1
|
Nimmerjahn F. Role of Antibody Glycosylation in Health, Disease, and Therapy. Handb Exp Pharmacol 2025. [PMID: 40119204 DOI: 10.1007/164_2025_744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Immunoglobulin G (IgG) antibodies are an essential component of humoral immunity protecting the host from recurrent infections. Among all antibody isotypes, IgG antibodies have a uniquely long half-life, can basically reach any tissue in the body, and have the ability to kill opsonized target cells, which has made them the molecule of choice for therapeutic interventions in cancer and autoimmunity. Moreover, IgG antibodies in the form of pooled serum IgG preparations from healthy donors are used to treat chronic inflammatory and autoimmune diseases, providing evidence that serum IgG antibodies can have an active immunomodulatory activity. Research over the last two decades has established that the single sugar moiety attached to each IgG heavy chain plays a very important role in modulating the pro- and anti-inflammatory activities of IgG. Moreover, specific sugar moieties such as sialic acid and galactose residues can serve as highly specific biomarkers for ongoing inflammatory processes. This chapter will summarize how different sugar residues in the IgG sugar moiety change upon inflammation and how such changes may translate to altered IgG function and hence maybe useful for optimizing or modulating the function of therapeutic antibodies.
Collapse
Affiliation(s)
- Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
2
|
Edwards DL, Huang M, Wang TT. Soluble Factors and Mechanisms Regulated by Sialylated IgG Signaling. Immunol Rev 2025; 330:e70021. [PMID: 40084926 DOI: 10.1111/imr.70021] [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: 11/01/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Inflammation is a complex biological response that can be both induced and actively suppressed by IgG-Fc gamma receptor (FcγR) interactions. This review explores the role of IgG sialylation in reducing or blocking inflammatory responses. We first revisit foundational studies that established the anti-inflammatory properties of sialylated IgG1 Fc. These early investigations revealed that the sialylated fraction is crucial for intravenous immunoglobulin's (IVIg's) ability to reduce inflammation in many autoinflammatory diseases and defined a paracrine signaling mechanism underlying this activity. Next, we discuss a recently identified mechanism whereby sialylated IgG directly induces RE1-Silencing Transcription Factor (REST) which functions as a transcriptional repressor of NF-κB1. This mechanism suggests a very broad role for sialylated IgG signaling in inflammation control since NF-κB is a central mediator of responses downstream of diverse activating receptors on both adaptive and innate immune cells. Finally, we review a set of soluble factors that are suppressed by sialylated IgG signaling in the murine airway and in purified human macrophages, providing additional insight into mechanisms by which sialylated IgG contributes to broad inflammatory control.
Collapse
Affiliation(s)
- Desmond L Edwards
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - Min Huang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
| | - Taia T Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA
- Department of Medicine, Division of Infectious Diseases, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
3
|
Lünemann JD. Moving beyond immunoglobulin therapy for CIDP with efgartigimod. Nat Rev Neurol 2025; 21:1-2. [PMID: 39609632 DOI: 10.1038/s41582-024-01045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Affiliation(s)
- Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University and University Hospital of Münster, Münster, Germany.
| |
Collapse
|
4
|
Du Y, Yan Q, Li C, Zhu W, Zhao C, Hao Y, Li L, Yao D, Zhou X, Li Y, Dang Y, Zhang R, Han L, Wang Y, Hou T, Li J, Li H, Jiang P, Wang P, Chen F, Zhu T, Liu J, Liu S, Gao L, Zhao Y, Zhang W. Efficacy and safety of combined low-dose rituximab regimen for chronic inflammatory demyelinating polyradiculoneuropathy. Ann Clin Transl Neurol 2025; 12:180-191. [PMID: 39660535 PMCID: PMC11752089 DOI: 10.1002/acn3.52270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE To determine the efficacy and safety of combined low-dose rituximab with conventional therapy for chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) treatment. METHODS Total 73 patients with CIDP were enrolled for the retrospective cohort study, and divided into conventional first-line therapy cohort (n = 40) and combined low-dose rituximab (100 mg per infusion) cohort (n = 33). The outcome measures include scores of I-RODS, mRS, INCAT, ONLS, TSS, and COMPASS 31 scale at baseline and regular four visits (4, 16, 28, and 52 weeks), as well as proportion of favorable response and outcome, corticosteroids dosage, and deterioration occurrence during follow-up. RESULTS Compared to conventional therapy cohort, combined rituximab cohort presented better improvements and higher proportion of favorable response in scales assessments at each visit, as well as significantly reduced corticosteroids dosage and deterioration occurrence during the follow-up. Analyses of subgroups showed better improvements in both typical CIDP and CIDP variants in combined rituximab cohort than those in conventional therapy cohort, but had no differences between each other. Early initiating combined rituximab regimen (<10 weeks) showed better improvements than delayed initiation (≥10 weeks) at the first three visits within 28 weeks, while had no difference in favorable prognoses at the last visit of 52 weeks after once reinfusion. No rituximab correlated serious adverse events were reported in our patients. INTERPRETATION Our simplified regimen of combined low-dose rituximab has been firstly demonstrated for the better efficacy and safety than conventional therapy in CIDP treatment.
Collapse
Affiliation(s)
- Ying Du
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Qi Yan
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Chuan Li
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Wenping Zhu
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
- Xi'an Medical UniversityXi'an710021ShaanxiChina
| | - Chao Zhao
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Yunfeng Hao
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Lin Li
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Dan Yao
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Xuan Zhou
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Ying Li
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Yuting Dang
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Rong Zhang
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Lin Han
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Yuanyuan Wang
- Department of Internal MedicineQianxian Traditional Chinese Medicine HospitalXianyang713300ShaanxiChina
| | - Tao Hou
- Department of NeurologyFuping County HospitalWeinan711700ShaanxiChina
| | - Juan Li
- Department of NeurologyLantian Country People's HospitalXi'an710500ShaanxiChina
| | - Hailin Li
- Department of NeurologyPingli County HospitalAnkang725500ShaanxiChina
| | - Panpan Jiang
- Department of NeurologyThe Second Hospital of WeinanWeinan711700ShaanxiChina
| | - Pei Wang
- Department of Internal MedicineBaishui County HospitalWeinan715600ShaanxiChina
| | - Fenying Chen
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Tingge Zhu
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| | - Juntong Liu
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
- Xi'an Medical UniversityXi'an710021ShaanxiChina
| | - Shuyu Liu
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
- Xi'an Medical UniversityXi'an710021ShaanxiChina
| | - Lan Gao
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
- Xi'an Medical UniversityXi'an710021ShaanxiChina
| | - Yingjun Zhao
- Department of Neurology and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, Institute of Neuroscience, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, School of MedicineXiamen UniversityXiamen361005FujianChina
| | - Wei Zhang
- Department of Neurology, Tangdu HospitalFourth Military Medical UniversityXi'an710038ShaanxiChina
| |
Collapse
|
5
|
Hematianlarki M, Nimmerjahn F. Immunomodulatory and anti-inflammatory properties of immunoglobulin G antibodies. Immunol Rev 2024; 328:372-386. [PMID: 39340138 PMCID: PMC11659946 DOI: 10.1111/imr.13404] [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: 09/30/2024]
Abstract
Antibodies provide an essential layer of protection from infection and reinfection with microbial pathogens. An impaired ability to produce antibodies results in immunodeficiency and necessitates the constant substitution with pooled serum antibodies from healthy donors. Among the five antibody isotypes in humans and mice, immunoglobulin G (IgG) antibodies are the most potent anti-microbial antibody isotype due to their long half-life, their ability to penetrate almost all tissues and due to their ability to trigger a wide variety of effector functions. Of note, individuals suffering from IgG deficiency frequently produce self-reactive antibodies, suggesting that a normal serum IgG level also may contribute to maintaining self-tolerance. Indeed, the substitution of immunodeficient patients with pooled serum IgG fractions from healthy donors, also referred to as intravenous immunoglobulin G (IVIg) therapy, not only protects the patient from infection but also diminishes autoantibody induced pathology, providing more direct evidence that IgG antibodies play an active role in maintaining tolerance during the steady state and during resolution of inflammation. The aim of this review is to discuss different conceptual models that may explain how serum IgG or IVIg can contribute to maintaining a balanced immune response. We will focus on pathways depending on the IgG fragment crystallizable (Fc) as pre-clinical data in various mouse model systems as well as human clinical data have demonstrated that the IgG Fc-domain recapitulates the ability of intact IVIg with respect to its ability to trigger resolution of inflammation. We will further discuss how the findings already have or are in the process of being translated to novel therapeutic approaches to substitute IVIg in treating autoimmune inflammation.
Collapse
Affiliation(s)
- Marjan Hematianlarki
- Division of Genetics, Department of BiologyFriedrich Alexander University Erlangen‐NürnbergErlangenGermany
| | - Falk Nimmerjahn
- Division of Genetics, Department of BiologyFriedrich Alexander University Erlangen‐NürnbergErlangenGermany
| |
Collapse
|
6
|
Jandus C, Jandus P. Effects of Intravenous Immunoglobulins on Human Innate Immune Cells: Collegium Internationale Allergologicum Update 2024. Int Arch Allergy Immunol 2024; 185:975-996. [PMID: 38852585 DOI: 10.1159/000539069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/22/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Intravenous immunoglobulin (IVIg) has been used for almost 40 years in the treatment of autoimmune and systemic inflammatory diseases. Numerous cells are involved in the innate immune response, including monocytes/macrophages, neutrophils, dendritic cells, mast cells, basophils, eosinophils, natural killer cells, and innate lymphoid cells. Many studies have investigated the mechanisms by which IVIg down-modulates inflammatory and autoimmune processes of innate immune cells. However, questions remain regarding the precise mechanism of action in autoimmune or inflammatory conditions. The aim of this work was to review the immunomodulatory effect of IVIg on only human innate immune cells. A narrative review approach was chosen to summarize key evidence on the immunomodulatory effects of commercially available and unmodified IVIg on human innate immune cells. SUMMARY Numerous different immunomodulatory effects of IVIg have been reported, with some very different effects depending on the immune cell type and disease. Several limitations of the different studies were identified. Of the 77 studies identified and reviewed, 29 (37.7%) dealt with autoimmune or inflammatory diseases. Otherwise, the immunomodulatory effects of IVIg were studied only in healthy donors using an in vitro experimental approach. Some of the documented effects showed disease-specific effects, such as in Kawasaki disease. Various methodological limitations have also been identified that may reduce the validity of some studies. KEY MESSAGE As further insights have been gained into the various inflammatory cascades activated in immunological diseases, interesting insights have also been gained into the mechanism of action of IVIg. We are still far from discovering all the immunomodulatory mechanisms of IVIg.
Collapse
Affiliation(s)
- Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Centre in Onco-Haematology (CRTOH), Geneva, Switzerland
| | - Peter Jandus
- Division of Immunology and Allergology, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
7
|
Ozdag Acarli AN, Tuzun E, Sanli E, Koral G, Akbayir E, Cakar A, Sirin NG, Soysal A, Aysal F, Durmus H, Parman Y, Yilmaz V. Disease activity in chronic inflammatory demyelinating polyneuropathy: association between circulating B-cell subsets, cytokine levels, and clinical outcomes. Clin Exp Immunol 2024; 215:65-78. [PMID: 37638717 PMCID: PMC10776240 DOI: 10.1093/cei/uxad103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP), a common and treatable autoimmune neuropathy, is frequently misdiagnosed. The aim of this study is to evaluate the relationship between immunological markers and clinical outcome measures in a mixed cohort of patients with typical CIDP and CIDP variants at different disease stages. Twenty-three typical, 16 multifocal and five distal CIDP patients were included. Twenty-five sex and age-matched healthy controls and 12 patients with Charcot-Marie-Tooth type 1A (CMT1A) disease served as controls. Peripheral B-cell populations were analyzed by flow cytometry. IL6, IL10, TNFA mRNA and mir-21, mir-146a, and mir-155-5p expression levels were evaluated by real-time polymerase chain reaction in peripheral blood mononuclear cells (PBMC) and/or skin biopsy specimens. Results were then assessed for a possible association with clinical disability scores and intraepidermal nerve fiber densities (IENFD) in the distal leg. We detected a significant reduction in naive B cells (P ≤ 0.001), plasma cells (P ≤ 0.001) and regulatory B cells (P < 0.05), and an elevation in switched memory B cells (P ≤ 0.001) in CIDP compared to healthy controls. CMT1A and CIDP patients had comparable B-cell subset distribution. CIDP cases had significantly higher TNFA and IL10 gene expression levels in PBMC compared to healthy controls (P < 0.05 and P ≤ 0.01, respectively). IENFDs in the distal leg showed a moderate negative correlation with switched memory B-cell ratios (r = -0.51, P < 0.05) and a moderate positive correlation with plasma cell ratios (r = 0.46, P < 0.05). INCAT sum scores showed a moderate positive correlation with IL6 gene expression levels in PBMC (r = 0.54, P < 0.05). Altered B-cell homeostasis and IL10 and TNFA gene expression levels imply chronic antigen exposure and overactivity in the humoral immune system, and seem to be a common pathological pathway in both typical CIDP and CIDP variants.
Collapse
Affiliation(s)
- Ayse Nur Ozdag Acarli
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Erdem Tuzun
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Elif Sanli
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Gizem Koral
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ece Akbayir
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Arman Cakar
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nermin Gorkem Sirin
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- Department of Neurology, Bakirkoy Mazhar Osman Mental Health and Neurological Diseases Education and Research Hospital, Istanbul, Turkey
| | - Aysun Soysal
- Department of Neurology, Bakirkoy Mazhar Osman Mental Health and Neurological Diseases Education and Research Hospital, Istanbul, Turkey
| | - Fikret Aysal
- Department of Neurology, Bakirkoy Mazhar Osman Mental Health and Neurological Diseases Education and Research Hospital, Istanbul, Turkey
| | - Hacer Durmus
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yesim Parman
- Neuromuscular Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Vuslat Yilmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
8
|
Svačina MKR, Meißner A, Schweitzer F, Ladwig A, Pitarokoili K, Kofler DM, Sprenger‐Svačina A, Schneider C, Kohle F, Klein I, Wüstenberg H, Lehmann HC. Immunomodulatory effects of intravenous and subcutaneous immunoglobulin in chronic inflammatory demyelinating polyneuropathy: An observational study. Eur J Neurol 2024; 31:e16079. [PMID: 37789648 PMCID: PMC11235934 DOI: 10.1111/ene.16079] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/23/2023] [Accepted: 09/14/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND AND PURPOSE It is not known whether the route of administration affects the mechanisms of action of therapeutic immunoglobulin in chronic inflammatory demyelinating polyneuropathy (CIDP). The aim of this study, therefore, was to compare the immunomodulatory effects of intravenous (IVIg) and subcutaneous immunoglobulin (SCIg) in patients with CIDP and in IVIg-treated common variable immunodeficiency (CVID) patients. METHODS Serum and peripheral blood mononuclear cell samples were obtained from 30 CIDP patients receiving IVIg, 10 CIDP patients receiving SCIg, and 15 patients with CVID receiving IVIg. Samples and clinical data were obtained prior to IVIg/SCIg and at 3 days, 7 days, and, in CIDP patients receiving IVIg, 21 days post-administration. Serum cytokines were assessed by Luminex-based multiplex assay and enzyme-linked immunosorbent assay. Immune cells were characterized by flow cytometry. RESULTS Immune cell profiles of CIDP and CVID patients differed in frequencies of myeloid dendritic cells and cytotoxic natural killer cells. During treatment with IVIg or SCIg in CIDP patients, cellular immunomarkers were largely similar. CIDP patients receiving IVIg had higher macrophage inflammatory protein (MIP)-1α (p = 0.01), interleukin (IL)-4 (p = 0.04), and IL-33 (p = 0.04) levels than SCIg recipients. IVIg treatment more broadly modulated cytokines in CIDP than SCIg treatment. CONCLUSIONS Our study demonstrates that the modulation of cellular immunomarkers in CIDP is independent of the application route of therapeutic immunoglobulin. Minor differences were observed between CIDP and CVID patients. In contrast, cytokines were differentially modulated by IVIg and SCIg in CIDP.
Collapse
Affiliation(s)
- Martin K. R. Svačina
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Anika Meißner
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Finja Schweitzer
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Anne Ladwig
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | | | - David M. Kofler
- Medical Clinic I, Department of Immunology and RheumatologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Alina Sprenger‐Svačina
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Christian Schneider
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
- Department of NeurologySt. Katharinen HospitalFrechenGermany
| | - Felix Kohle
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Ines Klein
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Hauke Wüstenberg
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
| | - Helmar C. Lehmann
- Department of NeurologyFaculty of Medicine, University Hospital of CologneCologneGermany
- Department of NeurologyClinic of Leverkusen gGmbHLeverkusenGermany
| |
Collapse
|
9
|
Svačina MKR, Sprenger-Svačina A, Tsakmaklis A, Rüb AM, Klein I, Wüstenberg H, Fink GR, Lehmann HC, Vehreschild MJGT, Farowski F. The gut microbiome in intravenous immunoglobulin-treated chronic inflammatory demyelinating polyneuropathy. Eur J Neurol 2023; 30:3551-3556. [PMID: 36651357 DOI: 10.1111/ene.15679] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND PURPOSE The gut microbiome is involved in autoimmunity. Data on its composition in chronic inflammatory demyelinating polyneuropathy (CIDP), the most common chronic autoimmune disorder of peripheral nerves, are currently lacking. METHODS In this monocentric exploratory pilot study, stool samples were prospectively collected from 16 CIDP patients (mean age 58 ± 10 years, 25% female) before and 1 week after administration of intravenous immunoglobulin (IVIg). Gut microbiota were analyzed via bacterial 16S rRNA gene sequencing and compared to 15 age-matched healthy subjects (mean age 59 ± 15 years, 66% female). RESULTS The gut microbiota of CIDP patients showed an increased alpha-diversity (p = 0.005) and enrichment of Firmicutes, such as Blautia (p = 0.0004), Eubacterium hallii (p = 0.0004), or Ruminococcus torques (p = 0.03), and of Actinobacteriota (p = 0.03) compared to healthy subjects. IVIg administration did not alter the gut microbiome composition in CIDP in this short-term observation (p = 0.95). CONCLUSIONS The gut microbiome in IVIg-treated CIDP shows distinct features, with increased bacterial diversity and enrichment of short-chain fatty acid producing Firmicutes. IVIg had no short-term impact on the gut microbiome in CIDP patients. As the main limitation of this exploratory pilot study was small cohort size, future studies also including therapy-naïve patients are warranted to verify our findings and to explore the impact of long-term IVIg treatment on the gut microbiome in CIDP.
Collapse
Affiliation(s)
- Martin K R Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Alina Sprenger-Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Anastasia Tsakmaklis
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Alina M Rüb
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Ines Klein
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Hauke Wüstenberg
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Gereon R Fink
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
- Cognitive Neuroscience, Research Center Juelich, Institute of Neuroscience and Medicine (INM-3), Juelich, Germany
| | - Helmar C Lehmann
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Maria J G T Vehreschild
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
- Department of Internal Medicine II, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| | - Fedja Farowski
- Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
- Department of Internal Medicine II, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Braunschweig, Germany
| |
Collapse
|
10
|
Beneduce C, Nguyen S, Washburn N, Schaeck J, Meccariello R, Holte K, Ortiz D, Manning AM, Bosques CJ, Kurtagic E. Inhibitory Fc-Gamma IIb Receptor Signaling Induced by Multivalent IgG-Fc Is Dependent on Sialylation. Cells 2023; 12:2130. [PMID: 37681862 PMCID: PMC10486564 DOI: 10.3390/cells12172130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023] Open
Abstract
Immunoglobulin (IgG) Fc glycosylation has been shown to be important for the biological activity of antibodies. Fc sialylation is important for the anti-inflammatory activity of IgGs. However, evaluating the structure-activity relationship (SAR) of antibody Fc glycosylation has been hindered using simplified in vitro models in which antibodies are often displayed in monomeric forms. Presenting antibodies in monomeric forms may not accurately replicate the natural environment of the antibodies when binding their antigen in vivo. To address these limitations, we used different Fc-containing molecules, displaying their Fc domains in monovalent and multivalent fashion. Given the inhibitory role of Fc gamma receptor IIb (FcγRIIb) in autoimmune and inflammatory diseases, we focused on evaluating the impact of Fc sialylation on the activation of FcγRIIb. We report for the first time that in human cellular systems, sialic acid mediates the induction of FcγRIIb phosphorylation by IgG-Fc when the IgG-Fc is displayed in a multivalent fashion. This effect was observed with different types of therapeutic agents such as sialylated anti-TNFα antibodies, sialylated IVIg and sialylated recombinant multivalent Fc products. These studies represent the first report of the specific effects of Fc sialylation on FcγRIIb signaling on human immune cells and may help in the characterization of the anti-inflammatory activity of Fc-containing therapeutic candidates.
Collapse
Affiliation(s)
- Christopher Beneduce
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | | | - Nathaniel Washburn
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | - John Schaeck
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | - Robin Meccariello
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| | | | - Daniel Ortiz
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
| | | | | | - Elma Kurtagic
- Momenta Pharmaceuticals Inc., Cambridge, MA 02142, USA
- Janssen Research & Development, Cambridge, MA 02142, USA
| |
Collapse
|
11
|
Svačina MKR, Meißner A, Schweitzer F, Sprenger-Svačina A, Klein I, Wüstenberg H, Kohle F, Walter HL, Schroeter M, Lehmann HC. CIDP: Analysis of Immunomarkers During COVID-19 mRNA-Vaccination and IVIg-Immunomodulation: An Exploratory Study. J Neuroimmune Pharmacol 2023; 18:208-214. [PMID: 36929282 PMCID: PMC10018581 DOI: 10.1007/s11481-023-10058-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/16/2023] [Indexed: 03/18/2023]
Abstract
Availability of COVID-19 mRNA vaccine for patients with chronic inflammatory demyelinating polyneuropathy (CIDP) treated with intravenous immunoglobulin (IVIg) raises the question of whether COVID-19 mRNA vaccine influences disease activity or IVIg-mediated immunomodulation in CIDP. In this exploratory study, blood samples of CIDP patients on IVIg treatment were longitudinally analyzed before and after vaccination with a COVID-19 mRNA vaccine. A total of 44 samples of eleven patients were characterized at four timepoints by ELISA and flow cytometry in terms of immunomarkers for disease activity and IVIg-immunomodulation. Apart from a significantly lower expression of CD32b on naïve B cells after vaccination, no significant alteration of immunomarkers for CIDP or IVIg-mediated immunomodulation was observed. Our exploratory study suggests that COVID-19 mRNA vaccine does not have a relevant impact on immune activity in CIDP. In addition, immunomodulatory effects of IVIg in CIDP are not altered by COVID-19 mRNA vaccine. This study was registered in the German clinical trial register (DRKS00025759). Overview over the study design. Blood samples of CIDP patients on recurrent IVIg treatment and vaccination with a COVID-19 mRNA vaccine were obtained at four timepoints for cytokine ELISA and flow cytometry, to assess key cytokines and cellular immunomarkers for disease activity and IVIg-immunomodulation in CIDP.
Collapse
Affiliation(s)
- Martin K R Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Anika Meißner
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Finja Schweitzer
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Alina Sprenger-Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Ines Klein
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Hauke Wüstenberg
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Felix Kohle
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Helene L Walter
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Michael Schroeter
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Helmar C Lehmann
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Kerpener Straße 62, Cologne, 50937, Germany.
- Department of Neurology, Städtisches Klinikum Leverkusen, Leverkusen, Germany.
| |
Collapse
|
12
|
Shastri A, Al Aiyan A, Kishore U, Farrugia ME. Immune-Mediated Neuropathies: Pathophysiology and Management. Int J Mol Sci 2023; 24:7288. [PMID: 37108447 PMCID: PMC10139406 DOI: 10.3390/ijms24087288] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Dysfunction of the immune system can result in damage of the peripheral nervous system. The immunological mechanisms, which include macrophage infiltration, inflammation and proliferation of Schwann cells, result in variable degrees of demyelination and axonal degeneration. Aetiology is diverse and, in some cases, may be precipitated by infection. Various animal models have contributed and helped to elucidate the pathophysiological mechanisms in acute and chronic inflammatory polyradiculoneuropathies (Guillain-Barre Syndrome and chronic inflammatory demyelinating polyradiculoneuropathy, respectively). The presence of specific anti-glycoconjugate antibodies indicates an underlying process of molecular mimicry and sometimes assists in the classification of these disorders, which often merely supports the clinical diagnosis. Now, the electrophysiological presence of conduction blocks is another important factor in characterizing another subgroup of treatable motor neuropathies (multifocal motor neuropathy with conduction block), which is distinct from Lewis-Sumner syndrome (multifocal acquired demyelinating sensory and motor neuropathy) in its response to treatment modalities as well as electrophysiological features. Furthermore, paraneoplastic neuropathies are also immune-mediated and are the result of an immune reaction to tumour cells that express onconeural antigens and mimic molecules expressed on the surface of neurons. The detection of specific paraneoplastic antibodies often assists the clinician in the investigation of an underlying, sometimes specific, malignancy. This review aims to discuss the immunological and pathophysiological mechanisms that are thought to be crucial in the aetiology of dysimmune neuropathies as well as their individual electrophysiological characteristics, their laboratory features and existing treatment options. Here, we aim to present a balance of discussion from these diverse angles that may be helpful in categorizing disease and establishing prognosis.
Collapse
Affiliation(s)
- Abhishek Shastri
- Central and North West London NHS Foundation Trust, London NW1 3AX, UK
| | - Ahmad Al Aiyan
- Department of Veterinary Medicine, UAE University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Uday Kishore
- Department of Veterinary Medicine, UAE University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Maria Elena Farrugia
- Department of Neurology, Institute of Neurological Sciences, Southern General Hospital, Glasgow G51 4TF, UK
| |
Collapse
|
13
|
Spatola M, Chuquisana O, Jung W, Lopez JA, Wendel EM, Ramanathan S, Keller CW, Hahn T, Meinl E, Reindl M, Dale RC, Wiendl H, Lauffenburger DA, Rostásy K, Brilot F, Alter G, Lünemann JD. Humoral signatures of MOG-antibody-associated disease track with age and disease activity. Cell Rep Med 2023; 4:100913. [PMID: 36669487 PMCID: PMC9975090 DOI: 10.1016/j.xcrm.2022.100913] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/26/2022] [Accepted: 12/24/2022] [Indexed: 01/20/2023]
Abstract
Myelin oligodendrocyte glycoprotein (MOG)-antibody (Ab)-associated disease (MOGAD) is an inflammatory demyelinating disease of the CNS. Although MOG is encephalitogenic in different mammalian species, the mechanisms by which human MOG-specific Abs contribute to MOGAD are poorly understood. Here, we use a systems-level approach combined with high-dimensional characterization of Ab-associated immune features to deeply profile humoral immune responses in 123 patients with MOGAD. We show that age is a major determinant for MOG-antibody-related immune signatures. Unsupervised clustering additionally identifies two dominant immunological endophenotypes of MOGAD. The pro-inflammatory endophenotype characterized by increased binding affinities for activating Fcγ receptors (FcγRs), capacity to activate innate immune cells, and decreased frequencies of galactosylated and sialylated immunoglobulin G (IgG) glycovariants is associated with clinically active disease. Our data support the concept that FcγR-mediated effector functions control the pathogenicity of MOG-specific IgG and suggest that FcγR-targeting therapies should be explored for their therapeutic potential in MOGAD.
Collapse
Affiliation(s)
- Marianna Spatola
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA.
| | - Omar Chuquisana
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, WWU, Münster 48149, Germany
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA; Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joseph A Lopez
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Specialty of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Eva-Maria Wendel
- Department of Pediatric Neurology, Olgahospital/Klinikum Stuttgart, 70174 Stuttgart, Germany
| | - Sudarshini Ramanathan
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia; Department of Neurology, Concord Hospital, Sydney, NSW 2139, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Christian W Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, WWU, Münster 48149, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, 48149 Münster, Germany
| | - Edgar Meinl
- Institute of Clinical Neuroimmunology, Biomedical Center and University Hospital, Ludwig-Maximilians-Universität München, 82152 Munich, Germany
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Russell C Dale
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Specialty of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, WWU, Münster 48149, Germany; Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Kevin Rostásy
- Department of Pediatric Neurology, Children's Hospital Datteln, University Witten/Herdecke, 45711 Datteln, Germany
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, NSW 2145, Australia; Specialty of Child and Adolescent Health, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard Medical School, Cambridge, MA 02139, USA
| | - Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, WWU, Münster 48149, Germany.
| |
Collapse
|
14
|
Ballow M. Immunoglobulin Therapy: Replacement and Immunomodulation. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00082-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
15
|
Svačina MKR, Meißner A, Schweitzer F, Ladwig A, Sprenger‐Svačina A, Klein I, Wüstenberg H, Kohle F, Schneider C, Grether NB, Wunderlich G, Fink GR, Klein F, Di Cristanziano V, Lehmann HC. Antibody response after COVID-19 vaccination in intravenous immunoglobulin-treated immune neuropathies. Eur J Neurol 2022; 29:3380-3388. [PMID: 35842740 PMCID: PMC9349681 DOI: 10.1111/ene.15508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 06/24/2022] [Accepted: 07/13/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE This study assessed the prevalence of anti-SARS-CoV-2 antibodies in therapeutic immunoglobulin and their impact on serological response to COVID-19 mRNA vaccine in patients with intravenous immunoglobulin (IVIg)-treated chronic immune neuropathies. METHODS Forty-six samples of different brands or lots of IVIg or subcutaneous IgG were analyzed for anti-SARS-CoV-2 IgG using enzyme-linked immunosorbent assay and chemiluminescent microparticle immunoassay. Blood sera from 16 patients with immune neuropathies were prospectively analyzed for anti-SARS-CoV-2 IgA, IgG, and IgM before and 1 week after IVIg infusion subsequent to consecutive COVID-19 mRNA vaccine doses and after 12 weeks. These were compared to 42 healthy subjects. RESULTS Twenty-four (52%) therapeutic immunoglobulin samples contained anti-SARS-CoV-2 IgG. All patients with immune neuropathies (mean age = 65 ± 16 years, 25% female) were positive for anti-SARS-CoV-2 IgG after COVID-19 vaccination. Anti-SARS-CoV-2 IgA titers significantly decreased 12-14 weeks after vaccination (p = 0.02), whereas IgG titers remained stable (p = 0.2). IVIg did not significantly reduce intraindividual anti-SARS-CoV-2 IgA/IgG serum titers in immune neuropathies (p = 0.69). IVIg-derived anti-SARS-CoV-2 IgG did not alter serum anti-SARS-CoV-2 IgG decrease after IVIg administration (p = 0.67). CONCLUSIONS Our study indicates that IVIg does not impair the antibody response to COVID-19 mRNA vaccine in a short-term observation, when administered a minimum of 2 weeks after each vaccine dose. The infusion of current IVIg preparations that contain anti-SARS-CoV-2 IgG does not significantly alter serum anti-SARS-CoV-2 IgG titers.
Collapse
Affiliation(s)
- Martin K. R. Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Anika Meißner
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Finja Schweitzer
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Anne Ladwig
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Alina Sprenger‐Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Ines Klein
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Hauke Wüstenberg
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Felix Kohle
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Christian Schneider
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Nicolai B. Grether
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Gilbert Wunderlich
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Gereon R. Fink
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- Cognitive Neuroscience, Research Center JuelichInstitute of Neuroscience and Medicine (INM‐3)JuelichGermany
| | - Florian Klein
- Institute of Virology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
- German Center for Infection Research (DZIF), partner site Bonn‐CologneCologneGermany
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| | - Helmar C. Lehmann
- Department of Neurology, Faculty of Medicine and University Hospital of CologneUniversity of CologneCologneGermany
| |
Collapse
|
16
|
Dalakas MC, Latov N, Kuitwaard K. Intravenous immunoglobulin in chronic inflammatory demyelinating polyradiculoneuropathy (CIDP): mechanisms of action and clinical and genetic considerations. Expert Rev Neurother 2022; 22:953-962. [PMID: 36645654 DOI: 10.1080/14737175.2022.2169134] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune peripheral nerve disorder that is characterized by subacute onset, progressive or relapsing weakness, and sensory deficits. Proven treatments include intravenous immunoglobulin (IVIg), corticosteroids, and plasma exchange. This review focuses on the mechanisms of action, pharmacodynamics, genetic variations, and disease characteristics that can affect the efficacy of IVIg. AREAS COVERED The proposed mechanisms of action of IVIg that can mediate its therapeutic effects are reviewed. These include anti-idiotypic interactions, inhibition of neonatal Fc receptors (FcRn), anti-complement activity, upregulation of inhibitory FcγRIIB receptors, and downregulation of macrophage activation or co-stimulatory and adhesion molecules. Clinical and genetic factors that can affect the therapeutic response include misdiagnosis, degree of axonal damage, pharmacokinetic variability, and genetic variations. EXPERT OPINION The mechanisms of action of IVIg in CIDP and their relative contribution to its efficacy are subject of ongoing investigation. Studies in other autoimmune neurological conditions, in addition, highlight the role of key immunopathological pathways and factors that are likely to be affected. Further investigation into the pathogenesis of CIDP and the mechanisms of action of IVIg may lead to the development of improved diagnostics, better utilization of IVIg, and more targeted and effective therapies.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson Neuroimmunology Unit, Philadelphia, PA and National and Department of Pathophysiology, Kapodistrian University of Athens, Greece
| | - Norman Latov
- Neuroimmunology Unit, Weill Cornell Medical College, New York, NY, USA
| | - Krista Kuitwaard
- Department of Neurology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Neurology, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| |
Collapse
|
17
|
Wang C, Gong Y, You R, Zhi-Ying L, Ming-Hui Z, Chen M. Down-regulated FcγRII expression on plasma cells is associated with the disease activity of ANCA-associated vasculitis. Rheumatology (Oxford) 2022; 62:1998-2004. [PMID: 36111855 DOI: 10.1093/rheumatology/keac524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
Objectives
Inhibitory FcγRIIB/CD32B on B cells are critical for immunity regulation to help maintain peripheral tolerance. Altered FcγRIIB expression on B cells has been observed in several autoimmune diseases, and animal studies suggested that FcγRIIB on B cells participates in the pathogenesis of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). Here, we investigated the expression of FcγRII (FcγRIIB) on various B cell subsets and the correlation of FcγRII/CD32 expression with disease activity in AAV patients.
Methods
Blood samples of patients with AAV in active stage and in remission were collected. FcγRII/CD32 expressions on various B cell subsets of the whole blood were detected by flow cytometry, and their correlation with clinical and pathological data was analyzed.
Results
The expression of FcγRII/CD32 on plasma cells was significantly lower in AAV patients in active stage than those in both AAV patients in remission and healthy donors. Furthermore, the expression of FcγRII/CD32 on plasma cells negatively correlated with Birmingham Vasculitis Activity Scores and percentages of cellular crescents in renal biopsies.
Conclusion
Hence there is a down-regulation of FcγRIIB/CD32B expression on B cells in patients with AAV, which is associated with the disease activity of AAV.
Collapse
Affiliation(s)
- Chen Wang
- Peking University First Hospital; Peking University Institute of Nephrology Renal Division, Department of Medicine, , Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China , Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) , Ministry of Education, Beijing, China
| | - Yan Gong
- Department of Clinical Laboratory, Peking University First Hospital , Beijing, China
| | - Ran You
- Department of Clinical Laboratory, Peking University First Hospital , Beijing, China
| | - Li Zhi-Ying
- Peking University First Hospital; Peking University Institute of Nephrology Renal Division, Department of Medicine, , Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China , Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) , Ministry of Education, Beijing, China
| | - Zhao Ming-Hui
- Peking University First Hospital; Peking University Institute of Nephrology Renal Division, Department of Medicine, , Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China , Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) , Ministry of Education, Beijing, China
- Peking-Tsinghua Center for Life Sciences , Beijing, China
| | - Min Chen
- Peking University First Hospital; Peking University Institute of Nephrology Renal Division, Department of Medicine, , Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China , Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University) , Ministry of Education, Beijing, China
| |
Collapse
|
18
|
Keller CW, Chuquisana O, Derdelinckx J, Gross CC, Berger K, Robinson J, Nimmerjahn F, Wiendl H, Willcox N, Lünemann JD. Impaired B cell Expression of the Inhibitory Fcγ Receptor IIB in Myasthenia Gravis. Ann Neurol 2022; 92:1046-1051. [PMID: 36094152 DOI: 10.1002/ana.26507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022]
Abstract
Myasthenia gravis (MG) is an autoimmune disease in which pathogenic immunoglobulin G (IgG) antibodies (Abs) bind to acetylcholine receptors (AChR) or to functionally related molecules at the neuromuscular junction. B cell expression of the inhibitory IgG receptor, FcγRIIB, maintains peripheral immune tolerance and its absence renders B cells hyperresponsive to autoantigen. Here, we report that FcγRIIB expression levels are substantially reduced in B lineage cells derived from immunotherapy-naïve patients with AChR-Ab+ early-onset MG (EOMG). In contrast, genetic variants associated with impaired FcγRIIB expression are not enriched in MG, indicating post-transcriptional dysregulation. FcγR-targeted therapies could have therapeutic benefits in MG. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Christian W Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Omar Chuquisana
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Judith Derdelinckx
- Department of Neurology, Faculty of Medicine and Health Sciences, Antwerp University Hospital, Antwerp, Belgium.,Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Catharina C Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Klaus Berger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - James Robinson
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, LIGHT Laboratories, Leeds, UK.,National Institute of Health Research-Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, LS7 4SA, UK
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Erlangen, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Nick Willcox
- Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| |
Collapse
|
19
|
Zis P, Liampas A, Pozotou T, Parperis K, Artemiadis A, Hadjigeorgiou G. Immunoglobulin Use for the Management of Painful Peripheral Neuropathy: A Systematic Review and Meta-Analysis. Pain Ther 2022; 11:1219-1227. [PMID: 35925489 PMCID: PMC9633877 DOI: 10.1007/s40122-022-00416-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Immunoglobulins (IG) are widely used for the treatment of a variety of immune-mediated diseases. The exact mechanism of action remains unknown, but IG modulate the expression and function of Fc receptors, interfere with complement activation and production of cytokines, neutralize pathogenic autoantibodies, and affect the activation and effector functions of B and T lymphocytes. Immunoglobulins are usually delivered intravenously, and are effective in ameliorating motor symptoms, and/or preventing disease progression in immune-mediated neuropathies, including Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy. OBJECTIVE The aim of this systematic review and meta-analysis was to study the potential of IG for the treatment of painful peripheral neuropathy (PPN). The outcome of interest was the percentage of patients with PPN who achieved pain relief following IG administration. METHODS We performed a systematic literature search on March 17, 2022, in the PubMed database without any publication date restrictions. We also looked for unpublished or ongoing trials in clinicaltrials.org. Pain reduction following IG treatment had to be within the aims (primary or secondary). RESULTS The aforementioned literature search strategy revealed five studies (two open-label, three randomized placebo-controlled) eligible to be included. The pooled estimate of the percentage of patients with PPN who received immunoglobulins and reported pain relief was found to be 65% (95% CI 58-71%). The likelihood of achieving pain relief with immunoglobulin treatment was 2.9 times higher (95% CI 1.6-5.2) compared to placebo (p = 0.0003). CONCLUSION The use of IG for the treatment of pain due to peripheral neuropathy has a potential therapeutic benefit. Further studies across patients with different types of painful peripheral neuropathy are needed to better characterize this effect. Registration number on PROSPERO: CRD42022319614.
Collapse
Affiliation(s)
- Panagiotis Zis
- Second Department of Neurology, School of Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece. .,Medical School, University of Cyprus, Nicosia, Cyprus.
| | - Andreas Liampas
- Department of Neurology, Nicosia New General Hospital, Nicosia, Cyprus
| | - Theodora Pozotou
- Department of Neurology, Nicosia New General Hospital, Nicosia, Cyprus
| | - Konstantinos Parperis
- Medical School, University of Cyprus, Nicosia, Cyprus.,Division of Rheumatology, Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
| | | | | |
Collapse
|
20
|
Delpire B, Van Loon E, Naesens M. The Role of Fc Gamma Receptors in Antibody-Mediated Rejection of Kidney Transplants. Transpl Int 2022; 35:10465. [PMID: 35935272 PMCID: PMC9346079 DOI: 10.3389/ti.2022.10465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/08/2022] [Indexed: 11/25/2022]
Abstract
For the past decades, complement activation and complement-mediated destruction of allograft cells were considered to play a central role in anti-HLA antibody-mediated rejection (AMR) of kidney transplants. However, also complement-independent mechanisms are relevant in the downstream immune activation induced by donor-specific antibodies, such as Fc-gamma receptor (FcγR)-mediated direct cellular activation. This article reviews the literature regarding FcγR involvement in AMR, and the potential contribution of FcγR gene polymorphisms to the risk for antibody mediated rejection of kidney transplants. There is large heterogeneity between the studies, both in the definition of the clinical phenotypes and in the technical aspects. The study populations were generally quite small, except for two larger study cohorts, which obviates drawing firm conclusions regarding the associations between AMR and specific FcγR polymorphisms. Although FcγR are central in the pathophysiology of AMR, it remains difficult to identify genetic risk factors for AMR in the recipient’s genome, independent of clinical risk factors, independent of the donor-recipient genetic mismatch, and in the presence of powerful immunosuppressive agents. There is a need for larger, multi-center studies with standardised methods and endpoints to identify potentially relevant FcγR gene polymorphisms that represent an increased risk for AMR after kidney transplantation.
Collapse
Affiliation(s)
- Boris Delpire
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Naesens
- University Hospitals Leuven, Leuven, Belgium
- Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
- *Correspondence: Maarten Naesens,
| |
Collapse
|
21
|
Vattepu R, Sneed SL, Anthony RM. Sialylation as an Important Regulator of Antibody Function. Front Immunol 2022; 13:818736. [PMID: 35464485 PMCID: PMC9021442 DOI: 10.3389/fimmu.2022.818736] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/17/2022] [Indexed: 12/14/2022] Open
Abstract
Antibodies play a critical role in linking the adaptive immune response to the innate immune system. In humans, antibodies are categorized into five classes, IgG, IgM, IgA, IgE, and IgD, based on constant region sequence, structure, and tropism. In serum, IgG is the most abundant antibody, comprising 75% of antibodies in circulation, followed by IgA at 15%, IgM at 10%, and IgD and IgE are the least abundant. All human antibody classes are post-translationally modified by sugars. The resulting glycans take on many divergent structures and can be attached in an N-linked or O-linked manner, and are distinct by antibody class, and by position on each antibody. Many of these glycan structures on antibodies are capped by sialic acid. It is well established that the composition of the N-linked glycans on IgG exert a profound influence on its effector functions. However, recent studies have described the influence of glycans, particularly sialic acid for other antibody classes. Here, we discuss the role of glycosylation, with a focus on terminal sialylation, in the biology and function across all antibody classes. Sialylation has been shown to influence not only IgG, but IgE, IgM, and IgA biology, making it an important and unappreciated regulator of antibody function.
Collapse
Affiliation(s)
- Ravi Vattepu
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sunny Lyn Sneed
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Robert M Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Stathopoulos P, Dalakas MC. Evolution of Anti-B Cell Therapeutics in Autoimmune Neurological Diseases. Neurotherapeutics 2022; 19:691-710. [PMID: 35182380 PMCID: PMC9294112 DOI: 10.1007/s13311-022-01196-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2022] [Indexed: 02/08/2023] Open
Abstract
B cells have an ever-increasing role in the etiopathology of a number of autoimmune neurological disorders, acting as antigen-presenting cells facilitating antibody production but also as sensors, coordinators, and regulators of the immune response. In particular, B cells can regulate the T cell activation process through their participation in antigen presentation, production of proinflammatory cytokines (bystander activation or suppression), and contribution to ectopic lymphoid aggregates. Such an important interplay between B and T cells makes therapeutic depletion of B cells an attractive treatment strategy. The last decade, anti-B cell therapies using monoclonal antibodies against B cell surface molecules have evolved into a rational approach for successfully treating autoimmune neurological disorders, even when T cells seem to be the main effector cells. The paper summarizes basic aspects of B cell biology, discusses the roles of B cells in neurological autoimmunities, and highlights how the currently available or under development anti-B cell therapeutics exert their action in the wide spectrum and immunologically diverse neurological disorders. The efficacy of the various anti-B cell therapies and practical issues on induction and maintenance therapy is specifically detailed for the treatment of patients with multiple sclerosis, neuromyelitis-spectrum disorders, autoimmune encephalitis and hyperexcitability CNS disorders, autoimmune neuropathies, myasthenia gravis, and inflammatory myopathies. The success of anti-B cell therapies in inducing long-term remission in IgG4 neuroautoimmunities is also highlighted pointing out potential biomarkers for follow-up infusions.
Collapse
Affiliation(s)
- Panos Stathopoulos
- 1st Department of Neurology, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos C Dalakas
- Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
23
|
Dalakas MC. Autoimmune Neurological Disorders with IgG4 Antibodies: a Distinct Disease Spectrum with Unique IgG4 Functions Responding to Anti-B Cell Therapies. Neurotherapeutics 2022; 19:741-752. [PMID: 35290608 PMCID: PMC9294117 DOI: 10.1007/s13311-022-01210-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2022] [Indexed: 11/26/2022] Open
Abstract
The main IgG4 antibody-mediated neurological disorders (IgG4-ND) include MuSK myasthenia; CIDP with nodal/paranodal antibodies to Neurofascin-155, contactin-1/caspr-1, or pan-neurofascins; anti-LGI1 and CASPR2-associated limbic encephalitis, Morvan syndrome, or neuromyotonia; and several cases of the anti-IgLON5 and anti-DPPX-spectrum CNS diseases. The paper is centered on the clinical spectrum of IgG4-ND and their immunopathogenesis highlighting the unique functional effects of the IgG4 subclass compared to IgG1-3 antibody subclasses. The IgG4 antibodies exert pathogenic effects on their targeted antigens by blocking enzymatic activity or disrupting protein-protein interactions affecting signal transduction pathways, but not by activating complement, binding to inhibitory FcγRIIb receptor or engaging in cross-linking of the targeted antigen with immune complex formation as the IgG1-IgG3 antibody subclasses do. IgG4 can even inhibit the classical complement pathway by affecting the affinity of IgG1-2 subclasses to C1q binding. Because the IgG4 antibodies do not trigger inflammatory processes or complement-mediated immune responses, the conventional anti-inflammatory therapies, especially with IVIg, immunosuppressants, and plasmapheresis, are ineffective or not sufficiently effective in inducing long-term remissions. In contrast, aiming at the activated plasmablasts connected with IgG4 antibody production is a meaningful therapeutic target in IgG4-ND. Indeed, data from large series of patients with MuSK myasthenia, CIDP with nodal/paranodal antibodies, and anti-LGI1 and CASPR2-associated syndromes indicate that B cell depletion therapy with rituximab exerts long-lasting clinical remissions by targeting memory B cells and IgG4-producing CD20-positive short-lived plasma cells. Because IgG4 antibody titers seem reduced in remissions and increased in exacerbation, they may serve as potential biomarkers of treatment response supporting further the pathogenic role of self-reacting B cells. Controlled trials are needed in IgG4-ND not only with rituximab but also with the other anti-B cell agents that target CD19/20, especially those like obexelimab and obinutuzumab, that concurrently activate the inhibitory FcγRIIb receptors which have low binding affinity to IgG4, exerting a more prolonged anti-B cell action affecting also antigen presentation and cytotoxic T cells. Antibody therapies targeting FcRn, testing those anti-FcRn inhibitors that effectively catabolize the IgG4 antibody subclass, may be especially promising.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
24
|
Dalakas MC, Meisel A. Immunomodulatory effects and clinical benefits of intravenous immunoglobulin in myasthenia gravis. Expert Rev Neurother 2022; 22:313-318. [PMID: 35350948 DOI: 10.1080/14737175.2022.2057223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Myasthenia gravis (MG) is an antibody-mediated disease that develops in the majority of patients mainly as a result of acetylcholine receptor (AChR) autoantibodies. This process is mediated by a series of immunoregulatory events. Therapeutic targets for MG include suppression of circulating antibodies or antibody production, suppression of complement activation, and immunomodulation of cytokines or T cells. Intravenous immunoglobulin (IVIg) has an effect on all of these mechanisms. AREAS COVERED This narrative review explores the broad immunomodulatory effects of IVIg in MG and provides an update on IVIg treatment for MG. EXPERT OPINION IVIg has a range of immunomodulatory effects on therapeutic targets relevant to the immunopathogenesis of MG. An emerging area of research is the pharmacogenomics of IVIg in MG related to FcRn and IgG catabolism. New data indicate that the FcRn VNTR3 genotype can affect the efficacy of IVIg in certain MG patients and may have an impact on IgG kinetics and selected dosing. Immune globulin 10% caprylate/chromatography purified (IVIg-C) has been shown to reverse the symptoms of severe acute exacerbation in patients with MG. Available data support the use of IVIg-C as an effective and safe treatment for this severely ill subgroup of patients during a relapse.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andreas Meisel
- Department of Neurology, Integrated Center for Myasthenia Gravis, Charité - University Medicine Berlin, Germany
| |
Collapse
|
25
|
Svačina MKR, Lehmann HC. Chronic Inflammatory Demyelinating Polyneuropathy (CIDP): Current Therapies and Future Approaches. Curr Pharm Des 2022; 28:854-862. [PMID: 35339172 DOI: 10.2174/1381612828666220325102840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/08/2022] [Indexed: 11/22/2022]
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is an acquired immune-mediated polyradiculoneuropathy leading to disability via inflammatory demyelination of peripheral nerves. Various therapeutic approaches with different mechanisms of action are established for the treatment of CIDP. Of those, corticosteroids, intravenous or subcutaneous immunoglobulin, or plasma exchange are established first-line therapies as suggested by the recently revised EAN/PNS guidelines for the management of CIDP. In special cases, immunosuppressants or rituximab may be used. Novel therapeutic approaches currently undergoing clinical studies include molecules or monoclonal antibodies interacting with Fc receptors on immune cells to alleviate immune-mediated neuronal damage. Despite various established therapies and the current development of novel therapeutics, treatment of CIDP remains challenging due to an inter-individually heterogeneous disease course and the lack of surrogate parameters to predict the risk of clinical deterioration.
Collapse
Affiliation(s)
- Martin K R Svačina
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Helmar C Lehmann
- Department of Neurology, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
26
|
Potential therapeutic strategies in chronic inflammatory demyelinating polyradiculoneuropathy. Clin Exp Rheumatol 2022; 21:103032. [PMID: 34999243 DOI: 10.1016/j.autrev.2022.103032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/01/2022] [Indexed: 11/23/2022]
Abstract
Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an autoimmune neuropathy involving peripheral nerve and nerve roots. The pathological hallmark of CIDP is macrophage-induced demyelination. Antibodies against nerve fibers, complement decomposition, abnormalities in plasma and cerebrospinal fluid cytokine profile, and changes of peripheral blood cell proportion were also reported in CIDP patients. These findings in immunopathology provide support for the introduction of potential therapeutic options for the treatment of CIDP. In this review, we systematically listed the potential therapeutic strategies targeting different components of the immune system by comparing the treatment of other autoimmune inflammatory diseases of the nervous system. Several ongoing clinical trials will assess the efficacy and safety of potential CIDP treatments.
Collapse
|
27
|
Dalakas MC. IgG4-Mediated Neurologic Autoimmunities: Understanding the Pathogenicity of IgG4, Ineffectiveness of IVIg, and Long-Lasting Benefits of Anti-B Cell Therapies. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 9:9/1/e1116. [PMID: 34845096 PMCID: PMC8630661 DOI: 10.1212/nxi.0000000000001116] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND OBJECTIVES Describe the unique functions of immunoglobulin G4 (IgG4) in IgG4-neurologic disorders (IgG4-ND) and explain why, in contrast to their IgG1-counterparts, they respond poorly to intravenous immune globulin (IVIg) but effectively to anti-B cell therapies. METHODS The IgG4 structure and isotype switch, B cells and plasmablasts relevant to IgG4 production, and IgG4-induced disruption of the targeted antigens are reviewed and compared with IgG1-mediated autoimmune ND, where IVIg inhibits IgG1-triggered inflammatory effects. RESULTS The main IgG4-ND include muscle-specific kinase myasthenia; nodal/paranodal chronic inflammatory demyelinating polyradiculoneuropathy with antibodies to neurofascin-155, contactin-1/caspr-1, or pan-neurofascins; antileucine-rich, glioma-inactivated-1 and contactin-associated protein-like 2 associated-limbic encephalitis, Morvan syndrome, or neuromyotonia; and anti-IgLON5 disorder. The IgG4, because of its unique structural features in the hinge region, has noninflammatory properties being functionally monovalent and bispecific, unable to engage in cross-linking and internalization of the targeted antigen. In contrast to IgG1 subclass which is bivalent and monospecific, IgG4 does not activate complement and cannot bind to inhibitory Fcγ receptor (FcγRIIb) to activate cellular and complement-mediated immune responses, the key functions inhibited by IVIg. Because IVIg contains only 0.7%-2.6% IgG4, its idiotypes are of IgG1 subclass and cannot effectively neutralize IgG4 or sufficiently enhance IgG4 catabolism by saturating FcRn. In contrast, rituximab, by targeting memory B cells and IgG4-producing CD20-positive short-lived plasma cells, induces long-lasting clinical benefits. DISCUSSION Rituximab is the preferred treatment in IgG4-ND patients with severe disease by effectively targeting the production of pathogenic IgG-4 antibodies. In contrast, IVIG is ineffective because it inhibits immunoinflammatory functions irrelevant to the mechanistic effects of IgG4 and contains IgG-1 idiotypes that cannot sufficiently neutralize or possibly catabolize IgG4. Controlled studies with anti-CD19/20 monoclonals that also activate FcγRIIb may be more promising in treating IgG4-ND.
Collapse
Affiliation(s)
- Marinos C Dalakas
- From Thomas Jefferson University, Philadelphia, PA; and the University of Athens Medical School, Greece.
| |
Collapse
|
28
|
Querol L, Lleixà C. Novel Immunological and Therapeutic Insights in Guillain-Barré Syndrome and CIDP. Neurotherapeutics 2021; 18:2222-2235. [PMID: 34549385 PMCID: PMC8455117 DOI: 10.1007/s13311-021-01117-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammatory neuropathies are a heterogeneous group of rare diseases of the peripheral nervous system that include acute and chronic diseases, such as Guillain-Barré syndrome (GBS) and chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). The etiology and pathophysiological mechanisms of inflammatory neuropathies are only partly known, but are considered autoimmune disorders in which an aberrant immune response, including cellular and humoral components, is directed towards components of the peripheral nerve causing demyelination and axonal damage. Therapy of these disorders includes broad-spectrum immunomodulatory and immunosuppressive treatments, such as intravenous immunoglobulin, corticosteroids, or plasma exchange. However, a significant proportion of patients do not respond to any of these therapies, and treatment selection is not optimized according to disease pathophysiology. Therefore, research on disease pathophysiology aiming to reveal clinically and functionally relevant disease mechanisms and the development of new treatment approaches are needed to optimize disease outcomes in CIDP and GBS. This topical review describes immunological progress that may help guide therapeutic strategies in the future in these two disorders.
Collapse
Affiliation(s)
- Luis Querol
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas 90, 08041, Barcelona, Spain.
- Centro Para La Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Madrid, Spain.
| | - Cinta Lleixà
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de La Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Mas Casanovas 90, 08041, Barcelona, Spain
| |
Collapse
|
29
|
Dalakas MC. Update on Intravenous Immunoglobulin in Neurology: Modulating Neuro-autoimmunity, Evolving Factors on Efficacy and Dosing and Challenges on Stopping Chronic IVIg Therapy. Neurotherapeutics 2021; 18:2397-2418. [PMID: 34766257 PMCID: PMC8585501 DOI: 10.1007/s13311-021-01108-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
In the last 25 years, intravenous immunoglobulin (IVIg) has had a major impact in the successful treatment of previously untreatable or poorly controlled autoimmune neurological disorders. Derived from thousands of healthy donors, IVIg contains IgG1 isotypes of idiotypic antibodies that have the potential to bind pathogenic autoantibodies or cross-react with various antigenic peptides, including proteins conserved among the "common cold"-pre-pandemic coronaviruses; as a result, after IVIg infusions, some of the patients' sera may transiently become positive for various neuronal antibodies, even for anti-SARS-CoV-2, necessitating caution in separating antibodies derived from the infused IVIg or acquired humoral immunity. IVIg exerts multiple effects on the immunoregulatory network by variably affecting autoantibodies, complement activation, FcRn saturation, FcγRIIb receptors, cytokines, and inflammatory mediators. Based on randomized controlled trials, IVIg is approved for the treatment of GBS, CIDP, MMN and dermatomyositis; has been effective in, myasthenia gravis exacerbations, and stiff-person syndrome; and exhibits convincing efficacy in autoimmune epilepsy, neuromyelitis, and autoimmune encephalitis. Recent evidence suggests that polymorphisms in the genes encoding FcRn and FcγRIIB may influence the catabolism of infused IgG or its anti-inflammatory effects, impacting on individualized dosing or efficacy. For chronic maintenance therapy, IVIg and subcutaneous IgG are effective in controlled studies only in CIDP and MMN preventing relapses and axonal loss up to 48 weeks; in practice, however, IVIg is continuously used for years in all the aforementioned neurological conditions, like is a "forever necessary therapy" for maintaining stability, generating challenges on when and how to stop it. Because about 35-40% of patients on chronic therapy do not exhibit objective neurological signs of worsening after stopping IVIg but express subjective symptoms of fatigue, pains, spasms, or a feeling of generalized weakness, a conditioning effect combined with fear that discontinuing chronic therapy may destabilize a multi-year stability status is likely. The dilemmas of continuing chronic therapy, the importance of adjusting dosing and scheduling or periodically stopping IVIg to objectively assess necessity, and concerns in accurately interpreting IVIg-dependency are discussed. Finally, the merit of subcutaneous IgG, the ineffectiveness of IVIg in IgG4-neurological autoimmunities, and genetic factors affecting IVIg dosing and efficacy are addressed.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA.
- Neuroimmunology Unit, Dept. of Pathophysiology, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
30
|
Werner A, Schäfer S, Zaytseva O, Albert H, Lux A, Krištić J, Pezer M, Lauc G, Winkler T, Nimmerjahn F. Targeting B cells in the pre-phase of systemic autoimmunity globally interferes with autoimmune pathology. iScience 2021; 24:103076. [PMID: 34585117 PMCID: PMC8455742 DOI: 10.1016/j.isci.2021.103076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/21/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is characterized by a loss of self-tolerance, systemic inflammation, and multi-organ damage. While a variety of therapeutic interventions are available, it has become clear that an early diagnosis and treatment may be key to achieve long lasting therapeutic responses and to limit irreversible organ damage. Loss of humoral tolerance including the appearance of self-reactive antibodies can be detected years before the actual onset of the clinical autoimmune disease, representing a potential early point of intervention. Not much is known, however, about how and to what extent this pre-phase of disease impacts the onset and development of subsequent autoimmunity. By targeting the B cell compartment in the pre-disease phase of a spontaneous mouse model of SLE we now show, that resetting the humoral immune system during the clinically unapparent phase of the disease globally alters immune homeostasis delaying the downstream development of systemic autoimmunity. The clinically unapparent pre-phase of SLE impacts clinical disease Autoreactive IgM antibodies represent a biomarker for early therapeutic intervention Pre-phase B cells orchestrate clinical disease Depleting pre-phase B cells diminishes disease pathology
Collapse
Affiliation(s)
- Anja Werner
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Simon Schäfer
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Olga Zaytseva
- Genos Ltd, Glycoscience Research Laboratory, Borongajska 83H, 10000 Zagreb, Croatia
| | - Heike Albert
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Anja Lux
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erwin-Rommelstr. 3, 91058 Erlangen, Germany
| | - Jasminka Krištić
- Genos Ltd, Glycoscience Research Laboratory, Borongajska 83H, 10000 Zagreb, Croatia
| | - Marija Pezer
- Genos Ltd, Glycoscience Research Laboratory, Borongajska 83H, 10000 Zagreb, Croatia
| | - Gordan Lauc
- Genos Ltd, Glycoscience Research Laboratory, Borongajska 83H, 10000 Zagreb, Croatia
| | - Thomas Winkler
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erwin-Rommelstr. 3, 91058 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Falk Nimmerjahn
- Chair of Genetics, Department of Biology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erwin-Rommelstr. 3, 91058 Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
31
|
Maverakis E, Merleev AA, Park D, Kailemia MJ, Xu G, Ruhaak LR, Kim K, Hong Q, Li Q, Leung P, Liakos W, Wan YJY, Bowlus CL, Marusina AI, Lal NN, Xie Y, Luxardi G, Lebrilla CB. Glycan biomarkers of autoimmunity and bile acid-associated alterations of the human glycome: Primary biliary cirrhosis and primary sclerosing cholangitis-specific glycans. Clin Immunol 2021; 230:108825. [PMID: 34403816 DOI: 10.1016/j.clim.2021.108825] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022]
Abstract
We have recently introduced multiple reaction monitoring (MRM) mass spectrometry as a novel tool for glycan biomarker research and discovery. Herein, we employ this technique to characterize the site-specific glycan alterations associated with primary biliary cirrhosis (PBC) and primary sclerosing cholangitis (PSC). Glycopeptides associated with disease severity were also identified. Multinomial regression modelling was employed to construct and validate multi-analyte diagnostic models capable of accurately distinguishing PBC, PSC, and healthy controls from one another (AUC = 0.93 ± 0.03). Finally, to investigate how disease-relevant environmental factors can influence glycosylation, we characterized the ability of bile acids known to be differentially expressed in PBC to alter glycosylation. We hypothesize that this could be a mechanism by which altered self-antigens are generated and become targets for immune attack. This work demonstrates the utility of the MRM method to identify diagnostic site-specific glycan classifiers capable of distinguishing even related autoimmune diseases from one another.
Collapse
Affiliation(s)
- Emanual Maverakis
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA.
| | - Alexander A Merleev
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Dayoung Park
- Department of Chemistry, University of California Davis, Davis, CA, USA; Department of Surgery, Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | - Gege Xu
- Department of Chemistry, University of California Davis, Davis, CA, USA
| | - L Renee Ruhaak
- Department of Chemistry, University of California Davis, Davis, CA, USA; Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Center, ZA, Leiden, the Netherlands
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - Qiuting Hong
- Department of Chemistry, University of California Davis, Davis, CA, USA
| | - Qiongyu Li
- Department of Chemistry, University of California Davis, Davis, CA, USA
| | - Patrick Leung
- Department of Internal Medicine, Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis School of Medicine, Davis, CA, USA
| | - William Liakos
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Christopher L Bowlus
- Division of Gastroenterology and Hepatology, UC Davis School of Medicine, CA, USA
| | - Alina I Marusina
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Nelvish N Lal
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Yixuan Xie
- Department of Chemistry, University of California Davis, Davis, CA, USA
| | - Guillaume Luxardi
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California Davis, Davis, CA, USA; Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA; Foods for Health Institute, University of California Davis, Davis, CA, USA
| |
Collapse
|
32
|
Freeman S, Grinstein S. Promoters and Antagonists of Phagocytosis: A Plastic and Tunable Response. Annu Rev Cell Dev Biol 2021; 37:89-114. [PMID: 34152790 DOI: 10.1146/annurev-cellbio-120219-055903] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent observations indicate that, rather than being an all-or-none response, phagocytosis is finely tuned by a host of developmental and environmental factors. The expression of key phagocytic determinants is regulated via transcriptional and epigenetic means that confer memory on the process. Membrane traffic, the cytoskeleton, and inside-out signaling control the activation of phagocytic receptors and their ability to access their targets. An exquisite extra layer of complexity is introduced by the coexistence of distinct "eat-me" and "don't-eat-me" signals on targets and of corresponding "eat" and "don't-eat" receptors on the phagocyte surface. Moreover, assorted physical barriers constitute "don't-come-close-to-me" hurdles that obstruct the engagement of ligands by receptors. The expression, mobility, and accessibility of all these determinants can be modulated, conferring extreme plasticity on phagocytosis and providing attractive targets for therapeutic intervention in cancer, atherosclerosis, and dementia. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Spencer Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario M5G0A4, Canada; , .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario M5G0A4, Canada; , .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
33
|
Hayashi T, Nukui T, Piao JL, Sugimoto T, Anada R, Matsuda N, Yamamoto M, Konishi H, Dougu N, Nakatsuji Y. Serum neurofilament light chain in chronic inflammatory demyelinating polyneuropathy. Brain Behav 2021; 11:e02084. [PMID: 33617139 PMCID: PMC8119854 DOI: 10.1002/brb3.2084] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES Neurofilament light chain (NfL) levels have been suggested as reflecting axonal damage in various inflammatory and neurodegenerative disorders, including acquired peripheral neuropathies. We aimed to investigate if serum NfL (sNfL) levels can be a biomarker of disease activity and treatment response in patients with chronic inflammatory demyelinating polyneuropathy (CIDP). MATERIALS AND METHODS The sNfL levels of eleven newly diagnosed patients with CIDP were retrospectively assayed and compared with seven healthy volunteers. The levels were assayed before and after intravenous immunoglobulin treatment in patients with CIDP and were also assayed in the remission period. RESULTS Baseline sNfL levels in patients with CIDP before treatment were significantly higher than those in healthy controls. The levels significantly decreased overtime after one month of treatment and in remission period. There were significant negative correlations between the sNfL levels and the disease duration (the interval between the onset of the disease and the time of sampling), and weak correlations between the sNfL levels and overall neuropathy limitations scale. CONCLUSIONS sNfL may be a potential biomarker reflecting the disease activity in patients with CIDP.
Collapse
Affiliation(s)
- Tomohiro Hayashi
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Takamasa Nukui
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Jin-Lan Piao
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | | | - Ryoko Anada
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Noriyuki Matsuda
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Mamoru Yamamoto
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Hirofumi Konishi
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Nobuhiro Dougu
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Yuji Nakatsuji
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
34
|
Hagen KM, Ousman SS. The immune response and aging in chronic inflammatory demyelinating polyradiculoneuropathy. J Neuroinflammation 2021; 18:78. [PMID: 33752693 PMCID: PMC7983397 DOI: 10.1186/s12974-021-02113-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/16/2021] [Indexed: 12/26/2022] Open
Abstract
Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) consists of various autoimmune subtypes in which the peripheral nervous system (PNS) is attacked. CIDP can follow a relapsing-remitting or progressive course where the resultant demyelination caused by immune cells (e.g., T cells, macrophages) and antibodies can lead to disability in patients. Importantly, the age of CIDP patients has a role in their symptomology and specific variants have been associated with differing ages of onset. Furthermore, older patients have a decreased frequency of functional recovery after CIDP insult. This may be related to perturbations in immune cell populations that could exacerbate the disease with increasing age. In the present review, the immune profile of typical CIDP will be discussed followed by inferences into the potential role of relevant aging immune cell populations. Atypical variants will also be briefly reviewed followed by an examination of the available studies on the immunology underlying them.
Collapse
Affiliation(s)
- Kathleen M Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Shalina S Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
35
|
Kuitwaard K, van Doorn PA, Bengrine T, van Rijs W, Baas F, Nagelkerke SQ, Kuijpers TW, Fokkink WJR, Bunschoten C, Broers MC, Willemsen SP, Jacobs BC, Huizinga R. Genetic biomarkers for intravenous immunoglobulin response in chronic inflammatory demyelinating polyradiculoneuropathy. Eur J Neurol 2021; 28:1677-1683. [PMID: 33460483 PMCID: PMC8247870 DOI: 10.1111/ene.14742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/07/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a clinical and electrophysiological heterogeneous immune-mediated polyneuropathy. Intravenous immunoglobulin (IVIg), corticosteroids, and plasma exchange are proven effective treatments for CIDP. The clinical response to IVIg is variable between patients and currently unexplained. Finding biomarkers related to treatment response can help to understand the diversity of CIDP and personalise treatment choice. METHODS We investigated whether genetic variation between patients may explain some of these differences in treatment response. Based on previous publications, we selected six candidate genes that might affect immune and axonal functions, IVIg metabolism, and treatment response in CIDP. Genetic variants were assessed in 172 CIDP patients treated with at least one course of IVIg (2 g/kg). A response to IVIg was defined by ≥1 grade improvement on the modified Rankin Scale. Blood samples were tested for variations in CNTN2, PRF1, FCGRT, FCGR2B, GJB1, and SH2D2A genes. RESULTS In univariate analysis, patients with the FCGR2B promoter variant 2B.4/2B.1 responded more often to IVIg than patients with the 2B.1/2B.1 variant (odds ratio [OR] = 6.9, 95% confidence interval [CI] = 1.6-30; p = 0.003). Patients with the p.(Ala91Val) variant of PRF1 were less often IVIg responsive (OR = 0.34, 95% CI = 0.13-0.91; p = 0.038). In multivariate analysis, both PRF1 and FCGR2B showed discriminative ability to predict the chance of IVIg response (area under the curve = 0.67). CONCLUSIONS Variations in PRF1 and the promoter region of FCGR2B are associated with the response to IVIg in CIDP. These findings, which require validation, are a first step towards the understanding of the heterogeneity in the treatment response in CIDP.
Collapse
Affiliation(s)
- Krista Kuitwaard
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Neurology, Albert Schweitzer hospital, Dordrecht, the Netherlands
| | - Pieter A van Doorn
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Thiziri Bengrine
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Wouter van Rijs
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Immunology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Sietse Q Nagelkerke
- Department of Blood Cell Research, Landsteiner Laboratory, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands.,Department of Pediatric Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Landsteiner Laboratory, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands.,Department of Pediatric Immunology and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Willem-Jan R Fokkink
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Immunology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Carina Bunschoten
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Merel C Broers
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Sten P Willemsen
- Department of Epidemiology and Biostatistics, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Bart C Jacobs
- Department of Neurology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands.,Department of Immunology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Ruth Huizinga
- Department of Immunology, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
36
|
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
|
37
|
Chio JCT, Wang J, Surendran V, Li L, Zavvarian MM, Pieczonka K, Fehlings MG. Delayed administration of high dose human immunoglobulin G enhances recovery after traumatic cervical spinal cord injury by modulation of neuroinflammation and protection of the blood spinal cord barrier. Neurobiol Dis 2020; 148:105187. [PMID: 33249350 DOI: 10.1016/j.nbd.2020.105187] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/INTRODUCTION The neuroinflammatory response plays a major role in the secondary injury cascade after traumatic spinal cord injury (SCI). To date, systemic anti-inflammatory medications such as methylprednisolone sodium succinate (MPSS) have shown promise in SCI. However, systemic immunosuppression can have detrimental side effects. Therefore, immunomodulatory approaches including the use of human immunoglobulin G (hIgG) could represent an attractive alternative. While emerging preclinical data suggests that hIgG is neuroprotective after SCI, the optimal time window of administration and the mechanism of action remain incompletely understood. These knowledge gaps were the focus of this research study. METHODS Female adult Wistar rats received a clip compression-contusion SCI at the C7/T1 level of the spinal cord. Injured rats were randomized, in a blinded manner, to receive a single intravenous bolus of hIgG (2 g/kg) or control buffer at 15 minutes (min), 1 hour (h) or 4 h post-SCI. At 24 h and 8 weeks post-SCI, molecular, histological and neurobehavioral analyses were undertaken. RESULTS At all 3 administration time points, hIgG (2 g/kg) resulted in significantly better short-term and long-term outcomes as compared to control buffer. No significant differences were observed when comparing outcomes between the different time points of administration. At 24 h post-injury, hIgG (2 g/kg) administration enhanced the integrity of the blood spinal cord barrier (BSCB) by increasing expression of tight junction proteins and reducing inflammatory enzyme expression. Improvements in BSCB integrity were associated with reduced immune cell infiltration, lower amounts of albumin and Evans Blue in the injured spinal cord and greater expression of anti-inflammatory cytokines. Furthermore, hIgG (2 g/kg) increased expression of neutrophil chemoattractants in the spleen and sera. After hIgG (2 g/kg) treatment, there were more neutrophils in the spleen and fewer neutrophils in the blood. hIgG also co-localized with endothelial cell ligands that mediate neutrophil extravasation into the injured spinal cord. Importantly, short-term effects of delayed hIgG (2 g/kg) administration were associated with enhanced tissue and neuron preservation, as well as neurobehavioral and sensory recovery at 8 weeks post-SCI. DISCUSSION AND CONCLUSION hIgG (2 g/kg) shows promise as a therapeutic approach for SCI. The anti-inflammatory effects mediated by hIgG (2 g/kg) in the injured spinal cord might be explained in twofold. First, hIgG might antagonize neutrophil infiltration into the spinal cord by co-localizing with endothelial cell ligands that mediate various steps in neutrophil extravasation. Second, hIgG could traffic neutrophils towards the spleen by increasing expression of neutrophil chemoattractants in the spleen and sera. Overall, we demonstrate that delayed administration of hIgG (2 g/kg) at 1 and 4-h post-injury enhances short-term and long-term benefits after SCI by modulating local and systemic neuroinflammatory cascades.
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.
| | - Jian Wang
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Vithushan Surendran
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Lijun Li
- Division of Translational and Experimental Neuroscience, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.
| | - Mohammad-Masoud Zavvarian
- 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.
| | - Katarzyna Pieczonka
- 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.
| | - 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
|
38
|
Markina YV, Gerasimova EV, Markin AM, Glanz VY, Wu WK, Sobenin IA, Orekhov AN. Sialylated Immunoglobulins for the Treatment of Immuno-Inflammatory Diseases. Int J Mol Sci 2020; 21:ijms21155472. [PMID: 32751832 PMCID: PMC7432344 DOI: 10.3390/ijms21155472] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/20/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Immunoglobulins are the potent effector proteins of the humoral immune response. In the course of evolution, immunoglobulins have formed extremely diverse types of molecular structures with antigen-recognizing, antigen-binding, and effector functions embedded in a single molecule. Polysaccharide moiety of immunoglobulins plays the essential role in immunoglobulin functioning. There is growing evidence that the carbohydrate composition of immunoglobulin-linked glycans, and especially their terminal sialic acid residues, provide a key effect on the effector functions of immunoglobulins. Possibly, sialylation of Fc glycan is a common mechanism of IgG anti-inflammatory action in vivo. Thus, the post-translational modification (glycosylation) of immunoglobulins opens up significant possibilities in the diagnosis of both immunological and inflammatory disorders and in their therapies. This review is focused on the analysis of glycosylation of immunoglobulins, which can be a promising addition to improve existing strategies for the diagnosis and treatment of various immuno-inflammatory diseases.
Collapse
Affiliation(s)
- Yuliya V. Markina
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russia; (A.M.M.); (V.Y.G.); (I.A.S.); (A.N.O.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Correspondence: ; Tel.: +7-905-336-67-76
| | - Elena V. Gerasimova
- Department of Systemic Rheumatic Diseases, V.A. Nasonova Research Institute of Rheumatology, 34A Kashirskoe Shosse, 115522 Moscow, Russia;
| | - Alexander M. Markin
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russia; (A.M.M.); (V.Y.G.); (I.A.S.); (A.N.O.)
| | - Victor Y. Glanz
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russia; (A.M.M.); (V.Y.G.); (I.A.S.); (A.N.O.)
| | - Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital, Bei-Hu Branch, Taipei 108, Taiwan;
| | - Igor A. Sobenin
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russia; (A.M.M.); (V.Y.G.); (I.A.S.); (A.N.O.)
- Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Cellular and Molecular Pathology of the Cardiovascular System, Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russia; (A.M.M.); (V.Y.G.); (I.A.S.); (A.N.O.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, 125315 Moscow, Russia
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
| |
Collapse
|
39
|
Shock A, Humphreys D, Nimmerjahn F. Dissecting the mechanism of action of intravenous immunoglobulin in human autoimmune disease: Lessons from therapeutic modalities targeting Fcγ receptors. J Allergy Clin Immunol 2020; 146:492-500. [PMID: 32721416 DOI: 10.1016/j.jaci.2020.06.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 12/18/2022]
Abstract
Since the first description of the administration of high doses of pooled serum IgG, also referred to as intravenous IgG (IVIg) therapy, as being able to ameliorate various autoimmune diseases, researchers have been investigating which molecular and cellular pathways underlie IVIg activity. Apart from trying to understand the obvious conundrum that IgG can trigger both autoimmune pathology and resolution of inflammation, the rapidly expanding use of IVIg has led to a lack of availability of this primary blood product, providing a strong rationale for developing recombinant alternatives. During the last decade, a tremendous number of novel insights into IVIg activity brought the goal of replacing IVIg within reach, at least in select indications, and has led to the initiation of several clinical trials. At the forefront of this effort is the modulation of autoantibody half-life and blocking access of autoantibodies to fragment cystallizable γ receptors (Fcγ receptors). In this rostrum article, we will briefly discuss current models of IVIg activity, followed by a more specific focus on novel therapeutic avenues that are entering the clinic and may replace IVIg in the future.
Collapse
Affiliation(s)
| | | | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany; Medical Immunology Campus Erlangen, Erlangen, Germany.
| |
Collapse
|
40
|
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
|
41
|
Danzer H, Glaesner J, Baerenwaldt A, Reitinger C, Lux A, Heger L, Dudziak D, Harrer T, Gessner A, Nimmerjahn F. Human Fcγ-receptor IIb modulates pathogen-specific versus self-reactive antibody responses in lyme arthritis. eLife 2020; 9:55319. [PMID: 32613944 PMCID: PMC7438111 DOI: 10.7554/elife.55319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022] Open
Abstract
Pathogen-specific antibody responses need to be tightly regulated to generate protective but limit self-reactive immune responses. While loss of humoral tolerance has been associated with microbial infections, the pathways involved in balancing protective versus autoreactive antibody responses in humans are incompletely understood. Studies in classical mouse model systems have provided evidence that balancing of immune responses through inhibitory receptors is an important quality control checkpoint. Genetic differences between inbred mouse models and the outbred human population and allelic receptor variants not present in mice; however, argue for caution when directly translating these findings to the human system. By studying Borrelia burgdorferi infection in humanized mice reconstituted with human hematopoietic stem cells from donors homozygous for a functional or a non-functional FcγRIIb allele, we show that the human inhibitory FcγRIIb is a critical checkpoint balancing protective and autoreactive immune responses, linking infection with induction of autoimmunity in the human immune system.
Collapse
Affiliation(s)
- Heike Danzer
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Joachim Glaesner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Anne Baerenwaldt
- Laboratory for Cancer Immunotherapy, University Hospital Basel, Basel, Switzerland
| | - Carmen Reitinger
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anja Lux
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lukas Heger
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Erlangen, Germany
| | - Diana Dudziak
- Department of Dermatology, Laboratory of Dendritic Cell Biology, University Hospital Erlangen, Erlangen, Germany.,Medical Immunology Campus Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Harrer
- Medical Department 3, University Hospital Erlangen, Erlangen, Germany
| | - André Gessner
- Institute of Medical Microbiology and Hygiene, University Regensburg, Regensburg, Germany
| | - Falk Nimmerjahn
- Institute of Genetics, Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
42
|
Ohyama Y, Nakajima K, Renfrow MB, Novak J, Takahashi K. Mass spectrometry for the identification and analysis of highly complex glycosylation of therapeutic or pathogenic proteins. Expert Rev Proteomics 2020; 17:275-296. [PMID: 32406805 DOI: 10.1080/14789450.2020.1769479] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Protein glycosylation influences characteristics such as folding, stability, protein interactions, and solubility. Therefore, glycan moieties of therapeutic proteins and proteins that are likely associated with disease pathogenesis should be analyzed in-depth, including glycan heterogeneity and modification sites. Recent advances in analytical methods and instrumentation have enabled comprehensive characterization of highly complex glycosylated proteins. AREA COVERED The following aspects should be considered when analyzing glycosylated proteins: sample preparation, chromatographic separation, mass spectrometry (MS) and fragmentation methods, and bioinformatics, such as software solutions for data analyses. Notably, analysis of glycoproteins with heavily sialylated glycans or multiple glycosylation sites requires special considerations. Here, we discuss recent methodological advances in MS that provide detailed characterization of heterogeneous glycoproteins. EXPERT OPINION As characterization of complex glycosylated proteins is still analytically challenging, the function or pathophysiological significance of these proteins is not fully understood. To reproducibly produce desired forms of therapeutic glycoproteins or to fully elucidate disease-specific patterns of protein glycosylation, a highly reproducible and robust analytical platform(s) should be established. In addition to advances in MS instrumentation, optimization of analytical and bioinformatics methods and utilization of glycoprotein/glycopeptide standards is desirable. Ultimately, we envision that an automated high-throughput MS analysis will provide additional power to clinical studies and precision medicine.
Collapse
Affiliation(s)
- Yukako Ohyama
- Department of Nephrology, Fujita Health University School of Medicine , Toyoake, Japan.,Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine , Toyoake, Japan
| | - Kazuki Nakajima
- Center for Research Promotion and Support, Fujita Health University , Toyoake, Japan
| | - Matthew B Renfrow
- Departments of Biochemistry and Molecular Genetics and Microbiology, University of Alabama at Birmingham , Birmingham, AL, USA
| | - Jan Novak
- Departments of Biochemistry and Molecular Genetics and Microbiology, University of Alabama at Birmingham , Birmingham, AL, USA
| | - Kazuo Takahashi
- Department of Nephrology, Fujita Health University School of Medicine , Toyoake, Japan.,Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine , Toyoake, Japan.,Departments of Biochemistry and Molecular Genetics and Microbiology, University of Alabama at Birmingham , Birmingham, AL, USA
| |
Collapse
|
43
|
Wang TT, Ravetch JV. Functional diversification of IgGs through Fc glycosylation. J Clin Invest 2020; 129:3492-3498. [PMID: 31478910 DOI: 10.1172/jci130029] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
IgG antibodies are secreted from B cells and bind to a variety of pathogens to control infections as well as contribute to inflammatory diseases. Many of the functions of IgGs are mediated through Fcγ receptors (FcγRs), which transduce interactions with immune complexes, leading to a variety of cellular outcomes depending on the FcγRs and cell types engaged. Which FcγRs and cell types will be engaged during an immune response depends on the structure of Fc domains within immune complexes that are formed when IgGs bind to cognate antigen(s). Recent studies have revealed an unexpected degree of structural variability in IgG Fc domains among people, driven primarily by differences in IgG subclasses and N-linked glycosylation of the CH2 domain. This translates, in turn, to functional immune diversification through type I and type II FcγR-mediated cellular functions. For example, Fc domain sialylation triggers conformational changes of IgG1 that enable interactions with type II FcγRs; these receptors mediate cellular functions including antiinflammatory activity or definition of thresholds for B cell selection based on B cell receptor affinity. Similarly, presence or absence of a core fucose alters type I FcγR binding of IgG1 by modulating the Fc's affinity for FcγRIIIa, thereby altering its proinflammatory activity. How heterogeneity in IgG Fc domains contributes to human immune diversity is now being elucidated, including impacts on vaccine responses and susceptibility to disease and its sequelae during infections. Here, we discuss how Fc structures arising from sialylation and fucosylation impact immunity, focusing on responses to vaccination and infection. We also review work defining individual differences in Fc glycosylation, regulation of Fc glycosylation, and clinical implications of these pathways.
Collapse
Affiliation(s)
- Taia T Wang
- Department of Medicine, Division of Infectious Diseases, and Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.,Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, New York, USA
| |
Collapse
|
44
|
Rojas M, Rodríguez Y, Monsalve DM, Acosta-Ampudia Y, Camacho B, Gallo JE, Rojas-Villarraga A, Ramírez-Santana C, Díaz-Coronado JC, Manrique R, Mantilla RD, Shoenfeld Y, Anaya JM. Convalescent plasma in Covid-19: Possible mechanisms of action. Autoimmun Rev 2020. [PMID: 32380316 DOI: 10.1016/j.autrev.2020.102554.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible of the coronavirus disease 2019 (COVID-19) pandemic. Therapeutic options including antimalarials, antivirals, and vaccines are under study. Meanwhile the current pandemic has called attention over old therapeutic tools to treat infectious diseases. Convalescent plasma (CP) constitutes the first option in the current situation, since it has been successfully used in other coronaviruses outbreaks. Herein, we discuss the possible mechanisms of action of CP and their repercussion in COVID-19 pathogenesis, including direct neutralization of the virus, control of an overactive immune system (i.e., cytokine storm, Th1/Th17 ratio, complement activation) and immunomodulation of a hypercoagulable state. All these benefits of CP are expected to be better achieved if used in non-critically hospitalized patients, in the hope of reducing morbidity and mortality.
Collapse
Affiliation(s)
- Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Bernardo Camacho
- Instituto Distrital de Ciencia Biotecnología e Investigación en Salud, IDCBIS, Bogota, Colombia
| | | | | | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | | | - Rubén Manrique
- Epidemiology and Biostatistics Research Group, Universidad CES, Medellin, Colombia
| | - Ruben D Mantilla
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to Tel-Aviv University, Tel Aviv, Israel; Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia.
| |
Collapse
|
45
|
The Effects of Pre-Existing Antibodies on Live-Attenuated Viral Vaccines. Viruses 2020; 12:v12050520. [PMID: 32397218 PMCID: PMC7290594 DOI: 10.3390/v12050520] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Live-attenuated vaccines (LAVs) have achieved remarkable successes in controlling virus spread, as well as for other applications such as cancer immunotherapy. However, with rapid increases in international travel, globalization, geographic spread of viral vectors, and widespread use of vaccines, there is an increasing need to consider how pre-exposure to viruses which share similar antigenic regions can impact vaccine efficacy. Pre-existing antibodies, derived from either from maternal–fetal transmission, or by previous infection or vaccination, have been demonstrated to interfere with vaccine immunogenicity of measles, adenovirus, and influenza LAVs. Immune interference of LAVs can be caused by the formation of virus–antibody complexes that neutralize virus infection in antigen-presenting cells, or by the cross-linking of the B-cell receptor with the inhibitory receptor, FcγRIIB. On the other hand, pre-existing antibodies can augment flaviviral LAV efficacy such as that of dengue and yellow fever virus, especially when pre-existing antibodies are present at sub-neutralizing levels. The increased vaccine immunogenicity can be facilitated by antibody-dependent enhancement of virus infection, enhancing virus uptake in antigen-presenting cells, and robust induction of innate immune responses that promote vaccine immunogenicity. This review examines the literature on this topic and examines the circumstances where pre-existing antibodies can inhibit or enhance LAV efficacy. A better knowledge of the underlying mechanisms involved could allow us to better manage immunization in seropositive individuals and even identify possibilities that could allow us to exploit pre-existing antibodies to boost vaccine-induced responses for improved vaccine efficacy.
Collapse
|
46
|
Rojas M, Rodríguez Y, Monsalve DM, Acosta-Ampudia Y, Camacho B, Gallo JE, Rojas-Villarraga A, Ramírez-Santana C, Díaz-Coronado JC, Manrique R, Mantilla RD, Shoenfeld Y, Anaya JM. Convalescent plasma in Covid-19: Possible mechanisms of action. Autoimmun Rev 2020; 19:102554. [PMID: 32380316 PMCID: PMC7198427 DOI: 10.1016/j.autrev.2020.102554] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 12/17/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible of the coronavirus disease 2019 (COVID-19) pandemic. Therapeutic options including antimalarials, antivirals, and vaccines are under study. Meanwhile the current pandemic has called attention over old therapeutic tools to treat infectious diseases. Convalescent plasma (CP) constitutes the first option in the current situation, since it has been successfully used in other coronaviruses outbreaks. Herein, we discuss the possible mechanisms of action of CP and their repercussion in COVID-19 pathogenesis, including direct neutralization of the virus, control of an overactive immune system (i.e., cytokine storm, Th1/Th17 ratio, complement activation) and immunomodulation of a hypercoagulable state. All these benefits of CP are expected to be better achieved if used in non-critically hospitalized patients, in the hope of reducing morbidity and mortality. Coronavirus disease 19 (COVID-19) is an emerging viral threat with major repercussions for public health. There is not specific treatment for COVID-19. Convalescent plasma (CP) emerges as the first option of management for hospitalized patients with COVID-19. Transference of neutralizing antibodies helps to control COVID-19 infection and modulates inflammatory response. Other plasma components may enhance the antiviral and anti-inflammatory properties of CP.
Collapse
Affiliation(s)
- Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Bernardo Camacho
- Instituto Distrital de Ciencia Biotecnología e Investigación en Salud, IDCBIS, Bogota, Colombia
| | | | | | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | | | - Rubén Manrique
- Epidemiology and Biostatistics Research Group, Universidad CES, Medellin, Colombia
| | - Ruben D Mantilla
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to Tel-Aviv University, Tel Aviv, Israel; Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia.
| |
Collapse
|
47
|
Wabnitz H, Khan R, Lazarus AH. The use of IVIg in fetal and neonatal alloimmune thrombocytopenia- Principles and mechanisms. Transfus Apher Sci 2019; 59:102710. [PMID: 31926738 DOI: 10.1016/j.transci.2019.102710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a rare neonatal disorder that is caused by alloimmunization against platelet antigens during pregnancy. Although rare, affecting only 1 in 1000 live births, it can cause intracranial hemorrhage and other bleeding complications that can lead to miscarriage, stillbirth and life-long neurological complications. One of the gold-standard therapies for at risk pregnancies is the administration of IVIg. Although IVIg has been used in a variety of different disorders for over 40 years, its exact mechanism of action is still unknown. In FNAIT, the majority of its therapeutic effect is thought the be mediated through the neonatal Fc receptor, however other mechanisms cannot be excluded. Due to safety, supply and other concerns that are associated with IVIg use, alternative therapies that could replace IVIg are additionally being investigated. This includes the possibility of a prophylaxis regimen for FNAIT, similarly to what has been successfully used in hemolytic disease of the fetus and newborn for over 50 years.
Collapse
Affiliation(s)
- Hanna Wabnitz
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada; Keenan Research Centre, Department of Laboratory Medicine, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada; Toronto Platelet Immunobiology Group (TPIG), Toronto, ON, M5B 1T8, Canada
| | - Ramsha Khan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada; Keenan Research Centre, Department of Laboratory Medicine, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada; Toronto Platelet Immunobiology Group (TPIG), Toronto, ON, M5B 1T8, Canada; Canadian Blood Services, Centre for Innovation, Ottawa, ON, K1G 4J5, Canada
| | - Alan H Lazarus
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada; Keenan Research Centre, Department of Laboratory Medicine, St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada; Toronto Platelet Immunobiology Group (TPIG), Toronto, ON, M5B 1T8, Canada; Canadian Blood Services, Centre for Innovation, Ottawa, ON, K1G 4J5, Canada; Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
48
|
Abstract
Glycosylation of IgG Fc domains is a central mechanism in the diversification of antibody function. Modifications to the core Fc glycan impact antibody function by shifting the balance of Type I and Type II Fc gamma receptors (FcγR) that will be engaged by immune complexes. This, in turn, modulates the effector cells and functions that can be recruited during immune activation. Critically, humans have evolved to regulate Fc glycan modifications for immune homeostasis. Dysregulation in Fc glycan modifications can lead to loss of immune tolerance, symptomatic autoimmunity, and susceptibility to infectious diseases. Here, we discuss IgG Fc glycosylation and its role in human health and disease.
Collapse
Affiliation(s)
- Taia T Wang
- Department of Medicine, Division of Infectious Diseases, Department of Microbiology and Immunology, Program in Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
49
|
Rodríguez Y, Vatti N, Ramírez-Santana C, Chang C, Mancera-Páez O, Gershwin ME, Anaya JM. Chronic inflammatory demyelinating polyneuropathy as an autoimmune disease. J Autoimmun 2019; 102:8-37. [DOI: 10.1016/j.jaut.2019.04.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/13/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022]
|
50
|
Hoefsmit EP, Rozeman EA, Haanen JBAG, Blank CU. Susceptible loci associated with autoimmune disease as potential biomarkers for checkpoint inhibitor-induced immune-related adverse events. ESMO Open 2019; 4:e000472. [PMID: 31423333 PMCID: PMC6677983 DOI: 10.1136/esmoopen-2018-000472] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/01/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Unprecedented successes regarding cancer immunotherapy have been achieved, in which therapeutic agents are used to target immune cells rather than cancer cells. The most effective immunotherapy to date is the group of immune checkpoint inhibitors (CPI), targeting, for example, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) or programmed cell death protein (PD-1). TThe combination of these therapies (anti-PD-1 with anti-CTLA-4) induces high response rates, and seem to be increased further when applied in early-stage disease. However, combined CTLA-4 plus PD-1 blockade causes frequent high-grade immune-related adverse events (irAE). To date, research on biological mechanism of irAEs is scarce and no widely accepted biomarkers predicting onset of severe irAEs have been identified. The similarity of irAEs to autoimmune disorders fuels the hypothesis that irAEs may be linked to susceptible genetic loci related to various autoimmune diseases. In this review, we extensively searched for susceptible loci associated with various autoimmune diseases, and pooled them in groups most likely to be associated with CPI-induced irAEs. These sets could be used in future research on predicting irAEs and guide physicians in a more refined and personal manner.
Collapse
Affiliation(s)
- Esmée P Hoefsmit
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elisa A Rozeman
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Medical Oncology Department, Netherlands Cancer Institute-Antoni van Leeuwenhoek Ziekenhuis, Amsterdam, The Netherlands
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Medical Oncology Department, Netherlands Cancer Institute-Antoni van Leeuwenhoek Ziekenhuis, Amsterdam, The Netherlands
| | - Christian U Blank
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands.,Medical Oncology Department, Netherlands Cancer Institute-Antoni van Leeuwenhoek Ziekenhuis, Amsterdam, The Netherlands
| |
Collapse
|