1
|
Loriamini M, Lewis‐Bakker MM, Binnington B, Kotra LP, Branch DR. A novel small molecule phagocytosis inhibitor, KB-208, ameliorates ITP in mouse models with similar efficacy as IVIG. Transfusion 2024; 64:2233-2240. [PMID: 39511877 PMCID: PMC11637249 DOI: 10.1111/trf.18060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND The characteristic feature of immune cytopenias involves the process of extravascular phagocytosis, wherein macrophages in the spleen and/or liver engage in the destruction of blood cells that have been opsonized by auto- or alloantibodies. Therefore, new treatments that prevent phagocytosis will be advantageous, especially for short-term usage along with alternative options. STUDY DESIGN AND METHODS KB-208, a small molecule drug, previously shown to be efficacious for the in vitro inhibition of phagocytosis was synthesized. A passive antibody mouse model of immune thrombocytopenia (ITP) was used. Three different mouse strains (BALB/c, C57BL/6, CD1) were used to determine the efficacy of KB-208 compared with IVIG to ameliorate the ITP. Toxicity was investigated after 60-day chronic administration of KB-208 by a biochemistry panel, gross necroscopy and histopathology. RESULTS KB-208 showed similar efficacy to ameliorate the thrombocytopenia compared with IVIG in all three mouse strains. This small molecule drug was effective at 1 mg/kg in ameliorating ITP, in comparison with IVIG at 1000-2500 mg/kg. KB-208 did not affect other blood parameters or elevate serum biochemistry markers of toxicity nor were any abnormal histopathological findings found. CONCLUSION KB-208 is similar to IVIG for the amelioration of ITP in multiple mouse strains. Chronic administration of KB-208 for 60 days did not demonstrate in vivo toxicity. These findings indicate that KB-208 is efficacious, without significant in vivo toxicities in mice, and is a potential small molecule candidate for further evaluation to be used in the treatment of ITP and possibly all immune cytopenias where phagocytosis is responsible for the pathophysiology.
Collapse
Affiliation(s)
- Melika Loriamini
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
- Centre for InnovationCanadian Blood ServicesTorontoOntarioCanada
| | | | - Beth Binnington
- Centre for InnovationCanadian Blood ServicesTorontoOntarioCanada
| | - Lakshmi P. Kotra
- Krembil Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
| | - Donald R. Branch
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
- Centre for InnovationCanadian Blood ServicesTorontoOntarioCanada
- Department of MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
2
|
Krištić J, Lauc G. The importance of IgG glycosylation-What did we learn after analyzing over 100,000 individuals. Immunol Rev 2024; 328:143-170. [PMID: 39364834 PMCID: PMC11659926 DOI: 10.1111/imr.13407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
All four subclasses of immunoglobulin G (IgG) antibodies have glycan structures attached to the protein part of the IgG molecules. Glycans linked to the Fc portion of IgG are found in all IgG antibodies, while about one-fifth of IgG antibodies in plasma also have glycans attached to the Fab portion of IgG. The IgG3 subclass is characterized by more complex glycosylation compared to other IgG subclasses. In this review, we discuss the significant influence that glycans exert on the structural and functional properties of IgG. We provide a comprehensive overview of how the composition of these glycans can affect IgG's effector functions by modulating its interactions with Fcγ receptors and other molecules such as the C1q component of complement, which in turn influence various immune responses triggered by IgG, including antibody-dependent cell-mediated cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). In addition, the importance of glycans for the efficacy of therapeutics like monoclonal antibodies and intravenous immunoglobulin (IVIg) therapy is discussed. Moreover, we offer insights into IgG glycosylation characteristics and roles derived from general population, disease-specific, and interventional studies. These studies indicate that IgG glycans are important biomarkers and functional effectors in health and disease.
Collapse
Affiliation(s)
| | - Gordan Lauc
- Genos Glycoscience Research LaboratoryZagrebCroatia
- Faculty of Pharmacy and BiochemistryUniversity of ZagrebZagrebCroatia
| |
Collapse
|
3
|
Beyze A, Larroque C, Le Quintrec M. The role of antibody glycosylation in autoimmune and alloimmune kidney diseases. Nat Rev Nephrol 2024; 20:672-689. [PMID: 38961307 DOI: 10.1038/s41581-024-00850-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 07/05/2024]
Abstract
Immunoglobulin glycosylation is a pivotal mechanism that drives the diversification of antibody functions. The composition of the IgG glycome is influenced by environmental factors, genetic traits and inflammatory contexts. Differential IgG glycosylation has been shown to intricately modulate IgG effector functions and has a role in the initiation and progression of various diseases. Analysis of IgG glycosylation is therefore a promising tool for predicting disease severity. Several autoimmune and alloimmune disorders, including critical and potentially life-threatening conditions such as systemic lupus erythematosus, anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculitis and antibody-mediated kidney graft rejection, are driven by immunoglobulin. In certain IgG-driven kidney diseases, including primary membranous nephropathy, IgA nephropathy and lupus nephritis, particular glycome characteristics can enhance in situ complement activation and the recruitment of innate immune cells, resulting in more severe kidney damage. Hypofucosylation, hypogalactosylation and hyposialylation are the most common IgG glycosylation traits identified in these diseases. Modulating IgG glycosylation could therefore be a promising therapeutic strategy for regulating the immune mechanisms that underlie IgG-driven kidney diseases and potentially reduce the burden of immunosuppressive drugs in affected patients.
Collapse
Affiliation(s)
- Anaïs Beyze
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France.
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France.
- University of Montpellier, Montpellier, France.
| | - Christian Larroque
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France
- University of Montpellier, Montpellier, France
| | - Moglie Le Quintrec
- Institute of Regenerative Medicine and Biotherapy, IRMB U1183, Montpellier, France.
- Department of Nephrology, Dialysis and Transplantation, Montpellier University Hospital, Montpellier, France.
- University of Montpellier, Montpellier, France.
| |
Collapse
|
4
|
Bauer-Smith H, Sudol ASL, Beers SA, Crispin M. Serum immunoglobulin and the threshold of Fc receptor-mediated immune activation. Biochim Biophys Acta Gen Subj 2023; 1867:130448. [PMID: 37652365 PMCID: PMC11032748 DOI: 10.1016/j.bbagen.2023.130448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Antibodies can mediate immune recruitment or clearance of immune complexes through the interaction of their Fc domain with cellular Fc receptors. Clustering of antibodies is a key step in generating sufficient avidity for efficacious receptor recognition. However, Fc receptors may be saturated with prevailing, endogenous serum immunoglobulin and this raises the threshold by which cellular receptors can be productively engaged. Here, we review the factors controlling serum IgG levels in both healthy and disease states, and discuss how the presence of endogenous IgG is encoded into the functional activation thresholds for low- and high-affinity Fc receptors. We discuss the circumstances where antibody engineering can help overcome these physiological limitations of therapeutic antibodies. Finally, we discuss how the pharmacological control of Fc receptor saturation by endogenous IgG is emerging as a feasible mechanism for the enhancement of antibody therapeutics.
Collapse
Affiliation(s)
- Hannah Bauer-Smith
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stephen A Beers
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
5
|
García-Alija M, van Moer B, Sastre DE, Azzam T, Du JJ, Trastoy B, Callewaert N, Sundberg EJ, Guerin ME. Modulating antibody effector functions by Fc glycoengineering. Biotechnol Adv 2023; 67:108201. [PMID: 37336296 PMCID: PMC11027751 DOI: 10.1016/j.biotechadv.2023.108201] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Antibody based drugs, including IgG monoclonal antibodies, are an expanding class of therapeutics widely employed to treat cancer, autoimmune and infectious diseases. IgG antibodies have a conserved N-glycosylation site at Asn297 that bears complex type N-glycans which, along with other less conserved N- and O-glycosylation sites, fine-tune effector functions, complement activation, and half-life of antibodies. Fucosylation, galactosylation, sialylation, bisection and mannosylation all generate glycoforms that interact in a specific manner with different cellular antibody receptors and are linked to a distinct functional profile. Antibodies, including those employed in clinical settings, are generated with a mixture of glycoforms attached to them, which has an impact on their efficacy, stability and effector functions. It is therefore of great interest to produce antibodies containing only tailored glycoforms with specific effects associated with them. To this end, several antibody engineering strategies have been developed, including the usage of engineered mammalian cell lines, in vitro and in vivo glycoengineering.
Collapse
Affiliation(s)
- Mikel García-Alija
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain
| | - Berre van Moer
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium
| | - Diego E Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Tala Azzam
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jonathan J Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Beatriz Trastoy
- Structural Glycoimmunology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia, 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| | - Nico Callewaert
- VIB Center for Medical Biotechnology, VIB, Zwijnaarde, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium; Department of Biochemistry and Microbiology, Ghent University, Technologiepark 71, 9052 Ghent (Zwijnaarde), Belgium.
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Marcelo E Guerin
- Structural Glycobiology Laboratory, Biocruces Health Research Institute, Barakaldo, Bizkaia 48903, Spain; Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain.
| |
Collapse
|
6
|
Bayry J, Ahmed EA, Toscano-Rivero D, Vonniessen N, Genest G, Cohen CG, Dembele M, Kaveri SV, Mazer BD. Intravenous Immunoglobulin: Mechanism of Action in Autoimmune and Inflammatory Conditions. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1688-1697. [PMID: 37062358 DOI: 10.1016/j.jaip.2023.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/18/2023]
Abstract
Intravenous immunoglobulin (IVIG) is the mainstay of therapy for humoral immune deficiencies and numerous inflammatory disorders. Although the use of IVIG may be supplanted by several targeted therapies to cytokines, the ability of polyclonal normal IgG to act as an effector molecule as well as a regulatory molecule is a clear example of the polyfunctionality of IVIG. This article will address the mechanism of action of IVIG in a number of important conditions that are otherwise resistant to treatment. In this commentary, we will highlight mechanistic studies that shed light on the action of IVIG. This will be approached by identifying effects that are both common and disease-specific, targeting actions that have been demonstrated on cells and processes that represent both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France; Department of Biological Sciences and Engineering, Indian Institute of Technology Palakkad, Palakkad, India.
| | - Eisha A Ahmed
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Diana Toscano-Rivero
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Nicholas Vonniessen
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Genevieve Genest
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Casey G Cohen
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Marieme Dembele
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Srini V Kaveri
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - Bruce D Mazer
- Research Institute of McGill University Health Centre, Translational Program in Respiratory Diseases and Department of Pediatrics, McGill University Faculty of Medicine, Montreal, Quebec, Canada.
| |
Collapse
|
7
|
Abstract
Glycosylation has a profound influence on protein activity and cell biology through a variety of mechanisms, such as protein stability, receptor interactions and signal transduction. In many rheumatic diseases, a shift in protein glycosylation occurs, and is associated with inflammatory processes and disease progression. For example, the Fc-glycan composition on (auto)antibodies is associated with disease activity, and the presence of additional glycans in the antigen-binding domains of some autoreactive B cell receptors can affect B cell activation. In addition, changes in synovial fibroblast cell-surface glycosylation can alter the synovial microenvironment and are associated with an altered inflammatory state and disease activity in rheumatoid arthritis. The development of our understanding of the role of glycosylation of plasma proteins (particularly (auto)antibodies), cells and tissues in rheumatic pathological conditions suggests that glycosylation-based interventions could be used in the treatment of these diseases.
Collapse
Affiliation(s)
- Theresa Kissel
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - René E M Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Thomas W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
8
|
Mimura Y, Mimura-Kimura Y, Saldova R, Rudd PM, Jefferis R. Enhanced Immunomodulatory Effect of Intravenous Immunoglobulin by Fc Galactosylation and Nonfucosylation. Front Immunol 2022; 13:818382. [PMID: 35154135 PMCID: PMC8831331 DOI: 10.3389/fimmu.2022.818382] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is used as an immunomodulatory agent in the treatment of various autoimmune/inflammatory diseases although its mechanism of action remains elusive. Recently, nonfucosylated IgG has been shown to be preferentially bound to Fcγ receptor IIIa (FcγRIIIa) on circulating natural killer cells; therefore, we hypothesized that nonfucosylated IVIG may modulate immune responses through FcγRIIIa blockade. Here, homogeneous fucosylated or nonfucosylated glycoforms of normal polyclonal IgG bearing sialylated, galactosylated or nongalactosylated Fc oligosaccharides were generated by chemoenzymatic glycoengineering to investigate whether the IgG glycoforms can inhibit antibody-dependent cellular cytotoxicity (ADCC). Among the six IgG glycoforms, galactosylated, nonfucosylated IgG [(G2)2] had the highest affinity to FcγRIIIa and 20 times higher potency to inhibit ADCC than native IgG. A pilot study of IVIG treatment in mice with collagen antibody-induced arthritis highlighted the low-dose (G2)2 glycoform of IVIG (0.1 g/kg) as an effective immunomodulatory agent as the 10-fold higher dose of native IVIG. These preliminary results suggest that the anti-inflammatory activity of IVIG is in part mediated via activating FcγR blockade by galactosylated, nonfucosylated IgG and that such nonfucosylated IgG glycoforms bound to FcγRs on immune cells play immunomodulatory roles in health and disease. This study provides insights into improved therapeutic strategies for autoimmune/inflammatory diseases using glycoengineered IVIG and recombinant Fc.
Collapse
Affiliation(s)
- Yusuke Mimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
- *Correspondence: Yusuke Mimura,
| | - Yuka Mimura-Kimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Dublin, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Dublin, Ireland
| | - Pauline M. Rudd
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Dublin, Ireland
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Centros, Singapore
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
9
|
New insights into IVIg mechanisms and alternatives in autoimmune and inflammatory diseases. Curr Opin Hematol 2021; 27:392-398. [PMID: 32868670 DOI: 10.1097/moh.0000000000000609] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Intravenous immunoglobulin (IVIg) is an effective treatment for an increasing number of autoimmune and inflammatory conditions. However, IVIg continues to be limited by problems of potential shortages and cost. A number of mechanisms have been described for IVIg, which have been captured in newly emergent IVIg mimetic and IVIg alternative therapies. This review discusses the recent developments in IVIg mimetics and alternatives. RECENT FINDINGS Newly emergent IVIg mimetics and alternatives capture major proposed mechanisms of IVIg, including FcγR blockade, FcRn inhibition, complement inhibition, immune complex mimetics and sialylated IgG. Many of these emergent therapies have promising preclinical and clinical trial results. SUMMARY Significant research has been undertaken into the mechanism of IVIg in the treatment of autoimmune and inflammatory disease. Understanding the major IVIg mechanisms has allowed for rational development of IVIg mimetics and alternatives for several IVIg-treatable diseases.
Collapse
|
10
|
Chio JCT, Xu KJ, Popovich P, David S, Fehlings MG. Neuroimmunological therapies for treating spinal cord injury: Evidence and future perspectives. Exp Neurol 2021; 341:113704. [PMID: 33745920 DOI: 10.1016/j.expneurol.2021.113704] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022]
Abstract
Spinal cord injury (SCI) has a complex pathophysiology. Following the initial physical trauma to the spinal cord, which may cause vascular disruption, hemorrhage, mechanical injury to neural structures and necrosis, a series of biomolecular cascades is triggered to evoke secondary injury. Neuroinflammation plays a major role in the secondary injury after traumatic SCI. To date, the administration of systemic immunosuppressive medications, in particular methylprednisolone sodium succinate, has been the primary pharmacological treatment. This medication is given as a complement to surgical decompression of the spinal cord and maintenance of spinal cord perfusion through hemodynamic augmentation. However, the impact of neuroinflammation is complex with harmful and beneficial effects. The use of systemic immunosuppressants is further complicated by the natural onset of post-injury immunosuppression, which many patients with SCI develop. It has been hypothesized that immunomodulation to attenuate detrimental aspects of neuroinflammation after SCI, while avoiding systemic immunosuppression, may be a superior approach. To accomplish this, a detailed understanding of neuroinflammation and the systemic immune responses after SCI is required. Our review will strive to achieve this goal by first giving an overview of SCI from a clinical and basic science context. The role that neuroinflammation plays in the pathophysiology of SCI will be discussed. Next, the positive and negative attributes of the innate and adaptive immune systems in neuroinflammation after SCI will be described. With this background established, the currently existing immunosuppressive and immunomodulatory therapies for treating SCI will be explored. We will conclude with a summary of topics that can be explored by neuroimmunology research. These concepts will be complemented by points to be considered by neuroscientists developing therapies for SCI and other injuries to the central nervous system.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| | - Katherine Jiaxi Xu
- Human Biology Program, University of Toronto, Wetmore Hall, 300 Huron St., Room 105, Toronto, Ontario M5S 3J6, Canada.
| | - Phillip Popovich
- Department of Neuroscience, Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Neurological Institute, The Ohio State University, Wexner Medical Center, 410 W. 10(th) Ave., Columbus 43210, USA.
| | - Samuel David
- Centre for Research in Neuroscience and BRaIN Program, The Research Institute of the McGill University Health Centre, 1650 Cedar Ave., Montreal, Quebec H3G 1A4, Canada.
| | - Michael G Fehlings
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Chen Y, Hu J, Chen Y. Platelet desialylation and TFH cells-the novel pathway of immune thrombocytopenia. Exp Hematol Oncol 2021; 10:21. [PMID: 33722280 PMCID: PMC7958461 DOI: 10.1186/s40164-021-00214-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/07/2021] [Indexed: 12/15/2022] Open
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by immune-mediated destruction of one's own platelets. The progression of thrombocytopenia involves an imbalance of platelet production and clearance. B cells can induce autoantibodies, and T cells contribute to the pathological progression as well. Some patients with ITP have a poor response to common first-line therapies. Recent studies have shown that a novel Fc-independent platelet clearance pathway is associated with poor prognosis in these patients. By this pathway, desialylated platelets can be cleared by Ashwell-Morell receptor (AMR) on hepatocytes. Research has demonstrated that patients with refractory ITP usually have a high level of desialylation, indicating the important role of sialylation on platelet membrane glycoprotein (GP) in patients with primary immune thrombocytopenia, and neuraminidase 1(NEU1) translocation might be involved in this process. Patients with ITP who are positive for anti-GPIbα antibodies have a poor prognosis, which indicates that anti-GPIbα antibodies are associated with this Fc-independent platelet clearance pathway. Experiments have proven that these antibodies could lead to the desialylation of GPs on platelets. The T follicular helper (TFH) cell level is related to the expression of the anti-GPIbα antibody, which indicates its role in the progression of desialylation. This review will discuss platelet clearance and production, especially the role of the anti-GPIbα antibody and desialylation in the pathophysiology of ITP and therapy for this disease.
Collapse
Affiliation(s)
- Yuwen Chen
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, No.29 Xinquan Road, 350001, Fuzhou, Fujian, China
| | - Jianda Hu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, No.29 Xinquan Road, 350001, Fuzhou, Fujian, China
| | - Yingyu Chen
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, No.29 Xinquan Road, 350001, Fuzhou, Fujian, China.
| |
Collapse
|
12
|
Zhou X, Motta F, Selmi C, Ridgway WM, Gershwin ME, Zhang W. Antibody glycosylation in autoimmune diseases. Autoimmun Rev 2021; 20:102804. [PMID: 33727152 DOI: 10.1016/j.autrev.2021.102804] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023]
Abstract
The glycosylation of the fragment crystallizable (Fc) region of immunoglobulins (Ig) is critical for the modulation of antibody effects on inflammation. Moreover, antibody glycosylation may induce pathologic modifications and ultimately contribute to the development of autoimmune diseases. Thanks to progress in the analysis of glycosylation, more data are available on IgG and its subclass structures in the context of autoimmune diseases. In this review, we focused on the impact of Ig glycosylation in autoimmunity, describing how it modulates the immune response and how glycome profiles can be used as biomarkers of disease activity. The analysis of antibody glycosylation demonstrated specific features in human autoimmune and chronic inflammatory conditions, including rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel disease and autoimmune liver diseases, among others. Within the same disease, different patterns are associated with disease severity and treatment options. Future research may increase the information available on the distinct glycome profiles and expand their potential role as biomarkers and as targets for treatment, ultimately favoring an individualized approach.
Collapse
Affiliation(s)
- Xing Zhou
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA 95616, USA; Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Francesca Motta
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - William M Ridgway
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA 95616, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA 95616, USA.
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA 95616, USA.
| |
Collapse
|
13
|
Li D, Lou Y, Zhang Y, Liu S, Li J, Tao J. Sialylated immunoglobulin G: a promising diagnostic and therapeutic strategy for autoimmune diseases. Am J Cancer Res 2021; 11:5430-5446. [PMID: 33859756 PMCID: PMC8039950 DOI: 10.7150/thno.53961] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
Human immunoglobulin G (IgG), especially autoantibodies, has major implications for the diagnosis and management of a wide range of autoimmune diseases. However, some healthy individuals also have autoantibodies, while a portion of patients with autoimmune diseases test negative for serologic autoantibodies. Recent advances in glycomics have shown that IgG Fc N-glycosylations are more reliable diagnostic and monitoring biomarkers than total IgG autoantibodies in a wide variety of autoimmune diseases. Furthermore, these N-glycosylations of IgG Fc, particularly sialylation, have been reported to exert significant anti-inflammatory effects by upregulating inhibitory FcγRIIb on effector macrophages and reducing the affinity of IgG for either complement protein or activating Fc gamma receptors. Therefore, sialylated IgG is a potential therapeutic strategy for attenuating pathogenic autoimmunity. IgG sialylation-based therapies for autoimmune diseases generated through genetic, metabolic or chemoenzymatic modifications have made some advances in both preclinical studies and clinical trials.
Collapse
|
14
|
Neuraminidase Inhibitor Zanamivir Ameliorates Collagen-Induced Arthritis. Int J Mol Sci 2021; 22:ijms22031428. [PMID: 33572654 PMCID: PMC7867009 DOI: 10.3390/ijms22031428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/27/2021] [Accepted: 01/27/2021] [Indexed: 01/20/2023] Open
Abstract
Altered sialylation patterns play a role in chronic autoimmune diseases such as rheumatoid arthritis (RA). Recent studies have shown the pro-inflammatory activities of immunoglobulins (Igs) with desialylated sugar moieties. The role of neuraminidases (NEUs), enzymes which are responsible for the cleavage of terminal sialic acids (SA) from sialoglycoconjugates, is not fully understood in RA. We investigated the impact of zanamivir, an inhibitor of the influenza virus neuraminidase, and mammalian NEU2/3 on clinical outcomes in experimental arthritides studies. The severity of arthritis was monitored and IgG titers were measured by ELISA. (2,6)-linked SA was determined on IgG by ELISA and on cell surfaces by flow cytometry. Zanamivir at a dose of 100 mg/kg (zana-100) significantly ameliorated collagen-induced arthritis (CIA), whereas zana-100 was ineffective in serum transfer-induced arthritis. Systemic zana-100 treatment reduced the number of splenic CD138+/TACI+ plasma cells and CD19+ B cells, which was associated with lower IgG levels and an increased sialylation status of IgG compared to controls. Our data reveal the contribution of NEU2/3 in CIA. Zanamivir down-modulated the T and B cell-dependent humoral immune response and induced an anti-inflammatory milieu by inhibiting sialic acid degradation. We suggest that neuraminidases might represent a promising therapeutic target for RA and possibly also for other antibody-mediated autoimmune diseases.
Collapse
|
15
|
Nimmerjahn F, Werner A. Sweet Rules: Linking Glycosylation to Antibody Function. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:365-393. [PMID: 34687017 DOI: 10.1007/978-3-030-76912-3_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Antibodies produced upon infections with pathogenic microorganisms are essential for clearing primary infections and for providing the host with long-lasting immunity. Moreover, antibodies have become the most widely used platform for developing novel therapies against cancer and autoimmunity, requiring an in-depth understanding of how antibodies mediate their activity in vivo and which factors modulate pro- or anti-inflammatory antibody activities. Since the discovery that select residues present in the sugar domain attached to the immunoglobulin G (IgG) fragment crystallizable (Fc) region can modulate both, pro- and anti-inflammatory effector functions, a wealth of studies has focused on understanding how IgG glycosylation is regulated and how this knowledge can be used to optimize therapeutic antibody activity. With the introduction of glycoengineered afucosylated antibodies in cancer therapy and the initiation of clinical testing of highly sialylated anti-inflammatory antibodies the proof-of-concept that understanding antibody glycosylation can lead to clinical innovation has been provided. The focus of this review is to summarize recent insights into how antibody glycosylation is regulated in vivo and how select sugar residues impact IgG function.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Chair of Genetics, Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany.
- Medical Immunology Campus Erlangen, Erlangen, Germany.
| | - Anja Werner
- Chair of Genetics, Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Abstract
Changes in immunoglobulin G (IgG) glycosylation pattern have been observed in a vast array of auto- and alloimmune, infectious, cardiometabolic, malignant, and other diseases. This chapter contains an updated catalog of over 140 studies within which IgG glycosylation analysis was performed in a disease setting. Since the composition of IgG glycans is known to modulate its effector functions, it is suggested that a changed IgG glycosylation pattern in patients might be involved in disease development and progression, representing a predisposition and/or a functional effector in disease pathology. In contrast to the glycopattern of bulk serum IgG, which likely relates to the systemic inflammatory background, the glycosylation profile of antigen-specific IgG probably plays a direct role in disease pathology in several infectious and allo- and autoimmune antibody-dependent diseases. Depending on the specifics of any given disease, IgG glycosylation read-out might therefore in the future be developed into a useful clinical biomarker or a supplementary to currently used biomarkers.
Collapse
Affiliation(s)
- Marija Pezer
- Glycoscience Research Laboratory, Genos Ltd., Zagreb, Croatia.
| |
Collapse
|
17
|
Mimura Y, Saldova R, Mimura-Kimura Y, Rudd PM, Jefferis R. Importance and Monitoring of Therapeutic Immunoglobulin G Glycosylation. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:481-517. [PMID: 34687020 DOI: 10.1007/978-3-030-76912-3_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The complex diantennary-type oligosaccharides at Asn297 residues of the IgG heavy chains have a profound impact on the safety and efficacy of therapeutic IgG monoclonal antibodies (mAbs). Fc glycosylation of a mAb is an established critical quality attribute (CQA), and its oligosaccharide profile is required to be thoroughly characterized by state-of-the-art analytical methods. The Fc oligosaccharides are highly heterogeneous, and the differentially glycosylated species (glycoforms) of IgG express unique biological activities. Glycoengineering is a promising approach for the production of selected mAb glycoforms with improved effector functions, and non- and low-fucosylated mAbs exhibiting enhanced antibody-dependent cellular cytotoxicity activity have been approved or are under clinical evaluation for treatment of cancers, autoimmune/chronic inflammatory diseases, and infection. Recently, the chemoenzymatic glycoengineering method that allows for the transfer of structurally defined oligosaccharides to Asn-linked GlcNAc residues with glycosynthase has been developed for remodeling of IgG-Fc oligosaccharides with high efficiency and flexibility. Additionally, various glycoengineering methods have been developed that utilize the Fc oligosaccharides of IgG as reaction handles to conjugate cytotoxic agents by "click chemistry", providing new routes to the design of antibody-drug conjugates (ADCs) with tightly controlled drug-antibody ratios (DARs) and homogeneity. This review focuses on current understanding of the biological relevance of individual IgG glycoforms and advances in the development of next-generation antibody therapeutics with improved efficacy and safety through glycoengineering.
Collapse
Affiliation(s)
- Yusuke Mimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan.
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin, Ireland
- UCD School of Medicine, College of Health and Agricultural Science, University College Dublin, Belfield, Dublin, Ireland
| | - Yuka Mimura-Kimura
- Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Pauline M Rudd
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin, Ireland
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Centros, Singapore
| | - Roy Jefferis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
18
|
Dall'Olio F, Malagolini N. Immunoglobulin G Glycosylation Changes in Aging and Other Inflammatory Conditions. EXPERIENTIA SUPPLEMENTUM (2012) 2021; 112:303-340. [PMID: 34687015 DOI: 10.1007/978-3-030-76912-3_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Among the multiple roles played by protein glycosylation, the fine regulation of biological interactions is one of the most important. The asparagine 297 (Asn297) of IgG heavy chains is decorated by a diantennary glycan bearing a number of galactose and sialic acid residues on the branches ranging from 0 to 2. In addition, the structure can present core-linked fucose and/or a bisecting GlcNAc. In many inflammatory and autoimmune conditions, as well as in metabolic, cardiovascular, infectious, and neoplastic diseases, the IgG Asn297-linked glycan becomes less sialylated and less galactosylated, leading to increased expression of glycans terminating with GlcNAc. These conditions alter also the presence of core-fucose and bisecting GlcNAc. Importantly, similar glycomic alterations are observed in aging. The common condition, shared by the above-mentioned pathological conditions and aging, is a low-grade, chronic, asymptomatic inflammatory state which, in the case of aging, is known as inflammaging. Glycomic alterations associated with inflammatory diseases often precede disease onset and follow remission. The aberrantly glycosylated IgG glycans associated with inflammation and aging can sustain inflammation through different mechanisms, fueling a vicious loop. These include complement activation, Fcγ receptor binding, binding to lectin receptors on antigen-presenting cells, and autoantibody reactivity. The complex molecular bases of the glycomic changes associated with inflammation and aging are still poorly understood.
Collapse
Affiliation(s)
- Fabio Dall'Olio
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy.
| | - Nadia Malagolini
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
19
|
Cobb BA. The history of IgG glycosylation and where we are now. Glycobiology 2020; 30:202-213. [PMID: 31504525 DOI: 10.1093/glycob/cwz065] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 12/15/2022] Open
Abstract
IgG glycosylation is currently at the forefront of both immunology and glycobiology, likely due in part to the widespread and growing use of antibodies as drugs. For over four decades, it has been recognized that the conserved N-linked glycan on asparagine 297 found within the second Ig domain of the heavy chain (CH2) that helps to comprise Fc region of IgG plays a special role in IgG structure and function. Changes in galactosylation, fucosylation and sialylation are now well-established factors, which drive differential IgG function, ranging from inhibitory/anti-inflammatory to activating complement and promoting antibody-dependent cellular cytotoxicity. Thus, if we are to truly understand how to design and deploy antibody-based drugs with maximal efficacy and evaluate proper vaccine responses from a protective and functional perspective, a deep understanding of IgG glycosylation is essential. This article is intended to provide a comprehensive review of the IgG glycosylation field and the impact glycans have on IgG function, beginning with the earliest findings over 40 years ago, in order to provide a robust foundation for moving forward.
Collapse
Affiliation(s)
- Brian A Cobb
- Department of Pathology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
20
|
Gillespie ER, Ruitenberg MJ. Neuroinflammation after SCI: Current Insights and Therapeutic Potential of Intravenous Immunoglobulin. J Neurotrauma 2020; 39:320-332. [PMID: 32689880 DOI: 10.1089/neu.2019.6952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Traumatic spinal cord injury (SCI) elicits a complex cascade of cellular and molecular inflammatory events. Although certain aspects of the inflammatory response are essential to wound healing and repair, post-SCI inflammation is, on balance, thought to be detrimental to recovery by causing "bystander damage" and the spread of pathology into spared but vulnerable regions of the spinal cord. Much of the research to date has therefore focused on understanding the inflammatory drivers of secondary tissue loss after SCI, to define therapeutic targets and positively modulate this response. Numerous experimental studies have demonstrated that modulation of the inflammatory response to SCI can indeed lead to significant neuroprotection and improved recovery. However, it is now also recognized that broadscale immunosuppression is not necessarily beneficial and may even carry the risk of contributing to the development of serious adverse events. Immune modulation rather than suppression is therefore now considered a more promising approach to target harmful post-traumatic inflammation following a major neurotraumatic event such as SCI. One promising immunomodulatory agent is intravenous immunoglobulin (IVIG), a plasma product that contains mostly immunoglobulin G (IgG) from thousands of healthy donors. IVIG is currently already widely used to treat a range of autoimmune diseases, but recent studies have found that it also holds great promise for treating acute neurological conditions, including SCI. This review provides an overview of the inflammatory response to SCI, immunomodulatory approaches that are currently in clinical trials, proposed mechanisms of action for IVIG therapy, and the putative relevance of these in the context of neurotraumatic events.
Collapse
Affiliation(s)
- Ellen R Gillespie
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Trauma, Critical Care, and Recovery, Brisbane Diamantina Health Partners, Brisbane, Australia
| |
Collapse
|
21
|
Liu X, Cao W, Li T. High-Dose Intravenous Immunoglobulins in the Treatment of Severe Acute Viral Pneumonia: The Known Mechanisms and Clinical Effects. Front Immunol 2020; 11:1660. [PMID: 32760407 PMCID: PMC7372093 DOI: 10.3389/fimmu.2020.01660] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022] Open
Abstract
The current outbreak of viral pneumonia, caused by novel coronavirus SARS-CoV-2, is the focus of worldwide attention. The WHO declared the COVID-19 outbreak a pandemic event on Mar 12, 2020, and the number of confirmed cases is still on the rise worldwide. While most infected individuals only experience mild symptoms or may even be asymptomatic, some patients rapidly progress to severe acute respiratory failure with substantial mortality, making it imperative to develop an efficient treatment for severe SARS-CoV-2 pneumonia alongside supportive care. So far, the optimal treatment strategy for severe COVID-19 remains unknown. Intravenous immunoglobulin (IVIg) is a blood product pooled from healthy donors with high concentrations of immunoglobulin G (IgG) and has been used in patients with autoimmune and inflammatory diseases for more than 30 years. In this review, we aim to highlight the known mechanisms of immunomodulatory effects of high-dose IVIg therapy, the immunopathological hypothesis of viral pneumonia, and the clinical evidence of IVIg therapy in viral pneumonia. We then make cautious therapeutic inferences about high-dose IVIg therapy in treating severe COVID-19. These inferences may provide relevant and useful insights in order to aid treatment for COVID-19.
Collapse
Affiliation(s)
- Xiaosheng Liu
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Wei Cao
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Taisheng Li
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Department of Infectious Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Lewis BJ, Branch DR. Mouse Models of Rheumatoid Arthritis for Studies on Immunopathogenesis and Preclinical Testing of Fc Receptor-Targeting Biologics. Pharmacology 2020; 105:618-629. [DOI: 10.1159/000508239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/27/2020] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation, swelling, and pain in the joints and involves systemic complications. Mouse models of RA have been extensively used to model the pathogenesis of RA and to develop effective therapies. Although many components of the immune system have been studied in these models, the role of crystallizable fragment (Fc) gamma receptors (FcγRs) in RA has been sorely neglected. The aim of this review was to introduce the different mouse models of RA and to describe the different drug development strategies that have been tested in these models to target FcγR function, with the focus being on drugs that have been made from the Fc of immunoglobulin G (IgG). <b><i>Summary:</i></b> Evidence suggests that FcγRs play a major role in immune complex-induced inflammation in autoimmune diseases, such as RA. However, there is limited knowledge on the importance of FcγRs in the human disease even though there has been extensive work in mouse models of RA. Numerous mouse models of RA are available, with each model depicting certain aspects of the disease. Induced models of RA have nonspecific immune activation with cartilage-directed autoimmunity, whereas spontaneous models of RA develop without immunization, which results in a more chronic form of arthritis. These models have been used to test FcγR-targeting monoclonal antibodies, intravenous immunoglobulin (IVIg), subcutaneously administered IVIg, and recombinant Fcs for their ability to interact with and modify FcγR function. Recombinant Fcs avidly bind FcγRs and exhibit enhanced therapeutic efficacy in mouse models of RA. <b><i>Key Message:</i></b> The therapeutic utility of targeting FcγRs with recombinant Fcs is great and should be explored in human clinical trials for autoimmune diseases, such as RA.
Collapse
|
23
|
Graeter S, Simon HU, von Gunten S. Granulocyte death mediated by specific antibodies in intravenous immunoglobulin (IVIG). Pharmacol Res 2020; 154:104168. [DOI: 10.1016/j.phrs.2019.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 12/23/2022]
|
24
|
Das M, Karnam A, Stephen-Victor E, Gilardin L, Bhatt B, Kumar Sharma V, Rambabu N, Patil V, Lecerf M, Käsermann F, Bruneval P, Narayanaswamy Balaji K, Benveniste O, Kaveri SV, Bayry J. Intravenous immunoglobulin mediates anti-inflammatory effects in peripheral blood mononuclear cells by inducing autophagy. Cell Death Dis 2020; 11:50. [PMID: 31974400 PMCID: PMC6978335 DOI: 10.1038/s41419-020-2249-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 12/11/2022]
Abstract
Autophagy plays an important role in the regulation of autoimmune and autoinflammatory responses of the immune cells. Defective autophagy process is associated with various autoimmune and inflammatory diseases. Moreover, in many of these diseases, the therapeutic use of normal immunoglobulin G or intravenous immunoglobulin (IVIG), a pooled normal IgG preparation, is well documented. Therefore, we explored if IVIG immunotherapy exerts therapeutic benefits via induction of autophagy in the immune cells. Here we show that IVIG induces autophagy in peripheral blood mononuclear cells (PBMCs). Further dissection of this process revealed that IVIG-induced autophagy is restricted to inflammatory cells like monocytes, dendritic cells, and M1 macrophages but not in cells associated with Th2 immune response like M2 macrophages. IVIG induces autophagy by activating AMP-dependent protein kinase, beclin-1, class III phosphoinositide 3-kinase and p38 mitogen-activated protein kinase and by inhibiting mammalian target of rapamycin. Mechanistically, IVIG-induced autophagy is F(ab')2-dependent but sialylation independent, and requires endocytosis of IgG by innate cells. Inhibition of autophagy compromised the ability of IVIG to suppress the inflammatory cytokines in innate immune cells. Moreover, IVIG therapy in inflammatory myopathies such as dermatomyositis, antisynthetase syndrome and immune-mediated necrotizing myopathy induced autophagy in PBMCs and reduced inflammatory cytokines in the circulation, thus validating the translational importance of these results. Our data provide insight on how circulating normal immunoglobulins maintain immune homeostasis and explain in part the mechanism by which IVIG therapy benefits patients with autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Mrinmoy Das
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France
| | - Emmanuel Stephen-Victor
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France
| | - Laurent Gilardin
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France.,Département de Médecine Interne et Immunologie Clinique, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Bharat Bhatt
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
| | - Varun Kumar Sharma
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France
| | - Naresh Rambabu
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France
| | - Veerupaxagouda Patil
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France
| | - Maxime Lecerf
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| | - Fabian Käsermann
- CSL Behring, Research, CSL Biologics Research Center, 3014, Bern, Switzerland
| | - Patrick Bruneval
- Service d'anatomie pathologique, Hôpital Européen Georges Pompidou, 75015, Paris, France
| | | | - Olivier Benveniste
- Département de Médecine Interne et Immunologie Clinique, Hôpital Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,Institut National de la Santé et de la Recherche Médicale Unité 974, Sorbonne Université, 75013, Paris, France
| | - Srini V Kaveri
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe- Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, 75006, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France.
| |
Collapse
|
25
|
Chen YY, Zhou YQ, Zhao N, Zhang Y, Xu WQ, Tang YM. Evaluation of IVIG response in relation to Th1/Th2 cytokines in pediatricm immune thrombocytopenia. Cytokine 2019; 120:234-241. [DOI: 10.1016/j.cyto.2019.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/10/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022]
|
26
|
Abstract
The precise mechanisms underlying anti-inflammatory effects of intravenous immunoglobulin (IVIg) therapies remain elusive. The sialylated IgG fraction within IVIg has been shown to be therapeutically more active in mouse models. Functionally, it has been suggested that IgG undergoes conformational changes upon Fc-sialylation which sterically impede binding to conventional FcγRs, but simultaneously allow binding to human DC-SIGN (SIGN-R1 in mice) and also CD23. These latter C-type lectins have been proposed responsible for the immunomodulatory effects in mouse models. However, there is conflicting evidence supporting direct interactions between sialylated human IgG and CD23/DC-SIGN. While cells expressing human CD23 and DC-SIGN in their native configuration bound their natural ligands IgE and ICAM-3, respectively, no IgG binding was observed, regardless of Fc-glycan sialylation in any context (with or without bisection and/or fucosylation) or presence of sialylated Fab-glycans. This was tested by both by FACS and a novel cellular Surface Plasmon Resonance imaging (cSPRi) approach allowing for monitoring low-affinity but high-avidity interactions. In summary, we find no evidence for human CD23 or DC-SIGN being bona fide receptors to human IgG, regardless of IgG Fc- or Fab-glycosylation status. However, these results do not exclude the possibility that either IgG glycosylation or C-type lectins affect IVIg therapies.
Collapse
|
27
|
Lewis BJB, Leontyev D, Neschadim A, Blacquiere M, Branch DR. GM-CSF and IL-4 are not involved in IVIG-mediated amelioration of ITP in mice: a role for IL-11 cannot be ruled out. Clin Exp Immunol 2019; 193:293-301. [PMID: 29704458 DOI: 10.1111/cei.13144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2018] [Indexed: 12/14/2022] Open
Abstract
Previously, we have reported that interleukin (IL)-4, granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-11, but not IL-33, are up-regulated in two strains of mice with immune thrombocytopenia (ITP) that are responsive to intravenous immunoglobulin (IVIg) treatment. Previously, IL-4 was ruled out in the mechanism of IVIg; however, other publications have suggested this cytokine as a major player in the mechanism of IVIg action. Thus, we sought to further investigate a role for IL-4 and, in addition, GM-CSF and IL-11 in the mechanism of action of IVIg using a murine model of ITP. A passive platelet antibody model was used to generate ITP in IL-4 receptor knock-out (IL-4R-/- ), IL-11 receptor knock-out (IL-11Rα-/- ) and GM-CSF knock-out (Csf2-/- ) mice. We also used a neutralizing antibody to IL-11 and recombinant human IL-11 (rhIL-11) in addition to depleting basophils in vivo to study the effect of IVIg to ameliorate ITP. Our results showed that basophils, IL-4 and GM-CSF were unimportant in both ITP induction and its amelioration by IVIg. The role of IL-11 in these processes was less clear. Even though IL-11Rα-/- mice with ITP responded to IVIg similarly to wild-type (WT) mice, treatment of ITP WT mice with rhIL-11 instead of IVIg showed an increase in platelet numbers and WT mice administered anti-IL-11 showed a significant reduction in the ability of IVIg to ameliorate the ITP. Our findings indicate that neither IL-4, basophils or GM-CSF have roles in IVIg amelioration of ITP; however, a role for IL-11 requires further study.
Collapse
Affiliation(s)
- B J B Lewis
- Department of Laboratory Medicine and Pathobiology, University of Toronto.,Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - D Leontyev
- Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - A Neschadim
- Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - M Blacquiere
- Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - D R Branch
- Department of Laboratory Medicine and Pathobiology, University of Toronto.,Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Kozicky LK, Menzies SC, Hotte N, Madsen KL, Sly LM. Intravenous immunoglobulin (IVIg) or IVIg-treated macrophages reduce DSS-induced colitis by inducing macrophage IL-10 production. Eur J Immunol 2019; 49:1251-1268. [PMID: 31054259 DOI: 10.1002/eji.201848014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/24/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022]
Abstract
Intravenous immunoglobulin (IVIg) is used to treat immune-mediated diseases but its mechanism of action is poorly understood. We have reported that co-treatment with IVIg and lipopolysaccharide activates macrophages to produce large amounts of anti-inflammatory IL-10 in vitro. Thus, we asked whether IVIg-treated macrophages or IVIg could reduce intestinal inflammation in mice during dextran sulfate sodium (DSS)-induced colitis by inducing macrophage IL-10 production in vivo. Adoptive transfer of IVIg-treated macrophages reduces intestinal inflammation in mice and collagen accumulation post-DSS. IVIg treatment also reduces DSS-induced intestinal inflammation and its activity is dependent on the Fc portion of the antibody. Ex vivo, IVIg induces IL-10 production and reduces IL-12/23p40 and IL-1β production in colon explant cultures. Co-staining tissues for mRNA, we demonstrate that macrophages are the source of IL-10 in IVIg-treated mice; and using IL-10-GFP reporter mice, we demonstrate that IVIg induces IL-10 production by intestinal macrophages. Finally, IVIg-mediated protection is lost in mice deficient in macrophage IL-10 production (LysMcre+/- IL-10fl/fl mice). Together, our data demonstrate a novel, in vivo mechanism of action for IVIg. IVIg-treated macrophages or IVIg could be used to treat people with intestinal inflammation and may be particularly useful for people with inflammatory bowel disease, who are refractory to therapy.
Collapse
Affiliation(s)
- Lisa K Kozicky
- Department of Pediatrics, Division of Gastroenterology, BC Children's Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Susan C Menzies
- Department of Pediatrics, Division of Gastroenterology, BC Children's Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Naomi Hotte
- Department of Medicine, Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada
| | - Karen L Madsen
- Department of Medicine, Division of Gastroenterology, University of Alberta, Edmonton, Alberta, Canada
| | - Laura M Sly
- Department of Pediatrics, Division of Gastroenterology, BC Children's Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
29
|
Zhang X, Owens J, Olsen HS, So E, Burch E, McCroskey MC, Li X, Weber GL, Bennett D, Rybin D, Zhou H, Hao H, Mérigeon EY, Block DS, LaRosa G, Strome SE. A recombinant human IgG1 Fc multimer designed to mimic the active fraction of IVIG in autoimmunity. JCI Insight 2019; 4:121905. [PMID: 30674715 DOI: 10.1172/jci.insight.121905] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 12/11/2018] [Indexed: 12/30/2022] Open
Abstract
The antiinflammatory effects of i.v. Ig (IVIG) in the treatment of autoimmune disease are due, in part, to the Fc fragments of Ig aggregates. In order to capitalize on the known antiinflammatory and tolerogenic properties of Ig Fc aggregates, we created a recombinant human IgG1 Fc multimer, GL-2045. In vitro, GL-2045 demonstrated high-avidity binding to Fc receptors, blocked the binding of circulating immune complexes from patients with rheumatoid arthritis to human Fcγ receptors (FcγRs), and inhibited antibody-mediated phagocytosis at log order-lower concentrations than IVIG. In vivo, administration of GL-2045 conferred partial protection against antibody-mediated platelet loss in a murine immune thrombocytopenic purpura (ITP) model. GL-2045 also suppressed disease activity in a therapeutic model of murine collagen-induced arthritis (CIA), which was associated with reduced circulating levels of IL-6. Furthermore, GL-2045 administration to nonhuman primates (NHPs) transiently increased systemic levels of the antiinflammatory cytokines IL-10 and IL-1RA, reduced the proinflammatory cytokine IL-8, and decreased surface expression of CD14 and HLA-DR on monocytes. These findings demonstrate the immunomodulatory properties of GL-2045 and suggest that it has potential as a treatment for autoimmune and inflammatory diseases, as a recombinant alternative to IVIG.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jane Owens
- Pfizer Inc., Cambridge, Massachusetts, USA
| | | | - Edward So
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Erin Burch
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | - Hua Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Haiping Hao
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Scott E Strome
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Galeotti C, Kaveri SV, Bayry J. IVIG-mediated effector functions in autoimmune and inflammatory diseases. Int Immunol 2019; 29:491-498. [PMID: 28666326 DOI: 10.1093/intimm/dxx039] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) is a pooled preparation of normal IgG obtained from several thousand healthy donors. It is widely used in the immunotherapy of a large number of autoimmune and inflammatory diseases. The mechanisms of action of IVIG are complex and, as discussed in this review, experimental and clinical data provide an indicator that the therapeutic benefit of IVIG therapy is due to several mutually non-exclusive mechanisms affecting soluble mediators as well as cellular components of the immune system. These mechanisms depend on Fc and/or F(ab')2 fragments. A better understanding of the effector functions of IVIG should help in identification of biomarkers of responses to IVIG in autoimmune patients.
Collapse
Affiliation(s)
- Caroline Galeotti
- Institut National de la Santé et de la Recherche Médicale Unité, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Centre de Recherche des Cordeliers, Equipe -Immunopathologie et Immunointervention Thérapeutique, Paris, France.,Department of Pediatric Rheumatology, National Referral Centre of Auto-inflammatory Diseases, CHU de Bicêtre, France
| | - Srini V Kaveri
- Institut National de la Santé et de la Recherche Médicale Unité, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Centre de Recherche des Cordeliers, Equipe -Immunopathologie et Immunointervention Thérapeutique, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale Unité, France.,Sorbonne Universités, UPMC Univ Paris 06, Paris, France.,Centre de Recherche des Cordeliers, Equipe -Immunopathologie et Immunointervention Thérapeutique, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| |
Collapse
|
31
|
Brückner C, Lehmann C, Dudziak D, Nimmerjahn F. Sweet SIGNs: IgG glycosylation leads the way in IVIG-mediated resolution of inflammation. Int Immunol 2019; 29:499-509. [PMID: 29300958 DOI: 10.1093/intimm/dxx053] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022] Open
Abstract
A hallmark of many chronic inflammatory and autoimmune diseases is that there is an impaired resolution of inflammation and return to the steady state. The infusion of high doses of pooled serum IgG preparations from thousands of donors [intravenous immunoglobulin (IVIG) therapy] has been shown to induce resolution of inflammation in a variety of chronic inflammatory and autoimmune diseases, suggesting that IgG molecules can instruct the immune system to stop inflammatory processes and initiate the return to the steady state. The aim of this review is to discuss how insights into the mechanism of IVIG activity may help to understand the molecular and cellular pathways underlying resolution of inflammation. We will put a special emphasis on pathways dependent on the IgG FC domain and IgG sialylation, as several recent studies have provided new insights into how this glycosylation-dependent pathway modulates innate and adaptive immune responses through different sets of C-type or I-type lectins.
Collapse
Affiliation(s)
- Christin Brückner
- Chair of Genetics, Department of Biology, Friedrich Alexander University Erlangen Nürnberg (FAU), Germany
| | - Christian Lehmann
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Germany.,Medical Immunology Campus Erlangen, Germany
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, Friedrich Alexander University Erlangen Nürnberg (FAU), Germany.,Medical Immunology Campus Erlangen, Germany
| |
Collapse
|
32
|
|
33
|
Gudelj I, Lauc G, Pezer M. Immunoglobulin G glycosylation in aging and diseases. Cell Immunol 2018; 333:65-79. [DOI: 10.1016/j.cellimm.2018.07.009] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 01/01/2023]
|
34
|
Spirig R, Campbell IK, Koernig S, Chen CG, Lewis BJB, Butcher R, Muir I, Taylor S, Chia J, Leong D, Simmonds J, Scotney P, Schmidt P, Fabri L, Hofmann A, Jordi M, Spycher MO, Cattepoel S, Brasseit J, Panousis C, Rowe T, Branch DR, Baz Morelli A, Käsermann F, Zuercher AW. rIgG1 Fc Hexamer Inhibits Antibody-Mediated Autoimmune Disease via Effects on Complement and FcγRs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:2542-2553. [PMID: 29531170 PMCID: PMC5890536 DOI: 10.4049/jimmunol.1701171] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 02/13/2018] [Indexed: 12/27/2022]
Abstract
Activation of Fc receptors and complement by immune complexes is a common important pathogenic trigger in many autoimmune diseases and so blockade of these innate immune pathways may be an attractive target for treatment of immune complex-mediated pathomechanisms. High-dose IVIG is used to treat autoimmune and inflammatory diseases, and several studies demonstrate that the therapeutic effects of IVIG can be recapitulated with the Fc portion. Further, recent data indicate that recombinant multimerized Fc molecules exhibit potent anti-inflammatory properties. In this study, we investigated the biochemical and biological properties of an rFc hexamer (termed Fc-μTP-L309C) generated by fusion of the IgM μ-tailpiece to the C terminus of human IgG1 Fc. Fc-μTP-L309C bound FcγRs with high avidity and inhibited FcγR-mediated effector functions (Ab-dependent cell-mediated cytotoxicity, phagocytosis, respiratory burst) in vitro. In addition, Fc-μTP-L309C prevented full activation of the classical complement pathway by blocking C2 cleavage, avoiding generation of inflammatory downstream products (C5a or sC5b-9). In vivo, Fc-μTP-L309C suppressed inflammatory arthritis in mice when given therapeutically at approximately a 10-fold lower dose than IVIG, which was associated with reduced inflammatory cytokine production and complement activation. Likewise, administration of Fc-μTP-L309C restored platelet counts in a mouse model of immune thrombocytopenia. Our data demonstrate a potent anti-inflammatory effect of Fc-μTP-L309C in vitro and in vivo, likely mediated by blockade of FcγRs and its unique inhibition of complement activation.
Collapse
Affiliation(s)
| | - Ian K Campbell
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Sandra Koernig
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Chao-Guang Chen
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Bonnie J B Lewis
- Centre for Innovation Canadian Blood Services, Toronto, Ontario K1G 4J5, Canada; and
- Department of Medicine, University of Toronto, Toronto, Ontario M5G 2M1, Canada
| | - Rebecca Butcher
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Ineke Muir
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Shirley Taylor
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Jenny Chia
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - David Leong
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Jason Simmonds
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Pierre Scotney
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Peter Schmidt
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Louis Fabri
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | | | | | | | | | | | - Con Panousis
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Tony Rowe
- CSL Ltd., Bio21 Institute, Parkville, Victoria 3010, Australia
| | - Donald R Branch
- Centre for Innovation Canadian Blood Services, Toronto, Ontario K1G 4J5, Canada; and
- Department of Medicine, University of Toronto, Toronto, Ontario M5G 2M1, Canada
| | | | | | | |
Collapse
|
35
|
Jones MB. IgG and leukocytes: Targets of immunomodulatory α2,6 sialic acids. Cell Immunol 2018; 333:58-64. [PMID: 29685495 DOI: 10.1016/j.cellimm.2018.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/30/2018] [Indexed: 12/27/2022]
Abstract
ST6Gal1 is a critical sialyltransferase enzyme that controls the addition of α2,6-linked sialic acids to the termini of glycans. Attachment of sialic acids to glycoproteins as a posttranslational modification influences cellular responses, and is a well-known modifier of immune cell behavior. ST6Gal1 activity impacts processes such as: effector functions of immunoglobulin G via Fc sialylation, hematopoietic capacity by hematopoietic stem and progenitor cell surface sialylation, and lymphocyte activation thresholds though CD22 engagement and inhibition of galectins. This review summarizes recent studies that suggest α2,6 sialylation by ST6Gal1 has an immunoregulatory effect on immune reactions.
Collapse
Affiliation(s)
- Mark B Jones
- Case Western Reserve University, School of Medicine, Department of Pathology, Cleveland, OH 44106, United States.
| |
Collapse
|
36
|
Dekkers G, Rispens T, Vidarsson G. Novel Concepts of Altered Immunoglobulin G Galactosylation in Autoimmune Diseases. Front Immunol 2018; 9:553. [PMID: 29616041 PMCID: PMC5867308 DOI: 10.3389/fimmu.2018.00553] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/05/2018] [Indexed: 12/22/2022] Open
Abstract
The composition of the conserved N297 glycan in immunoglobulin G (IgG) has been shown to affect antibody effector functions via C1q of the complement system and Fc gamma receptors (FcγR) on immune cells. Changes in the general levels of IgG-glycoforms, such as lowered total IgG galactosylation observed in many autoimmune diseases have been associated with elevated disease severity. Agalactosyslated IgG has therefore been regarded and classified by many as pro-inflammatory. However, and somewhat counterintuitively, agalactosylation has been shown by several groups to decrease affinity for FcγRIII and decrease C1q binding and downstream activation, which seems at odds with this proposed pro-inflammatory nature. In this review, we discuss these circumstances where altered IgG galactosylation/glycosylation is found. We propose a novel model based on these observations and current biochemical evidence, where the levels of IgG galactosylation found in the total bulk IgG affect the threshold required to achieve immune activation by autoantibodies through either C1q or FcγR. Although this model needs experimental verification, it is supported by several clinical observations and reconciles apparent discrepancies in the literature, and suggests a general mechanism in IgG-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Gillian Dekkers
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands.,Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Department of Immunopathology, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| | - Gestur Vidarsson
- Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Centre, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
37
|
Finke JM, Banks WA. Modulators of IgG penetration through the blood-brain barrier: Implications for Alzheimer's disease immunotherapy. Hum Antibodies 2018; 25:131-146. [PMID: 28035915 DOI: 10.3233/hab-160306] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review serves to highlight approaches that may improve the access of antibody drugs to regions of the brain affected by Alzheimer's Disease. While previous antibody drugs have been unsuccessful in treating Alzheimer's disease, recent work demonstrates that Alzheimer's pathology can be modified if these drugs can penetrate the brain parenchyma with greater efficacy. Research in antibody blood-brain barrier drug delivery predominantly follows one of three distinct directions: (1) enhancing influx with reduced antibody size, addition of Trojan horse modules, or blood-brain barrier disruption; (2) modulating trancytotic equilibrium and/or kinetics of the neonatal Fc Receptor; and (3) manipulation of antibody glycan carbohydrate composition. In addition to these topics, recent studies are discussed that reveal a role of glycan sialic acid in suppressing antibody efflux from the brain.
Collapse
Affiliation(s)
- John M Finke
- Division of Sciences and Mathematics, Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, USA
| | - William A Banks
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.,Department of Geriatric Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
38
|
Mimura Y, Katoh T, Saldova R, O'Flaherty R, Izumi T, Mimura-Kimura Y, Utsunomiya T, Mizukami Y, Yamamoto K, Matsumoto T, Rudd PM. Glycosylation engineering of therapeutic IgG antibodies: challenges for the safety, functionality and efficacy. Protein Cell 2018; 9:47-62. [PMID: 28597152 PMCID: PMC5777974 DOI: 10.1007/s13238-017-0433-3] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/22/2017] [Indexed: 12/25/2022] Open
Abstract
Glycosylation of the Fc region of IgG has a profound impact on the safety and clinical efficacy of therapeutic antibodies. While the biantennary complex-type oligosaccharide attached to Asn297 of the Fc is essential for antibody effector functions, fucose and outer-arm sugars attached to the core heptasaccharide that generate structural heterogeneity (glycoforms) exhibit unique biological activities. Hence, efficient and quantitative glycan analysis techniques have been increasingly important for the development and quality control of therapeutic antibodies, and glycan profiles of the Fc are recognized as critical quality attributes. In the past decade our understanding of the influence of glycosylation on the structure/function of IgG-Fc has grown rapidly through X-ray crystallographic and nuclear magnetic resonance studies, which provides possibilities for the design of novel antibody therapeutics. Furthermore, the chemoenzymatic glycoengineering approach using endoglycosidase-based glycosynthases may facilitate the development of homogeneous IgG glycoforms with desirable functionality as next-generation therapeutic antibodies. Thus, the Fc glycans are fertile ground for the improvement of the safety, functionality, and efficacy of therapeutic IgG antibodies in the era of precision medicine.
Collapse
Affiliation(s)
- Yusuke Mimura
- Department of Clinical Research, NHO Yamaguchi-Ube Medical Center, 685 Higashi-Kiwa, Ube, 755-0241, Japan.
| | - Toshihiko Katoh
- Laboratory of Molecular Biology and Bioresponse, Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kitashirakawa, Oiwake-Cho, Sakyo-Ku, Kyoto, 606-8502, Japan
| | - Radka Saldova
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin 4, Ireland
| | - Roisin O'Flaherty
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin 4, Ireland
| | - Tomonori Izumi
- Center for Regenerative Medicine, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami Kogushi, Ube, 755-8505, Japan
| | - Yuka Mimura-Kimura
- Department of Clinical Research, NHO Yamaguchi-Ube Medical Center, 685 Higashi-Kiwa, Ube, 755-0241, Japan
| | - Toshiaki Utsunomiya
- Department of Clinical Research, NHO Yamaguchi-Ube Medical Center, 685 Higashi-Kiwa, Ube, 755-0241, Japan
| | - Yoichi Mizukami
- Center for Gene Research, Yamaguchi University, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Kenji Yamamoto
- Research Institute for Bioresources and Biotechnology, Ishikawa Prefectural University, 1-308 Suematsu, Nonoichi, Ishikawa, 921-8836, Japan
| | - Tsuneo Matsumoto
- Department of Clinical Research, NHO Yamaguchi-Ube Medical Center, 685 Higashi-Kiwa, Ube, 755-0241, Japan
| | - Pauline M Rudd
- NIBRT GlycoScience Group, National Institute for Bioprocessing Research and Training, Mount Merrion, Blackrock, Dublin 4, Ireland
| |
Collapse
|
39
|
Effects of Acyclovir and IVIG on Behavioral Outcomes after HSV1 CNS Infection. Behav Neurol 2017; 2017:5238402. [PMID: 29358844 PMCID: PMC5735307 DOI: 10.1155/2017/5238402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/06/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Herpes simplex virus 1 (HSV) encephalitis (HSE) has serious neurological complications, involving behavioral and cognitive impairments that cause significant morbidity and a reduced quality of life. We showed that HSE results from dysregulated central nervous system (CNS) inflammatory responses. We hypothesized that CNS inflammation is casually involved in behavioral abnormalities after HSE and that treatment with ACV and pooled human immunoglobulin (IVIG), an immunomodulatory drug, would improve outcomes compared to mice treated with phosphate buffered saline (PBS) or ACV alone. Anxiety levels were high in HSV-infected PBS and ACV-treated mice compared to mice treated with ACV + IVIG, consistent with reports implicating inflammation in anxiety induced by lipopolysaccharide (LPS) or stress. Female, but not male, PBS-treated mice were cognitively impaired, and unexpectedly, ACV was protective, while the inclusion of IVIG surprisingly antagonized ACV's beneficial effects. Distinct serum proteomic profiles were observed for male and female mice, and the antagonistic effects of ACV and IVIG on behavior were paralleled by similar changes in the serum proteome of ACV- and ACV + IVIG-treated mice. We conclude that inflammation and other factors mediate HSV-induced behavioral impairments and that the effects of ACV and IVIG on behavior involve novel mechanisms.
Collapse
|
40
|
Sordé L, Spindeldreher S, Palmer E, Karle A. Massive immune response against IVIg interferes with response against other antigens in mice: A new mode of action? PLoS One 2017; 12:e0186046. [PMID: 29023507 PMCID: PMC5638328 DOI: 10.1371/journal.pone.0186046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/25/2017] [Indexed: 11/19/2022] Open
Abstract
Administration of high dose intravenous immunoglobulin (IVIg) is widely used in the clinic to treat autoimmune and severe inflammatory diseases. However, its mechanisms of action remain poorly understood. We assessed the impact of IVIg on immune cell populations using an in vivo ovalbumin (Ova)-immunization mouse model. High dose IVIg significantly reduced the Ova-specific antibody response. Intriguingly, the results obtained indicate an immediate and massive immune reaction against IVIg, as shown by the activation and expansion of B cells and CD4+ T cells in the spleen and draining lymph nodes and the production of IVIg-specific antibodies. We propose that IVIg competes at the T-cell level with the response against Ova to explain the immunomodulatory properties of IVIg. Two monoclonal antibodies did not succeeded in reproducing the effects of IVIg. This suggests that in addition to the mouse response against human constant domains, the enormous sequence diversity of IVIg may significantly contribute to this massive immune response against IVIg. While correlation of these findings to IVIg-treated patients remains to be explored, our data demonstrate for the first time that IVIg re-directs the immune response towards IVIg and away from a specific antigen response.
Collapse
Affiliation(s)
- Laetitia Sordé
- Novartis Pharma AG, Integrated Biologics Profiling Unit, Immunogenicity Risk Assessment, Basel, Switzerland
| | - Sebastian Spindeldreher
- Novartis Institute for Biomedical Research, Drug Metabolism and Pharmacokinetics, Biologics, Basel, Switzerland
| | - Ed Palmer
- University Hospital Basel, Department of Biomedicine, Transplantation Immunology and Nephrology, Basel, Switzerland
| | - Anette Karle
- Novartis Pharma AG, Integrated Biologics Profiling Unit, Immunogenicity Risk Assessment, Basel, Switzerland
- * E-mail:
| |
Collapse
|
41
|
Seeling M, Brückner C, Nimmerjahn F. Differential antibody glycosylation in autoimmunity: sweet biomarker or modulator of disease activity? Nat Rev Rheumatol 2017; 13:621-630. [DOI: 10.1038/nrrheum.2017.146] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Kao D, Lux A, Schaffert A, Lang R, Altmann F, Nimmerjahn F. IgG subclass and vaccination stimulus determine changes in antigen specific antibody glycosylation in mice. Eur J Immunol 2017; 47:2070-2079. [PMID: 28771702 DOI: 10.1002/eji.201747208] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 06/30/2017] [Accepted: 08/01/2017] [Indexed: 11/11/2022]
Abstract
Immunoglobulin G (IgG) glycosylation can modulate antibody effector functions. Depending on the precise composition of the sugar moiety attached to individual IgG glycovariants either pro- or anti-inflammatory effector pathways can be initiated via differential binding to type I or type II Fc-receptors. However, an in depth understanding of how individual IgG subclasses are glycosylated during the steady state and how their glycosylation pattern changes during vaccination is missing. To monitor IgG subclass glycosylation during the steady state and upon vaccination of mice with different T-cell dependent and independent antigens, tryptic digests of serum, and antigen-specific IgG preparations were analyzed by reversed phase-liquid chromatography-mass spectrometry. We show that there is a remarkable difference with respect to how individual IgG subclasses are glycosylated during the steady state. More importantly, upon T-cell dependent and independent vaccinations, individual antigen-specific IgG subclasses reacted differently with respect to changes in individual glycoforms, suggesting that the IgG subclass itself is a major determinant of restricting or allowing alterations in specific IgG glycovariants.
Collapse
Affiliation(s)
- Daniela Kao
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Lux
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Schaffert
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Roland Lang
- Chair of Microbiology and Infection Immunology, University Hospital Erlangen, Erlangen, Germany
| | - Friedrich Altmann
- Department of Biochemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
43
|
Schneider C, Wicki S, Graeter S, Timcheva TM, Keller CW, Quast I, Leontyev D, Djoumerska-Alexieva IK, Käsermann F, Jakob SM, Dimitrova PA, Branch DR, Cummings RD, Lünemann JD, Kaufmann T, Simon HU, von Gunten S. IVIG regulates the survival of human but not mouse neutrophils. Sci Rep 2017; 7:1296. [PMID: 28465620 PMCID: PMC5430961 DOI: 10.1038/s41598-017-01404-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) are purified IgG preparations made from the pooled plasma from thousands of healthy donors and are being tested in preclinical mouse models. Inherent challenges, however, are the pluripotency of IVIG and its xenogeneicity in animals. IVIG can alter the viability of human neutrophils via agonistic antibodies to Fas and Siglec-9. In this study, we compared the effects of IVIG on human and mouse neutrophils using different death assays. Different commercial IVIG preparations similarly induced cytokine-dependent death in human neutrophils, whereas they had no effects on the survival of either peripheral blood or bone marrow neutrophils from C57BL/6 or BALB/c mice. F(ab’)2 but not Fc fragments of IVIG induced death of human neutrophils, whereas neither of these IVIG fragments, nor agonistic monoclonal antibodies to human Fas or Siglec-9 affected the viability of mouse neutrophils. Pooled mouse IgG, which exhibited a different immunoprofile compared to IVIG, also had no effect on mouse cells. Together, these observations demonstrate that effects of IVIG on neutrophil survival are not adequately reflected in current mouse models, despite the key role of these cells in human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
| | - Simone Wicki
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stefanie Graeter
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | - Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Isaak Quast
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Danila Leontyev
- Department of Medicine, University of Toronto and Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Iglika K Djoumerska-Alexieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Stephan M Jakob
- Department of Intensive Care Medicine, University Hospital Bern (Inselspital), University of Bern, Bern, Switzerland
| | - Petya A Dimitrova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Donald R Branch
- Department of Medicine, University of Toronto and Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
44
|
Abstract
Taking advantage of the "World Apheresis Association/Société Française d'Hémaphérèse" meeting held in Paris in April 2016, this article reviews the current knowledge on the mechanisms of action of intravenous immunoglobulins. Immunoglobulins are a plasma-derived drug, which have been initially used as a replacement therapy for patients with antibody deficiency. Since 1980 they have also been used for their anti-inflammatory and immunomodulating efficacy in auto-immune diseases. Herein, we review the requirements for their production and composition before giving a specific attention to their mechanisms of action including substitution and immunomodulation.
Collapse
Affiliation(s)
- Benjamin Chaigne
- Université Paris Descartes, Faculté de Médecine, Service de Médecine Interne, Centre de référence pour les vascularites nécrosantes et la sclérodermie systémique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Paris, France
| | - Luc Mouthon
- Université Paris Descartes, Faculté de Médecine, Service de Médecine Interne, Centre de référence pour les vascularites nécrosantes et la sclérodermie systémique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Paris, France.
| |
Collapse
|
45
|
Catera M, Borelli V, Malagolini N, Chiricolo M, Venturi G, Reis CA, Osorio H, Abruzzo PM, Capri M, Monti D, Ostan R, Franceschi C, Dall'Olio F. Identification of novel plasma glycosylation-associated markers of aging. Oncotarget 2016; 7:7455-68. [PMID: 26840264 PMCID: PMC4884931 DOI: 10.18632/oncotarget.7059] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/23/2016] [Indexed: 12/14/2022] Open
Abstract
The pro- or anti-inflammatory activities of immunoglobulins G (IgGs) are controlled by the structure of the glycan N-linked to Asn297 of their heavy chain. The age-associated low grade inflammation (inflammaging) is associated with increased plasmatic levels of agalactosylated IgGs terminating with N-acetylglucosamine (IgG-G0) whose biogenesis has not been fully explained. Although the biosynthesis of glycans is in general mediated by glycosyltransferases associated with internal cell membranes, the extracellular glycosylation of circulating glycoproteins mediated by plasmatic glycosyltransferases has been recently demonstrated. In this study we have investigated the relationship between plasmatic glycosyltransferases, IgG glycosylation and inflammatory and aging markers. In cohorts of individuals ranging from infancy to centenarians we determined the activity of plasmatic β4 galactosyltransferase(s) (B4GALTs) and of α2,6-sialyltransferase ST6GAL1, the glycosylation of IgG, the GlycoAge test (a glycosylation-based marker of aging) and the plasma level of inflammatory and liver damage markers. Our results show that: 1) plasmatic B4GALTs activity is a new marker of aging, showing a linear increase throughout the whole age range. 2) plasmatic ST6GAL1 was high only in children and in people above 80, showing a quadratic relationship with age. 3) Neither plasmatic glycosyltransferase correlated with markers of liver damage. 4) plasmatic ST6GAL1 showed a positive association with acute phase proteins in offspring of short lived parents, but not in centenarians or in their offspring. 5) Although the glycosylation of IgGs was not correlated with the level of the two plasmatic glycosyltransferases, it showed progressive age-associated changes consistent with a shift toward a pro-inflammatory glycotype.
Collapse
Affiliation(s)
- Mariangela Catera
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Vincenzo Borelli
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Nadia Malagolini
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Mariella Chiricolo
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Giulia Venturi
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, and Institute of Molecular Pathology and Immunology of The University of Porto IPATIMUP), Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Faculty of Medicine of The University of Porto, Al. Prof. Hernâni Monteiro, Porto, Portugal
| | - Hugo Osorio
- Instituto de Investigação e Inovação em Saúde (Institute for Research and Innovation in Health), University of Porto, and Institute of Molecular Pathology and Immunology of The University of Porto IPATIMUP), Porto, Portugal.,Faculty of Medicine of The University of Porto, Al. Prof. Hernâni Monteiro, Porto, Portugal
| | - Provvidenza M Abruzzo
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Miriam Capri
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Daniela Monti
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", University of Florence, Florence, Italy
| | - Rita Ostan
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| | - Fabio Dall'Olio
- Dipartimento di Medicina Specialistica Diagnostica e Sperimentale (DIMES) University of Bologna, Bologna, Italy
| |
Collapse
|
46
|
Späth PJ, Schneider C, von Gunten S. Clinical Use and Therapeutic Potential of IVIG/SCIG, Plasma-Derived IgA or IgM, and Other Alternative Immunoglobulin Preparations. Arch Immunol Ther Exp (Warsz) 2016; 65:215-231. [DOI: 10.1007/s00005-016-0422-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022]
|
47
|
Le NPL, Bowden TA, Struwe WB, Crispin M. Immune recruitment or suppression by glycan engineering of endogenous and therapeutic antibodies. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1860:1655-68. [PMID: 27105835 PMCID: PMC4922387 DOI: 10.1016/j.bbagen.2016.04.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/15/2016] [Accepted: 04/16/2016] [Indexed: 12/20/2022]
Abstract
Human serum IgG contains multiple glycoforms which exhibit a range of binding properties to effector molecules such as cellular Fc receptors. Emerging knowledge of how the Fc glycans contribute to the antibody structure and effector functions has opened new avenues for the exploitation of defined antibody glycoforms in the treatment of diseases. Here, we review the structure and activity of antibody glycoforms and highlight developments in antibody glycoengineering by both the manipulation of the cellular glycosylation machinery and by chemoenzymatic synthesis. We discuss wide ranging applications of antibody glycoengineering in the treatment of cancer, autoimmunity and inflammation. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.
Collapse
Affiliation(s)
- Ngoc Phuong Lan Le
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | - Thomas A Bowden
- Division of Structural Biology, University of Oxford, Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Weston B Struwe
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | - Max Crispin
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom.
| |
Collapse
|
48
|
van de Bovenkamp FS, Hafkenscheid L, Rispens T, Rombouts Y. The Emerging Importance of IgG Fab Glycosylation in Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:1435-41. [PMID: 26851295 DOI: 10.4049/jimmunol.1502136] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human IgG is the most abundant glycoprotein in serum and is crucial for protective immunity. In addition to conserved IgG Fc glycans, ∼15-25% of serum IgG contains glycans within the variable domains. These so-called "Fab glycans" are primarily highly processed complex-type biantennary N-glycans linked to N-glycosylation sites that emerge during somatic hypermutation. Specific patterns of Fab glycosylation are concurrent with physiological and pathological conditions, such as pregnancy and rheumatoid arthritis. With respect to function, Fab glycosylation can significantly affect stability, half-life, and binding characteristics of Abs and BCRs. Moreover, Fab glycans are associated with the anti-inflammatory activity of IVIgs. Consequently, IgG Fab glycosylation appears to be an important, yet poorly understood, process that modulates immunity.
Collapse
Affiliation(s)
- Fleur S van de Bovenkamp
- Department of Immunopathology, Sanquin Research, 1066 CX Amsterdam, the Netherlands; Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Lise Hafkenscheid
- Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, 1066 CX Amsterdam, the Netherlands; Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands;
| | - Yoann Rombouts
- Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and Université Lille, CNRS, UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, F 59 000 Lille, France
| |
Collapse
|
49
|
Ohmi Y, Ise W, Harazono A, Takakura D, Fukuyama H, Baba Y, Narazaki M, Shoda H, Takahashi N, Ohkawa Y, Ji S, Sugiyama F, Fujio K, Kumanogoh A, Yamamoto K, Kawasaki N, Kurosaki T, Takahashi Y, Furukawa K. Sialylation converts arthritogenic IgG into inhibitors of collagen-induced arthritis. Nat Commun 2016; 7:11205. [PMID: 27046227 PMCID: PMC4822049 DOI: 10.1038/ncomms11205] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/02/2016] [Indexed: 01/08/2023] Open
Abstract
Rheumatoid arthritis (RA)-associated IgG antibodies such as anti-citrullinated protein antibodies (ACPAs) have diverse glycosylation variants; however, key sugar chains modulating the arthritogenic activity of IgG remain to be clarified. Here, we show that reduced sialylation is a common feature of RA-associated IgG in humans and in mouse models of arthritis. Genetically blocking sialylation in activated B cells results in exacerbation of joint inflammation in a collagen-induced arthritis (CIA) model. On the other hand, artificial sialylation of anti-type II collagen antibodies, including ACPAs, not only attenuates arthritogenic activity, but also suppresses the development of CIA in the antibody-infused mice, whereas sialylation of other IgG does not prevent CIA. Thus, our data demonstrate that sialylation levels control the arthritogenicity of RA-associated IgG, presenting a potential target for antigen-specific immunotherapy.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Autoantibodies/chemistry
- Autoantibodies/immunology
- Autoantibodies/metabolism
- Carbohydrate Sequence
- Collagen Type II/immunology
- Collagen Type II/metabolism
- Humans
- Immunoglobulin G/chemistry
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Molecular Sequence Data
- Protein Processing, Post-Translational
- Sialic Acids/immunology
- Sialic Acids/metabolism
Collapse
Affiliation(s)
- Yuhsuke Ohmi
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Wataru Ise
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center and Graduate School of Frontier Biosciences, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Akira Harazono
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Daisuke Takakura
- Laboratory of Proteome Science, Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Hidehiro Fukuyama
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yoshihiro Baba
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center and Graduate School of Frontier Biosciences, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nobunori Takahashi
- Department of Orthopedics, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
| | - Yuki Ohkawa
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, 1200 Matsumoto, Kasugai 487-8501, Japan
| | - Shuting Ji
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Ten-no-dai, Tsukuba 305-8575, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Nana Kawasaki
- Division of Biological Chemistry and Biologicals, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
- Laboratory of Proteome Science, Graduate School of Medical Life Science, Yokohama City University, 1-7-29, Suehiro, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center and Graduate School of Frontier Biosciences, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehirocho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Koichi Furukawa
- Department of Biochemistry II, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-0065, Japan
- Department of Biomedical Sciences, Chubu University College of Life and Health Sciences, 1200 Matsumoto, Kasugai 487-8501, Japan
| |
Collapse
|
50
|
Crow AR, Lazarus AH. Mechanistic properties of intravenous immunoglobulin in murine immune thrombocytopenia: support for FcγRIIB falls by the wayside. Semin Hematol 2016; 53 Suppl 1:S20-2. [DOI: 10.1053/j.seminhematol.2016.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|