101
|
Longitudinal monitoring of immunoglobulin A glycosylation during pregnancy by simultaneous MALDI-FTICR-MS analysis of N- and O-glycopeptides. Sci Rep 2016; 6:27955. [PMID: 27302155 PMCID: PMC4908400 DOI: 10.1038/srep27955] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/24/2016] [Indexed: 12/13/2022] Open
Abstract
Immunoglobulin A (IgA) is a glycoprotein of which altered glycosylation has been associated with several pathologies. Conventional methods for IgA N- and O-glycosylation analysis are tedious, thus limiting such analyses to small sample sizes. Here we present a high-throughput strategy for the simultaneous analysis of serum-derived IgA1 N- and O-glycopeptides using matrix-assisted laser/desorption ionisation Fourier transform ion cyclotron resonance (MALDI-FTICR) mass spectrometry (MS). Six non-fucosylated diantennary complex type glycoforms were detected on the Asn144-containing glycopeptide. Thirteen distinct glycoforms were identified for the Asn340-containing tailpiece glycopeptide, mainly of the diantennary complex type, and low amounts of triantennary glycoforms. Simultaneously with these N-glycopeptides, 53 compositional glycoforms of the hinge region O-glycopeptide were profiled in a single high resolution MALDI-FTICR spectrum. Since many pregnancy associated changes have been recognized for immunoglobulin G, we sought to demonstrate the clinical applicability of this method in a cohort of 29 pregnant women, from whom samples were collected at three time points during pregnancy and three time points after delivery. Pregnancy associated changes of N-glycan bisection were different for IgA1 as compared to IgG-Fc described earlier. We foresee further applications of the developed method for larger patient cohorts to study IgA N- and O-glycosylation changes in pathologies.
Collapse
|
102
|
Knoppova B, Reily C, Maillard N, Rizk DV, Moldoveanu Z, Mestecky J, Raska M, Renfrow MB, Julian BA, Novak J. The Origin and Activities of IgA1-Containing Immune Complexes in IgA Nephropathy. Front Immunol 2016; 7:117. [PMID: 27148252 PMCID: PMC4828451 DOI: 10.3389/fimmu.2016.00117] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 03/15/2016] [Indexed: 12/12/2022] Open
Abstract
IgA nephropathy (IgAN) is the most common primary glomerulonephritis, frequently leading to end-stage renal disease, as there is no disease-specific therapy. IgAN is diagnosed from pathological assessment of a renal biopsy specimen based on predominant or codominant IgA-containing immunodeposits, usually with complement C3 co-deposits and with variable presence of IgG and/or IgM. The IgA in these renal deposits is galactose-deficient IgA1, with less than a full complement of galactose residues on the O-glycans in the hinge region of the heavy chains. Research from the past decade led to the definition of IgAN as an autoimmune disease with a multi-hit pathogenetic process with contributing genetic and environmental components. In this process, circulating galactose-deficient IgA1 (autoantigen) is bound by antiglycan IgG or IgA (autoantibodies) to form immune complexes. Some of these circulating complexes deposit in glomeruli, and thereby activate mesangial cells and induce renal injury through cellular proliferation and overproduction of extracellular matrix components and cytokines/chemokines. Glycosylation pathways associated with production of the autoantigen and the unique characteristics of the corresponding autoantibodies in patients with IgAN have been uncovered. Complement likely plays a significant role in the formation and the nephritogenic activities of these complexes. Complement activation is mediated through the alternative and lectin pathways and probably occurs systemically on IgA1-containing circulating immune complexes as well as locally in glomeruli. Incidence of IgAN varies greatly by geographical location; the disease is rare in central Africa but accounts for up to 40% of native-kidney biopsies in eastern Asia. Some of this variation may be explained by genetically determined influences on the pathogenesis of the disease. Genome-wide association studies to date have identified several loci associated with IgAN. Some of these loci are associated with the increased prevalence of IgAN, whereas others, such as deletion of complement factor H-related genes 1 and 3, are protective against the disease. Understanding the molecular mechanisms and genetic and biochemical factors involved in formation and activities of pathogenic IgA1-containing immune complexes will enable the development of future disease-specific therapies as well as identification of non-invasive disease-specific biomarkers.
Collapse
Affiliation(s)
- Barbora Knoppova
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Colin Reily
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicolas Maillard
- Université Jean Monnet, Saint Etienne, France
- PRES Université de Lyon, Lyon, France
| | - Dana V. Rizk
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zina Moldoveanu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jiri Mestecky
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Milan Raska
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, Czech Republic
| | - Matthew B. Renfrow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bruce A. Julian
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
103
|
Borrok MJ, Luheshi NM, Beyaz N, Davies GC, Legg JW, Wu H, Dall'Acqua WF, Tsui P. Enhancement of antibody-dependent cell-mediated cytotoxicity by endowing IgG with FcαRI (CD89) binding. MAbs 2016; 7:743-51. [PMID: 25970007 DOI: 10.1080/19420862.2015.1047570] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Fc effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cell-mediated phagocytosis (ADCP) are crucial to the efficacy of many antibody therapeutics. In addition to IgG, antibodies of the IgA isotype can also promote cell killing through engagement of myeloid lineage cells via interactions between the IgA-Fc and FcαRI (CD89). Herein, we describe a unique, tandem IgG1/IgA2 antibody format in the context of a trastuzumab variable domain that exhibits enhanced ADCC and ADCP capabilities. The IgG1/IgA2 tandem Fc format retains IgG1 FcγR binding as well as FcRn-mediated serum persistence, yet is augmented with myeloid cell-mediated effector functions via FcαRI/IgA Fc interactions. In this work, we demonstrate anti-human epidermal growth factor receptor-2 antibodies with the unique tandem IgG1/IgA2 Fc can better recruit and engage cytotoxic polymorphonuclear (PMN) cells than either the parental IgG1 or IgA2. Pharmacokinetics of IgG1/IgA2 in BALB/c mice are similar to the parental IgG, and far surpass the poor serum persistence of IgA2. The IgG1/IgA2 format is expressed at similar levels and with similar thermal stability to IgG1, and can be purified via standard protein A chromatography. The tandem IgG1/IgA2 format could potentially augment IgG-based immunotherapeutics with enhanced PMN-mediated cytotoxicity while avoiding many of the problems associated with developing IgAs.
Collapse
Key Words
- ADCC
- ADCC, antibody-dependent cell-mediated cytotoxicity
- ADCP, antibody-dependent cell-mediated phagocytosis
- AUC, area under the curve; CL, clearance rate
- CD89
- CDC, complement dependent cytotoxicity
- Cmax, maximum serum concentration
- DSC, differential scanning calorimetry
- E:T ratio, effector to target ratio
- FCM, flow cytometry
- FcRn, neonatal Fc receptor
- FcαRI
- FcγR, Fc gamma receptor
- HER2, human epithelial receptor two
- IgA
- IgA, immunoglobulin A
- IgG, immunoglobulin G
- LDH, lactate dehydrogenase
- MΦ, macrophage
- NK, natural killer
- PBMC, peripheral blood mononuclear cell
- PK, pharmacokinetics
- PMN, polymorphonuclear
- SPR, surface plasmon resonance
- TAA, tumor associated antigens
- T½, half-life
- Vss, central compartment volume of distribution
- macrophage
- monoclonal antibody
- neutrophil
- tandem
- trastuzumab
Collapse
Affiliation(s)
- M Jack Borrok
- a Antibody Discovery and Protein Engineering; Medimmune Ltd. ; Gaithersburg , MD , USA
| | | | | | | | | | | | | | | |
Collapse
|
104
|
Hinneburg H, Stavenhagen K, Schweiger-Hufnagel U, Pengelley S, Jabs W, Seeberger PH, Silva DV, Wuhrer M, Kolarich D. The Art of Destruction: Optimizing Collision Energies in Quadrupole-Time of Flight (Q-TOF) Instruments for Glycopeptide-Based Glycoproteomics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2016; 27:507-19. [PMID: 26729457 PMCID: PMC4756043 DOI: 10.1007/s13361-015-1308-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 05/12/2023]
Abstract
In-depth site-specific investigations of protein glycosylation are the basis for understanding the biological function of glycoproteins. Mass spectrometry-based N- and O-glycopeptide analyses enable determination of the glycosylation site, site occupancy, as well as glycan varieties present on a particular site. However, the depth of information is highly dependent on the applied analytical tools, including glycopeptide fragmentation regimes and automated data analysis. Here, we used a small set of synthetic disialylated, biantennary N-glycopeptides to systematically tune Q-TOF instrument parameters towards optimal energy stepping collision induced dissociation (CID) of glycopeptides. A linear dependency of m/z-ratio and optimal fragmentation energy was found, showing that with increasing m/z-ratio, more energy is required for glycopeptide fragmentation. Based on these optimized fragmentation parameters, a method combining lower- and higher-energy CID was developed, allowing the online acquisition of glycan and peptide-specific fragments within a single tandem MS experiment. We validated this method analyzing a set of human immunoglobulins (IgA1+2, sIgA, IgG1+2, IgE, IgD, IgM) as well as bovine fetuin. These optimized fragmentation parameters also enabled software-assisted glycopeptide assignment of both N- and O-glycopeptides including information about the most abundant glycan compositions, peptide sequence and putative structures. Twenty-six out of 30 N-glycopeptides and four out of five O-glycopeptides carrying >110 different glycoforms could be identified by this optimized LC-ESI tandem MS method with minimal user input. The Q-TOF based glycopeptide analysis platform presented here opens the way to a range of different applications in glycoproteomics research as well as biopharmaceutical development and quality control.
Collapse
Affiliation(s)
- Hannes Hinneburg
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Kathrin Stavenhagen
- Division of BioAnalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424, Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Arnimallee 22, 14195, Berlin, Germany
| | - Daniel Varón Silva
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424, Potsdam, Germany
| | - Manfred Wuhrer
- Division of BioAnalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424, Potsdam, Germany.
| |
Collapse
|
105
|
Virdi V, Juarez P, Boudolf V, Depicker A. Recombinant IgA production for mucosal passive immunization, advancing beyond the hurdles. Cell Mol Life Sci 2016; 73:535-45. [PMID: 26511868 PMCID: PMC11108522 DOI: 10.1007/s00018-015-2074-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 09/26/2015] [Accepted: 10/14/2015] [Indexed: 01/21/2023]
Abstract
Vaccination is a successful strategy to proactively develop immunity to a certain pathogen, but most vaccines fail to trigger a specific immune response at the mucosal surfaces, which are the first port of entry for infectious agents. At the mucosal surfaces, the predominant immunoglobulin is secretory IgA (SIgA) that specifically neutralizes viruses and prevents bacterial colonization. Mucosal passive immunization, i.e. the application of pathogen-specific SIgAs at the mucosae, can be an effective alternative to achieve mucosal protection. However, this approach is not straightforward, mainly because SIgAs are difficult to obtain from convalescent sources, while recombinant SIgA production is challenging due to its complex structure. This review provides an overview of manufacturing difficulties presented by the unique structural diversity of SIgAs, and the innovative solutions being explored for SIgA production in mammalian and plant expression systems.
Collapse
Affiliation(s)
- Vikram Virdi
- Department of Plant Systems Biology, VIB, 9052, Ghent, Belgium.
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052, Ghent, Belgium.
| | - Paloma Juarez
- Department of Plant Systems Biology, VIB, 9052, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Veronique Boudolf
- Department of Plant Systems Biology, VIB, 9052, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Ann Depicker
- Department of Plant Systems Biology, VIB, 9052, Ghent, Belgium.
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technologiepark 927, 9052, Ghent, Belgium.
| |
Collapse
|
106
|
Lohse S, Meyer S, Meulenbroek LAPM, Jansen JHM, Nederend M, Kretschmer A, Klausz K, Möginger U, Derer S, Rösner T, Kellner C, Schewe D, Sondermann P, Tiwari S, Kolarich D, Peipp M, Leusen JHW, Valerius T. An Anti-EGFR IgA That Displays Improved Pharmacokinetics and Myeloid Effector Cell Engagement In Vivo. Cancer Res 2015; 76:403-17. [PMID: 26634925 DOI: 10.1158/0008-5472.can-15-1232] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/07/2015] [Indexed: 11/16/2022]
Abstract
Antibodies of IgA isotype effectively engage myeloid effector cells for cancer immunotherapy. Here, we describe preclinical studies with an Fc engineered IgA2m(1) antibody containing the variable regions of the EGFR antibody cetuximab. Compared with wild-type IgA2m(1), the engineered molecule lacked two N-glycosylation sites (N166 and N337), two free cysteines (C311 and C472), and contained a stabilized heavy and light chain linkage (P221R mutation). This novel molecule displayed improved production rates and biochemical properties compared with wild-type IgA. In vitro, Fab- and Fc-mediated effector functions, such as inhibition of ligand binding, receptor modulation, and engagement of myeloid effector cells for antibody-dependent cell-mediated cytotoxicity, were similar between wild-type and engineered IgA2. The engineered antibody displayed lower levels of terminal galactosylation leading to reduced asialoglycoprotein-receptor binding and to improved pharmacokinetic properties. In a long-term in vivo model against EGFR-positive cancer cells, improved serum half-life translated into higher efficacy of the engineered molecule, which required myeloid cells expressing human FcαRI for its full efficacy. However, Fab-mediated effector functions contributed to the in vivo efficacy because the novel IgA antibody demonstrated therapeutic activity also in non-FcαRI transgenic mice. Together, these results demonstrate that engineering of an IgA antibody can significantly improve its pharmacokinetics and its therapeutic efficacy to inhibit tumor growth in vivo.
Collapse
Affiliation(s)
- Stefan Lohse
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Saskia Meyer
- Laboratory for Translational Immunology, Immunotherapy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laura A P M Meulenbroek
- Laboratory for Translational Immunology, Immunotherapy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J H Marco Jansen
- Laboratory for Translational Immunology, Immunotherapy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maaike Nederend
- Laboratory for Translational Immunology, Immunotherapy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anna Kretschmer
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Katja Klausz
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Uwe Möginger
- Department of Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Potsdam, Germany. Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Thies Rösner
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Denis Schewe
- Department of General Pediatrics, Christian-Albrechts-University, Kiel, Germany
| | | | - Sanjay Tiwari
- Molecular Imaging North Competence Center, Christian-Albrechts-University, Kiel, Germany
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Potsdam, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany
| | - Jeanette H W Leusen
- Laboratory for Translational Immunology, Immunotherapy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, 2 Department of Medicine, Christian-Albrechts-University, Kiel, Germany.
| |
Collapse
|
107
|
Jansen BC, Reiding KR, Bondt A, Hipgrave Ederveen AL, Palmblad M, Falck D, Wuhrer M. MassyTools: A High-Throughput Targeted Data Processing Tool for Relative Quantitation and Quality Control Developed for Glycomic and Glycoproteomic MALDI-MS. J Proteome Res 2015; 14:5088-98. [PMID: 26565759 DOI: 10.1021/acs.jproteome.5b00658] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The study of N-linked glycosylation has long been complicated by a lack of bioinformatics tools. In particular, there is still a lack of fast and robust data processing tools for targeted (relative) quantitation. We have developed modular, high-throughput data processing software, MassyTools, that is capable of calibrating spectra, extracting data, and performing quality control calculations based on a user-defined list of glycan or glycopeptide compositions. Typical examples of output include relative areas after background subtraction, isotopic pattern-based quality scores, spectral quality scores, and signal-to-noise ratios. We demonstrated MassyTools' performance on MALDI-TOF-MS glycan and glycopeptide data from different samples. MassyTools yielded better calibration than the commercial software flexAnalysis, generally showing 2-fold better ppm errors after internal calibration. Relative quantitation using MassyTools and flexAnalysis gave similar results, yielding a relative standard deviation (RSD) of the main glycan of ~6%. However, MassyTools yielded 2- to 5-fold lower RSD values for low-abundant analytes than flexAnalysis. Additionally, feature curation based on the computed quality criteria improved the data quality. In conclusion, we show that MassyTools is a robust automated data processing tool for high-throughput, high-performance glycosylation analysis. The package is released under the Apache 2.0 license and is freely available on GitHub ( https://github.com/Tarskin/MassyTools ).
Collapse
Affiliation(s)
- Bas C Jansen
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | - Karli R Reiding
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | - Albert Bondt
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands.,Department of Rheumatology, Erasmus University Medical Center , 3000 CA Rotterdam, The Netherlands
| | - Agnes L Hipgrave Ederveen
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | - Magnus Palmblad
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | - David Falck
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center , 2300 RC Leiden, The Netherlands.,Division of BioAnalytical Chemistry, VU University Amsterdam , 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
108
|
Clerc F, Reiding KR, Jansen BC, Kammeijer GSM, Bondt A, Wuhrer M. Human plasma protein N-glycosylation. Glycoconj J 2015; 33:309-43. [PMID: 26555091 PMCID: PMC4891372 DOI: 10.1007/s10719-015-9626-2] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 01/09/2023]
Abstract
Glycosylation is the most abundant and complex protein modification, and can have a profound structural and functional effect on the conjugate. The oligosaccharide fraction is recognized to be involved in multiple biological processes, and to affect proteins physical properties, and has consequentially been labeled a critical quality attribute of biopharmaceuticals. Additionally, due to recent advances in analytical methods and analysis software, glycosylation is targeted in the search for disease biomarkers for early diagnosis and patient stratification. Biofluids such as saliva, serum or plasma are of great use in this regard, as they are easily accessible and can provide relevant glycosylation information. Thus, as the assessment of protein glycosylation is becoming a major element in clinical and biopharmaceutical research, this review aims to convey the current state of knowledge on the N-glycosylation of the major plasma glycoproteins alpha-1-acid glycoprotein, alpha-1-antitrypsin, alpha-1B-glycoprotein, alpha-2-HS-glycoprotein, alpha-2-macroglobulin, antithrombin-III, apolipoprotein B-100, apolipoprotein D, apolipoprotein F, beta-2-glycoprotein 1, ceruloplasmin, fibrinogen, immunoglobulin (Ig) A, IgG, IgM, haptoglobin, hemopexin, histidine-rich glycoprotein, kininogen-1, serotransferrin, vitronectin, and zinc-alpha-2-glycoprotein. In addition, the less abundant immunoglobulins D and E are included because of their major relevance in immunology and biopharmaceutical research. Where available, the glycosylation is described in a site-specific manner. In the discussion, we put the glycosylation of individual proteins into perspective and speculate how the individual proteins may contribute to a total plasma N-glycosylation profile determined at the released glycan level.
Collapse
Affiliation(s)
- Florent Clerc
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Karli R Reiding
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Bas C Jansen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Guinevere S M Kammeijer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Albert Bondt
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.,Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands. .,Division of BioAnalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
109
|
Hu S, Bao H, Xu X, Zhou X, Qin W, Zeng C, Liu Z. Increased miR-374b promotes cell proliferation and the production of aberrant glycosylated IgA1 in B cells of IgA nephropathy. FEBS Lett 2015; 589:4019-25. [PMID: 26545495 DOI: 10.1016/j.febslet.2015.10.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/24/2015] [Accepted: 10/27/2015] [Indexed: 10/22/2022]
Abstract
The number of B cells is increased and the O-glycans of IgA1 are incompletely galactosylated in IgA nephropathy (IgAN). Here we report that expression of phosphatase and tensin homolog (PTEN) and Cosmc is decreased in B cells, and correlates with B cell number and the aberrant glycosylation of IgA1 in IgAN. Patients with IgAN exhibit higher miR-374b in B cells compared to controls. We show that miR-374b targets PTEN and Cosmc by luciferase assays and western blot analysis. Inhibition of miR-374b increased PTEN and Cosmc expression, and prevented cell proliferation and aberrant glycosylation of IgA1, thus representing a new therapeutic approach for IgAN.
Collapse
Affiliation(s)
- Shuai Hu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hao Bao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xianguang Zhou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Weisong Qin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Caihong Zeng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
| |
Collapse
|
110
|
Hong Q, Ruhaak LR, Stroble C, Parker E, Huang J, Maverakis E, Lebrilla CB. A Method for Comprehensive Glycosite-Mapping and Direct Quantitation of Serum Glycoproteins. J Proteome Res 2015; 14:5179-92. [PMID: 26510530 DOI: 10.1021/acs.jproteome.5b00756] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A comprehensive glycan map was constructed for the top eight abundant glycoproteins in plasma using both specific and nonspecific enzyme digestions followed by nano liquid chromatography (LC)-chip/quadrupole time-of-flight mass spectrometry (MS) analysis. Glycopeptides were identified using an in-house software tool, GPFinder. A sensitive and reproducible multiple reaction monitoring (MRM) technique on a triple quadrupole MS was developed and applied to quantify immunoglobulins G, A, M, and their site-specific glycans simultaneously and directly from human serum/plasma without protein enrichments. A total of 64 glycopeptides and 15 peptides were monitored for IgG, IgA, and IgM in a 20 min ultra high performance (UP)LC gradient. The absolute protein contents were quantified using peptide calibration curves. The glycopeptide ion abundances were normalized to the respective protein abundances to separate protein glycosylation from protein expression. This technique yields higher method reproducibility and less sample loss when compared with the quantitation method that involves protein enrichments. The absolute protein quantitation has a wide linear range (3-4 orders of magnitude) and low limit of quantitation (femtomole level). This rapid and robust quantitation technique, which provides quantitative information for both proteins and glycosylation, will further facilitate disease biomarker discoveries.
Collapse
Affiliation(s)
- Qiuting Hong
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| | - L Renee Ruhaak
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| | - Carol Stroble
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| | - Evan Parker
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| | - Jincui Huang
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| | - Emanual Maverakis
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| | - Carlito B Lebrilla
- Department of Chemistry and ‡Department of Dermatology, School of Medicine, University of California , Davis, California 95616, United States
| |
Collapse
|
111
|
Meyer S, Nederend M, Jansen JHM, Reiding KR, Jacobino SR, Meeldijk J, Bovenschen N, Wuhrer M, Valerius T, Ubink R, Boross P, Rouwendal G, Leusen JHW. Improved in vivo anti-tumor effects of IgA-Her2 antibodies through half-life extension and serum exposure enhancement by FcRn targeting. MAbs 2015; 8:87-98. [PMID: 26466856 PMCID: PMC4966554 DOI: 10.1080/19420862.2015.1106658] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Antibody therapy is a validated treatment approach for several malignancies. All currently clinically applied therapeutic antibodies (Abs) are of the IgG isotype. However, not all patients respond to this therapy and relapses can occur. IgA represents an alternative isotype for antibody therapy that engages FcαRI expressing myeloid effector cells, such as neutrophils and monocytes. IgA Abs have been shown to effectively kill tumor cells both in vitro and in vivo. However, due to the short half-life of IgA Abs in mice, daily injections are required to reach an effect comparable to IgG Abs. The relatively long half-life of IgG Abs and serum albumin arises from their capability of interacting with the neonatal Fc receptor (FcRn). As IgA Abs lack a binding site for FcRn, we generated IgA Abs with the variable regions of the Her2-specific Ab trastuzumab and attached an albumin-binding domain (ABD) to the heavy or light chain (HCABD/LCABD) to extend their serum half-life. These modified Abs were able to bind albumin from different species in vitro. Furthermore, tumor cell lysis of IgA-Her2-LCABD Abs in vitro was similar to unmodified IgA-Her2 Abs. Pharmacokinetic studies in mice revealed that the serum exposure and half-life of the modified IgA-Her2 Abs was extended. In a xenograft mouse model, the modified IgA1 Abs exhibited a slightly, but significantly, improved anti-tumor response compared to the unmodified Ab. In conclusion, empowering IgA Abs with albumin-binding capacity results in in vitro and in vivo functional Abs with an enhanced exposure and prolonged half-life.
Collapse
Affiliation(s)
- Saskia Meyer
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Maaike Nederend
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - J H Marco Jansen
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Karli R Reiding
- b Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden , The Netherlands
| | - Shamir R Jacobino
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Jan Meeldijk
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | - Niels Bovenschen
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands.,c Department of Pathology ; UMC Utrecht ; Utrecht , The Netherlands
| | - Manfred Wuhrer
- b Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden , The Netherlands
| | - Thomas Valerius
- d Division of Stem Cell Transplantation and Immunotherapy ; Department of Internal Medicine II; Christian-Albrechts-University ; Kiel , Germany
| | - Ruud Ubink
- e Synthon Biopharmaceuticals BV ; Nijmegen , The Netherlands
| | - Peter Boross
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| | | | - Jeanette H W Leusen
- a Laboratory of Translational Immunology; UMC Utrecht ; Utrecht , The Netherlands
| |
Collapse
|
112
|
Rouwendal GJ, van der Lee MM, Meyer S, Reiding KR, Schouten J, de Roo G, Egging DF, Leusen JH, Boross P, Wuhrer M, Verheijden GF, Dokter WH, Timmers M, Ubink R. A comparison of anti-HER2 IgA and IgG1 in vivo efficacy is facilitated by high N-glycan sialylation of the IgA. MAbs 2015; 8:74-86. [PMID: 26440530 DOI: 10.1080/19420862.2015.1102812] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Monomeric IgA has been proposed as an alternative antibody format for cancer therapy. Here, we present our studies on the production, purification and functional evaluation of anti-HER2 IgA antibodies as anti-cancer agents in comparison to the anti-HER2 IgG1 trastuzumab. MALDI-TOF MS analysis showed profound differences in glycosylation traits across the IgA isotypes and cell lines used for production, including sialylation and linkage thereof, fucosylation (both core and antennary) and the abundance of high-mannose type species. Increases in sialylation proved to positively correlate with in vivo plasma half-lives. The polymerization propensity of anti-HER2 IgA2m2 could be suppressed by an 18-aa deletion of the heavy chain tailpiece - coinciding with the loss of high-mannose type N-glycan species - as well as by 2 cysteine to serine mutations at positions 320 and 480. The HER2 F(ab')2-mediated anti-proliferative effect of the IgA2m1 and IgA2m2 subtypes was similar to IgG1, whereas the IgA1 isotype displayed considerably lower potency and efficacy. The Fc-mediated induction of antibody-dependent cell-mediated cytotoxicity (ADCC) using human whole blood ADCC assays did not demonstrate such clear differences between the IgA isotypes. However, the potency of the anti-HER2 IgA antibodies in these ADCC assays was found to be significantly lower than that of trastuzumab. In vivo anti-tumor activity of the anti-HER2 IgA antibodies was compared to that of trastuzumab in a BT-474 breast cancer xenograft model. Multiple dosing and sialylation of the IgA antibodies compensated for the short in vivo half-life of native IgA antibodies in mice compared to a single dose of IgG1. In the case of the IgA2m2 antibody, the resulting high plasma exposure levels were sufficient to cause clear tumor stasis comparable to that observed for trastuzumab at much lower plasma exposure levels.
Collapse
Affiliation(s)
| | | | - Saskia Meyer
- b Laboratory for Translational Immunology; University Medical Center Utrecht ; Utrecht ; The Netherlands
| | - Karli R Reiding
- c Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden ; The Netherlands
| | - Jan Schouten
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Guy de Roo
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - David F Egging
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Jeanette Hw Leusen
- b Laboratory for Translational Immunology; University Medical Center Utrecht ; Utrecht ; The Netherlands
| | - Peter Boross
- b Laboratory for Translational Immunology; University Medical Center Utrecht ; Utrecht ; The Netherlands
| | - Manfred Wuhrer
- c Center for Proteomics and Metabolomics; Leiden University Medical Center ; Leiden ; The Netherlands.,d Division of BioAnalytical Chemistry; VU University Amsterdam ; Amsterdam , The Netherlands
| | | | - Wim H Dokter
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Marco Timmers
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| | - Ruud Ubink
- a Synthon Biopharmaceuticals B.V. ; Nijmegen ; The Netherlands
| |
Collapse
|
113
|
The solution structures of native and patient monomeric human IgA1 reveal asymmetric extended structures: implications for function and IgAN disease. Biochem J 2015; 471:167-85. [PMID: 26268558 PMCID: PMC4692083 DOI: 10.1042/bj20150612] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/12/2015] [Indexed: 01/14/2023]
Abstract
Detailed analytical ultracentrifugation and X-ray/neutron scattering data and a new atomistic modelling approach revealed asymmetric extended solution structures for human IgA1 that account for its receptor-binding function. IgA1 with different hinge O-galactosylation patterns showed similar structures. Native IgA1, for which no crystal structure is known, contains an O-galactosylated 23-residue hinge region that joins its Fab and Fc regions. IgA nephropathy (IgAN) is a leading cause of chronic kidney disease in developed countries. Because IgA1 in IgAN often has a poorly O-galactosylated hinge region, the solution structures of monomeric IgA1 from a healthy subject and three IgAN patients with four different O-galactosylation levels were studied. Analytical ultracentrifugation showed that all four IgA1 samples were monomeric with similar sedimentation coefficients, s020,w. X-ray scattering showed that the radius of gyration (Rg) slightly increased with IgA1 concentration, indicating self-association, although their distance distribution curves, P(r), were unchanged with concentration. Neutron scattering indicated similar Rg values and P(r) curves, although IgA1 showed a propensity to aggregate in heavy water buffer. A new atomistic modelling procedure based on comparisons with 177000 conformationally-randomized IgA1 structures with the individual experimental scattering curves revealed similar extended Y-shaped solution structures for all four differentially-glycosylated IgA1 molecules. The final models indicated that the N-glycans at Asn263 were folded back against the Fc surface, the C-terminal tailpiece conformations were undefined and hinge O-galactosylation had little effect on the solution structure. The solution structures for full-length IgA1 showed extended hinges and the Fab and Fc regions were positioned asymmetrically to provide ample space for the functionally-important binding of two FcαR receptors to its Fc region. Whereas no link between O-galactosylation and the IgA1 solution structure was detected, an increase in IgA1 aggregation with reduced O-galactosylation may relate to IgAN.
Collapse
|
114
|
Rehman Z, Fahim A, Sadia H. Deciphering the mystery of hepatitis B virus receptors: A historical perspective. Virusdisease 2015; 26:97-104. [PMID: 26396975 DOI: 10.1007/s13337-015-0260-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 06/25/2015] [Indexed: 12/22/2022] Open
Abstract
Hepatitis B virus is one of the major reasons of viral hepatitis with an estimated 350 million infected patients worldwide. Although, the virus was discovered and cloned more than three decades ago, its entry mechanism has still been in investigation. Numerous potential candidates have been proposed and investigated rigorously to reveal HBV entry mechanism and to unveil the first door of viral entry to hepatocytes. This review provides a short account of role of receptors for entry of HBV into hepatocytes. The viral preS1 region of large surface protein is involved in the attachment of HBV to hepatocytes. The putative attachment site of HBV is located at amino acids 21-47 of preS1. So far, several proteins have been proposed to interact with these different regions of the preS1 domain which includes human immunoglobulin A receptor, glyceraldehyde-3-phosphate dehydrogenase, interleukin-6, a 31-kDa protein, HBV binding factor, asialoglycoprotein receptor, nascent polypeptide-associated complex α polypeptide, lipoprotein lipase, hepatocyte-associated heparan sulfate proteoglycans, glucose-regulated protein 75. However, none of them have appeared to be generally accepted as a true receptor for the virus until recently when sodium taurocholate cotransporting polypeptide identified as HBV entry receptor. Current review provides scientific historical perspective of various candidates known to be interacting with preS1 of HBV for their possible role in viral entry.
Collapse
Affiliation(s)
- Zaira Rehman
- Healthcare Biotechnology Department, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Sector H-12, Islamabad, Pakistan
| | - Ammad Fahim
- Healthcare Biotechnology Department, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Sector H-12, Islamabad, Pakistan
| | - Hajra Sadia
- Healthcare Biotechnology Department, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences & Technology (NUST), Sector H-12, Islamabad, Pakistan
| |
Collapse
|
115
|
Plomp R, Dekkers G, Rombouts Y, Visser R, Koeleman CAM, Kammeijer GSM, Jansen BC, Rispens T, Hensbergen PJ, Vidarsson G, Wuhrer M. Hinge-Region O-Glycosylation of Human Immunoglobulin G3 (IgG3). Mol Cell Proteomics 2015; 14:1373-84. [PMID: 25759508 PMCID: PMC4424406 DOI: 10.1074/mcp.m114.047381] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/06/2015] [Indexed: 01/18/2023] Open
Abstract
Immunoglobulin G (IgG) is one of the most abundant proteins present in human serum and a fundamental component of the immune system. IgG3 represents ∼8% of the total amount of IgG in human serum and stands out from the other IgG subclasses because of its elongated hinge region and enhanced effector functions. This study reports partial O-glycosylation of the IgG3 hinge region, observed with nanoLC-ESI-IT-MS(/MS) analysis after proteolytic digestion. The repeat regions within the IgG3 hinge were found to be in part O-glycosylated at the threonine in the triple repeat motif. Non-, mono- and disialylated core 1-type O-glycans were detected in various IgG3 samples, both poly- and monoclonal. NanoLC-ESI-IT-MS/MS with electron transfer dissociation fragmentation and CE-MS/MS with CID fragmentation were used to determine the site of IgG3 O-glycosylation. The O-glycosylation site was further confirmed by the recombinant production of mutant IgG3 in which potential O-glycosylation sites had been knocked out. For IgG3 samples from six donors we found similar O-glycan structures and site occupancies, whereas for the same samples the conserved N-glycosylation of the Fc CH2 domain showed considerable interindividual variation. The occupancy of each of the three O-glycosylation sites was found to be ∼10% in six serum-derived IgG3 samples and ∼13% in two monoclonal IgG3 allotypes.
Collapse
Affiliation(s)
- Rosina Plomp
- From the ‡Center for Proteomics and Metabolomics
| | | | - Yoann Rombouts
- From the ‡Center for Proteomics and Metabolomics, §Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Bas C Jansen
- From the ‡Center for Proteomics and Metabolomics
| | - Theo Rispens
- ¶¶Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Manfred Wuhrer
- From the ‡Center for Proteomics and Metabolomics, **Division of BioAnalytical Chemistry, VU University Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
116
|
Novak J, Rizk D, Takahashi K, Zhang X, Bian Q, Ueda H, Ueda Y, Reily C, Lai LY, Hao C, Novak L, Huang ZQ, Renfrow MB, Suzuki H, Julian BA. New Insights into the Pathogenesis of IgA Nephropathy. KIDNEY DISEASES 2015; 1:8-18. [PMID: 26568951 DOI: 10.1159/000382134] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND IgA nephropathy, a frequent cause of end-stage renal disease, is an autoimmune disease wherein immune complexes consisting of IgA1 with galactose-deficient O-glycans (autoantigen) and anti-glycan autoantibodies deposit in glomeruli and induce renal injury. Multiple genetic loci associated with disease risk have been identified. The prevalence of risk alleles varies geographically, highest in eastern Asia and northern Europe, fewer in other parts of Europe and North America, and the least in Africa. IgA nephropathy is diagnosed from pathological assessment of a renal biopsy specimen. Currently, therapy is not disease-targeted but rather is focused on maintaining control of blood pressure and proteinuria, ideally with suppression of angiotensin II. Possible additional approaches differ between countries. Disease-specific therapy as well as new tools for diagnosis, prognosis, and assessment of responses to therapy are needed. SUMMARY Glycosylation pathways associated with aberrant O-glycosylation of IgA1 and, thus, production of autoantigen, have been identified. Furthermore, unique characteristics of the autoantibodies in IgA nephropathy have been uncovered. Many of these biochemical features are shared by patients with IgA nephropathy and Henoch-Schönlein purpura nephritis, suggesting that the two diseases may represent opposite ends of a spectrum of a disease process. Understanding the molecular mechanisms involved in formation of pathogenic IgA1-containing immune complexes will enable development of disease-specific therapies as well as diagnostic and prognostic biomarkers. KEY MESSAGES IgA nephropathy is an autoimmune disease caused by glomerular deposition of nephritogenic circulating immune complexes consisting of galactose-deficient IgA1 (autoantigen) bound by anti-glycan autoantibodies. A better understanding of the multi-step process of pathogenesis of IgA nephropathy and the genetic and environmental contributing factors will lead to development of biomarkers to identify patients with progressive disease who would benefit from a future disease-specific therapy.
Collapse
Affiliation(s)
- Jan Novak
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dana Rizk
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kazuo Takahashi
- University of Alabama at Birmingham, Birmingham, AL, USA ; School of Medicine, Fujita Health University, Toyoake, Japan
| | - XianWen Zhang
- University of Alabama at Birmingham, Birmingham, AL, USA ; Longhua Hospital, Shanghai University of TCM, Shanghai, China
| | - Qi Bian
- University of Alabama at Birmingham, Birmingham, AL, USA ; Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hirouki Ueda
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yoshimi Ueda
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Colin Reily
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ling-Yun Lai
- University of Alabama at Birmingham, Birmingham, AL, USA ; Fudan University Huashan Hospital, Shanghai, China
| | | | - Lea Novak
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Hitoshi Suzuki
- University of Alabama at Birmingham, Birmingham, AL, USA ; Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Bruce A Julian
- University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
117
|
Chen XX, Chen YQ, Ye S. Measuring decreased serum IgG sialylation: a novel clinical biomarker of lupus. Lupus 2015; 24:948-54. [PMID: 25672371 DOI: 10.1177/0961203315570686] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 01/12/2015] [Indexed: 11/16/2022]
Abstract
Introduction The aim of this work was to develop a simple assay to distinguish between patients with rheumatoid arthritis (RA) and patients with systemic lupus erythematosus (SLE) by measuring the serum sialylated IgG (SAIgG). Methods Using a newly established SNA-based ELISA method, we compared the sialylation of immunoglobulin G (IgG) from a healthy control group ( n = 41), RA patients ( n = 30), SLE patients ( n = 45), patients with neuropsychiatric SLE (NPSLE) ( n = 30) and patients with juvenile idiopathic arthritis (JIA) ( n = 32). Results The average SAIgG level in healthy control individuals, RA patients, JIA patients, SLE patients and NPSLE patients was 0.64 ± 0.17, 0.82 ± 0.33, 0.69 ± 0.23, 0.12 ± 0.02 and 0.03 ± 0.01 mg/ml, respectively. The ratio of sialylated IgG to total IgG was significantly decreased in the SLE group (1.88 ± 0.32%) compared with the healthy population (4.64 ± 0.90%). Conclusion In summary, while the mean serum SAIgG level of RA and JIA patients was similar to that of the healthy population, there was a significant decrease in the serum SAIgG of both SLE groups tested, whereby the level of the NPSLE population group was the lowest.
Collapse
Affiliation(s)
- X-X Chen
- Department of Rheumatology, Renji Hospital, School of Medicine, Jiaotong University, Shanghai, China
| | - Y-Q Chen
- Department of Rheumatology, Renji Hospital, School of Medicine, Jiaotong University, Shanghai, China
| | - S Ye
- Department of Rheumatology, Renji Hospital, School of Medicine, Jiaotong University, Shanghai, China
| |
Collapse
|
118
|
Huang J, Guerrero A, Parker E, Strum JS, Smilowitz JT, German JB, Lebrilla CB. Site-specific glycosylation of secretory immunoglobulin A from human colostrum. J Proteome Res 2015; 14:1335-49. [PMID: 25629924 DOI: 10.1021/pr500826q] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Secretory immunoglobulin A (sIgA) is a major glycoprotein in milk and plays a key role in mediating immune protection of the gut mucosa. Although it is a highly glycosylated protein, its site-specific glycosylation and associated glycan micro-heterogeneity have still not been fully elucidated. In this study, the site-specific glycosylation of sIgA isolated from human colostrum (n = 3) was analyzed using a combination of LC-MS and LC-MS/MS and in-house software (Glycopeptide Finder). The majority of the glycans found are biantennary structures with one or more acidic Neu5Ac residues; however, a large fraction belonged to truncated complex structures with terminal GlcNAc. Multiple glycosites were identified with nearly 30 glycan compositions located at seven sites on the secretory component, six compositions at a single site on the J chain, and 16 compositions at five sites on the IgA heavy (H) chain. Site-specific heterogeneity and relative quantitation of each composition and the extent of occupation at each site were determined using nonspecific proteases. Additionally, 54 O-linked glycan compositions located at the IgA1 hinge region (HR) were identified by comparison against a theoretical O-glycopeptide library. This represents the most comprehensive report to date detailing the complexity of glycan micro-heterogeneity with relative quantitation of glycoforms for each glycosylation site on milk sIgA. This strategy further provides a general method for determining site-specific glycosylation in large protein complexes.
Collapse
Affiliation(s)
- Jincui Huang
- Department of Chemistry, ‡Foods for Health Institute, §Department of Food Science and Technology, and ∥Department of Biochemistry and Molecular Medicine, University of California , Davis, California 95616, United States
| | | | | | | | | | | | | |
Collapse
|
119
|
Abstract
The roles of human serum IgA, in contrast to that of mucosal IgA, are relatively unexplored. Previous studies have shown that IgA mediates either pro- or anti-inflammatory effects in innate immune cells. Serum IgA has been shown to interact with many proteins and glycoproteins of which the functions and mechanisms are not fully characterized. Here, we present fresh perspectives into the roles of serum IgA, describing novel IgA-protein interactions, the importance of its glycosylation status in normal functions, and the plausible role of IgA as a driver and regulator of autoimmune diseases/immune overactivation. Other potential roles, including the regulation of cytokines, effector cell function, and homeostasis, are considered in view of the maintenance of immune function. We anticipate future research to uncover new anti-inflammatory or pro-inflammatory roles of human serum IgA in immune functions and dysfunctions, with implications on systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Ka Wai Leong
- Department of Biological Sciences, National University of Singapore , Singapore , Singapore
| | | |
Collapse
|
120
|
Suzuki Y, Suzuki H, Yasutake J, Tomino Y. Paradigm shift in activity assessment of IgA nephropathy – optimizing the next generation of diagnostic and therapeutic maneuvers via glycan targeting. Expert Opin Biol Ther 2015; 15:583-93. [DOI: 10.1517/14712598.2015.1006624] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
121
|
|
122
|
|
123
|
|
124
|
|
125
|
Corfield AP. Mucins: A biologically relevant glycan barrier in mucosal protection. Biochim Biophys Acta Gen Subj 2015; 1850:236-52. [PMID: 24821013 DOI: 10.1016/j.bbagen.2014.05.003] [Citation(s) in RCA: 378] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 04/05/2014] [Accepted: 05/02/2014] [Indexed: 02/08/2023]
|
126
|
Novak J, Raska M, Mestecky J, Julian BA. IgA Nephropathy and Related Diseases. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00105-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
127
|
Zhou M, Ruprecht RM. Are anti-HIV IgAs good guys or bad guys? Retrovirology 2014; 11:109. [PMID: 25499540 PMCID: PMC4297362 DOI: 10.1186/s12977-014-0109-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/24/2014] [Indexed: 12/12/2022] Open
Abstract
An estimated 90% of all HIV transmissions occur mucosally. Immunoglobulin A (IgA) molecules are important components of mucosal fluids. In a vaccine efficacy study, in which virosomes displaying HIV gp41 antigens protected most rhesus monkeys (RMs) against simian-human immunodeficiency virus (SHIV), protection correlated with vaginal IgA capable of blocking HIV transcytosis in vitro. Furthermore, vaginal IgG exhibiting virus neutralization and/or antibody-dependent cellular cytotoxicity (ADCC) correlated with prevention of systemic infection. In contrast, plasma IgG had neither neutralizing nor ADCC activity. More recently, a passive mucosal immunization study provided the first direct proof that dimeric IgAs (dIgAs) can prevent SHIV acquisition in RMs challenged mucosally. This study compared dimeric IgA1 (dIgA1), dIgA2, or IgG1 versions of a human neutralizing monoclonal antibody (nmAb) targeting a conserved HIV Env epitope. While the nmAb neutralization profiles were identical in vitro, dIgA1 was significantly more protective in vivo than dIgA2. Protection was linked to a new mechanism: virion capture. Protection also correlated with inhibition of transcytosis of cell-free virus in vitro. While both of these primate model studies demonstrated protective effects of mucosal IgAs, the RV144 clinical trial identified plasma IgA responses to HIV Env as risk factors for increased HIV acquisition. In a secondary analysis of RV144, plasma IgA decreased the in vitro ADCC activity of vaccine-induced, Env-specific IgG with the same epitope specificity. Here we review the current literature regarding the potential of IgA – systemic as well as mucosal – in modulating virus acquisition and address the question whether anti-HIV IgA responses could help or harm the host.
Collapse
Affiliation(s)
- Mingkui Zhou
- Department of Virology & Immunology, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX, 78227, USA.
| | - Ruth M Ruprecht
- Department of Virology & Immunology, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX, 78227, USA. .,Southwest National Primate Research Center, 7620 NW Loop 410, San Antonio, TX, 78227, USA.
| |
Collapse
|
128
|
IgGA: A “Cross-Isotype” Engineered Human Fc Antibody Domain that Displays Both IgG-like and IgA-like Effector Functions. ACTA ACUST UNITED AC 2014; 21:1603-9. [DOI: 10.1016/j.chembiol.2014.10.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/15/2014] [Accepted: 10/22/2014] [Indexed: 02/04/2023]
|
129
|
Westerhof LB, Wilbers RHP, van Raaij DR, Nguyen DL, Goverse A, Henquet MGL, Hokke CH, Bosch D, Bakker J, Schots A. Monomeric IgA can be produced in planta as efficient as IgG, yet receives different N-glycans. PLANT BIOTECHNOLOGY JOURNAL 2014; 12:1333-42. [PMID: 25196296 DOI: 10.1111/pbi.12251] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/23/2014] [Accepted: 08/04/2014] [Indexed: 05/25/2023]
Abstract
The unique features of IgA, such as the ability to recruit neutrophils and suppress the inflammatory responses mediated by IgG and IgE, make it a promising antibody isotype for several therapeutic applications. However, in contrast to IgG, reports on plant production of IgA are scarce. We produced IgA1κ and IgG1κ versions of three therapeutic antibodies directed against pro-inflammatory cytokines in Nicotiana benthamiana: Infliximab and Adalimumab, directed against TNF-α, and Ustekinumab, directed against the interleukin-12p40 subunit. We evaluated antibody yield, quality and N-glycosylation. All six antibodies had comparable levels of expression between 3.5 and 9% of total soluble protein content and were shown to have neutralizing activity in a cell-based assay. However, IgA1κ-based Adalimumab and Ustekinumab were poorly secreted compared to their IgG counterparts. Infliximab was poorly secreted regardless of isotype backbone. This corresponded with the observation that both IgA1κ- and IgG1κ-based Infliximab were enriched in oligomannose-type N-glycan structures. For IgG1κ-based Ustekinumab and Adalimumab, the major N-glycan type was the typical plant complex N-glycan, biantennary with terminal N-acetylglucosamine, β1,2-xylose and core α1,3-fucose. In contrast, the major N-glycan on the IgA-based antibodies was xylosylated, but lacked core α1,3-fucose and one terminal N-acetylglucosamine. This type of N-glycan occurs usually in marginal percentages in plants and was never shown to be the main fraction of a plant-produced recombinant protein. Our data demonstrate that the antibody isotype may have a profound influence on the type of N-glycan an antibody receives.
Collapse
Affiliation(s)
- Lotte B Westerhof
- Plant Sciences Department, Laboratory of Nematology, Wageningen University and Research Centre, Wageningen, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Stuchlova Horynova M, Vrablikova A, Stewart TJ, Takahashi K, Czernekova L, Yamada K, Suzuki H, Julian BA, Renfrow MB, Novak J, Raska M. N-acetylgalactosaminide α2,6-sialyltransferase II is a candidate enzyme for sialylation of galactose-deficient IgA1, the key autoantigen in IgA nephropathy. Nephrol Dial Transplant 2014; 30:234-8. [PMID: 25281698 DOI: 10.1093/ndt/gfu308] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Galactose-deficient O-glycans in the hinge region (HR) of immunoglobulin A1 (IgA1) play a key role in the pathogenesis of IgA nephropathy (IgAN). O-Glycans of circulatory IgA1 consist of N-acetylgalactosamine (GalNAc) with a β1,3-linked galactose; both sugars may be sialylated. In patients with IgAN, α2,6-sialylated GalNAc is a frequent form of the galactose-deficient O-glycans. Prior analyses of IgA1-producing cells had indicated that α2,6-sialyltransferase II (ST6GalNAc-II) is likely responsible for sialylation of GalNAc of galactose-deficient IgA1, but direct evidence is missing. METHODS We produced a secreted variant of recombinant human ST6GalNAc-II and an IgA1 fragment comprised of Cα1-HR-Cα2. This IgA1 fragment and a synthetic HR peptide with enzymatically attached GalNAc residues served as acceptors. ST6GalNAc-II activity was assessed in vitro and the attachment of sialic acid to these acceptors was detected by lectin blot and mass spectrometry. RESULTS ST6GalNAc-II was active with both acceptors. High-resolution mass spectrometry analysis revealed that up to three sialic acid residues were added to the GalNAc residues of the HR glycopeptide. CONCLUSIONS Our data provide direct evidence that ST6GalNAc-II can sialylate GalNAc of galactose-deficient IgA1. As serum levels of galactose-deficient IgA1 with sialylated glycoforms are increased in IgAN patients, our data explain the corresponding part of the biosynthetic pathway.
Collapse
Affiliation(s)
- Milada Stuchlova Horynova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University, Olomouc 77515, Czech Republic Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alena Vrablikova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University, Olomouc 77515, Czech Republic Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tyler J Stewart
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kazuo Takahashi
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | - Lydie Czernekova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University, Olomouc 77515, Czech Republic Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Koshi Yamada
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
| | - Hitoshi Suzuki
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
| | - Bruce A Julian
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Matthew B Renfrow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Milan Raska
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University, Olomouc 77515, Czech Republic Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
131
|
Theodoratou E, Campbell H, Ventham NT, Kolarich D, Pučić-Baković M, Zoldoš V, Fernandes D, Pemberton IK, Rudan I, Kennedy NA, Wuhrer M, Nimmo E, Annese V, McGovern DPB, Satsangi J, Lauc G. The role of glycosylation in IBD. Nat Rev Gastroenterol Hepatol 2014; 11:588-600. [PMID: 24912389 DOI: 10.1038/nrgastro.2014.78] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A number of genetic and immunological studies give impetus for investigating the role of glycosylation in IBD. Experimental mouse models have helped to delineate the role of glycosylation in intestinal mucins and to explore the putative pathogenic role of glycosylation in colitis. These experiments have been extended to human studies investigating the glycosylation patterns of intestinal mucins as well as levels of glycans of serum glycoproteins and expression of glycan receptors. These early human studies have generated interesting hypotheses regarding the pathogenic role of glycans in IBD, but have generally been restricted to fairly small underpowered studies. Decreased glycosylation has been observed in the intestinal mucus of patients with IBD, suggesting that a defective inner mucus layer might lead to increased bacterial contact with the epithelium, potentially triggering inflammation. In sera, decreased galactosylation of IgG has been suggested as a diagnostic marker for IBD. Advances in glycoprofiling technology make it technically feasible and affordable to perform high-throughput glycan pattern analyses and to build on previous work investigating a much wider range of glycan parameters in large numbers of patients.
Collapse
Affiliation(s)
- Evropi Theodoratou
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, EH8 9AG, Edinburgh, UK
| | - Harry Campbell
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, EH8 9AG, Edinburgh, UK
| | - Nicholas T Ventham
- Centre for Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Edinburgh, UK
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1 OT Golm, 14476, Potsdam, Germany
| | | | - Vlatka Zoldoš
- University of Zagreb, Faculty of Science, Horvatovac 102a, 10000 Zagreb, Croatia
| | | | - Iain K Pemberton
- IP Research Consulting SAS, 34 Rue Carnot, 93160 Noisy-le-Grand, Paris, France
| | - Igor Rudan
- Centre for Population Health Sciences, University of Edinburgh, Teviot Place, EH8 9AG, Edinburgh, UK
| | - Nicholas A Kennedy
- Centre for Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Edinburgh, UK
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Elaine Nimmo
- Centre for Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Edinburgh, UK
| | - Vito Annese
- Department of Medical and Surgical Specialities, Division of Gastroenterology, AOU Careggi University Hospital, Largo Brambilla 13, 50139 Florence, Italy
| | - Dermot P B McGovern
- F.Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Suite D4063, Los Angeles, CA 90048, USA
| | - Jack Satsangi
- Centre for Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Edinburgh, UK
| | - Gordan Lauc
- Department of Biochemistry and Molecular Biology, University of Zagreb Faculty of Pharmacy and Biochemistry, Trg maršala Tita 14, 10000 Zagreb, Croatia
| |
Collapse
|
132
|
Abstract
PURPOSE OF REVIEW In this article, we review recent findings on the pathogenesis and genetics of immunoglobulin A (IgA) nephropathy. RECENT FINDINGS During the past 2 years, the understanding of the pathogenesis of IgA nephropathy has evolved as a result of progress in technology and new tools that have been developed. Since 1968, when IgA nephropathy was described as an IgA-IgG immune-complex disease, the knowledge base expanded to allow definition of IgA nephropathy as an autoimmune disease with a multihit pathogenetic process. Specifically, galactose-deficient immunoglobulin A1 (IgA1) is recognized by unique autoantibodies, resulting in the formation of pathogenic immune complexes that ultimately deposit in the glomerular mesangium and induce renal injury. New approaches using high-resolution mass spectrometry have provided unique insight at the molecular level into IgA1 O-glycosylation. Cutting-edge genome-wide association studies revealed multiple disease-associated risk loci and have mapped their geographic and racial distribution. SUMMARY Recent studies of molecular and genetic defects operating in IgA nephropathy can define new biomarkers specific for the disease that can be developed into clinical assays to aid in the diagnosis, assessment of prognosis, and monitoring of disease progression. Moreover, disease-specific targets are being discovered that may lead to development of new approaches for treatment.
Collapse
|
133
|
Sedzik J, Jastrzebski JP, Grandis M. Glycans of myelin proteins. J Neurosci Res 2014; 93:1-18. [PMID: 25213400 DOI: 10.1002/jnr.23462] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 06/19/2014] [Accepted: 07/07/2014] [Indexed: 01/04/2023]
Abstract
Human P0 is the main myelin glycoprotein of the peripheral nervous system. It can bind six different glycans, all linked to Asn(93) , the unique glycosylation site. Other myelin glycoproteins, also with a single glycosylation site (PMP22 at Asn(36) , MOG at Asn(31) ), bind only one glycan. The MAG has 10 glycosylation sites; the glycoprotein OMgp has 11 glycosylation sites. Aside from P0, no comprehensive data are available on other myelin glycoproteins. Here we review and analyze all published data on the physicochemical structure of the glycans linked to P0, PMP22, MOG, and MAG. Most data concern bovine P0, whose glycan moieties have an MW ranging from 1,294.56 Da (GP3) to 2,279.94 Da (GP5). The pI of glycosylated P0 protein varies from pH 9.32 to 9.46. The most charged glycan is MS2 containing three sulfate groups and one glucuronic acid; whereas the least charged one is the BA2 residue. All glycans contain one fucose and one galactose. The most mannose rich are the glycans MS2 and GP4, each of them has four mannoses; OPPE1 contains five N-acetylglucosamines and one sulfated glucuronic acid; GP4 contains one sialic acid. Furthermore, human P0 variants causing both gain and loss of glycosylation have been described and cause peripheral neuropathies with variable clinical severity. In particular, the substitution T(95) →M is a very common in Europe and is associated with a late-onset axonal neuropathy. Although peripheral myelin is made up largely of glycoproteins, mutations altering glycosylation have been described only in P0. This attractive avenue of research requires further study.
Collapse
Affiliation(s)
- Jan Sedzik
- Royal Institute of Technology, Department of Chemical Engineering, Protein Crystallization Facility, Stockholm, Sweden; National Institute of Physiological Sciences, Department of Neuroscience and Bioinformatics, Okazaki, Japan
| | | | | |
Collapse
|
134
|
Cellular signaling and production of galactose-deficient IgA1 in IgA nephropathy, an autoimmune disease. J Immunol Res 2014; 2014:197548. [PMID: 25152896 PMCID: PMC4134797 DOI: 10.1155/2014/197548] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/25/2014] [Indexed: 12/21/2022] Open
Abstract
Immunoglobulin A (IgA) nephropathy (IgAN), the leading cause of primary glomerulonephritis, is characterized by IgA1-containing immunodeposits in the glomeruli. IgAN is a chronic disease, with up to 40% of patients progressing to end-stage renal disease, with no disease-specific treatment. Multiple studies of the origin of the glomerular immunodeposits have linked elevated circulating levels of aberrantly glycosylated IgA1 (galactose-deficient in some O-glycans; Gd-IgA1) with formation of nephritogenic Gd-IgA1-containing immune complexes. Gd-IgA1 is recognized as an autoantigen in susceptible individuals by anti-glycan autoantibodies, resulting in immune complexes that may ultimately deposit in the kidney and induce glomerular injury. Genetic studies have revealed that an elevated level of Gd-IgA1 in the circulation of IgAN patients is a hereditable trait. Moreover, recent genome-wide association studies have identified several immunity-related loci that associated with IgAN. Production of Gd-IgA1 by IgA1-secreting cells of IgAN patients has been attributed to abnormal expression and activity of several key glycosyltransferases. Substantial evidence is emerging that abnormal signaling in IgA1-producing cells is related to the production of Gd-IgA1. As Gd-IgA1 is the key autoantigen in IgAN, understanding the genetic, biochemical, and environmental aspects of the abnormal signaling in IgA1-producing cells will provide insight into possible targets for future disease-specific therapy.
Collapse
|
135
|
Specificity of two monoclonal antibodies against a synthetic glycopeptide, an analogue to the hypo-galactosylated IgA1 hinge region. J Nephrol 2014; 28:181-6. [PMID: 25037591 PMCID: PMC4376957 DOI: 10.1007/s40620-014-0118-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 06/26/2014] [Indexed: 12/01/2022]
Abstract
Increased levels of hypo-galactosylated immunoglobulin (Ig)A1 (HG-IgA1) in IgA nephropathy (IgAN) have been detected using a Helix aspersa agglutinin lectin enzyme-linked immunosorbent assay (ELISA). In this study, we developed monoclonal antibodies to evaluate the HG-IgA1 in IgA nephropathy, aiming to gain a more consistent and reproducible assay. As an analogue to the HG-IgA1 hinge region, a 19 mer synthetic peptide with five GalNAc (sHGP) residues at positions 4, 7, 9, 11 and 15 [VPST(GalNAc)PPT(GalNAc)PS(GalNAc)PS(GalNAc)TPPT (GalNAc)PSPS-NH2] was synthesized. Two monoclonal antibodies against sHGP (35A12 and 44H8) that reacted with human IgA were developed. Also, their reactivities to serum IgA from IgAN patients (n = 49), patients with other forms of kidney diseases (OKD, n = 48), and healthy controls (HC, n = 41) were evaluated using ELISA assays. The binding levels of the two monoclonal antibodies against serum IgA were significantly higher (all comparisons, p < 0.0001, Steel–Dwass non-parametric test) in IgAN patients compared to HC and OKD patients. In each individual, there was a close correlation of IgA binding levels between 35A12 and 44H8 (R2 = 0.737). These results indicate that the monoclonal antibodies recognize similar epitopes in HG IgA1, which is found predominantly in IgAN patients. The developed antibodies are proposed as a clinically useful tool for IgAN screening.
Collapse
|
136
|
Takahashi K, Raska M, Stuchlova Horynova M, Hall SD, Poulsen K, Kilian M, Hiki Y, Yuzawa Y, Moldoveanu Z, Julian BA, Renfrow MB, Novak J. Enzymatic sialylation of IgA1 O-glycans: implications for studies of IgA nephropathy. PLoS One 2014; 9:e99026. [PMID: 24918438 PMCID: PMC4053367 DOI: 10.1371/journal.pone.0099026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/23/2014] [Indexed: 11/18/2022] Open
Abstract
Patients with IgA nephropathy (IgAN) have elevated circulating levels of IgA1 with some O-glycans consisting of galactose (Gal)-deficient N-acetylgalactosamine (GalNAc) with or without N-acetylneuraminic acid (NeuAc). We have analyzed O-glycosylation heterogeneity of naturally asialo-IgA1 (Ale) myeloma protein that mimics Gal-deficient IgA1 (Gd-IgA1) of patients with IgAN, except that IgA1 O-glycans of IgAN patients are frequently sialylated. Specifically, serum IgA1 of healthy controls has more α2,3-sialylated O-glycans (NeuAc attached to Gal) than α2,6-sialylated O-glycans (NeuAc attached to GalNAc). As IgA1-producing cells from IgAN patients have an increased activity of α2,6-sialyltransferase (ST6GalNAc), we hypothesize that such activity may promote premature sialylation of GalNAc and, thus, production of Gd-IgA1, as sialylation of GalNAc prevents subsequent Gal attachment. Distribution of NeuAc in IgA1 O-glycans may play an important role in the pathogenesis of IgAN. To better understand biological functions of NeuAc in IgA1, we established protocols for enzymatic sialylation leading to α2,3- or α2,6-sialylation of IgA1 O-glycans. Sialylation of Gal-deficient asialo-IgA1 (Ale) myeloma protein by an ST6GalNAc enzyme generated sialylated IgA1 that mimics the Gal-deficient IgA1 glycoforms in patients with IgAN, characterized by α2,6-sialylated Gal-deficient GalNAc. In contrast, sialylation of the same myeloma protein by an α2,3-sialyltransferase yielded IgA1 typical for healthy controls, characterized by α2,3-sialylated Gal. The GalNAc-specific lectin from Helix aspersa (HAA) is used to measure levels of Gd-IgA1. We assessed HAA binding to IgA1 sialylated at Gal or GalNAc. As expected, α2,6-sialylation of IgA1 markedly decreased reactivity with HAA. Notably, α2,3-sialylation also decreased reactivity with HAA. Neuraminidase treatment recovered the original HAA reactivity in both instances. These results suggest that binding of a GalNAc-specific lectin is modulated by sialylation of GalNAc as well as Gal in the clustered IgA1 O-glycans. Thus, enzymatic sialylation offers a useful model to test the role of NeuAc in reactivities of the clustered O-glycans with lectins.
Collapse
Affiliation(s)
- Kazuo Takahashi
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Milan Raska
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Faculty of Medicine and Dentistry, Department of Immunology, Palacky University in Olomouc, Olomouc, Czech Republic
| | - Milada Stuchlova Horynova
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Faculty of Medicine and Dentistry, Department of Immunology, Palacky University in Olomouc, Olomouc, Czech Republic
| | - Stacy D. Hall
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Knud Poulsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mogens Kilian
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Yoshiyuki Hiki
- Fujita Health University School of Health Sciences, Toyoake, Japan
| | - Yukio Yuzawa
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Zina Moldoveanu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Bruce A. Julian
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Matthew B. Renfrow
- UAB Biomedical FT-ICR MS Laboratory, Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
137
|
Abstract
IgA nephropathy (IgAN) represents the leading cause of kidney failure among East Asian populations and the most frequent form of primary glomerulonephritis among Europeans. Patients with IgAN develop characteristic IgA1-containing immune complexes that deposit in the glomerular mesangium, producing progressive kidney injury. Recent studies define IgAN as an autoimmune trait of complex architecture with a strong genetic determination. This Review summarizes new insights into the role of the O-glycosylation pathway, anti-glycan immune response, mucosal immunity, antigen processing and presentation, and the alternative complement pathway in the pathogenesis of IgAN.
Collapse
|
138
|
Chung AW, Alter G. Dissecting the antibody constant region protective immune parameters in HIV infection. Future Virol 2014. [DOI: 10.2217/fvl.14.19] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT: RV144 vaccine immune-correlates analysis has generated a renewed interest in understanding the potentially protective role of non-neutralizing antibodies in HIV infection and vaccine design. Antibodies consist of a variable region involved in antigen binding and a constant region. While both ends of the antibody collaborate to induce protective immunity, it is through the constant portion that an antibody provides instructions to the innate immune system on how the recognized antigen should be processed, contributing directly to antiviral immunity. Antibody constant regions, despite their name, are not uniform structures, but can vary both in protein sequence and glycosylation, together modulating antibody functionality via conformational changes that alter antibody affinity for Fc receptors, complement and so on. This review will focus on how the immune system naturally modulates the Fc domain of antibodies to achieve optimum protective Fc effector responses for vaccine and monoclonal therapeutic design efforts aimed at preventing or curing HIV infection.
Collapse
Affiliation(s)
- Amy W Chung
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology & Harvard, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology & Harvard, Boston, MA, USA
| |
Collapse
|
139
|
Narimatsu Y, Kuno A, Ito H, Kaji H, Kaneko S, Usui J, Yamagata K, Narimatsu H. IgA nephropathy caused by unusual polymerization of IgA1 with aberrant N-glycosylation in a patient with monoclonal immunoglobulin deposition disease. PLoS One 2014; 9:e91079. [PMID: 24651839 PMCID: PMC3961232 DOI: 10.1371/journal.pone.0091079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 02/07/2014] [Indexed: 11/18/2022] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is a form of chronic glomerulonephritis characterized by the deposition of IgA immune complexes in the glomerular region. The cause of IgAN is unknown, but multiple mechanisms have been suggested. We previously reported a rare case of mesangioproliferative glomerulonephritis in a patient with monoclonal immunoglobulin deposition disease associated with monoclonal IgA1. In this study, we performed the detailed analyses of serum IgA1 from this patient in comparison with those from patients with mIgA plasma cell disorder without renal involvement and healthy volunteers. We found unusual polymerization of IgA1 with additional N-glycosylation distinctive in this patient, which was different from known etiologies. Glycan profiling of IgA1 by the lectin microarray revealed an intense signal for Wisteria floribunda agglutinin (WFA). This signal was reduced by disrupting the native conformation of IgA1, suggesting that the distinct glycan profile was reflecting the conformational alteration of IgA1, including the glycan conformation detected as additional N-glycans on both the heavy and light chains. This unusually polymerized state of IgA1 would cause an increase of the binding avidity for lectins. WFA specifically recognized highly polymerized and glycosylated IgA1. Our results of analysis in the rare case of a patient with monoclonal immunoglobulin deposition disease suggest that the formation of unusually polymerized IgA1 is caused by divergent mechanisms including multiple structural alterations of glycans, which contributes to IgA1 deposition and mesangium proliferation.
Collapse
Affiliation(s)
- Yoshiki Narimatsu
- Research Center for Medical Glycoscience (RCMG), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Atsushi Kuno
- Research Center for Medical Glycoscience (RCMG), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Hiromi Ito
- Research Center for Medical Glycoscience (RCMG), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
- Department of Biochemistry, Life Sciences and Social Medicine, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hiroyuki Kaji
- Research Center for Medical Glycoscience (RCMG), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Syuzo Kaneko
- Department of Nephrology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Joichi Usui
- Department of Nephrology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kunihiro Yamagata
- Department of Nephrology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hisashi Narimatsu
- Research Center for Medical Glycoscience (RCMG), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
140
|
Kozak RP, Royle L, Gardner RA, Bondt A, Fernandes DL, Wuhrer M. Improved nonreductive O-glycan release by hydrazinolysis with ethylenediaminetetraacetic acid addition. Anal Biochem 2014; 453:29-37. [PMID: 24613257 DOI: 10.1016/j.ab.2014.02.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 02/23/2014] [Accepted: 02/26/2014] [Indexed: 10/25/2022]
Abstract
The study of protein O-glycosylation is receiving increasing attention in biological, medical, and biopharmaceutical research. Improved techniques are required to allow reproducible and quantitative analysis of O-glycans. An established approach for O-glycan analysis relies on their chemical release in high yield by hydrazinolysis, followed by fluorescent labeling at the reducing terminus and high-performance liquid chromatography (HPLC) profiling. However, an unwanted degradation known as "peeling" often compromises hydrazinolysis for O-glycan analysis. Here we addressed this problem using low-molarity solutions of ethylenediaminetetraacetic acid (EDTA) in hydrazine for O-glycan release. O-linked glycans from a range of different glycoproteins were analyzed, including bovine fetuin, bovine submaxillary gland mucin, and serum immunoglobulin A (IgA). The data for the O-glycans released by hydrazine with anhydrous EDTA or disodium salt dihydrate EDTA show high yields of the native O-glycans compared with the peeled product, resulting in a markedly increased robustness of the O-glycan profiling method. The presented method for O-glycan release demonstrates significant reduction in peeling and reduces the number of sample handling steps prior to release.
Collapse
Affiliation(s)
| | - Louise Royle
- Ludger, Culham Science Centre, Oxfordshire OX14 3EB, UK
| | | | - Albert Bondt
- Department of Rheumatology, Erasmus University Medical Center, 3000 CA Rotterdam, The Netherlands; Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | | | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands; Division of Bioanalytical Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
141
|
Kihara M, Ito K, Nakata J, Otani M, Tran NL, Morito N, Takahashi S, Wada Y, Izui S. O-linked glycosylation determines the nephritogenic potential of IgA rheumatoid factor. J Am Soc Nephrol 2014; 25:1282-90. [PMID: 24511137 DOI: 10.1681/asn.2013070771] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Deficient glycosylation of O-linked glycans in the IgA1 hinge region is associated with IgA nephropathy in humans, but the pathogenic contribution of the underlying structural aberrations remains incompletely understood. We previously showed that mice implanted with cells secreting the class-switch variant 6-19 IgA anti-IgG2a rheumatoid factor, but not 46-42 IgA anti-IgG2a rheumatoid factor, develop glomerular lesions resembling IgA nephropathy. Because the levels of O-linked glycosylation in the hinge region and the structures of N-linked glycans in the CH1 domain differ in 6-19 IgA and 46-42 IgA, we determined the respective contributions of O- and N-linked glycans to the nephritogenic potential of the 6-19 IgA rheumatoid factor in mice. Wild-type 6-19 IgA secreted by implanted cells induced significant formation of glomerular lesions, whereas poorly O-glycosylated 6-19 IgA glycovariants or a 6-19 IgA hinge mutant lacking O-linked glycans did not. However, we observed no apparent heterogeneity in the structure of N-linked glycans attached to three different sites of the Fc regions of nephritogenic and non-nephritogenic 6-19 IgAs. Collectively, our data suggest a critical role of O-linked glycans attached to the hinge region in the development of IgA nephropathy-like GN induced by 6-19 IgA rheumatoid factor in mice.
Collapse
Affiliation(s)
- Masao Kihara
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Kiyoaki Ito
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Junichiro Nakata
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Masako Otani
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Ngoc Lan Tran
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Satoru Takahashi
- Department of Anatomy and Embryology, Life System Medical Sciences, Faculty of Medicine, and International Institute for Integrative Sleep Medicine, University of Tsukuba, Ibaraki, Japan; and
| | - Yoshinao Wada
- Department of Molecular Medicine, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka, Japan
| | - Shozo Izui
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland;
| |
Collapse
|
142
|
Suzuki H, Raska M, Yamada K, Moldoveanu Z, Julian BA, Wyatt RJ, Tomino Y, Gharavi AG, Novak J. Cytokines alter IgA1 O-glycosylation by dysregulating C1GalT1 and ST6GalNAc-II enzymes. J Biol Chem 2014; 289:5330-9. [PMID: 24398680 PMCID: PMC3931088 DOI: 10.1074/jbc.m113.512277] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
IgA nephropathy (IgAN), the most common primary glomerulonephritis, is characterized by renal immunodeposits containing IgA1 with galactose-deficient O-glycans (Gd-IgA1). These immunodeposits originate from circulating immune complexes consisting of anti-glycan antibodies bound to Gd-IgA1. As clinical disease onset and activity of IgAN often coincide with mucosal infections and dysregulation of cytokines, we hypothesized that cytokines may affect IgA1 O-glycosylation. We used IgA1-secreting cells derived from the circulation of IgAN patients and healthy controls and assessed whether IgA1 O-glycosylation is altered by cytokines. Of the eight cytokines tested, only IL-6 and, to a lesser degree, IL-4 significantly increased galactose deficiency of IgA1; changes in IgA1 O-glycosylation were robust for the cells from IgAN patients. These cytokines reduced galactosylation of the O-glycan substrate directly via decreased expression of the galactosyltransferase C1GalT1 and, indirectly, via increased expression of the sialyltransferase ST6GalNAc-II, which prevents galactosylation by C1GalT1. These findings were confirmed by siRNA knockdown of the corresponding genes and by in vitro enzyme reactions. In summary, IL-6 and IL-4 accentuated galactose deficiency of IgA1 via coordinated modulation of key glycosyltransferases. These data provide a mechanism explaining increased immune-complex formation and disease exacerbation during mucosal infections in IgAN patients.
Collapse
Affiliation(s)
- Hitoshi Suzuki
- From the University of Alabama at Birmingham, Birmingham, Alabama 35294
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Cohen M, Varki A. Modulation of glycan recognition by clustered saccharide patches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 308:75-125. [PMID: 24411170 DOI: 10.1016/b978-0-12-800097-7.00003-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
All cells in nature are covered with a dense and complex array of glycan chains. Specific recognition and binding of glycans is a critical aspect of cellular interactions, both within and between species. Glycan-protein interactions tend to be of low affinity but high specificity, typically utilizing multivalency to generate the affinity required for biologically relevant binding. This review focuses on a higher level of glycan organization, the formation of clustered saccharide patches (CSPs), which can constitute unique ligands for highly specific interactions. Due to technical challenges, this aspect of glycan recognition remains poorly understood. We present a wealth of evidence for CSPs-mediated interactions, and discuss recent advances in experimental tools that are beginning to provide new insights into the composition and organization of CSPs. The examples presented here are likely the tip of the iceberg, and much further work is needed to elucidate fully this higher level of glycan organization.
Collapse
Affiliation(s)
- Miriam Cohen
- Department Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, California, USA.
| | - Ajit Varki
- Department of Medicine, University of California, San Diego, California, USA; Department Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, California, USA.
| |
Collapse
|
144
|
Brunke C, Lohse S, Derer S, Peipp M, Boross P, Kellner C, Beyer T, Dechant M, Royle L, Liew LP, Leusen JHW, Valerius T. Effect of a tail piece cysteine deletion on biochemical and functional properties of an epidermal growth factor receptor-directed IgA2m(1) antibody. MAbs 2013; 5:936-45. [PMID: 24492345 PMCID: PMC3896607 DOI: 10.4161/mabs.26396] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/04/2013] [Accepted: 09/06/2013] [Indexed: 02/06/2023] Open
Abstract
Antibodies of human IgA isotype are critical components of the mucosal immune system, but little is known about their immunotherapeutic potential. Compared with IgG antibodies, IgA molecules carry a C-terminal tail piece extension of 18 amino acids with a free cysteine at position 471. This cysteine is required for the formation of dimeric IgA antibodies, but may impair molecular characteristics of monomeric IgA antibodies as therapeutic reagents. Thus, we generated and characterized a d471-mutated antibody against the epidermal growth factor receptor (EGFR) and compared it to its respective IgA2m(1) wild type antibody. Both wild type and mutated IgA antibodies demonstrated similar EGFR binding and were similarly efficient in inhibiting EGF binding and in blocking EGF-mediated cell proliferation. Recruitment of Fc-mediated effector functions like antibody-dependent cell-mediated cytotoxicity by monocytes, macrophages or PMN was similar, but the d471-mutated IgA exhibited different biochemical properties compared with wild type antibody. As expected, mutated IgA did not form stable dimers in the presence of human joining (J)-chain, but we also observed reduced levels of dimeric aggregates in the absence of J-chain. Furthermore, glycoprofiling revealed different glycosylation patterns for both antibodies, including considerably less mannosylation of d471-mutated antibodies. Overall, our results demonstrate that the deletion of the C-terminal cysteine of IgA2 did not affect the investigated effector functions compared with wild type antibody, but it improved biochemical properties of an IgA2m(1) antibody against EGFR, and may thereby assist in exploring the immunotherapeutic potential of recombinant IgA antibodies.
Collapse
Affiliation(s)
- Christina Brunke
- Division of Stem Cell Transplantation and Immunotherapy, II; Department of Internal Medicine; Christian-Albrechts-University, Kiel, Germany
| | - Stefan Lohse
- Division of Stem Cell Transplantation and Immunotherapy, II; Department of Internal Medicine; Christian-Albrechts-University, Kiel, Germany
| | - Stefanie Derer
- Division of Stem Cell Transplantation and Immunotherapy, II; Department of Internal Medicine; Christian-Albrechts-University, Kiel, Germany
| | - Matthias Peipp
- Division of Stem Cell Transplantation and Immunotherapy, II; Department of Internal Medicine; Christian-Albrechts-University, Kiel, Germany
| | - Peter Boross
- Department of Immunology; Laboratory for Immunotherapy; University Medical Center Utrecht; Utrecht, the Netherlands
| | - Christian Kellner
- Division of Stem Cell Transplantation and Immunotherapy, II; Department of Internal Medicine; Christian-Albrechts-University, Kiel, Germany
| | - Thomas Beyer
- Department of Internal Medicine IV, Nephrology and Hypertension; Christian-Albrechts-University, Kiel, Germany
| | - Michael Dechant
- Department of Internal Medicine IV, Nephrology and Hypertension; Christian-Albrechts-University, Kiel, Germany
| | - Louise Royle
- Ludger Ltd; Culham Science Centre; Oxford, United Kingdom
| | - Li Phing Liew
- Ludger Ltd; Culham Science Centre; Oxford, United Kingdom
| | - Jeanette HW Leusen
- Department of Immunology; Laboratory for Immunotherapy; University Medical Center Utrecht; Utrecht, the Netherlands
| | - Thomas Valerius
- Division of Stem Cell Transplantation and Immunotherapy, II; Department of Internal Medicine; Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
145
|
Hastings MC, Moldoveanu Z, Suzuki H, Berthoux F, Julian BA, Sanders JT, Renfrow MB, Novak J, Wyatt RJ. Biomarkers in IgA nephropathy: relationship to pathogenetic hits. EXPERT OPINION ON MEDICAL DIAGNOSTICS 2013; 7:615-27. [PMID: 24175678 PMCID: PMC4557957 DOI: 10.1517/17530059.2013.856878] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION IgA nephropathy, the most prevalent glomerular disease in the world, requires a renal biopsy for diagnosis. Reliable biomarkers are needed for the non-invasive diagnosis of this disease and to more fully delineate its natural history and risk for progression. AREAS COVERED In this review, the authors examine serum levels of galactose-deficient IgA1 (Gd-IgA1) and glycan-specific IgG and IgA autoantibodies that are integral to pathogenesis of IgA nephropathy. They also explore biomarkers related to alternative and lectin pathways of complement activation and serum and urinary peptide biomarkers detected by mass spectrometric methods. The literature search included review of all publications having IgA nephropathy in the title that were cited in PubMed and Scopus over the past 10 years and a non-systematic review of abstracts published for the annual meetings of the American Society of Nephrology and the International Symposia on IgA Nephropathy. EXPERT OPINION Serum Gd-IgA1 level and glycan-specific autoantibody levels are prime candidates to become diagnostic biomarkers for IgA nephropathy because of their central role in the earliest stages of disease pathogenesis. Assays for serum levels of complement proteins C3 and factor H are readily available in clinical practice and deserve continued study, either alone or in tandem with total serum IgA or serum Gd-IgA1 levels, as prognostic biomarkers for patients with IgA nephropathy. Urinary peptidomic data are also reviewed because this approach can successfully differentiate patients with IgA nephropathy from healthy controls and from patients with other forms of renal disease.
Collapse
Affiliation(s)
- Margaret Colleen Hastings
- Children's Foundation Research Institute at Le Bonheur Children's Hospital , 50 North Dunlap, Room 520 Research Tower, Memphis, TN 38103-2893 , USA +1 901 287 5366 ; +1 901 287 6337 ;
| | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Rochereau N, Drocourt D, Perouzel E, Pavot V, Redelinghuys P, Brown GD, Tiraby G, Roblin X, Verrier B, Genin C, Corthésy B, Paul S. Dectin-1 is essential for reverse transcytosis of glycosylated SIgA-antigen complexes by intestinal M cells. PLoS Biol 2013; 11:e1001658. [PMID: 24068891 PMCID: PMC3775721 DOI: 10.1371/journal.pbio.1001658] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 08/08/2013] [Indexed: 01/27/2023] Open
Abstract
This work reports the long-awaited identification of Dectin-1 and Siglec-5 as the M cell co-receptors that mediate the reverse transcytosis of secretory IgA molecules to mount a gut immune response. Intestinal microfold (M) cells possess a high transcytosis capacity and are able to transport a broad range of materials including particulate antigens, soluble macromolecules, and pathogens from the intestinal lumen to inductive sites of the mucosal immune system. M cells are also the primary pathway for delivery of secretory IgA (SIgA) to the gut-associated lymphoid tissue. However, although the consequences of SIgA uptake by M cells are now well known and described, the mechanisms whereby SIgA is selectively bound and taken up remain poorly understood. Here we first demonstrate that both the Cα1 region and glycosylation, more particularly sialic acid residues, are involved in M cell–mediated reverse transcytosis. Second, we found that SIgA is taken up by M cells via the Dectin-1 receptor, with the possible involvement of Siglec-5 acting as a co-receptor. Third, we establish that transcytosed SIgA is taken up by mucosal CX3CR1+ dendritic cells (DCs) via the DC-SIGN receptor. Fourth, we show that mucosal and systemic antibody responses against the HIV p24-SIgA complexes administered orally is strictly dependent on the expression of Dectin-1. Having deciphered the mechanisms leading to specific targeting of SIgA-based Ag complexes paves the way to the use of such a vehicle for mucosal vaccination against various infectious diseases. Secretory IgA (SIgA) antibodies are secreted into the gut lumen and are considered to be a first line of defense in protecting the intestinal epithelium from gut pathogens. SIgA patrol the mucus and are usually known to help immune tolerance via entrapping dietary antigens and microorganisms and other mechanisms. SIgA, in complex with its antigens, can also be taken back up by the intestinal epithelium in a process known as reverse transcytosis. SIgA can thereby promote the uptake and delivery of antigens from the intestinal lumen to the Gut-Associated Lymphoid Tissues (GALT), influencing inflammatory responses. This reverse transcytosis of SIgA is mediated by specialized epithelial M cells. Because M cells possess the ability to take up antigens and are therefore important to the local immune system, they are a key target for the specific delivery of novel mucosal vaccines against various diseases. M cell receptors that take up the SIgA-antigen complexes, which serve as mucosal vaccine vehicles, represent an important aspect of this vaccine strategy. The identification of SIgA receptor(s) on the surface of M cells has, however, remained elusive for more than a decade. In this study, we now identify Dectin-1 and Siglec-5 as the key receptors for M cell–mediated reverse transcytosis of SIgA complexes. We further find that the glycosylation modification, and particularly sialylation, of SIgA is required for its uptake by M cells. We show that, when administered orally in complex with SIgA, the HIV p24 antigen is taken up in a strictly Dectin-1-dependent manner to stimulate a mucosal and systemic antibody response. These findings are considered important for understanding gut immunity.
Collapse
MESH Headings
- Animals
- Antigen-Antibody Complex/immunology
- Antigen-Antibody Complex/metabolism
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- CHO Cells
- CX3C Chemokine Receptor 1
- Caco-2 Cells
- Cell Adhesion Molecules/metabolism
- Cell Line
- Cricetulus
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Glycosylation
- HIV Core Protein p24/immunology
- HeLa Cells
- Humans
- Immunoglobulin A/immunology
- Immunoglobulin A, Secretory/immunology
- Intestinal Mucosa/cytology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestines/cytology
- Lectins/metabolism
- Lectins, C-Type/biosynthesis
- Lectins, C-Type/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- N-Acetylneuraminic Acid/chemistry
- Receptors, Cell Surface/metabolism
- Receptors, Chemokine/metabolism
- Transcytosis/immunology
Collapse
Affiliation(s)
- Nicolas Rochereau
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
| | | | | | - Vincent Pavot
- Institut de Biologie et Chimie des Protéines, FRE3310/CNRS, Université de Lyon, France
| | - Pierre Redelinghuys
- Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gordon D. Brown
- Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Xavier Roblin
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
| | - Bernard Verrier
- Institut de Biologie et Chimie des Protéines, FRE3310/CNRS, Université de Lyon, France
| | - Christian Genin
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
| | - Blaise Corthésy
- R&D Laboratory of the Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Stéphane Paul
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
- * E-mail:
| |
Collapse
|
147
|
Ju T, Wang Y, Aryal RP, Lehoux SD, Ding X, Kudelka MR, Cutler C, Zeng J, Wang J, Sun X, Heimburg-Molinaro J, Smith DF, Cummings RD. Tn and sialyl-Tn antigens, aberrant O-glycomics as human disease markers. Proteomics Clin Appl 2013; 7:618-31. [PMID: 23857728 DOI: 10.1002/prca.201300024] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 04/24/2013] [Indexed: 12/12/2022]
Abstract
In many different human disorders, the cellular glycome is altered. An interesting but poorly understood alteration occurs in the mucin-type O-glycome, in which there is aberrant expression of the truncated O-glycans Tn (GalNAcα1-Ser/Thr) and its sialylated version sialyl-Tn (STn) (Neu5Acα2,6GalNAcα1-Ser/Thr). Both Tn and STn are tumor-associated carbohydrate antigens and tumor biomarkers, since they are not expressed normally and appear early in tumorigenesis. Moreover, their expression is strongly associated with poor prognosis and tumor metastasis. The Tn and STn antigens are also expressed in other human diseases and disorders, such as Tn syndrome and IgA nephropathy. The major pathological mechanism for expression of the Tn and STn antigens is compromised T-synthase activity, resulting from alteration of the X-linked gene that encodes for Cosmc, a molecular chaperone specifically required for the correct folding of T-synthase to form active enzyme. This review will summarize our current understanding of the Tn and STn antigens in terms of their biochemistry and role in pathology.
Collapse
Affiliation(s)
- Tongzhong Ju
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yingchun Wang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Rajindra P Aryal
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Sylvain D Lehoux
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaokun Ding
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew R Kudelka
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher Cutler
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Junwei Zeng
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jianmei Wang
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Xiaodong Sun
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | | | - David F Smith
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard D Cummings
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
148
|
Targeted biomarker discovery by high throughput glycosylation profiling of human plasma alpha1-antitrypsin and immunoglobulin A. PLoS One 2013; 8:e73082. [PMID: 24039863 PMCID: PMC3767703 DOI: 10.1371/journal.pone.0073082] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 07/17/2013] [Indexed: 12/26/2022] Open
Abstract
Protein N-glycosylation patterns are known to show vast genetic as well as physiological and pathological variation and represent a large pool of potential biomarkers. Large-scale studies are needed for the identification and validation of biomarkers, and the analytical techniques required have recently been developed. Such methods have up to now mainly been applied to complex mixtures of glycoproteins in biofluids (e.g. plasma). Here, we analyzed N-glycosylation profiles of alpha1-antitrypsin (AAT) and immunoglobulin A (IgA) enriched fractions by 96-well microtitration plate based high-throughput immuno-affinity capturing and N-glycan analysis using multiplexed capillary gel electrophoresis with laser-induced fluorescence detection (CGE-LIF). Human plasma samples were from the Leiden Longevity Study comprising 2415 participants of different chronological and biological ages. Glycosylation patterns of AAT enriched fractions were found to be associated with chronological (calendar) age and they differed between females and males. Moreover, several glycans in the AAT enriched fraction were associated with physiological parameters marking cardiovascular and metabolic diseases. Pronounced differences were found between males and females in the glycosylation profiles of IgA enriched fractions. Our results demonstrate that large-scale immuno-affinity capturing of proteins from human plasma using a bead-based method combined with high-throughput N-glycan analysis is a powerful tool for the discovery of glycosylation-based biomarker candidates.
Collapse
|
149
|
Franc V, Řehulka P, Raus M, Stulík J, Novak J, Renfrow MB, Šebela M. Elucidating heterogeneity of IgA1 hinge-region O-glycosylation by use of MALDI-TOF/TOF mass spectrometry: role of cysteine alkylation during sample processing. J Proteomics 2013; 92:299-312. [PMID: 23891555 DOI: 10.1016/j.jprot.2013.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 06/19/2013] [Accepted: 07/11/2013] [Indexed: 12/22/2022]
Abstract
UNLABELLED Determining disease-associated changes in protein glycosylation provides a better understanding of pathogenesis. This work focuses on human immunoglobulin A1 (IgA1), where aberrant O-glycosylation plays a key role in the pathogenesis of IgA nephropathy (IgAN). Normal IgA1 hinge region carries 3 to 6 O-glycans consisting of N-acetylgalactosamine (GalNAc) and galactose (Gal); both sugars may be sialylated. In IgAN patients, some O-glycans on a fraction of IgA1 molecules are Gal-deficient. Here we describe a sample preparation protocol with optimized cysteine alkylation of a Gal-deficient polymeric IgA1 myeloma protein prior to in-gel digestion and analysis of the digest by MALDI-TOF/TOF mass spectrometry (MS). Following a novel strategy, IgA1 hinge-region O-glycopeptides were fractionated by reversed-phase liquid chromatography using a microgradient device and identified by MALDI-TOF/TOF tandem MS (MS/MS). The acquired MS/MS spectra were interpreted manually and by means of our own software. This allowed assigning up to six O-glycosylation sites and demonstration, for the first time, of the distribution of isomeric O-glycoforms having the same molecular mass, but a different glycosylation pattern. The most abundant Gal-deficient O-glycoforms were GalNAc4Gal3 and GalNAc5Gal4 with one Gal-deficient site and GalNAc5Gal3 and GalNAc4Gal2 with two Gal-deficient sites. The most frequent Gal-deficient sites were at Ser230 and/or Thr236. BIOLOGICAL SIGNIFICANCE In this work, we studied the O-glycosylation in the hinge region of human immunoglobulin A1 (IgA1). Aberrant glycosylation of the protein plays a key role in the pathogenesis of IgA nephropathy. Thus identification of the O-glycan composition of IgA1 is important for a deeper understanding of the disease mechanism, biomarker discovery and validation, and implementation and monitoring of disease-specific therapies. We developed a new procedure for elucidating the heterogeneity of IgA1 O-glycosylation. After running a polyacrylamide gel electrophoresis under denaturing conditions, the heavy chain of IgA1 was subjected to in-gel digestion by trypsin. O-glycopeptides were separated from the digest on capillary columns using a microgradient chromatographic device (replacing commonly used liquid chromatographs) and subjected to MALDI-TOF/TOF mass spectrometry (MS) and tandem mass spectrometry (MS/MS) involving post-source decay fragmentation. We show that the complete modification of cysteines by iodoacetamide prior to electrophoresis is critical for successful MS/MS analyses on the way to deciphering the microheterogeneity of O-glycosylation in IgA1. Similarly, the removal of the excess of the reagent is equally important. The acquired MS/MS allowed assigning up to six O-glycosylation sites and identification of isomeric O-glycoforms. We show that our simplified approach is efficient and has a high potential to provide a method for the rapid assessment of IgA1 heterogeneity that is a less expensive and yet corroborating alternative to LC-(high-resolution)-MS protocols. The novelty and biological significance reside in the demonstration, for the first time, of the distribution of the most abundant isoforms of HR O-glycopeptides of IgA1. As another new feature, we introduce a software solution for the interpretation of MS/MS data of O-glycopeptide isoforms, which provides the possibility of fast and easier data processing. This article is part of a Special Issue entitled: Posttranslational Protein modifications in biology and Medicine.
Collapse
Affiliation(s)
- Vojtěch Franc
- Department of Protein Biochemistry and Proteomics, Centre of the Region Haná for Biotechnological and Agricultural Research, Faculty of Science, Palacký University, Šlechtitelů 11, CZ-783 71 Olomouc, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
150
|
Moldt B, Saye-Francisco K, Schultz N, Burton DR, Hessell AJ. Simplifying the synthesis of SIgA: combination of dIgA and rhSC using affinity chromatography. Methods 2013; 65:127-32. [PMID: 23811333 DOI: 10.1016/j.ymeth.2013.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/17/2013] [Accepted: 06/18/2013] [Indexed: 01/30/2023] Open
Abstract
The mucosal epithelia together with adaptive immune responses, such as local production and secretion of dimeric and polymeric immunoglobulin A (IgA), are a crucial part of the first line of defense against invading pathogens. IgA is primarily secreted as SIgA and plays multiple roles in mucosal defense. The study of SIgA-mediated protection is an important area of research in mucosal immunity but an easy, fast and reproducible method to generate pathogen-specific SIgA in vitro has not been available. We report here a new method to produce SIgA by co-purification of dimeric IgA, containing J chain, and recombinant human SC expressed in CHO cells. We previously reported the generation, production and characterization of the human recombinant monoclonal antibody IgA2 b12. This antibody, derived from the variable regions of the neutralizing anti-HIV-1 mAb IgG1 b12, blocked viral attachment and uptake by epithelial cells in vitro. We used a cloned CHO cell line that expresses monomeric, dimeric and polymeric species of IgA2 b12 for large-scale production of dIgA2 b12. Subsequently, we generated a CHO cell line to express recombinant human secretory component (rhSC). Here, we combined dIgA2 b12 and CHO-expressed rhSC via column chromatography to produce SIgA2 b12 that remains fully intact upon elution with 0.1M citric acid, pH 3.0. We have performed biochemical analysis of the synthesized SIgA to confirm the species is of the expected size and retains the functional properties previously described for IgA2 b12. We show that SIgA2 b12 binds to the HIV-1 gp120 glycoprotein with similar apparent affinity to that of monomeric and dimeric forms of IgA2 b12 and neutralizes HIV-1 isolates with similar potency. An average yield of 6 mg of SIgA2 b12 was achieved from the combination of 20mg of purified dIgA2 b12 and 2L of rhSC-containing CHO cell supernatant. We conclude that synthesized production of stable SIgA can be generated by co-purification. This process introduces a simplified means of generating a variety of pathogen-specific SIgA antibodies for research and clinical applications.
Collapse
Affiliation(s)
- Brian Moldt
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Karen Saye-Francisco
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Niccole Schultz
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R Burton
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Boston, MA, USA
| | - Ann J Hessell
- Department of Immunology and Microbial Science and IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|