1
|
Wang W, Maliepaard JCL, Damelang T, Vidarsson G, Heck AJ, Reiding KR. Human IgG Subclasses Differ in the Structural Elements of Their N-Glycosylation. ACS CENTRAL SCIENCE 2024; 10:2048-2058. [PMID: 39634222 PMCID: PMC11613209 DOI: 10.1021/acscentsci.4c01157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 12/07/2024]
Abstract
Although immunoglobulin G (IgG) harbors just one N-glycosylation site per heavy chain, this glycosylation plays a key role in modulating its function. In human serum, IgG is classified into four subclasses (IgG1, IgG2, IgG3, IgG4), each characterized by unique features in their sequences, disulfide bridges and glycosylation signatures. While protein glycosylation is typically studied at the compositional level, this severely underestimates the complexity of the molecules involved. Glycan functionality heavily relies on the precise linkages and branching between monosaccharides, yet these features are challenging to study. Here, by development of a nanohydrophilic interaction chromatography (HILIC)-LC-MS/MS method, we reveal distinct structural glycosylation signatures for each of the four IgG subclasses, namely that IgG1 and IgG3 display predominant galactosylation of the 6-branched antenna, IgG2 instead of the 3-branched antenna, while IgG4 displays a balance. These and other subclass-specific glycostructural elements proved observable in both recombinant and endogenous IgGs as present in human plasma, in which interindividual differences and temporal stability could be demonstrated. Structural glycoproteomics is expected to fundamentally alter the way in which we study IgG, opening up a new layer of functional investigation and biomarker development, while also revealing new key structural differences between recombinant IgG subclasses in therapeutic applications.
Collapse
Affiliation(s)
- Weiwei Wang
- 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, 3584
CS Utrecht, The
Netherlands
- School
of Pharmaceutical Science, Shanghai Jiao
Tong University, 800
Dongchuan Road, 200240 Shanghai, People’s Republic
of China
| | - Joshua C. L. Maliepaard
- 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, 3584
CS Utrecht, The
Netherlands
| | - Timon Damelang
- 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
- Sanquin
Research, Department of Experimental Immunohematology
and Landsteiner Laboratory, Amsterdam 1006 AD, The Netherlands
- Sanquin
Research, Department of Immunopathology, Amsterdam 1006 AD, The Netherlands
| | - Gestur Vidarsson
- 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, 3584
CS Utrecht, The
Netherlands
- Sanquin
Research, Immunoglobulin Research Laboratory, Amsterdam 1006 AD, 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, 3584
CS Utrecht, The
Netherlands
| | - Karli R. Reiding
- 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, 3584
CS Utrecht, The
Netherlands
| |
Collapse
|
2
|
Hu Q, Wu HJ. Direct Glycan Analysis of Biological Samples and Intact Glycoproteins by Integrating Machine Learning-Driven Surface-Enhanced Raman Scattering and Boronic Acid Arrays. ACS MEASUREMENT SCIENCE AU 2024; 4:307-314. [PMID: 38910864 PMCID: PMC11191725 DOI: 10.1021/acsmeasuresciau.4c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/25/2024]
Abstract
Frequent monitoring of glycan patterns is a critical step in studying glycan-mediated cellular processes. However, the current glycan analysis tools are resource-intensive and less suitable for routine use in standard laboratories. We developed a novel glycan detection platform by integrating surface-enhanced Raman spectroscopy (SERS), boronic acid (BA) receptors, and machine learning tools. This sensor monitors the molecular fingerprint spectra of BA binding to cis-diol-containing glycans. Different types of BA receptors could yield different stereoselective reactions toward different glycans and exhibit unique vibrational spectra. By integration of the Raman spectra collected from different BA receptors, the structural information can be enriched, eventually improving the accuracy of glycan classification and quantification. Here, we established a SERS-based sensor incorporating multiple different BA receptors. This sensing platform could directly analyze the biological samples, including whole milk and intact glycoproteins (fetuin and asialofetuin), without tedious glycan release and purification steps. The results demonstrate the platform's ability to classify milk oligosaccharides with remarkable classification accuracy, despite the presence of other non-glycan constituents in the background. This sensor could also directly quantify sialylation levels of a fetuin/asialofetuin mixture without glycan release procedures. Moreover, by selecting appropriate BA receptors, the sensor exhibits an excellent performance of differentiating between α2,3 and α2,6 linkages of sialic acids. This low-cost, rapid, and highly accessible sensor will provide the scientific community with an invaluable tool for routine glycan screening in standard laboratories.
Collapse
Affiliation(s)
- Qiang Hu
- The Artie McFerrin Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Hung-Jen Wu
- The Artie McFerrin Department
of Chemical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
3
|
Rowe BA, Medina-Carle K, Chen K, Reese KJ, McCarthy KM, Concannon AA, Gunn GR, Gehman AP, Jiang Y, Meyer E. Unique challenges required reassessment and alterations to critical reagents to rescue a neutralizing antibody assay. Bioanalysis 2024; 16:735-745. [PMID: 38884331 PMCID: PMC11389750 DOI: 10.1080/17576180.2024.2360363] [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: 04/02/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
Aim: To redevelop a neutralizing antibody (NAb) assay to be much more drug tolerant, have a large dynamic range and have high inhibition when using high levels of positive control (PC).Materials & methods: Early assay data suggested that typical biotin labeling of the capture reagent (Drug 1, produced in a human cell line) was blocking it from binding with the PC or the detection target, and that the detection target was out competing the PC. Methodical biotin labeling experiments were performed at several challenge ratios and an Fc linker was added to the detection target.Results & conclusion: A larger dynamic range, high inhibition and higher drug tolerance were achieved by adding an acid dissociation step to the assay, performing atypical biotin labeling of Drug 1 and switching to a detection target that contained an Fc linker to increase steric hinderance and decrease its binding affinity to Drug 1.
Collapse
Affiliation(s)
- Blake A Rowe
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Katie Medina-Carle
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Keguan Chen
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Kimberly J Reese
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Kenneth M McCarthy
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Amy A Concannon
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - George R Gunn
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Andrew P Gehman
- GSK Research Statistics, Biostatistics, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| | - Yong Jiang
- Janssen Research & Development, 1400 McKean Rd, Lower Gwynedd Township, PA 19002,USA
| | - Erik Meyer
- GSK Precision Medicine, Biomarker & Bioanalytical Platforms, 1250 S Collegeville Rd, Collegeville, PA 19426, USA
| |
Collapse
|
4
|
Stanforth KJ, Zakhour MI, Chater PI, Wilcox MD, Adamson B, Robson NA, Pearson JP. The MUC2 Gene Product: Polymerisation and Post-Secretory Organisation-Current Models. Polymers (Basel) 2024; 16:1663. [PMID: 38932019 PMCID: PMC11207715 DOI: 10.3390/polym16121663] [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: 05/22/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
MUC2 mucin, the primary gel-forming component of intestinal mucus, is well researched and a model of polymerisation and post-secretory organisation has been published previously. Recently, several significant developments have been made which either introduce new ideas or challenge previous theories. New ideas include an overhaul of the MUC2 C-terminal globular structure which is proposed to harbour several previously unobserved domains, and include a site for an extra intermolecular disulphide bridge dimer between the cysteine 4379 of adjacent MUC2 C-termini. MUC2 polymers are also now thought to be secreted attached to the epithelial surface of goblet cells in the small intestine and removed following secretion via a metalloprotease meprin β-mediated cleavage of the von Willebrand D2 domain of the N-terminus. It remains unclear whether MUC2 forms intermolecular dimers, trimers, or both, at the N-termini during polymerisation, with several articles supporting either trimer or dimer formation. The presence of a firm inner mucus layer in the small intestine is similarly unclear. Considering this recent research, this review proposes an update to the previous model of MUC2 polymerisation and secretion, considers conflicting theories and data, and highlights the importance of this research to the understanding of MUC2 mucus layers in health and disease.
Collapse
Affiliation(s)
- Kyle J. Stanforth
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Maria I. Zakhour
- Biosciences Institute, Newcastle University Biosciences Institute, Catherine Cookson Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (M.I.Z.); (J.P.P.)
| | - Peter I. Chater
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Matthew D. Wilcox
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Beth Adamson
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Niamh A. Robson
- Aelius Biotech, The Medical School, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (P.I.C.); (M.D.W.); (B.A.); (N.A.R.)
| | - Jeffrey P. Pearson
- Biosciences Institute, Newcastle University Biosciences Institute, Catherine Cookson Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK; (M.I.Z.); (J.P.P.)
| |
Collapse
|
5
|
Bauer-Smith H, Sudol ASL, Beers SA, Crispin M. Serum immunoglobulin and the threshold of Fc receptor-mediated immune activation. Biochim Biophys Acta Gen Subj 2023; 1867:130448. [PMID: 37652365 PMCID: PMC11032748 DOI: 10.1016/j.bbagen.2023.130448] [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: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Antibodies can mediate immune recruitment or clearance of immune complexes through the interaction of their Fc domain with cellular Fc receptors. Clustering of antibodies is a key step in generating sufficient avidity for efficacious receptor recognition. However, Fc receptors may be saturated with prevailing, endogenous serum immunoglobulin and this raises the threshold by which cellular receptors can be productively engaged. Here, we review the factors controlling serum IgG levels in both healthy and disease states, and discuss how the presence of endogenous IgG is encoded into the functional activation thresholds for low- and high-affinity Fc receptors. We discuss the circumstances where antibody engineering can help overcome these physiological limitations of therapeutic antibodies. Finally, we discuss how the pharmacological control of Fc receptor saturation by endogenous IgG is emerging as a feasible mechanism for the enhancement of antibody therapeutics.
Collapse
Affiliation(s)
- Hannah Bauer-Smith
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stephen A Beers
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
6
|
García-Alija M, van Moer B, Sastre DE, Azzam T, Du JJ, Trastoy B, Callewaert N, Sundberg EJ, Guerin ME. Modulating antibody effector functions by Fc glycoengineering. Biotechnol Adv 2023; 67:108201. [PMID: 37336296 PMCID: PMC11027751 DOI: 10.1016/j.biotechadv.2023.108201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Antibody based drugs, including IgG monoclonal antibodies, are an expanding class of therapeutics widely employed to treat cancer, autoimmune and infectious diseases. IgG antibodies have a conserved N-glycosylation site at Asn297 that bears complex type N-glycans which, along with other less conserved N- and O-glycosylation sites, fine-tune effector functions, complement activation, and half-life of antibodies. Fucosylation, galactosylation, sialylation, bisection and mannosylation all generate glycoforms that interact in a specific manner with different cellular antibody receptors and are linked to a distinct functional profile. Antibodies, including those employed in clinical settings, are generated with a mixture of glycoforms attached to them, which has an impact on their efficacy, stability and effector functions. It is therefore of great interest to produce antibodies containing only tailored glycoforms with specific effects associated with them. To this end, several antibody engineering strategies have been developed, including the usage of engineered mammalian cell lines, in vitro and in vivo glycoengineering.
Collapse
Affiliation(s)
- Mikel García-Alija
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain
| | - Berre van Moer
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium
| | - Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tala Azzam
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jonathan J Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Beatriz Trastoy
- Structural Glycoimmunology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| | - Nico Callewaert
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium.
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| |
Collapse
|
7
|
Burock R, Cajic S, Hennig R, Buettner FFR, Reichl U, Rapp E. Reliable N-Glycan Analysis-Removal of Frequently Occurring Oligosaccharide Impurities by Enzymatic Degradation. Molecules 2023; 28:molecules28041843. [PMID: 36838829 PMCID: PMC9967028 DOI: 10.3390/molecules28041843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glycosylation, especially N-glycosylation, is one of the most common protein modifications, with immense importance at the molecular, cellular, and organismal level. Thus, accurate and reliable N-glycan analysis is essential in many areas of pharmaceutical and food industry, medicine, and science. However, due to the complexity of the cellular glycosylation process, in-depth glycoanalysis is still a highly challenging endeavor. Contamination of samples with oligosaccharide impurities (OSIs), typically linear glucose homo-oligomers, can cause further complications. Due to their physicochemical similarity to N-glycans, OSIs produce potentially overlapping signals, which can remain unnoticed. If recognized, suspected OSI signals are usually excluded in data evaluation. However, in both cases, interpretation of results can be impaired. Alternatively, sample preparation can be repeated to include an OSI removal step from samples. However, this significantly increases sample amount, time, and effort necessary. To overcome these issues, we investigated the option to enzymatically degrade and thereby remove interfering OSIs as a final sample preparation step. Therefore, we screened ten commercially available enzymes concerning their potential to efficiently degrade maltodextrins and dextrans as most frequently found OSIs. Of these enzymes, only dextranase from Chaetomium erraticum and glucoamylase P from Hormoconis resinae enabled a degradation of OSIs within only 30 min that is free of side reactions with N-glycans. Finally, we applied the straightforward enzymatic degradation of OSIs to N-glycan samples derived from different standard glycoproteins and various stem cell lysates.
Collapse
Affiliation(s)
- Robert Burock
- MPI for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
- glyXera GmbH, Brenneckestraße 20, 39120 Magdeburg, Germany
| | - Samanta Cajic
- MPI for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
- glyXera GmbH, Brenneckestraße 20, 39120 Magdeburg, Germany
| | - René Hennig
- MPI for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
- glyXera GmbH, Brenneckestraße 20, 39120 Magdeburg, Germany
- Correspondence:
| | - Falk F. R. Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Udo Reichl
- MPI for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
- Bioprocess Engineering, Otto-von-Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Erdmann Rapp
- MPI for Dynamics of Complex Technical Systems, Sandtorstraße 1, 39106 Magdeburg, Germany
- glyXera GmbH, Brenneckestraße 20, 39120 Magdeburg, Germany
| |
Collapse
|
8
|
Manuel LL, de los Ángeles César F, Pérez-Silva Nancy B, Celia PL, Elizabeth BR, Gonzalez Rosa O, Antonio GBJ, Jose S. Low-scale production and purification of a biologically active optimized form of the antitumor protein growth arrest specific 1 (GAS1) in a mammalian system for post-translational analysis. Biochem Eng J 2023. [DOI: 10.1016/j.bej.2023.108858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
9
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2019-2020. MASS SPECTROMETRY REVIEWS 2022:e21806. [PMID: 36468275 DOI: 10.1002/mas.21806] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
This review is the tenth update of the original article published in 1999 on the application of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2020. Also included are papers that describe methods appropriate to analysis by MALDI, such as sample preparation techniques, even though the ionization method is not MALDI. The review is basically divided into three sections: (1) general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation, quantification and the use of arrays. (2) Applications to various structural types such as oligo- and polysaccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals, and (3) other areas such as medicine, industrial processes and glycan synthesis where MALDI is extensively used. Much of the material relating to applications is presented in tabular form. The reported work shows increasing use of incorporation of new techniques such as ion mobility and the enormous impact that MALDI imaging is having. MALDI, although invented nearly 40 years ago is still an ideal technique for carbohydrate analysis and advancements in the technique and range of applications show little sign of diminishing.
Collapse
Affiliation(s)
- David J Harvey
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, UK
- Department of Chemistry, University of Oxford, Oxford, Oxfordshire, United Kingdom
| |
Collapse
|
10
|
Vattepu R, Sneed SL, Anthony RM. Sialylation as an Important Regulator of Antibody Function. Front Immunol 2022; 13:818736. [PMID: 35464485 PMCID: PMC9021442 DOI: 10.3389/fimmu.2022.818736] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/17/2022] [Indexed: 12/14/2022] Open
Abstract
Antibodies play a critical role in linking the adaptive immune response to the innate immune system. In humans, antibodies are categorized into five classes, IgG, IgM, IgA, IgE, and IgD, based on constant region sequence, structure, and tropism. In serum, IgG is the most abundant antibody, comprising 75% of antibodies in circulation, followed by IgA at 15%, IgM at 10%, and IgD and IgE are the least abundant. All human antibody classes are post-translationally modified by sugars. The resulting glycans take on many divergent structures and can be attached in an N-linked or O-linked manner, and are distinct by antibody class, and by position on each antibody. Many of these glycan structures on antibodies are capped by sialic acid. It is well established that the composition of the N-linked glycans on IgG exert a profound influence on its effector functions. However, recent studies have described the influence of glycans, particularly sialic acid for other antibody classes. Here, we discuss the role of glycosylation, with a focus on terminal sialylation, in the biology and function across all antibody classes. Sialylation has been shown to influence not only IgG, but IgE, IgM, and IgA biology, making it an important and unappreciated regulator of antibody function.
Collapse
Affiliation(s)
- Ravi Vattepu
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sunny Lyn Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert M Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
11
|
Glycoengineering of Therapeutic Antibodies with Small Molecule Inhibitors. Antibodies (Basel) 2021; 10:antib10040044. [PMID: 34842612 PMCID: PMC8628514 DOI: 10.3390/antib10040044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 01/22/2023] Open
Abstract
Monoclonal antibodies (mAbs) are one of the cornerstones of modern medicine, across an increasing range of therapeutic areas. All therapeutic mAbs are glycoproteins, i.e., their polypeptide chain is decorated with glycans, oligosaccharides of extraordinary structural diversity. The presence, absence, and composition of these glycans can have a profound effect on the pharmacodynamic and pharmacokinetic profile of individual mAbs. Approaches for the glycoengineering of therapeutic mAbs—the manipulation and optimisation of mAb glycan structures—are therefore of great interest from a technological, therapeutic, and regulatory perspective. In this review, we provide a brief introduction to the effects of glycosylation on the biological and pharmacological functions of the five classes of immunoglobulins (IgG, IgE, IgA, IgM and IgD) that form the backbone of all current clinical and experimental mAbs, including an overview of common mAb expression systems. We review selected examples for the use of small molecule inhibitors of glycan biosynthesis for mAb glycoengineering, we discuss the potential advantages and challenges of this approach, and we outline potential future applications. The main aim of the review is to showcase the expanding chemical toolbox that is becoming available for mAb glycoengineering to the biology and biotechnology community.
Collapse
|
12
|
Broszeit F, van Beek RJ, Unione L, Bestebroer TM, Chapla D, Yang JY, Moremen KW, Herfst S, Fouchier RAM, de Vries RP, Boons GJ. Glycan remodeled erythrocytes facilitate antigenic characterization of recent A/H3N2 influenza viruses. Nat Commun 2021; 12:5449. [PMID: 34521834 PMCID: PMC8440751 DOI: 10.1038/s41467-021-25713-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/18/2021] [Indexed: 01/04/2023] Open
Abstract
During circulation in humans and natural selection to escape antibody recognition for decades, A/H3N2 influenza viruses emerged with altered receptor specificities. These viruses lost the ability to agglutinate erythrocytes critical for antigenic characterization and give low yields and acquire adaptive mutations when cultured in eggs and cells, contributing to recent vaccine challenges. Examination of receptor specificities of A/H3N2 viruses reveals that recent viruses compensated for decreased binding of the prototypic human receptor by recognizing α2,6-sialosides on extended LacNAc moieties. Erythrocyte glycomics shows an absence of extended glycans providing a rationale for lack of agglutination by recent A/H3N2 viruses. A glycan remodeling approach installing functional receptors on erythrocytes, allows antigenic characterization of recent A/H3N2 viruses confirming the cocirculation of antigenically different viruses in humans. Computational analysis of HAs in complex with sialosides having extended LacNAc moieties reveals that mutations distal to the RBD reoriented the Y159 side chain resulting in an extended receptor binding site.
Collapse
MESH Headings
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- Antigens, Viral/metabolism
- Binding Sites
- Carbohydrate Sequence
- Erythrocytes/metabolism
- Erythrocytes/virology
- Glycomics/methods
- Glycosides/chemistry
- Glycosides/metabolism
- Hemagglutination Inhibition Tests
- Hemagglutinins, Viral/chemistry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/metabolism
- Host-Pathogen Interactions/genetics
- Humans
- Influenza A Virus, H3N2 Subtype/genetics
- Influenza A Virus, H3N2 Subtype/metabolism
- Influenza, Human/virology
- Microarray Analysis/methods
- Polysaccharides/chemistry
- Polysaccharides/metabolism
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- Protein Interaction Domains and Motifs
- Receptors, Virus/chemistry
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Sialic Acids/chemistry
- Sialic Acids/metabolism
Collapse
Affiliation(s)
- Frederik Broszeit
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Rosanne J van Beek
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Luca Unione
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Theo M Bestebroer
- Department of Viroscience, Erasmus MC, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Jeong-Yeh Yang
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Kelley W Moremen
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA
| | - Sander Herfst
- Department of Viroscience, Erasmus MC, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Ron A M Fouchier
- Department of Viroscience, Erasmus MC, P.O. Box 2040, Rotterdam, 3000 CA, The Netherlands
| | - Robert P de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, The Netherlands.
| | - Geert-Jan Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, The Netherlands.
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA, 30602, USA.
- Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands.
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
13
|
Baksmeier C, Blundell P, Steckel J, Schultz V, Gu Q, Da Silva Filipe A, Kohl A, Linnington C, Lu D, Dell A, Haslam S, Wang J, Czajkowsky D, Goebels N, Pleass RJ. Modified recombinant human IgG1-Fc is superior to natural intravenous immunoglobulin at inhibiting immune-mediated demyelination. Immunology 2021; 164:90-105. [PMID: 33880776 PMCID: PMC8358725 DOI: 10.1111/imm.13341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is an established treatment for numerous autoimmune conditions. Although Fc fragments derived from IVIG have shown efficacy in controlling immune thrombocytopenia in children, the mechanisms of action are unclear and controversial. The aim of this study was to dissect IVIG effector mechanisms using further adapted Fc fragments on demyelination in an ex vivo model of the central nervous system-immune interface. Using organotypic cerebellar slice cultures (OSCs) from transgenic mice, we induced extensive immune-mediated demyelination and oligodendrocyte loss with an antibody specific for myelin oligodendrocyte glycoprotein (MOG) and complement. Protective effects of adapted Fc fragments were assessed by live imaging of green fluorescent protein expression, immunohistochemistry and confocal microscopy. Cysteine- and glycan-adapted Fc fragments protected OSC from demyelination in a dose-dependent manner where equimolar concentrations of either IVIG or control Fc were ineffective. The protective effects of the adapted Fc fragments are partly attributed to interference with complement-mediated oligodendroglia damage. Transcriptome analysis ruled out signatures associated with inflammatory or innate immune responses. Taken together, our findings show that recombinant biomimetics can be made that are at least two hundred-fold more effective than IVIG in controlling demyelination by anti-MOG antibodies.
Collapse
Affiliation(s)
- Christine Baksmeier
- Department of NeurologyMedical FacultyHeinrich‐Heine‐University DuesseldorfDuesseldorfGermany
| | - Pat Blundell
- Department of Tropical Disease BiologyLiverpool School of Tropical MedicineLiverpoolUK
| | - Julia Steckel
- Department of NeurologyMedical FacultyHeinrich‐Heine‐University DuesseldorfDuesseldorfGermany
| | - Verena Schultz
- Institute of Infection, Immunity and InflammationCollege of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Quan Gu
- Institute of Infection, Immunity and InflammationCollege of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Ana Da Silva Filipe
- Institute of Infection, Immunity and InflammationCollege of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Alain Kohl
- Institute of Infection, Immunity and InflammationCollege of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Chris Linnington
- Institute of Infection, Immunity and InflammationCollege of Medical Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Dongli Lu
- Department of Life SciencesImperial College LondonLondonUK
| | - Anne Dell
- Department of Life SciencesImperial College LondonLondonUK
| | - Stuart Haslam
- Department of Life SciencesImperial College LondonLondonUK
| | - Jiabin Wang
- Shanghai Center for Systems BiomedicineKey Laboratory of Systems Biomedicine (Ministry of Education)Shanghai Jiao Tong UniversityShanghaiChina
| | - Dan Czajkowsky
- State Key Laboratory for Oncogenes and Related Genes and Bio‐ID CenterSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Norbert Goebels
- Department of NeurologyMedical FacultyHeinrich‐Heine‐University DuesseldorfDuesseldorfGermany
| | - Richard J. Pleass
- Department of Tropical Disease BiologyLiverpool School of Tropical MedicineLiverpoolUK
| |
Collapse
|
14
|
Savizi ISP, Motamedian E, E Lewis N, Jimenez Del Val I, Shojaosadati SA. An integrated modular framework for modeling the effect of ammonium on the sialylation process of monoclonal antibodies produced by CHO cells. Biotechnol J 2021; 16:e2100019. [PMID: 34021707 DOI: 10.1002/biot.202100019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Monoclonal antibodies (mABs) have emerged as one of the most important therapeutic recombinant proteins in the pharmaceutical industry. Their immunogenicity and therapeutic efficacy are influenced by post-translational modifications, specifically the glycosylation process. Bioprocess conditions can influence the intracellular process of glycosylation. Among all the process conditions that have been recognized to affect the mAB glycoforms, the detailed mechanism underlying how ammonium could perturb glycosylation remains to be fully understood. It was shown that ammonium induces heterogeneity in protein glycosylation by altering the sialic acid content of glycoproteins. Hence, understanding this mechanism would aid pharmaceutical manufacturers to ensure consistent protein glycosylation. METHODS Three different mechanisms have been proposed to explain how ammonium influences the sialylation process. In the first, the inhibition of CMP-sialic acid transporter, which transports CMP-sialic acid (sialylation substrate) into the Golgi, by an increase in UDP-GlcNAc content that is brought about by the augmented incorporation of ammonium into glucosamine formation. In the second, ammonia diffuses into the Golgi and raises its pH, thereby decreasing the sialyltransferase enzyme activity. In the third, the reduction of sialyltransferase enzyme expression level in the presence of ammonium. We employed these mechanisms in a novel integrated modular platform to link dynamic alteration in mAB sialylation process with extracellular ammonium concentration to elucidate how ammonium alters the sialic acid content of glycoproteins. RESULTS Our results show that the sialylation reaction rate is insensitive to the first mechanism. At low ammonium concentration, the second mechanism is the controlling mechanism in mAB sialylation and by increasing the ammonium level (< 8 mM) the third mechanism becomes the controlling mechanism. At higher ammonium concentrations (> 8 mM) the second mechanism becomes predominant again. CONCLUSION The presented model in this study provides a connection between extracellular ammonium and the monoclonal antibody sialylation process. This computational tool could help scientists to develop and formulate cell culture media. The model illustrated here can assist the researchers to select culture media that ensure consistent mAB sialylation.
Collapse
Affiliation(s)
- Iman Shahidi Pour Savizi
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Motamedian
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| | - Nathan E Lewis
- Department of Bioengineering, University of California, La Jolla, California, USA.,School of Medicine, Novo Nordisk Foundation Center for Biosustainability at the University of California, La Jolla, California, USA.,Department of Pediatrics, School of Medicine, University of California, La Jolla, California, USA
| | | | - Seyed Abbas Shojaosadati
- Faculty of Chemical Engineering, Biotechnology Department, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Donini R, Haslam SM, Kontoravdi C. Glycoengineering Chinese hamster ovary cells: a short history. Biochem Soc Trans 2021; 49:915-931. [PMID: 33704400 PMCID: PMC8106501 DOI: 10.1042/bst20200840] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 12/25/2022]
Abstract
Biotherapeutic glycoproteins have revolutionised the field of pharmaceuticals, with new discoveries and continuous improvements underpinning the rapid growth of this industry. N-glycosylation is a critical quality attribute of biotherapeutic glycoproteins that influences the efficacy, half-life and immunogenicity of these drugs. This review will focus on the advances and future directions of remodelling N-glycosylation in Chinese hamster ovary (CHO) cells, which are the workhorse of recombinant biotherapeutic production, with particular emphasis on antibody products, using strategies such as cell line and protein backbone engineering.
Collapse
Affiliation(s)
- Roberto Donini
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
| | - Cleo Kontoravdi
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| |
Collapse
|
16
|
Pleass RJ. The therapeutic potential of sialylated Fc domains of human IgG. MAbs 2021; 13:1953220. [PMID: 34288809 PMCID: PMC8296966 DOI: 10.1080/19420862.2021.1953220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/08/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Pathogens frequently use multivalent binding to sialic acid to infect cells or to modulate immunity through interactions with human sialic acid-binding immunoglobulin-type lectins (Siglecs). Molecules that interfere with these interactions could be of interest as diagnostics, anti-infectives or as immune modulators. This review describes the development of molecular scaffolds based on the crystallizable fragment (Fc) region of immunoglobulin (Ig) G that deliver high-avidity binding to innate immune receptors, including sialic acid-dependent receptors. The ways in which the sialylated Fc may be engineered as immune modulators that mimic the anti-inflammatory properties of intravenous polyclonal Ig or as blockers of sialic-acid-dependent infectivity by viruses are also discussed.
Collapse
Affiliation(s)
- Richard J. Pleass
- Department of Tropical Disease Biology, Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
17
|
IgE Antibodies against Cancer: Efficacy and Safety. Antibodies (Basel) 2020; 9:antib9040055. [PMID: 33081206 PMCID: PMC7709114 DOI: 10.3390/antib9040055] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Immunoglobulin E (IgE) antibodies are well known for their role in allergic diseases and for contributions to antiparasitic immune responses. Properties of this antibody class that mediate powerful effector functions may be redirected for the treatment of solid tumours. This has led to the rise of a new class of therapeutic antibodies to complement the armamentarium of approved tumour targeting antibodies, which to date are all IgG class. The perceived risk of type I hypersensitivity reactions following administration of IgE has necessitated particular consideration in the development of these therapeutic agents. Here, we bring together the properties of IgE antibodies pivotal to the hypothesis for superior antitumour activity compared to IgG, observations of in vitro and in vivo efficacy and mechanisms of action, and a focus on the safety considerations for this novel class of therapeutic agent. These include in vitro studies of potential hypersensitivity, selection of and observations from appropriate in vivo animal models and possible implications of the high degree of glycosylation of IgE. We also discuss the use of ex vivo predictive and monitoring clinical tools, as well as the risk mitigation steps employed in, and the preliminary outcomes from, the first-in-human clinical trial of a candidate anticancer IgE therapeutic.
Collapse
|