehydes to alcohols and exhibits a wide range of substrate specificities toward aldehydes, while excluding alcohols. The dimeric state of TmBDH was observed using rapid online buffer exchange native mass spectrometry. Additionally, we analyzed the coenzyme-binding sites and inferred the possible locations of the substrate-binding sites. These results provide insights that improve our understanding of BDHs.
Collapse
Affiliation(s)
- Xue Bai
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Ke Xu
- Department of Ophthalmology, The Fourth People's Hospital of Shenyang, Shenyang, China
| | - Zhidan Zhao
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Huiwen Qin
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, China
| | - Ki Hyun Nam
- College of General Education, Kookmin University, Seoul, South Korea
| | - Chunshan Quan
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| | - Nam-Chul Ha
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Yongbin Xu
- Department of Bioengineering, College of Life Science, Dalian Minzu University, Dalian, Liaoning, China
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, College of Life Science, Dalian Minzu University, Dalian, China
| |
Collapse
|
18
|
Wang Q, Wang Q, Qi Z, Moeller W, Wysocki VH, Sun L. Native Proteomics by Capillary Zone Electrophoresis-Mass Spectrometry. Angew Chem Int Ed Engl 2024; 63:e202408370. [PMID: 39196601 PMCID: PMC11646347 DOI: 10.1002/anie.202408370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/21/2024] [Accepted: 08/28/2024] [Indexed: 08/29/2024]
Abstract
Native proteomics measures endogenous proteoforms and protein complexes under a near physiological condition using native mass spectrometry (nMS) coupled with liquid-phase separations. Native proteomics should provide the most accurate bird's-eye view of proteome dynamics within cells, which is fundamental for understanding almost all biological processes. nMS has been widely employed to characterize well-purified protein complexes. However, there are only very few trials of utilizing nMS to measure proteoforms and protein complexes in a complex sample (i.e., a whole cell lysate). Here, we pioneer the native proteomics measurement of large proteoforms or protein complexes up to 400 kDa from a complex proteome via online coupling of native capillary zone electrophoresis (nCZE) to an ultra-high mass range (UHMR) Orbitrap mass spectrometer. The nCZE-MS technique enabled the measurement of a 115-kDa standard protein complex while consuming only about 0.1 ng of protein material. nCZE-MS analysis of an E.coli cell lysate detected 72 proteoforms or protein complexes in a mass range of 30-400 kDa in a single run while consuming only 50-ng protein material. The mass distribution of detected proteoforms or protein complexes agreed well with that from mass photometry measurement. This work represents a technical breakthrough in native proteomics for measuring complex proteomes.
Collapse
Affiliation(s)
- Qianyi Wang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
- Plant Resilience Institute, Michigan State University, East Lansing, Michigan 48824, USA
| | - Qianjie Wang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Zihao Qi
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
- Native MS Guided Structural Biology Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - William Moeller
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
- Native MS Guided Structural Biology Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio 43210, United States
- Native MS Guided Structural Biology Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, Michigan 48824, United States
| |
Collapse
|
19
|
Saikusa K. Advancing Native Mass Spectrometry Toward Cellular Biology. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5095. [PMID: 39445718 DOI: 10.1002/jms.5095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024]
Abstract
Protein structure, including various post-translational modifications and higher-order structures, regulates diverse biological functions. Native mass spectrometry (native MS) is a powerful analytical technique used to determine the masses of biomolecules, such as proteins and their complexes, while preserving their native folding in solution. This method provides structural information on the composition of monomers or complexes and the stoichiometry of subunits within each complex, significantly contributing to protein structural analysis. Native MS has evolved to incorporate top-down approaches, enabling the characterization of proteoforms and non-covalent interactions between metabolites or proteins and specific targets. This perspective highlights the advancements in native MS for intracellular proteins and protein complexes, and discusses future research directions toward cellular biology.
Collapse
Affiliation(s)
- Kazumi Saikusa
- National Metrology Institute of Japan (NMIJ), National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| |
Collapse
|
20
|
Sanders JD, Owen ON, Tran BH, Mosqueira JL, Marty MT. Coupling Online Size Exclusion Chromatography with Charge Detection-Mass Spectrometry Using Hadamard Transform Multiplexing. Anal Chem 2024; 96:16743-16749. [PMID: 39393347 DOI: 10.1021/acs.analchem.4c03248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Charge detection mass spectrometry (CD-MS) is a powerful technique for the analysis of large, heterogeneous biomolecules. By directly measuring the charge states of individual ions, CD-MS can measure the masses from spectra where conventional deconvolution approaches fail due to the lack of isotopic resolution or distinguishable charge states. However, CD-MS is inherently slow because hundreds or thousands of spectra need to be collected to produce adequate ion statistics. The slower speed of CD-MS complicates efforts to couple it with online separation techniques, which limit the number of spectra that can be acquired during a chromatographic peak. Here, we present the application of Hadamard transform multiplexing to online size exclusion chromatography (SEC) coupled with Orbitrap CD-MS, with a goal of using SEC for separating complex mixtures prior to CD-MS analysis. We developed a microcontroller to deliver pulsed injections from a large sample loop onto a SEC for online CD-MS analysis. Data showed a series of peaks spaced according to the pseudorandom injection sequence, which were demultiplexed with a Hadamard transform algorithm. The demultiplexed data revealed improved CD-MS signals while preserving retention time information. This multiplexing approach provides a general solution to the inherent incompatibilities of online separations and CD-MS detection that will enable a range of applications.
Collapse
Affiliation(s)
- James D Sanders
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| | - October N Owen
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| | - Brian H Tran
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| | - Jeffrey L Mosqueira
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| | - Michael T Marty
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
21
|
Tiambeng TN, Yan Y, Patel SK, Cotham VC, Wang S, Li N. Characterization of adeno-associated virus capsid proteins using denaturing size-exclusion chromatography coupled with mass spectrometry. J Pharm Biomed Anal 2024; 253:116524. [PMID: 39442445 DOI: 10.1016/j.jpba.2024.116524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/26/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
Recombinant adeno-associated viruses (AAVs) are a highly effective platform for gene delivery for the treatment of many human diseases. Characterization of AAV viral protein attributes (VP), such as serotype identity, VP stoichiometry, and VP post-translational modifications, is essential to ensure product and process consistency. While size-exclusion chromatography (SEC) coupled with mass spectrometry (MS) is commonly used in the biopharmaceutical industry for analyzing protein therapeutics, its application to intact AAV VP components has not gained traction, presumably due to difficulties in achieving adequate resolution of VP(1-3) monomers. Herein, we describe the development of a denaturing SEC method and optimization of SEC parameters, including stationary phase pore size, column temperature, and mobile phase composition, to achieve effective chromatographic separation of VP(1-3). We demonstrate that an optimized dSEC-MS method featuring MS-compatible formic acid, can effectively separate VP(1-3) across AAV1, 2, 5, 6, 8, and 9 serotypes using a single column and mobile phase condition. A case study was included to showcase successful application of the dSEC-MS method in analyzing changes across different AAV production processes, yielding similar conclusions to an orthogonal approach, such as hydrophilic interaction chromatography (HILIC)- MS. Additionally, dSEC integrated with fluorescence (FLR) and ultraviolet (UV) detection can be used to semi-quantitatively identify both AAV DNA and VP components from empty and full AAV samples. Overall, this robust and MS-friendly methodological advancement could greatly streamline the development and analytical quality control processes for AAV-based gene therapies, providing a highly sensitive method for intact VP characterization.
Collapse
Affiliation(s)
- Timothy N Tiambeng
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591-6707, USA
| | - Yuetian Yan
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591-6707, USA.
| | - Shailin K Patel
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591-6707, USA
| | - Victoria C Cotham
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591-6707, USA
| | - Shunhai Wang
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591-6707, USA.
| | - Ning Li
- Analytical Chemistry, Regeneron Pharmaceuticals Inc., Tarrytown, NY 10591-6707, USA
| |
Collapse
|
22
|
Shao X, Tian M, Yin J, Duan H, Tian Y, Wang H, Xia C, Wang Z, Zhu Y, Wang Y, Chaihu L, Tan M, Wang H, Huang Y, Wang J, Wang G. Biofunctionalized dissolvable hydrogel microbeads enable efficient characterization of native protein complexes. Nat Commun 2024; 15:8633. [PMID: 39366952 PMCID: PMC11452662 DOI: 10.1038/s41467-024-52948-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
The characterization of protein complex is vital for unraveling biological mechanisms in various life processes. Despite advancements in biophysical tools, the capture of non-covalent complexes and deciphering of their biochemical composition continue to present challenges for low-input samples. Here we introduce SNAP-MS, a Stationary-phase-dissolvable Native Affinity Purification and Mass Spectrometric characterization strategy. It allows for highly efficient purification and characterization from inputs at the pico-mole level. SNAP-MS replaces traditional elution with matrix dissolving during the recovery of captured targets, enabling the use of high-affinity bait-target pairs and eliminates interstitial voids. The purified intact protein complexes are compatible with native MS, which provides structural information including stoichiometry, topology, and distribution of proteoforms, size variants and interaction states. An algorithm utilizes the bait as a charge remover and mass corrector significantly enhances the accuracy of analyzing heterogeneously glycosylated complexes. With a sample-to-data time as brief as 2 hours, SNAP-MS demonstrates considerable versatility in characterizing native complexes from biological samples, including blood samples.
Collapse
Affiliation(s)
- Xinyang Shao
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- Changping Laboratory, Beijing, China
| | - Meng Tian
- School of Life Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center of Biological Structures, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Junlong Yin
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Haifeng Duan
- CYGNUS Bioscience (Beijing) Co. Ltd, Beijing, China
| | - Ye Tian
- Changping Laboratory, Beijing, China
| | - Hui Wang
- Department of Clinical Laboratories, Peking University People's Hospital, Beijing, China
| | - Changsheng Xia
- Department of Clinical Laboratories, Peking University People's Hospital, Beijing, China
| | - Ziwei Wang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yanxi Zhu
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
| | - Yifan Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Lingxiao Chaihu
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- School of Chemistry & Materials Science, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Minjie Tan
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Hongwei Wang
- School of Life Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center of Biological Structures, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yanyi Huang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- Changping Laboratory, Beijing, China
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, China
| | - Jianbin Wang
- Changping Laboratory, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
| | - Guanbo Wang
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China.
| |
Collapse
|
23
|
Wang Q, Wang Q, Qi Z, Moeller W, Wysocki VH, Sun L. Native Proteomics by Capillary Zone Electrophoresis-Mass Spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590970. [PMID: 38712154 PMCID: PMC11071496 DOI: 10.1101/2024.04.24.590970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Native proteomics measures endogenous proteoforms and protein complexes under a near physiological condition using native mass spectrometry (nMS) coupled with liquid-phase separations. Native proteomics should provide the most accurate bird's-eye view of proteome dynamics within cells, which is fundamental for understanding almost all biological processes. nMS has been widely employed to characterize well-purified protein complexes. However, there are only very few trials of utilizing nMS to measure proteoforms and protein complexes in a complex sample (i.e., a whole cell lysate). Here, we pioneer the native proteomics measurement of large proteoforms or protein complexes up to 400 kDa from a complex proteome via online coupling of native capillary zone electrophoresis (nCZE) to an ultra-high mass range (UHMR) Orbitrap mass spectrometer. The nCZE-MS technique enabled the measurement of a 115-kDa standard protein complex while consuming only about 0.1 ng of protein material. nCZE-MS analysis of an E . coli cell lysate detected 72 proteoforms or protein complexes in a mass range of 30-400 kDa in a single run while consuming only 50-ng protein material. The mass distribution of detected proteoforms or protein complexes agreed well with that from mass photometry measurement. This work represents a technical breakthrough in native proteomics for measuring complex proteomes.
Collapse
|
24
|
Szabo R, Gyemant G, Nagy C, Andrasi M, Gaspar A. Taylor-Aris Dispersion-Assisted Mass Spectrometry for the Analysis of Native Proteins. Anal Chem 2024; 96:11309-11317. [PMID: 38946421 DOI: 10.1021/acs.analchem.4c01270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
As has recently been shown, Taylor-Aris dispersion-assisted mass spectrometry (TADA-MS) can offer direct injection MS determinations in fields where the targets of the analyses are large molecules present in a matrix that would otherwise cause serious interferences. In the present study, we demonstrated the exceptional utility of TADA-MS in native protein analysis: (i) a dramatic improvement in detection sensitivity was found due to its ability to strongly reduce matrix interferences, (ii) more "native-like" conditions can be used during analyses, (iii) the direct injection of non-MS-compatible matrices is allowed into MS, and (iv) a considerable simplification and economization of the workflow is ensured. We investigated the behavior of different types of proteins and protein complexes present under native conditions, demonstrating the unambiguous benefits and simplicity of the method.
Collapse
Affiliation(s)
- Ruben Szabo
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Gyongyi Gyemant
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Cynthia Nagy
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Melinda Andrasi
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| | - Attila Gaspar
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Debrecen 4032, Hungary
| |
Collapse
|
25
|
Phan M, Chandrashekaran IR, Akhtar N, Konstantinidou E, Devine SM, Doak BC, Nebl T, Creek DJ, Scanlon MJ, Norton RS. Multiplexed Native Mass Spectrometry Determination of Ligand Selectivity for Fatty Acid-Binding Proteins. ACS Med Chem Lett 2024; 15:1071-1079. [PMID: 39015264 PMCID: PMC11247632 DOI: 10.1021/acsmedchemlett.4c00154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/08/2024] [Accepted: 06/12/2024] [Indexed: 07/18/2024] Open
Abstract
Although multiple approaches for characterizing protein-ligand interactions are available in target-based drug discovery, their throughput for determining selectivity is quite limited. Herein, we describe the application of native mass spectrometry for rapid, multiplexed screening of the selectivity of eight small-molecule ligands for five fatty acid-binding protein isoforms. Using high-resolution mass spectrometry, we were able to identify and quantify up to 20 different protein species in a single spectrum. We show that selectivity profiles generated by native mass spectrometry are in good agreement with those of traditional solution-phase techniques such as isothermal titration calorimetry and fluorescence polarization. Furthermore, we propose strategies for effective investigation of selectivity by native mass spectrometry, thus highlighting the potential of this technique to be used as an orthogonal method to traditional biophysical approaches for rapid, multiplexed screening of protein-ligand complexes.
Collapse
Affiliation(s)
- Michelle
Q. Phan
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Indu R. Chandrashekaran
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC
Centre for Fragment-Based Design, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Naureen Akhtar
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC
Centre for Fragment-Based Design, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Evgenia Konstantinidou
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC
Centre for Fragment-Based Design, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Shane M. Devine
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Bradley C. Doak
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC
Centre for Fragment-Based Design, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Thomas Nebl
- Biologics
Research and Development Group, Biomedical Manufacturing Program, CSIRO, Clayton, Victoria 3168, Australia
| | - Darren J. Creek
- Drug
Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Martin J. Scanlon
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC
Centre for Fragment-Based Design, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Raymond S. Norton
- Medicinal
Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- ARC
Centre for Fragment-Based Design, Monash Institute of Pharmaceutical
Sciences, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
26
|
Fischer MS, Rogers HT, Chapman EA, Chan HJ, Krichel B, Gao Z, Larson EJ, Ge Y. Online Mixed-Bed Ion Exchange Chromatography for Native Top-Down Proteomics of Complex Mixtures. J Proteome Res 2024; 23:2315-2322. [PMID: 38913967 PMCID: PMC11344481 DOI: 10.1021/acs.jproteome.4c00430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Native top-down mass spectrometry (nTDMS) allows characterization of protein structure and noncovalent interactions with simultaneous sequence mapping and proteoform characterization. The majority of nTDMS studies utilize purified recombinant proteins, with significant challenges hindering application to endogenous systems. To perform native top-down proteomics (nTDP), where endogenous proteins from complex biological systems are analyzed by nTDMS, it is essential to separate proteins under nondenaturing conditions. However, it remains difficult to achieve high resolution with MS-compatible online chromatography while preserving protein tertiary structure and noncovalent interactions. Herein, we report the use of online mixed-bed ion exchange chromatography (IEC) to enable separation of endogenous proteins from complex mixtures under nondenaturing conditions, preserving noncovalent interactions for nTDP analysis. We have successfully detected large proteins (>146 kDa) and identified endogenous metal-binding and oligomeric protein complexes in human heart tissue lysate. The use of a mixed-bed stationary phase allowed retention and elution of proteins over a wide range of isoelectric points without altering the sample or mobile phase pH. Overall, our method provides a simple online IEC-MS platform that can effectively separate proteins from complex mixtures under nondenaturing conditions and preserve higher-order structure for nTDP applications.
Collapse
Affiliation(s)
- Matthew S. Fischer
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Ave, Madison, WI, USA 53706
| | - Holden T. Rogers
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Ave, Madison, WI, USA 53706
| | - Emily A. Chapman
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Ave, Madison, WI, USA 53706
| | - Hsin-Ju Chan
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Ave, Madison, WI, USA 53706
| | - Boris Krichel
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, 1111 Highland Ave., Madison, WI, USA 53705
- School of Life Sciences, University of Siegen, Adolf-Reichwein Str. 2a, Siegen, Germany, 57076
| | - Zhan Gao
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, 1111 Highland Ave., Madison, WI, USA 53705
| | - Eli J. Larson
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Ave, Madison, WI, USA 53706
| | - Ying Ge
- Department of Chemistry, University of Wisconsin - Madison, 1101 University Ave, Madison, WI, USA 53706
- Department of Cell and Regenerative Biology, University of Wisconsin - Madison, 1111 Highland Ave., Madison, WI, USA 53705
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin - 1111 Highland Ave., Madison, WI, USA 53705
| |
Collapse
|
27
|
Cotham VC, Wang S, Li N. An Online Native Mass Spectrometry Approach for Fast, Sensitive, and Quantitative Assessment of Adeno-Associated Virus Capsid Content Ratios. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1567-1575. [PMID: 38888112 PMCID: PMC11228988 DOI: 10.1021/jasms.4c00151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Adeno-associated viruses (AAVs) have emerged as a leading platform for in vivo therapeutic gene delivery and offer tremendous potential in the treatment and prevention of human disease. The fast-paced development of this growing class of therapeutics, coupled with their intrinsic structural complexity, places a high demand on analytical methods capable of efficiently monitoring product quality to ensure safety and efficacy, as well as to support manufacturing and process optimization. Importantly, the presence and relative abundance of both empty and partially filled AAV capsid subpopulations are of principal concern, as these represent the most common product-related impurities in AAV manufacturing and have a direct impact on therapeutic potential. For this reason, the capsid content, or ratio of empty and partial capsids to those packaged with the full-length therapeutic genome, has been identified by regulatory agencies as a critical quality attribute (CQA) that must be carefully controlled to meet clinical specifications. Established analytical methods for the quantitation of capsid content ratios often suffer from long turnaround times, low throughput, and high sample demands that are not well-suited to the narrow timelines and limited sample availability typical of process development. In this study, we present an integrated online native mass spectrometry platform that aims to minimize sample handling and maximize throughput and robustness for rapid and sensitive quantitation of AAV capsid content ratios. The primary advantages of this platform for AAV analysis include the ability to perform online buffer exchange under low flow conditions to maintain sample stability with minimal sample dilution, as well as the ability to achieve online charge reduction via dopant-modified desolvation gas. By exploiting the latter, enhanced spectral resolution of signals arising from empty, partial, and full AAV capsids was accomplished in the m/z domain to facilitate improved spectral interpretation and quantitation that correlated well with the industry standard analytical ultracentrifugation (AUC) method for capsid content ratio determination. The utility of this approach was further demonstrated in several applications, including the rapid and universal screening of different AAV serotypes, evaluation of capsid content for in-process samples, and the monitoring of capsid stability when subjected to thermal stress conditions.
Collapse
Affiliation(s)
- Victoria C Cotham
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, New York 10591, United States
| | - Shunhai Wang
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, New York 10591, United States
| | - Ning Li
- Analytical Chemistry Group, Regeneron Pharmaceuticals Inc., Tarrytown, New York 10591, United States
| |
Collapse
|
28
|
Wu G, Yu C, Yin S, Du J, Zhang Y, Fu Z, Wang L, Wang J. A native SEC-MS workflow and validation for analyzing drug-to-antibody ratio and drug load distribution in cysteine-linked antibody-drug conjugates. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1241:124167. [PMID: 38823148 DOI: 10.1016/j.jchromb.2024.124167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
The development and optimization of Antibody-Drug Conjugates (ADCs) hinge on enhanced analytical and bioanalytical characterization, particularly in assessing critical quality attributes (CQAs). The ADC's potency is largely determined by the average number of drugs attached to the monoclonal antibody (mAb), known as the drug-to-antibody ratio (DAR). Furthermore, the drug load distribution (DLD) influences the therapeutic window of the ADC, defining the range of dosages effective in treating diseases without causing toxic effects. Among CQAs, DAR and DLD are vital; their control is essential for ensuring manufacturing consistency and product quality. Typically, hydrophobic interaction chromatography (HIC) or reversed-phase liquid chromatography (RPLC) with UV detector have been used to quantitate DAR and DLD in quality control (QC) environment. Recently, Native size-exclusion chromatography-mass spectrometry (nSEC-MS) proves the potential as a platformable quantitative method for characterizing DAR and DLD across various cysteine-linked ADCs in research or early preclinical development. In this work, we established and assessed a streamlined nSEC-MS workflow with a benchtop LC-MS platform, to quantitatively monitor DAR and DLD of different chemotype and drug load level cysteine-linked ADCs. Moreover, to deploy this workflow in QC environment, complete method validation was conducted in three independent laboratories, adhering to the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) Q2(R1) guidelines. The results met the predefined analytical target profile (ATP) and performance criteria, encompassing specificity/selectivity, accuracy, precision, linearity, range, quantification/detection limit, and robustness. Finally, the method validation design offers a reference for other nSEC-MS methods that are potentially used to determine the DAR and DLD on cysteine-linker ADCs. To the best of our knowledge, this study is the first reported systematic validation of the nSEC-MS method for detecting DAR and DLD. The results indicated that the co-validated nSEC-MS workflow is suitable for DAR and DLD routine analysis in ADC quality control, release, and stability testing.
Collapse
Affiliation(s)
- Gang Wu
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Chuanfei Yu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, China
| | - Sicheng Yin
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China
| | - Jialiang Du
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, China
| | - Yifan Zhang
- Shanghai Fudan-Zhangjiang Bio-Pharmaceutical Co., Ltd., Shanghai, China
| | - Zhihao Fu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, China
| | - Lan Wang
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, China
| | - Junzhi Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
29
|
Matney R, Gadkari VV. Recent advances in gas phase unfolding: Instrumentation and applications. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5059. [PMID: 38894609 DOI: 10.1002/jms.5059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024]
Abstract
Broader adoption of native mass spectrometry (MS) and ion mobility-mass spectrometry (IM-MS) has propelled the development of several techniques which take advantage of the selectivity, sensitivity, and speed of MS to make measurements of complex biological molecules in the gas phase. One such method, collision induced unfolding (CIU), has risen to prominence in recent years, due to its well documented capability to detect shifts in structural stability of biological molecules in response to external stimuli (e.g., mutations, stress, non-covalent interactions, sample conditions etc.). This increase in reported CIU measurements is enabled partly due to advances in IM-MS instrumentation by vendors, and also innovative method development by researchers. This perspective highlights a few of these advances and concludes with a look forward toward the future of the gas phase unfolding field.
Collapse
Affiliation(s)
- Rowan Matney
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| | - Varun V Gadkari
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
30
|
Yin V, Deslignière E, Mokiem N, Gazi I, Lood R, de Haas CJC, Rooijakkers SHM, Heck AJR. Not All Arms of IgM Are Equal: Following Hinge-Directed Cleavage by Online Native SEC-Orbitrap-Based CDMS. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:1320-1329. [PMID: 38767111 PMCID: PMC11157650 DOI: 10.1021/jasms.4c00094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
Immunoglobulins M (IgM) are key natural antibodies produced initially in humoral immune response. Due to their large molecular weights and extensive glycosylation loads, IgMs represent a challenging target for conventional mass analysis. Charge detection mass spectrometry (CDMS) may provide a unique approach to tackle heterogeneous IgM assemblies, although this technique can be quite laborious and technically challenging. Here, we describe the use of online size exclusion chromatography (SEC) to automate buffer exchange and sample introduction, and demonstrate its adaptability with Orbitrap-based CDMS. We discuss optimal experimental parameters for online SEC-CDMS experiments, including ion activation, choice of column, and resolution. Using this approach, CDMS histograms containing hundreds of individual ion signals can be obtained in as little as 5 min from single injections of <1 μg of sample. To demonstrate the unique utility of online SEC-CDMS, we performed real-time kinetic monitoring of pentameric IgM digestion by the protease IgMBRAZOR, which cleaves specifically in the hinge region of IgM. Several digestion intermediates corresponding to processive losses of F(ab')2 subunits could be mass-resolved and identified by SEC-CDMS. Interestingly, we find that for the J-chain linked IgM pentamer, cleavage of one of the F(ab')2 subunits is much slower than the other four F(ab')2 subunits, which we attribute to the symmetry-breaking interactions of the J-chain within the pentameric IgM structure. The online SEC-CDMS methodologies described here open new avenues into the higher throughput automated analysis of heterogeneous, high-mass protein assemblies by CDMS.
Collapse
Affiliation(s)
- Victor Yin
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584
CH Utrecht, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, 3584 CH Utrecht, The Netherlands
| | - Evolène Deslignière
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584
CH Utrecht, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, 3584 CH Utrecht, The Netherlands
| | - Nadia Mokiem
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584
CH Utrecht, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, 3584 CH Utrecht, The Netherlands
| | - Inge Gazi
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584
CH Utrecht, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, 3584 CH Utrecht, The Netherlands
| | - Rolf Lood
- Genovis
AB, Scheelevägen
2, 223 63 Lund, Sweden
| | - Carla J. C. de Haas
- Department
of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Suzan H. M. Rooijakkers
- Department
of Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Albert J. R. Heck
- Biomolecular
Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular
Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584
CH Utrecht, The Netherlands
- Netherlands
Proteomics Center, Padualaan
8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
31
|
Quek YJ, Tay A. Nanoscale Methods for Longitudinal Extraction of Intracellular Contents. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314184. [PMID: 38459829 DOI: 10.1002/adma.202314184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Longitudinal analysis of intracellular contents including gene and protein expression is crucial for deciphering the fundamentally dynamic nature of cells. This offers invaluable insights into complex tissue composition and behavior, and drives progress in disease diagnosis, biomarker discovery, and drug development. Traditional longitudinal analysis workflows, involving the destruction of cells at various timepoints, limit insights to singular moments and fail to account for cellular heterogeneity. Current non-destructive approaches, like temporal modeling with single-cell ribonucleic acid sequencing (RNA-seq) and live-cell fluorescence imaging, either rely on biological assumptions or possess the risk of cellular perturbation. Recent advances in nanoscale technologies for non-destructive intracellular content extraction offer a promising solution to these challenges. These novel methods work at the nanoscale to non-destructively access cellular membranes and can be broadly classified into three mechanisms: tip-facilitated aspiration, membrane-based, and probe-based methods. This perspective focuses on these emerging nanotechnologies for repeated intracellular content extraction. Their potential in longitudinal analysis is discussed, the critical requirements for effective repeated sampling are addressed, and the suitability of each technique for various applications is explored. Furthermore, unresolved challenges in repeated sampling are highlighted to encourage further research in this growing field.
Collapse
Affiliation(s)
- Ying Jie Quek
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, 138648, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Tissue Engineering Programme, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
32
|
Ivanov DG, Cheung K, Kaltashov IA. Probing the Architecture of Multisubunit Protein Complexes with In-line Disulfide Reduction and Native MS Analysis. Anal Chem 2024; 96:8243-8248. [PMID: 38733603 DOI: 10.1021/acs.analchem.4c00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Native mass spectrometry (MS) continues to enjoy growing popularity as a means of providing a wealth of information on noncovalent biopolymer assemblies ranging from composition and binding stoichiometry to characterization of the topology of these assemblies. The latter frequently relies on supplementing MS measurements with limited fragmentation of the noncovalent complexes in the gas phase to identify the pairs of neighboring subunits. While this approach has met with much success in the past two decades, its implementation remains difficult (and the success record relatively modest) within one class of noncovalent assemblies: protein complexes in which at least one binding partner has multiple subunits cross-linked by disulfide bonds. We approach this problem by inducing chemical reduction of disulfide bonds under nondenaturing conditions in solution followed by native MS analysis with online buffer exchange to remove unconsumed reagents that are incompatible with the electrospray ionization process. While this approach works well with systems comprised of thiol-linked subunits that remain stable upon reduction of the disulfide bridges (such as immunoglobulins), chemical reduction frequently gives rise to species that are unstable (prone to aggregation). This problem is circumvented by taking advantage of the recently introduced cross-path reactive chromatography platform (XPRC), which allows the disulfide reduction to be carried out in-line, thereby minimizing the loss of metastable protein subunits and their noncovalent complexes with the binding partners prior to MS analysis. The feasibility of this approach is demonstrated using hemoglobin complexes with haptoglobin 1-1, a glycoprotein consisting of four polypeptide chains cross-linked by disulfide bonds.
Collapse
Affiliation(s)
- Daniil G Ivanov
- Department of Chemistry, University of Massachusetts─Amherst, Amherst, Massachusetts 01002, United States
| | - Kevin Cheung
- Department of Chemistry, University of Massachusetts─Amherst, Amherst, Massachusetts 01002, United States
| | - Igor A Kaltashov
- Department of Chemistry, University of Massachusetts─Amherst, Amherst, Massachusetts 01002, United States
| |
Collapse
|
33
|
D'Atri V, Imiołek M, Quinn C, Finny A, Lauber M, Fekete S, Guillarme D. Size exclusion chromatography of biopharmaceutical products: From current practices for proteins to emerging trends for viral vectors, nucleic acids and lipid nanoparticles. J Chromatogr A 2024; 1722:464862. [PMID: 38581978 DOI: 10.1016/j.chroma.2024.464862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
The 21st century has been particularly productive for the biopharmaceutical industry, with the introduction of several classes of innovative therapeutics, such as monoclonal antibodies and related compounds, gene therapy products, and RNA-based modalities. All these new molecules are susceptible to aggregation and fragmentation, which necessitates a size variant analysis for their comprehensive characterization. Size exclusion chromatography (SEC) is one of the reference techniques that can be applied. The analytical techniques for mAbs are now well established and some of them are now emerging for the newer modalities. In this context, the objective of this review article is: i) to provide a short historical background on SEC, ii) to suggest some clear guidelines on the selection of packing material and mobile phase for successful method development in modern SEC; and iii) to highlight recent advances in SEC, such as the use of narrow-bore and micro-bore columns, ultra-wide pore columns, and low-adsorption column hardware. Some important innovations, such as recycling SEC, the coupling of SEC with mass spectrometry, and the use of alternative detectors such as charge detection mass spectrometry and mass photometry are also described. In addition, this review discusses the use of SEC in multidimensional setups and shows some of the most recent advances at the preparative scale. In the third part of the article, the possibility of SEC for the characterization of new modalities is also reviewed. The final objective of this review is to provide a clear summary of opportunities and limitations of SEC for the analysis of different biopharmaceutical products.
Collapse
Affiliation(s)
- Valentina D'Atri
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland
| | | | | | - Abraham Finny
- Waters Corporation, Wyatt Technology, Santa Barbara, CA, USA
| | - Matthew Lauber
- Waters Corporation, Wyatt Technology, Santa Barbara, CA, USA
| | | | - Davy Guillarme
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland; School of Pharmaceutical Sciences, University of Geneva, CMU - Rue Michel Servet 1,4, 1211 Geneva, Switzerland.
| |
Collapse
|
34
|
Thibert S, Reid DJ, Wilson JW, Varikoti R, Maltseva N, Schultz KJ, Kruel A, Babnigg G, Joachimiak A, Kumar N, Zhou M. Native Mass Spectrometry Dissects the Structural Dynamics of an Allosteric Heterodimer of SARS-CoV-2 Nonstructural Proteins. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:912-921. [PMID: 38535992 PMCID: PMC11066969 DOI: 10.1021/jasms.3c00453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 05/02/2024]
Abstract
Structure-based drug design, which relies on precise understanding of the target protein and its interaction with the drug candidate, is dramatically expedited by advances in computational methods for candidate prediction. Yet, the accuracy needs to be improved with more structural data from high throughput experiments, which are challenging to generate, especially for dynamic and weak associations. Herein, we applied native mass spectrometry (native MS) to rapidly characterize ligand binding of an allosteric heterodimeric complex of SARS-CoV-2 nonstructural proteins (nsp) nsp10 and nsp16 (nsp10/16), a complex essential for virus survival in the host and thus a desirable drug target. Native MS showed that the dimer is in equilibrium with monomeric states in solution. Consistent with the literature, well characterized small cosubstrate, RNA substrate, and product bind with high specificity and affinity to the dimer but not the free monomers. Unsuccessfully designed ligands bind indiscriminately to all forms. Using neutral gas collision, the nsp16 monomer with bound cosubstrate can be released from the holo dimer complex, confirming the binding to nsp16 as revealed by the crystal structure. However, we observed an unusual migration of the endogenous zinc ions bound to nsp10 to nsp16 after collisional dissociation. The metal migration can be suppressed by using surface collision with reduced precursor charge states, which presumably resulted in minimal gas-phase structural rearrangement and highlighted the importance of complementary techniques. With minimal sample input (∼μg), native MS can rapidly detect ligand binding affinities and locations in dynamic multisubunit protein complexes, demonstrating the potential of an "all-in-one" native MS assay for rapid structural profiling of protein-to-AI-based compound systems to expedite drug discovery.
Collapse
Affiliation(s)
- Stephanie
M. Thibert
- Environmental
Molecular Sciences Laboratory, Pacific Northwest
National Laboratory, Richland, Washington 99354, United States
| | - Deseree J. Reid
- Chemical
and Biological Signature Sciences, Pacific
Northwest National Laboratory, Richland, Washington 99354, United States
| | - Jesse W. Wilson
- Environmental
Molecular Sciences Laboratory, Pacific Northwest
National Laboratory, Richland, Washington 99354, United States
| | - Rohith Varikoti
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Natalia Maltseva
- Center
for Structural Biology of Infectious Diseases, Consortium for Advanced
Science and Engineering, University of Chicago, Chicago, Illinois 60637, United States
- Structural
Biology Center, X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Katherine J. Schultz
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Agustin Kruel
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Gyorgy Babnigg
- Center
for Structural Biology of Infectious Diseases, Consortium for Advanced
Science and Engineering, University of Chicago, Chicago, Illinois 60637, United States
- Biosciences
Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Andrzej Joachimiak
- Center
for Structural Biology of Infectious Diseases, Consortium for Advanced
Science and Engineering, University of Chicago, Chicago, Illinois 60637, United States
- Structural
Biology Center, X-ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Neeraj Kumar
- Biological
Sciences Division, Pacific Northwest National
Laboratory, Richland, Washington 99354, United States
| | - Mowei Zhou
- Environmental
Molecular Sciences Laboratory, Pacific Northwest
National Laboratory, Richland, Washington 99354, United States
| |
Collapse
|
35
|
Wong DCJ, Wang Z, Perkins J, Jin X, Marsh GE, John EG, Peakall R. The road less taken: Dihydroflavonol 4-reductase inactivation and delphinidin anthocyanin loss underpins a natural intraspecific flower colour variation. Mol Ecol 2024:e17334. [PMID: 38651763 DOI: 10.1111/mec.17334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/22/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024]
Abstract
Visual cues are of critical importance for the attraction of animal pollinators, however, little is known about the molecular mechanisms underpinning intraspecific floral colour variation. Here, we combined comparative spectral analysis, targeted metabolite profiling, multi-tissue transcriptomics, differential gene expression, sequence analysis and functional analysis to investigate a bee-pollinated orchid species, Glossodia major with common purple- and infrequent white-flowered morphs. We found uncommon and previously unreported delphinidin-based anthocyanins responsible for the conspicuous and pollinator-perceivable colour of the purple morph and three genetic changes underpinning the loss of colour in the white morph - (1) a loss-of-function (LOF; frameshift) mutation affecting dihydroflavonol 4-reductase (DFR1) coding sequence due to a unique 4-bp insertion, (2) specific downregulation of functional DFR1 expression and (3) the unexpected discovery of chimeric Gypsy transposable element (TE)-gene (DFR) transcripts with potential consequences to the genomic stability and post-transcriptional or epigenetic regulation of DFR. This is one of few known cases where regulatory changes and LOF mutation in an anthocyanin structural gene, rather than transcription factors, are important. Furthermore, if TEs prove to be a frequent source of mutation, the interplay between environmental stress-induced TE evolution and pollinator-mediated selection for adaptive colour variation may be an overlooked mechanism maintaining floral colour polymorphism in nature.
Collapse
Affiliation(s)
- Darren C J Wong
- Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Zemin Wang
- State Key Laboratory of Aridland Crop Science, Gansu Agricultural University, Lanzhou, China
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - James Perkins
- Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Xin Jin
- State Key Laboratory of Aridland Crop Science, Gansu Agricultural University, Lanzhou, China
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Grace Emma Marsh
- Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Emma Grace John
- Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Rod Peakall
- Ecology and Evolution, Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
36
|
Chapman EA, Li BH, Krichel B, Chan HJ, Buck KM, Roberts DS, Ge Y. Native Top-Down Mass Spectrometry for Characterizing Sarcomeric Proteins Directly from Cardiac Tissue Lysate. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:738-745. [PMID: 38422011 PMCID: PMC11098619 DOI: 10.1021/jasms.3c00430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Native top-down mass spectrometry (nTDMS) has emerged as a powerful structural biology tool that can localize post-translational modifications (PTMs), explore ligand-binding interactions, and elucidate the three-dimensional structure of proteins and protein complexes in the gas-phase. Fourier-transform ion cyclotron resonance (FTICR) MS offers distinct capabilities for nTDMS, owing to its ultrahigh resolving power, mass accuracy, and robust fragmentation techniques. Previous nTDMS studies using FTICR have mainly been applied to overexpressed recombinant proteins and protein complexes. Here, we report the first nTDMS study that directly analyzes human heart tissue lysate by direct infusion FTICR MS without prior chromatographic separation strategies. We have achieved comprehensive nTDMS characterization of cardiac contractile proteins that play critical roles in heart contraction and relaxation. Specifically, our results reveal structural insights into ventricular myosin light chain 2 (MLC-2v), ventricular myosin light chain 1 (MLC-1v), and alpha-tropomyosin (α-Tpm) in the sarcomere, the basic contractile unit of cardiac muscle. Furthermore, we verified the calcium (Ca2+) binding domain in MLC-2v. In summary, our nTDMS platform extends the application of FTICR MS to directly characterize the structure, PTMs, and metal-binding of endogenous proteins from heart tissue lysate without prior separation methods.
Collapse
Affiliation(s)
- Emily A. Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Brad H. Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Boris Krichel
- School of Life Sciences, University of Siegen, 57076, Germany
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| | - Hsin-Ju Chan
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Kevin M. Buck
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - David S. Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin 53705, USA
| |
Collapse
|
37
|
Sarin D, Krishna K, Nejadnik MR, Suryanarayanan R, Rathore AS. Impact of Excipient Extraction and Buffer Exchange on Recombinant Monoclonal Antibody Stability. Mol Pharm 2024; 21:1872-1883. [PMID: 38422397 PMCID: PMC10988557 DOI: 10.1021/acs.molpharmaceut.3c01157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
The foundation of a biosimilar manufacturer's regulatory filing is the demonstration of analytical and functional similarity between the biosimilar product and the pertinent originator product. The excipients in the formulation may interfere with characterization using typical analytical and functional techniques during this biosimilarity exercise. Consequently, the producers of biosimilar products resort to buffer exchange to isolate the biotherapeutic protein from the drug product formulation. However, the impact that this isolation has on the product stability is not completely known. This study aims to elucidate the extent to which mAb isolation via ultrafiltration-diafiltration-based buffer exchange impacts mAb stability. It has been demonstrated that repeated extraction cycles do result in significant changes in higher-order structure (red-shift of 5.0 nm in fluorescence maxima of buffer exchanged samples) of the mAb and also an increase in formation of basic variants from 19.1 to 26.7% and from 32.3 to 36.9% in extracted innovator and biosimilar Tmab samples, respectively. It was also observed that under certain conditions of tertiary structure disruptions, Tmab could be restabilized depending on formulation composition. Thus, mAb isolation through extraction with buffer exchange impacts the product stability. Based on the observations reported in this paper, we recommend that biosimilar manufacturers take into consideration these effects of excipients on protein stability when performing biosimilarity assessments.
Collapse
Affiliation(s)
- Deepika Sarin
- Department
of Chemical Engineering, Indian Institute
of Technology, Delhi, Hauz Khas, New Delhi 110016, India
| | - Kunal Krishna
- School
of Interdisciplinary Research, Indian Institute
of Technology, Delhi, Hauz Khas, New Delhi 110016, India
| | - M. Reza Nejadnik
- Department
of Pharmaceutical Sciences & Experimental Therapeutics, College
of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States
| | - Raj Suryanarayanan
- Department
of Pharmaceutics, College of Pharmacy, University
of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Anurag S. Rathore
- Department
of Chemical Engineering, Indian Institute
of Technology, Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
38
|
Harvey SR, Gadkari VV, Ruotolo BT, Russell DH, Wysocki VH, Zhou M. Expanding Native Mass Spectrometry to the Masses. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:646-652. [PMID: 38303101 PMCID: PMC11821079 DOI: 10.1021/jasms.3c00352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
At the 33rd ASMS Sanibel Meeting, on Membrane Proteins and Their Complexes, a morning roundtable discussion was held discussing the current challenges facing the field of native mass spectrometry and approaches to expanding the field to nonexperts. This Commentary summarizes the discussion and current initiatives to address these challenges.
Collapse
Affiliation(s)
- Sophie R. Harvey
- Department of Chemistry and Biochemistry, Native Mass Spectrometry Guided Structural Biology Center, The Ohio State University, Columbus, OH, USA, 43210
| | - Varun V. Gadkari
- Department of Chemistry, University of Minnesota, Minneapolis, MN, USA, 55455
| | - Brandon T. Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA, 48109
| | - David H. Russell
- Department of Chemistry, Texas A&M University, College Station, TX, USA, 77844
| | - Vicki H. Wysocki
- Department of Chemistry and Biochemistry, Native Mass Spectrometry Guided Structural Biology Center, The Ohio State University, Columbus, OH, USA, 43210
| | - Mowei Zhou
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China, 310058
| |
Collapse
|
39
|
Williams JD, Pu F, Sawicki JW, Elsen NL. Ultra-high-throughput mass spectrometry in drug discovery: fundamentals and recent advances. Expert Opin Drug Discov 2024; 19:291-301. [PMID: 38111363 DOI: 10.1080/17460441.2023.2293153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
INTRODUCTION Ultra-high-throughput mass spectrometry, uHT-MS, is a technology that utilizes ionization and sample delivery technologies optimized to enable sampling from well plates at > 1 sample per second. These technologies do not need a chromatographic separation step and can be utilized in a wide variety of assays to detect a broad range of analytes including small molecules, lipids, and proteins. AREAS COVERED This manuscript provides a brief historical review of high-throughput mass spectrometry and the recently developed technologies that have enabled uHT-MS. The report also provides examples and references on how uHT-MS has been used in biochemical and chemical assays, nuisance compound profiling, protein analysis and high throughput experimentation for chemical synthesis. EXPERT OPINION The fast analysis time provided by uHT-MS is transforming how biochemical and chemical assays are performed in drug discovery. The potential to associate phenotypic responses produced by 1000's of compound treatments with changes in endogenous metabolite and lipid signals is becoming feasible. With the augmentation of simple, fast, high-throughput sample preparation, the scope of uHT-MS usage will increase. However, it likely will not supplant LC-MS for analyses that require low detection limits from complex matrices or characterization of complex biotherapeutics such as antibody-drug conjugates.
Collapse
Affiliation(s)
| | - Fan Pu
- Abbvie Discovery Research, North Chicago, IL, USA
| | | | | |
Collapse
|
40
|
Lanzillotti MB, Brodbelt JS. A High-Throughput Workflow for Mass Spectrometry Analysis of Nucleic Acids by Nanoflow Desalting. Anal Chem 2024. [PMID: 38330443 PMCID: PMC11382294 DOI: 10.1021/acs.analchem.3c05428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Broad interest in nucleic acids, both their therapeutic capabilities and understanding the nuances of their structure and resulting function, has increased in recent years. Post-transcriptional modifications, in particular, have become an important analysis target, as these covalent modifications to the sugars, nitrogenous bases, and phosphate backbone impart differential functionality to synthetic and biological nucleic acids. Characterizing these post-transcriptional modifications can be difficult with traditional sequencing workflows; however, advancements in top-down mass spectrometry address these challenges. Online desalting platforms have enabled facile sample cleanup and reliable ionization of increasingly large (100 nt) oligonucleotides, and application of existing tandem mass spectrometry techniques has yielded information-rich spectra which can be used to interrogate primary sequences. To extend the capabilities of top-down MS and its analysis of nucleic acids, we have developed a nanoflow desalting platform for high-throughput and low sample-use desalting coupled with collision-induced dissociation (CID), 213 nm ultraviolet photodissociation (UVPD), and activated-ion electron photodetachment dissociation (a-EPD) to yield high-quality MS/MS spectra. Fragments identified using an m/z-domain isotope matching strategy yielded high sequence coverage (>70%) of a yeast phenylalanine tRNA.
Collapse
Affiliation(s)
- Michael B Lanzillotti
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer S Brodbelt
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
41
|
Zhou Y, Tan C, Zenobi R. Rapid Profiling of the Glycosylation Effects on the Binding of SARS-CoV-2 Spike Protein to Angiotensin-Converting Enzyme 2 Using MALDI-MS with High Mass Detection. Anal Chem 2024; 96:1898-1905. [PMID: 38279913 DOI: 10.1021/acs.analchem.3c03930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024]
Abstract
The spike protein receptor-binding domain (RBD) of SARS-CoV-2 binds directly to angiotensin-converting enzyme 2 (ACE2), mediating the host cell entry of SARS-CoV-2. Both spike protein and ACE2 are highly glycosylated, which can regulate the binding. Here, we utilized high-mass MALDI-MS with chemical cross-linking for profiling the glycosylation effects on the binding between RBD and ACE2. Overall, it was found that ACE2 glycosylation affects the binding more strongly than does RBD glycosylation. The binding affinity was improved after desialylation or partial deglycosylation (N690) of ACE2, while it decreased after degalactosylation. ACE2 can form dimers in solution, which bind more tightly to the RBD than the ACE2 monomers. The ACE2 dimerization and the binding of RBD to dimeric ACE2 can also be improved by the desialylation or deglycosylation of ACE2. Partial deglycosylation of ACE2 increased the dimerization of ACE2 and the binding affinity of RBD and ACE2 by more than a factor of 2, suggesting its high potential for neutralizing SARS-CoV-2. The method described in the work provided a simple way to analyze the protein-protein interaction without sample purification. It can be widely used for rapid profiling of glycosylation effects on protein-protein interaction for glycosylation-related diseases and the study of multiple interactions between protein and protein aggregates in a single system.
Collapse
Affiliation(s)
- Yuye Zhou
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), CH-8093 Zürich, Switzerland
- School of Engineering Sciences in Chemistry, Biotechnology and Health, Department of Chemistry, Division of Applied Physical Chemistry, Analytical Chemistry, KTH Royal Institute of Technology, SE-10044 Stockholm, Sweden
| | - Congrui Tan
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), CH-8093 Zürich, Switzerland
| | - Renato Zenobi
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH), CH-8093 Zürich, Switzerland
| |
Collapse
|
42
|
Chen H, Qiu D, Shi J, Wang N, Li M, Wu Y, Tian Y, Bu X, Liu Q, Jiang Y, Hamilton SE, Han P, Sun S. In-Depth Structure and Function Characterization of Heterogeneous Interchain Cysteine-Conjugated Antibody-Drug Conjugates. ACS Pharmacol Transl Sci 2024; 7:212-221. [PMID: 38230295 PMCID: PMC10789146 DOI: 10.1021/acsptsci.3c00235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024]
Abstract
Antibody-drug conjugates (ADCs), integrating high specificity of antigen-targeting antibodies and high potency of cell-killing chemical drugs, have become one of the most rapidly expanding therapeutic biologics in oncology. Although ADCs were widely studied from multiple aspects, overall structural elucidation with comprehensive understanding of variants is scarcely reported. Here, for the first time, we present a holistic and in-depth characterization of an interchain cysteine-conjugated ADC, focusing on conjugation and charge heterogeneity, and in vitro biological activities. Conjugation mapping utilized a bottom-up approach, unraveled positional isomer composition, provided insights into the conjugation process, and elucidated how conjugation affects the physicochemical and biological properties of an ADC. Charge profiling combined bottom-up and top-down approaches to interrogate the origin of charge heterogeneity, its impact on function, and best practice for characterization. Specifically, we pioneered the utilization of capillary isoelectric focusing-mass spectrometry to decode not only critical post-translational modifications but also drug load and positional isomer distribution. The study design provides general guidance for in-depth characterization of ADCs, and the analytical findings in turn benefit the discovery and development of future ADCs.
Collapse
Affiliation(s)
- Huijie Chen
- Analytical
Sciences, WuXi Biologics, 31 Yiwei Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Danye Qiu
- Analytical
Sciences, WuXi Biologics, 1150 Lanfeng Road, Fengxian District, Shanghai 201403, China
| | - Jian Shi
- Analytical
Sciences, WuXi Biologics, 31 Yiwei Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Ningning Wang
- WuXi
Process Development Analytical Science, WuXi Biologics, 200
Meiliang Road, Binhu District, WuXi, Jiangsu 214092, China
| | - Muchen Li
- WuXi
Process Development Analytical Science, WuXi Biologics, 200
Meiliang Road, Binhu District, WuXi, Jiangsu 214092, China
| | - Ying Wu
- Analytical
Sciences, WuXi Biologics, 31 Yiwei Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Yu Tian
- Biologics
Innovation & Discovery, WuXi Biologics, 227 Meisheng Road, Waigaoqiao Free
Trade Zone, Shanghai 200131, China
| | - Xiaodong Bu
- Analytical
Research & Development, Merck &
Co., Inc., 126 E. Lincoln
Avenue, Rahway, New Jersey 07065, United States
| | - Qingyuan Liu
- Analytical
Research & Development, Merck &
Co., Inc., 2000 Galloping
Hill Road, Kenilworth, New
Jersey 07033, United States
| | - Yanrui Jiang
- Analytical
Research & Development, MSD, Industrie Nord 1, Schachen (Luzern) CH-6105, Switzerland
| | - Simon E. Hamilton
- Analytical
Research & Development, MSD, 120 Moorgate, London EC2M 6UR, U.K.
| | - Ping Han
- Analytical
Research & Development, Merck &
Co., Inc., 2000 Galloping
Hill Road, Kenilworth, New
Jersey 07033, United States
| | - Shuwen Sun
- Analytical
Research & Development, Merck &
Co., Inc., 126 E. Lincoln
Avenue, Rahway, New Jersey 07065, United States
| |
Collapse
|
43
|
Stiving AQ, Foreman DJ, VanAernum ZL, Durr E, Wang S, Vlasak J, Galli J, Kafader JO, Tsukidate T, Li X, Schuessler HA, Richardson DD. Dissecting the Heterogeneous Glycan Profiles of Recombinant Coronavirus Spike Proteins with Individual Ion Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:62-73. [PMID: 38032172 DOI: 10.1021/jasms.3c00309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Surface-embedded glycoproteins, such as the spike protein trimers of coronaviruses MERS, SARS-CoV, and SARS-CoV-2, play a key role in viral function and are the target antigen for many vaccines. However, their significant glycan heterogeneity poses an analytical challenge. Here, we utilized individual ion mass spectrometry (I2MS), a multiplexed charge detection measurement with similarities to charge detection mass spectrometry (CDMS), in which a commercially available Orbitrap analyzer is used to directly produce mass profiles of these heterogeneous coronavirus spike protein trimers under native-like conditions. Analysis by I2MS shows that glycosylation contributes to the molecular mass of each protein trimer more significantly than expected by bottom-up techniques, highlighting the importance of obtaining complementary intact mass information when characterizing glycosylation of such heterogeneous proteins. Enzymatic dissection to remove sialic acid or N-linked glycans demonstrates that I2MS can be used to better understand the glycan profile from a native viewpoint. Deglycosylation of N-glycans followed by I2MS analysis indicates that the SARS-CoV-2 spike protein trimer contains glycans that are more difficult to remove than its MERS and SARS-CoV counterparts, and these differences are correlated with solvent accessibility. I2MS technology enables characterization of protein mass and intact glycan profile and is orthogonal to traditional mass analysis methods such as size exclusion chromatography-multiangle light scattering (SEC-MALS) and field flow fractionation-multiangle light scattering (FFF-MALS). An added advantage of I2MS is low sample use, requiring 100-fold less than other methodologies. This work highlights how I2MS technology can enable efficient development of vaccines and therapeutics for pharmaceutical development.
Collapse
Affiliation(s)
- Alyssa Q Stiving
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - David J Foreman
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Zachary L VanAernum
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Eberhard Durr
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Shiyi Wang
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Josef Vlasak
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Jennifer Galli
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jared O Kafader
- Departments of Chemistry and Molecular Biosciences, The Chemistry of Life Processes Institute, The Proteomics Center of Excellence at Northwestern University, Evanston, Illinois 60208, United States
| | - Taku Tsukidate
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Xuanwen Li
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Hillary A Schuessler
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| | - Douglas D Richardson
- Analytical Research and Development, Merck & Co., Inc., 126 E. Lincoln Avenue, Rahway, New Jersey 07065, United States
| |
Collapse
|
44
|
Chapman EA, Roberts DS, Tiambeng TN, Andrews J, Wang MD, Reasoner EA, Melby JA, Li BH, Kim D, Alpert AJ, Jin S, Ge Y. Structure and dynamics of endogenous cardiac troponin complex in human heart tissue captured by native nanoproteomics. Nat Commun 2023; 14:8400. [PMID: 38110393 PMCID: PMC10728164 DOI: 10.1038/s41467-023-43321-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/07/2023] [Indexed: 12/20/2023] Open
Abstract
Protein complexes are highly dynamic entities that display substantial diversity in their assembly, post-translational modifications, and non-covalent interactions, allowing them to play critical roles in various biological processes. The heterogeneity, dynamic nature, and low abundance of protein complexes in their native states present challenges to study using conventional structural biology techniques. Here we develop a native nanoproteomics strategy for the enrichment and subsequent native top-down mass spectrometry (nTDMS) analysis of endogenous cardiac troponin (cTn) complex directly from human heart tissue. The cTn complex is enriched and purified using peptide-functionalized superparamagnetic nanoparticles under non-denaturing conditions to enable the isotopic resolution of cTn complex, revealing their complex structure and assembly. Moreover, nTDMS elucidates the stoichiometry and composition of the cTn complex, localizes Ca2+ binding domains, defines cTn-Ca2+ binding dynamics, and provides high-resolution mapping of the proteoform landscape. This native nanoproteomics strategy opens a paradigm for structural characterization of endogenous native protein complexes.
Collapse
Affiliation(s)
- Emily A Chapman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - David S Roberts
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Timothy N Tiambeng
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jãán Andrews
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Man-Di Wang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Emily A Reasoner
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Jake A Melby
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Brad H Li
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Donguk Kim
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Song Jin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
45
|
Liu W, Jayasekera HS, Sanders JD, Zhang G, Viner R, Marty MT. Online Buffer Exchange Enables Automated Membrane Protein Analysis by Native Mass Spectrometry. Anal Chem 2023; 95:17212-17219. [PMID: 37963237 PMCID: PMC10696660 DOI: 10.1021/acs.analchem.3c02164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Membrane proteins represent the majority of clinical drug targets and are actively involved in a range of cellular processes. However, the complexity of membrane mimetics for membrane protein solubilization poses challenges for native mass spectrometry (MS) analyses. The most common approach for native MS analyses of membrane proteins remains offline buffer exchange into native MS-compatible buffers prior to manual sample loading into static nano-ESI emitters. This laborious process requires relatively high sample consumption and optimization for the individual proteins. Here, we developed online buffer exchange coupled to native mass spectrometry (OBE-nMS) for analyzing membrane proteins in different membrane mimetics, including detergent micelles and nanodiscs. Detergent screening for OBE-nMS reveals that mobile phases containing ammonium acetate with lauryl-dimethylamine oxide are most universal for characterizing both bacterial and mammalian membrane proteins in detergent. Membrane proteins in nanodiscs simply require ammonium acetate as the mobile phase. To preserve the intact nanodiscs, a novel switching electrospray approach was used to capture the high-flow separation on the column with a low-flow injection to MS. Rapid OBE-nMS completes each membrane protein measurement within minutes and thus enables higher-throughput assessment of membrane protein integrity prior to its structural elucidation.
Collapse
Affiliation(s)
- Weijing Liu
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, CA 95134
| | - Hiruni S. Jayasekera
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721
| | - James D. Sanders
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721
| | - Guozhi Zhang
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721
| | - Rosa Viner
- Thermo Fisher Scientific, 355 River Oaks Parkway, San Jose, CA 95134
| | - Michael T. Marty
- Department of Chemistry and Biochemistry and Bio5 Institute, University of Arizona, Tucson, Arizona 85721
| |
Collapse
|
46
|
Juliano BR, Keating JW, Li HW, Anders AG, Xie Z, Ruotolo BT. Development of an Automated, High-Throughput Methodology for Native Mass Spectrometry and Collision-Induced Unfolding. Anal Chem 2023; 95:16717-16724. [PMID: 37924308 PMCID: PMC11081713 DOI: 10.1021/acs.analchem.3c03788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2023]
Abstract
Native ion mobility mass spectrometry (nIM-MS) has emerged as a useful technology for the rapid evaluation of biomolecular structures. When combined with collisional activation in a collision-induced unfolding (CIU) experiment, nIM-MS experimentation can be leveraged to gain greater insight into biomolecular conformation and stability. However, nIM-MS and CIU remain throughput limited due to nonautomated sample preparation and introduction. Here, we explore the use of a RapidFire robotic sample handling system to develop an automated, high-throughput methodology for nMS and CIU. We describe native RapidFire-MS (nRapidFire-MS) capable of performing online desalting and sample introduction in as little as 10 s per sample. When combined with CIU, our nRapidFire-MS approach can be used to collect CIU fingerprints in 30 s following desalting by using size exclusion chromatography cartridges. When compared to nMS and CIU data collected using standard approaches, ion signals recorded by nRapidFire-MS exhibit identical ion collision cross sections, indicating that the same conformational populations are tracked by the two approaches. Our data further suggest that nRapidFire-MS can be extended to study a variety of biomolecular classes, including proteins and protein complexes ranging from 5 to 300 kDa and oligonucleotides. Furthermore, nRapidFire-MS data acquired for biotherapeutics suggest that nRapidFire-MS has the potential to enable high-throughput nMS analyses of biopharmaceutical samples. We conclude by discussing the potential of nRapidFire-MS for enabling the development of future CIU assays capable of catalyzing breakthroughs in protein engineering, inhibitor discovery, and formulation development for biotherapeutics.
Collapse
Affiliation(s)
- Brock R Juliano
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Joseph W Keating
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry W Li
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Anna G Anders
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhuoer Xie
- Attribute Sciences, Process Development, Amgen, Thousand Oaks, California 91320, United States
| | - Brandon T Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
47
|
Di Marco F, Blümel G, Blöchl C, Wuhrer M, Huber CG. A semi-automated hybrid HPLC-MS approach for in-depth characterization of intact non-covalent heterodimer glycoforms of gonadotropin biopharmaceuticals. Anal Chim Acta 2023; 1274:341574. [PMID: 37455084 DOI: 10.1016/j.aca.2023.341574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/07/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Gonadotropins are a class of heavily glycosylated protein hormones, thus extremely challenging to characterize by mass spectrometry. As biopharmaceuticals, gonadotropins are prescribed for the treatment of infertility and are derived from different sources: either from pooled urine of pregnant women or upon production in genetically modified Chinese Hamster Ovary cells. Human chorionic gonadotropin (hCG) is sold as a biopharmaceutical under the name Pregnyl® (urinary hCG, u-hCG) and Ovitrelle® (recombinant hCG, r-hCG), and recombinant human follicle stimulating hormone (r-hFSH) is marketed as Gonal-f®. Recently, we reported the exhaustive characterization of r-hCG at different structural levels. RESULTS We implement size exclusion (SE) HPLC-MS to automatize the acquisition of native mass spectra of r-hCG dimer, but also u-hCG and r-hFSH, comparing the drug products up to intact heterodimer level. A hybrid HPLC-MS approach was employed for the characterization of r-hCG, u-hCG and r-hFSH drug products at different structural levels. Released glycans were analyzed by porous graphitized carbon (PGC)-HPLC-MS/MS, glycopeptides by reversed-phase (RP)-HPLC-MS/MS, subunits by RP-HPLC-MS and finally the intact native heterodimers by semi-automated online buffer exchange SE-HPLC-MS. The data were integrated using bioinformatic tools, to finally unravel the composition of 1481 co-existing dimeric glycoforms for r-hCG, 1167 glycoforms for u-hCG, and 1440 glycoforms for r-hFSH, and to compare critical quality attributes of the different drug products such as their degree of sialylation and O-glycosylation. SIGNIFICANCE AND NOVELTY The strong alliance of bioanalytics and bioinformatics data integration at the different structural levels allowed the identification of more than thousand different glycoforms of r-hCG, u-hCG, and r-hFSH. The results showed that these biopharmaceuticals differ considerably in their glycosylation patterns and highlight the importance of in-depth characterization of biopharmaceuticals for quality control. © 2017 Elsevier Inc. All rights reserved.
Collapse
Affiliation(s)
- Fiammetta Di Marco
- Department of Biosciences and Medical Biology, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Gabriele Blümel
- Department of Biosciences and Medical Biology, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Constantin Blöchl
- Department of Biosciences and Medical Biology, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria; Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Christian G Huber
- Department of Biosciences and Medical Biology, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria.
| |
Collapse
|
48
|
Lermyte F, Habeck T, Brown K, Des Soye B, Lantz C, Zhou M, Alam N, Hossain MA, Jung W, Keener J, Volny M, Wilson J, Ying Y, Agar J, Danis P, Ge Y, Kelleher N, Li H, Loo J, Marty M, Pasa-Tolic L, Sandoval W. Top-down mass spectrometry of native proteoforms and their complexes: A community study. RESEARCH SQUARE 2023:rs.3.rs-3228472. [PMID: 37674709 PMCID: PMC10479449 DOI: 10.21203/rs.3.rs-3228472/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
The combination of native electrospray ionisation with top-down fragmentation in mass spectrometry allows simultaneous determination of the stoichiometry of noncovalent complexes and identification of their component proteoforms and co-factors. While this approach is powerful, both native mass spectrometry and top-down mass spectrometry are not yet well standardised, and only a limited number of laboratories regularly carry out this type of research. To address this challenge, the Consortium for Top-Down Proteomics (CTDP) initiated a study to develop and test protocols for native mass spectrometry combined with top-down fragmentation of proteins and protein complexes across eleven instruments in nine laboratories. The outcomes are summarised in this report to provide robust benchmarks and a valuable entry point for the scientific community.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jeffrey Agar
- Department of Chemistry and Chemical Biology, Northeastern University
| | | | - Ying Ge
- University of Wisconsin-Madison
| | | | | | | | | | | | | |
Collapse
|
49
|
Phung W, Bakalarski CE, Hinkle TB, Sandoval W, Marty MT. UniDec Processing Pipeline for Rapid Analysis of Biotherapeutic Mass Spectrometry Data. Anal Chem 2023; 95:11491-11498. [PMID: 37478487 DOI: 10.1021/acs.analchem.3c02010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Recent advances in native mass spectrometry (MS) and denatured intact protein MS have made these techniques essential for biotherapeutic characterization. As MS analysis has increased in throughput and scale, new data analysis workflows are needed to provide rapid quantitation from large datasets. Here, we describe the UniDec processing pipeline (UPP) for the analysis of batched biotherapeutic intact MS data. UPP is built into the UniDec software package, which provides fast processing, deconvolution, and peak detection. The user and programming interfaces for UPP read a spreadsheet that contains the data file names, deconvolution parameters, and quantitation settings. After iterating through the spreadsheet and analyzing each file, it returns a spreadsheet of results and HTML reports. We demonstrate the use of UPP to measure the correct pairing percentage on a set of bispecific antibody data and to measure drug-to-antibody ratios from antibody-drug conjugates. Moreover, because the software is free and open-source, users can easily build on this platform to create customized workflows and calculations. Thus, UPP provides a flexible workflow that can be deployed in diverse settings and for a wide range of biotherapeutic applications.
Collapse
Affiliation(s)
- Wilson Phung
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc., South San Francisco, California 94080, United States
| | - Corey E Bakalarski
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc., South San Francisco, California 94080, United States
| | - Trent B Hinkle
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc., South San Francisco, California 94080, United States
| | - Wendy Sandoval
- Microchemistry, Proteomics, and Lipidomics Department, Genentech, Inc., South San Francisco, California 94080, United States
| | - Michael T Marty
- Department of Chemistry and Biochemistry and the Bio5 Institute, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
50
|
Ventouri IK, Veelders S, Passamonti M, Endres P, Roemling R, Schoenmakers PJ, Somsen GW, Haselberg R, Gargano AFG. Micro-flow size-exclusion chromatography for enhanced native mass spectrometry of proteins and protein complexes. Anal Chim Acta 2023; 1266:341324. [PMID: 37244657 DOI: 10.1016/j.aca.2023.341324] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/29/2023] [Accepted: 05/03/2023] [Indexed: 05/29/2023]
Abstract
Size-exclusion chromatography (SEC) employing aqueous mobile phases with volatile salts at neutral pH combined with native mass spectrometry (nMS) is a valuable tool to characterize proteins and protein aggregates in their native state. However, the liquid-phase conditions (high salt concentrations) frequently used in SEC-nMS hinder the analysis of labile protein complexes in the gas phase, necessitating higher desolvation-gas flow and source temperature, leading to protein fragmentation/dissociation. To overcome this issue, we investigated narrow SEC columns (1.0 mm internal diameter, I.D.) operated at 15-μL/min flow rates and their coupling to nMS for the characterization of proteins, protein complexes and higher-order structures (HOS). The reduced flow rate resulted in a significant increase in the protein-ionization efficiency, facilitating the detection of low-abundant impurities and HOS up to 230 kDa (i.e., the upper limit of the Orbitrap-MS instrument used). More-efficient solvent evaporation and lower desolvation energies allowed for softer ionization conditions (e.g., lower gas temperatures), ensuring little or no structural alterations of proteins and their HOS during transfer into the gas phase. Furthermore, ionization suppression by eluent salts was decreased, permitting the use of volatile-salt concentrations up to 400 mM. Band broadening and loss of resolution resulting from the introduction of injection volumes exceeding 3% of the column volume could be circumvented by incorporating an online trap-column containing a mixed-bed ion-exchange (IEX) material. The online IEX-based solid-phase extraction (SPE) or "trap-and-elute" set-up provided on-column focusing (sample preconcentration). This allowed the injection of large sample volumes on the 1-mm I.D. SEC column without compromising the separation. The enhanced sensitivity attained by the micro-flow SEC-MS, along with the on-column focusing achieved by the IEX precolumn, provided picogram detection limits for proteins.
Collapse
Affiliation(s)
- Iro K Ventouri
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands.
| | - Sharene Veelders
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands
| | - Marta Passamonti
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands
| | - Patrick Endres
- Tosoh Bioscience GmbH, Im Leuschnerpark 4, 64347, Griesheim, Germany
| | - Regina Roemling
- Tosoh Bioscience GmbH, Im Leuschnerpark 4, 64347, Griesheim, Germany
| | - Peter J Schoenmakers
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands
| | - Govert W Somsen
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Division of Bioanalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081, HV Amsterdam, the Netherlands
| | - Rob Haselberg
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Division of Bioanalytical Chemistry, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081, HV Amsterdam, the Netherlands
| | - Andrea F G Gargano
- Analytical Chemistry group, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands; Centre for Analytical Sciences Amsterdam, van't Hoff Insititute for Molecular Sciences (HIMS), University of Amsterdam, Science Park 904, 1098XH, Amsterdam, the Netherlands.
| |
Collapse
|