1
|
Harnessing IgG Fc glycosylation for clinical benefit. Curr Opin Immunol 2022; 77:102231. [PMID: 35797920 PMCID: PMC9870045 DOI: 10.1016/j.coi.2022.102231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 01/26/2023]
Abstract
The effector activity of IgG antibodies is regulated at several levels, including IgG subclass, modifications of the Fc glycan, and the distribution of Type I and II Fcγ receptors (FcγR) on effector cells. Here, we explore how Fc glycosylation, particularly sialylation and fucosylation, tunes cellular responses to immune complexes. We review the current understanding of the pathways and mechanisms underlying this biology, address FcγR in antigen presentation, and discuss aspects of the clinical understanding of Fc glycans in therapies and disease.
Collapse
|
2
|
Zuo Y, Deng GM. Fc Gamma Receptors as Regulators of Bone Destruction in Inflammatory Arthritis. Front Immunol 2021; 12:688201. [PMID: 34248975 PMCID: PMC8262610 DOI: 10.3389/fimmu.2021.688201] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/09/2021] [Indexed: 12/29/2022] Open
Abstract
Bone erosion is one of the primary features of inflammatory arthritis and is caused by excessive differentiation and activation of osteoclasts. Fc gamma receptors (FcγRs) have been implicated in osteoclastogenesis. Our recent studies demonstrate that joint-deposited lupus IgG inhibited RANKL-induced osteoclastogenesis. FcγRI is required for RANKL-induced osteoclastogenesis and lupus IgG-induced signaling transduction. We reviewed the results of studies that analyzed the association between FcγRs and bone erosion in inflammatory arthritis. The analysis revealed the dual roles of FcγRs in bone destruction in inflammatory arthritis. Thus, IgG/FcγR signaling molecules may serve as potential therapeutic targets against bone erosion.
Collapse
Affiliation(s)
- Yuyue Zuo
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guo-Min Deng
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Sun K, Li YY, Jin J. A double-edged sword of immuno-microenvironment in cardiac homeostasis and injury repair. Signal Transduct Target Ther 2021; 6:79. [PMID: 33612829 PMCID: PMC7897720 DOI: 10.1038/s41392-020-00455-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/14/2020] [Accepted: 11/15/2020] [Indexed: 02/07/2023] Open
Abstract
The response of immune cells in cardiac injury is divided into three continuous phases: inflammation, proliferation and maturation. The kinetics of the inflammatory and proliferation phases directly influence the tissue repair. In cardiac homeostasis, cardiac tissue resident macrophages (cTMs) phagocytose bacteria and apoptotic cells. Meanwhile, NK cells prevent the maturation and transport of inflammatory cells. After cardiac injury, cTMs phagocytose the dead cardiomyocytes (CMs), regulate the proliferation and angiogenesis of cardiac progenitor cells. NK cells prevent the cardiac fibrosis, and promote vascularization and angiogenesis. Type 1 macrophages trigger the cardioprotective responses and promote tissue fibrosis in the early stage. Reversely, type 2 macrophages promote cardiac remodeling and angiogenesis in the late stage. Circulating macrophages and neutrophils firstly lead to chronic inflammation by secreting proinflammatory cytokines, and then release anti-inflammatory cytokines and growth factors, which regulate cardiac remodeling. In this process, dendritic cells (DCs) mediate the regulation of monocyte and macrophage recruitment. Recruited eosinophils and Mast cells (MCs) release some mediators which contribute to coronary vasoconstriction, leukocyte recruitment, formation of new blood vessels, scar formation. In adaptive immunity, effector T cells, especially Th17 cells, lead to the pathogenesis of cardiac fibrosis, including the distal fibrosis and scar formation. CMs protectors, Treg cells, inhibit reduce the inflammatory response, then directly trigger the regeneration of local progenitor cell via IL-10. B cells reduce myocardial injury by preserving cardiac function during the resolution of inflammation.
Collapse
Affiliation(s)
- Kang Sun
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Yi-Yuan Li
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, China.
| | - Jin Jin
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
- Sir Run Run Shaw Hospital, College of Medicine Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
4
|
Rijnink WF, Ottenhoff THM, Joosten SA. B-Cells and Antibodies as Contributors to Effector Immune Responses in Tuberculosis. Front Immunol 2021; 12:640168. [PMID: 33679802 PMCID: PMC7930078 DOI: 10.3389/fimmu.2021.640168] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/29/2021] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is still a major threat to mankind, urgently requiring improved vaccination and therapeutic strategies to reduce TB-disease burden. Most present vaccination strategies mainly aim to induce cell-mediated immunity (CMI), yet a series of independent studies has shown that B-cells and antibodies (Abs) may contribute significantly to reduce the mycobacterial burden. Although early studies using B-cell knock out animals did not support a major role for B-cells, more recent studies have provided new evidence that B-cells and Abs can contribute significantly to host defense against Mtb. B-cells and Abs exist in many different functional subsets, each equipped with unique functional properties. In this review, we will summarize current evidence on the contribution of B-cells and Abs to immunity toward Mtb, their potential utility as biomarkers, and their functional contribution to Mtb control.
Collapse
Affiliation(s)
- Willemijn F Rijnink
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
5
|
Khare P, Sun W, Ramakrishnan S, Swiercz R, Hao G, Lo ST, Nham K, Sun X, Ober RJ, Ward ES. Selective depletion of radiolabeled HER2-specific antibody for contrast improvement during PET. MAbs 2021; 13:1976705. [PMID: 34592895 PMCID: PMC8489906 DOI: 10.1080/19420862.2021.1976705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/22/2021] [Accepted: 09/01/2021] [Indexed: 12/20/2022] Open
Abstract
The prolonged in vivo persistence of antibodies results in high background and poor contrast during their use as molecular imaging agents for positron emission tomography (PET). We have recently described a class of engineered Fc fusion proteins that selectively deplete antigen-specific antibodies without affecting the levels of antibodies of other specificities. Here, we demonstrate that these Fc fusions (called Seldegs, for selective degradation) can be used to clear circulating, radiolabeled HER2-specific antibody during diagnostic imaging of HER2-positive tumors in mice. The analyses show that Seldegs have considerable promise for the reduction of whole-body exposure to radiolabel and improvement of contrast during PET.
Collapse
Affiliation(s)
- Priyanka Khare
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Wei Sun
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Sreevidhya Ramakrishnan
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Rafal Swiercz
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
- Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Guiyang Hao
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Su-Tang Lo
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kien Nham
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Raimund J. Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
- Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - E. Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
- Cancer Sciences Unit, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
6
|
Zhang X, Owens J, Olsen HS, So E, Burch E, McCroskey MC, Li X, Weber GL, Bennett D, Rybin D, Zhou H, Hao H, Mérigeon EY, Block DS, LaRosa G, Strome SE. A recombinant human IgG1 Fc multimer designed to mimic the active fraction of IVIG in autoimmunity. JCI Insight 2019; 4:121905. [PMID: 30674715 DOI: 10.1172/jci.insight.121905] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 12/11/2018] [Indexed: 12/30/2022] Open
Abstract
The antiinflammatory effects of i.v. Ig (IVIG) in the treatment of autoimmune disease are due, in part, to the Fc fragments of Ig aggregates. In order to capitalize on the known antiinflammatory and tolerogenic properties of Ig Fc aggregates, we created a recombinant human IgG1 Fc multimer, GL-2045. In vitro, GL-2045 demonstrated high-avidity binding to Fc receptors, blocked the binding of circulating immune complexes from patients with rheumatoid arthritis to human Fcγ receptors (FcγRs), and inhibited antibody-mediated phagocytosis at log order-lower concentrations than IVIG. In vivo, administration of GL-2045 conferred partial protection against antibody-mediated platelet loss in a murine immune thrombocytopenic purpura (ITP) model. GL-2045 also suppressed disease activity in a therapeutic model of murine collagen-induced arthritis (CIA), which was associated with reduced circulating levels of IL-6. Furthermore, GL-2045 administration to nonhuman primates (NHPs) transiently increased systemic levels of the antiinflammatory cytokines IL-10 and IL-1RA, reduced the proinflammatory cytokine IL-8, and decreased surface expression of CD14 and HLA-DR on monocytes. These findings demonstrate the immunomodulatory properties of GL-2045 and suggest that it has potential as a treatment for autoimmune and inflammatory diseases, as a recombinant alternative to IVIG.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jane Owens
- Pfizer Inc., Cambridge, Massachusetts, USA
| | | | - Edward So
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Erin Burch
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | - Hua Zhou
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Haiping Hao
- Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | | | - Scott E Strome
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Ulrichts P, Guglietta A, Dreier T, van Bragt T, Hanssens V, Hofman E, Vankerckhoven B, Verheesen P, Ongenae N, Lykhopiy V, Enriquez FJ, Cho J, Ober RJ, Ward ES, de Haard H, Leupin N. Neonatal Fc receptor antagonist efgartigimod safely and sustainably reduces IgGs in humans. J Clin Invest 2018; 128:4372-4386. [PMID: 30040076 DOI: 10.1172/jci97911] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/03/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Intravenous Ig (IVIg), plasma exchange, and immunoadsorption are frequently used in the management of severe autoimmune diseases mediated by pathogenic IgG autoantibodies. These approaches modulating IgG levels can, however, be associated with some severe adverse reactions and a substantial burden to patients. Targeting the neonatal Fc receptor (FcRn) presents an innovative and potentially more effective, safer, and more convenient alternative for clearing pathogenic IgGs. METHODS A randomized, double-blind, placebo-controlled first-in-human study was conducted in 62 healthy volunteers to explore single and multiple ascending intravenous doses of the FcRn antagonist efgartigimod. The study objectives were to assess safety, tolerability, pharmacokinetics, pharmacodynamics, and immunogenicity. The findings of this study were compared with the pharmacodynamics profile elicited by efgartigimod in cynomolgus monkeys. RESULTS Efgartigimod treatment resulted in a rapid and specific clearance of serum IgG levels in both cynomolgus monkeys and healthy volunteers. In humans, single administration of efgartigimod reduced IgG levels up to 50%, while multiple dosing further lowered IgGs on average by 75% of baseline levels. Approximately 8 weeks following the last administration, IgG levels returned to baseline. Efgartigimod did not alter the homeostasis of albumin or Igs other than IgG, and no serious adverse events related to efgartigimod infusion were observed. CONCLUSION Antagonizing FcRn using efgartigimod is safe and results in a specific, profound, and sustained reduction of serum IgG levels. These results warrant further evaluation of this therapeutic approach in IgG-driven autoimmune diseases. TRIAL REGISTRATION Clinicaltrials.gov NCT03457649. FUNDING argenx BVBA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - JunHaeng Cho
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA.,Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - E Sally Ward
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas, USA.,Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, Bryan, Texas, USA
| | | | | |
Collapse
|
8
|
Liu P, Li L, Fan P, Zheng J, Zhao D. High-dose of intravenous immunoglobulin modulates immune tolerance in premature infants. BMC Pediatr 2018; 18:74. [PMID: 29466960 PMCID: PMC5822672 DOI: 10.1186/s12887-018-1055-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 02/07/2018] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Intravenous immunoglobulin (IVIG) is commonly used to improve the immunomodulatory effects, although its regulatory effect on premature Treg cells is unclear. The purpose of this study is to study the effect of high dose of IVIG (HD-IVIG) on Treg cells expression and cytokine profile in premature birth. METHODS Fifty-two premature infants were enrolled in this study and thirty-one premature infants who were suspected to have intrauterine infection received HD-IVIG (1-2 g/kg) at the first day of birth; the remaining 21 premature infants were assigned as the control group. The peripheral blood CD4 + T and foxp3+ Treg cells were checked by flow cytometry, and cytokine concentrations were detected by cytometric bead array. RESULTS With the gestational age growth, peripheral blood CD4 + T and foxp3+ Treg cells of prematurity gradually declined from 50% to 35% and from 8% to 6%, respectively. Meanwhile, HD-IVIG increased the percentage of CD4 + T and foxp3+ Treg cells compared with their baseline levels (p < 0.001). HD-IVIG demonstrated different regulating effects on cytokines secretion, increased IL-17 and TGF-β, and inhibited IL-6 secretion. CONCLUSION Our results demonstrated that HD-IVIG not only enhanced the premature immune tolerance, but also suppressed the excessive inflammation response mediated by IL-6. TRIAL REGISTRATION This study was under the clinical study registration (ChiCTR-ORC-16008872, date of registration, 2016-07-21).
Collapse
Affiliation(s)
- Pin Liu
- Pediatrics and Neonatology Department, Zhongnan Hospital of Wuhan University, Donghu road 169, Wuhan, 430071 China
| | - Lijun Li
- Pediatrics and Neonatology Department, Zhongnan Hospital of Wuhan University, Donghu road 169, Wuhan, 430071 China
| | - Panpan Fan
- Pediatrics and Neonatology Department, Zhongnan Hospital of Wuhan University, Donghu road 169, Wuhan, 430071 China
| | - Junwen Zheng
- Pediatrics and Neonatology Department, Zhongnan Hospital of Wuhan University, Donghu road 169, Wuhan, 430071 China
| | - Dongchi Zhao
- Pediatrics and Neonatology Department, Zhongnan Hospital of Wuhan University, Donghu road 169, Wuhan, 430071 China
| |
Collapse
|
9
|
Pagan JD, Kitaoka M, Anthony RM. Engineered Sialylation of Pathogenic Antibodies In Vivo Attenuates Autoimmune Disease. Cell 2018; 172:564-577.e13. [PMID: 29275858 PMCID: PMC5849077 DOI: 10.1016/j.cell.2017.11.041] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/20/2017] [Accepted: 11/21/2017] [Indexed: 12/31/2022]
Abstract
Self-reactive IgGs contribute to the pathology of autoimmune diseases, including systemic lupus erythematosus and rheumatoid arthritis. Paradoxically, IgGs are used to treat inflammatory diseases in the form of high-dose intravenous immunoglobulin (IVIG). Distinct glycoforms on the IgG crystallizable fragment (Fc) dictate these divergent functions. IgG anti-inflammatory activity is attributed to sialylation of the Fc glycan. We therefore sought to convert endogenous IgG to anti-inflammatory mediators in vivo by engineering solubilized glycosyltransferases that attach galactose or sialic acid. When both enzymes were administered in a prophylactic or therapeutic fashion, autoimmune inflammation was markedly attenuated in vivo. The enzymes worked through a similar pathway to IVIG, requiring DC-SIGN, STAT6 signaling, and FcγRIIB. Importantly, sialylation was highly specific to pathogenic IgG at the site of inflammation, driven by local platelet release of nucleotide-sugar donors. These results underscore the therapeutic potential of glycoengineering in vivo.
Collapse
Affiliation(s)
- Jose D Pagan
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Maya Kitaoka
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Robert M Anthony
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
10
|
Effects of Acyclovir and IVIG on Behavioral Outcomes after HSV1 CNS Infection. Behav Neurol 2017; 2017:5238402. [PMID: 29358844 PMCID: PMC5735307 DOI: 10.1155/2017/5238402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/06/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Herpes simplex virus 1 (HSV) encephalitis (HSE) has serious neurological complications, involving behavioral and cognitive impairments that cause significant morbidity and a reduced quality of life. We showed that HSE results from dysregulated central nervous system (CNS) inflammatory responses. We hypothesized that CNS inflammation is casually involved in behavioral abnormalities after HSE and that treatment with ACV and pooled human immunoglobulin (IVIG), an immunomodulatory drug, would improve outcomes compared to mice treated with phosphate buffered saline (PBS) or ACV alone. Anxiety levels were high in HSV-infected PBS and ACV-treated mice compared to mice treated with ACV + IVIG, consistent with reports implicating inflammation in anxiety induced by lipopolysaccharide (LPS) or stress. Female, but not male, PBS-treated mice were cognitively impaired, and unexpectedly, ACV was protective, while the inclusion of IVIG surprisingly antagonized ACV's beneficial effects. Distinct serum proteomic profiles were observed for male and female mice, and the antagonistic effects of ACV and IVIG on behavior were paralleled by similar changes in the serum proteome of ACV- and ACV + IVIG-treated mice. We conclude that inflammation and other factors mediate HSV-induced behavioral impairments and that the effects of ACV and IVIG on behavior involve novel mechanisms.
Collapse
|
11
|
van de Bovenkamp FS, Hafkenscheid L, Rispens T, Rombouts Y. The Emerging Importance of IgG Fab Glycosylation in Immunity. THE JOURNAL OF IMMUNOLOGY 2016; 196:1435-41. [PMID: 26851295 DOI: 10.4049/jimmunol.1502136] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human IgG is the most abundant glycoprotein in serum and is crucial for protective immunity. In addition to conserved IgG Fc glycans, ∼15-25% of serum IgG contains glycans within the variable domains. These so-called "Fab glycans" are primarily highly processed complex-type biantennary N-glycans linked to N-glycosylation sites that emerge during somatic hypermutation. Specific patterns of Fab glycosylation are concurrent with physiological and pathological conditions, such as pregnancy and rheumatoid arthritis. With respect to function, Fab glycosylation can significantly affect stability, half-life, and binding characteristics of Abs and BCRs. Moreover, Fab glycans are associated with the anti-inflammatory activity of IVIgs. Consequently, IgG Fab glycosylation appears to be an important, yet poorly understood, process that modulates immunity.
Collapse
Affiliation(s)
- Fleur S van de Bovenkamp
- Department of Immunopathology, Sanquin Research, 1066 CX Amsterdam, the Netherlands; Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Lise Hafkenscheid
- Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research, 1066 CX Amsterdam, the Netherlands; Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands;
| | - Yoann Rombouts
- Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and Université Lille, CNRS, UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle, F 59 000 Lille, France
| |
Collapse
|
12
|
Abstract
Antibodies against surface molecules of human tumors are now frequently administered in combination with strong chemotherapy, increasing therapeutic efficacy but making the task of elucidating immunological events more difficult. Experiments on genetically manipulated mice indicate that antibody efficacy is greatest when IgG antibody coating tumor cells is engaged by the Fcγ-receptors of effector cells, chiefly the monocyte/macrophage lineage. Evidence suggests lesser roles for NK cells, neutrophils, receptor-mediated cytotoxicity and complement-mediated cytotoxicity. The classical mode of killing employed by macrophages is phagocytosis, but much has to be learned about optimally activating macrophages for this task, and about any other modes of cytotoxicity used. There is renewed interest in antigenic modulation, which implies removal of therapeutic antibody linked with antigen from target-cell surfaces. It is now apparent that this removal of immune complexes can be achieved either by internalization by the target cell, or by transfer of the complexes to another cell by trogocytosis. In trials, anti-idiotype antibodies surprisingly proved therapeutically more effective than anti-CD20, despite anti-idiotype being more effectively removed from target-cell surfaces by antigenic modulation. This anomalous result might reflect the fact that persistence of anti-CD20 immune complexes in large amounts induces serious effector modulation, which paralyzes macrophage attacks on antibody-coated cells. The case for effector modulation is argued by analogy with the therapeutic suppression of autoimmune inflammation by effector modulation, achieved by infusion either of normal IgG in large amounts, or of anti-red cell IgG in relatively small amounts.
Collapse
Affiliation(s)
- George T Stevenson
- University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| |
Collapse
|
13
|
Epelman S, Liu PP, Mann DL. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat Rev Immunol 2015; 15:117-29. [PMID: 25614321 DOI: 10.1038/nri3800] [Citation(s) in RCA: 439] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the advances that have been made in developing new therapeutics, cardiovascular disease remains the leading cause of worldwide mortality. Therefore, understanding the mechanisms underlying cardiovascular tissue injury and repair is of prime importance. Following cardiac tissue injury, the immune system has an important and complex role in driving both the acute inflammatory response and the regenerative response. This Review summarizes the role of the immune system in cardiovascular disease - focusing on the idea that the immune system evolved to promote tissue homeostasis following injury and/or infection, and that the inherent cost of this evolutionary development is unwanted inflammatory damage.
Collapse
Affiliation(s)
- Slava Epelman
- Toronto Medical Discovery Tower, 101 College Street, TMDT 3903 Toronto, Ontario, M5G 1L7, Canada
| | - Peter P Liu
- University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario, K1Y 4W7, Canada
| | - Douglas L Mann
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
14
|
Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, Flynn J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin Immunol 2014; 26:588-600. [PMID: 25458990 PMCID: PMC4314354 DOI: 10.1016/j.smim.2014.10.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 12/24/2022]
Abstract
Mycobacterium tuberculosis remains a major public health burden. It is generally thought that while B cell- and antibody-mediated immunity plays an important role in host defense against extracellular pathogens, the primary control of intracellular microbes derives from cellular immune mechanisms. Studies on the immune regulatory mechanisms during infection with M. tuberculosis, a facultative intracellular organism, has established the importance of cell-mediated immunity in host defense during tuberculous infection. Emerging evidence suggest a role for B cell and humoral immunity in the control of intracellular pathogens, including obligatory species, through interactions with the cell-mediated immune compartment. Recent studies have shown that B cells and antibodies can significantly impact on the development of immune responses to the tubercle bacillus. In this review, we present experimental evidence supporting the notion that the importance of humoral and cellular immunity in host defense may not be entirely determined by the niche of the pathogen. A comprehensive approach that examines both humoral and cellular immunity could lead to better understanding of the immune response to M. tuberculosis.
Collapse
Affiliation(s)
- John Chan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Simren Mehta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sushma Bharrhan
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yong Chen
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jacqueline M Achkar
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Arturo Casadevall
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - JoAnne Flynn
- Departments of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
15
|
Domínguez-Soto A, de las Casas-Engel M, Bragado R, Medina-Echeverz J, Aragoneses-Fenoll L, Martín-Gayo E, van Rooijen N, Berraondo P, Toribio ML, Moro MA, Cuartero I, Castrillo A, Sancho D, Sánchez-Torres C, Bruhns P, Sánchez-Ramón S, Corbí AL. Intravenous immunoglobulin promotes antitumor responses by modulating macrophage polarization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:5181-9. [PMID: 25326025 DOI: 10.4049/jimmunol.1303375] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intravenous Igs (IVIg) therapy is widely used as an immunomodulatory strategy in inflammatory pathologies and is suggested to promote cancer regression. Because progression of tumors depends on their ability to redirect the polarization state of tumor-associated macrophages (from M1/immunogenic/proinflammatory to M2/anti-inflammatory), we have evaluated whether IVIg limits tumor progression and dissemination through modulation of macrophage polarization. In vitro, IVIg inhibited proinflammatory cytokine production from M1 macrophages and induced a M2-to-M1 polarization switch on human and murine M2 macrophages. In vivo, IVIg modified the polarization of tumor-associated myeloid cells in a Fcεr1γ chain-dependent manner, modulated cytokine blood levels in tumor-bearing animals, and impaired tumor progression via FcγRIII (CD16), FcγRIV, and FcRγ engagement, the latter two effects being macrophage mediated. Therefore, IVIg immunomodulatory activity is dependent on the polarization state of the responding macrophages, and its ability to trigger a M2-to-M1 macrophage polarization switch might be therapeutically useful in cancer, in which proinflammatory or immunogenic functions should be promoted.
Collapse
Affiliation(s)
- Angeles Domínguez-Soto
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain;
| | - Mateo de las Casas-Engel
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain
| | - Rafael Bragado
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Madrid 28040, Spain
| | | | - Laura Aragoneses-Fenoll
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain
| | - Enrique Martín-Gayo
- Centro de Biología Molecular, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Nico van Rooijen
- Department of Molecular Cell Biology, Free University Medical Center, Amsterdam 1081, the Netherlands
| | | | - María L Toribio
- Centro de Biología Molecular, Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - María A Moro
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Isabel Cuartero
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Consejo Superior de Investigaciones Cientificas-Universidad Autónoma de Madrid, Madrid 28029, Spain; Unidad Asociada de Biomedicina, Consejo Superior de Investigaciones Cientificas-Universidad de Las Palmas de Gran Canarias 35001, Spain
| | - David Sancho
- Fundación Centro Nacional de Investigaciones Cardiovasculares, Centro Nacional de Investigaciones Cardiovasculares, Madrid 28029, Spain
| | - Carmen Sánchez-Torres
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico
| | - Pierre Bruhns
- Institut Pasteur, Département d'Immunologie, Unité des Anticorps en Thérapie et Pathologie, Paris 75015, France; INSERM U760, Paris 75015, France; and
| | | | - Angel L Corbí
- Centro de Investigaciones Biológicas/Consejo Superior de Investigaciones Cientificas, Madrid 28040, Spain;
| |
Collapse
|
16
|
Swiercz R, Chiguru S, Tahmasbi A, Ramezani SM, Hao G, Challa DK, Lewis MA, Kulkarni PV, Sun X, Ober RJ, Mason RP, Ward ES. Use of Fc-Engineered Antibodies as Clearing Agents to Increase Contrast During PET. J Nucl Med 2014; 55:1204-7. [PMID: 24868106 DOI: 10.2967/jnumed.113.136481] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/02/2014] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Despite promise for the use of antibodies as molecular imaging agents in PET, their long in vivo half-lives result in poor contrast and radiation damage to normal tissue. This study describes an approach to overcome these limitations. METHODS Mice bearing human epidermal growth factor receptor type 2 (HER2)-overexpressing tumors were injected with radiolabeled ((124)I, (125)I) HER2-specific antibody (pertuzumab). Pertuzumab injection was followed 8 h later by the delivery of an engineered, antibody-based inhibitor of the receptor, FcRn. Biodistribution analyses and PET were performed at 24 and 48 h after pertuzumab injection. RESULTS The delivery of the engineered, antibody-based FcRn inhibitor (or Abdeg, for antibody that enhances IgG degradation) results in improved tumor-to-blood ratios, reduced systemic exposure to radiolabel, and increased contrast during PET. CONCLUSION Abdegs have considerable potential as agents to stringently regulate antibody dynamics in vivo, resulting in increased contrast during molecular imaging with PET.
Collapse
Affiliation(s)
- Rafal Swiercz
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Srinivas Chiguru
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Amir Tahmasbi
- Department of Electrical Engineering, University of Texas at Dallas, Richardson, Texas
| | - Saleh M Ramezani
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Guiyang Hao
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Dilip K Challa
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Matthew A Lewis
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Padmakar V Kulkarni
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Xiankai Sun
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - Raimund J Ober
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas Department of Electrical Engineering, University of Texas at Dallas, Richardson, Texas
| | - Ralph P Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas; and
| | - E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
17
|
Ward ES, Velmurugan R, Ober RJ. Targeting FcRn for therapy: from live cell imaging to in vivo studies in mice. Immunol Lett 2014; 160:158-62. [PMID: 24572175 DOI: 10.1016/j.imlet.2014.02.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 12/18/2022]
Abstract
The role of FcRn in regulating antibody levels and transport in the body is well documented. The use of fluorescence microscopy to investigate the subcellular trafficking behavior of FcRn and its IgG ligand has led to insight into the function of this receptor, including the identification of new intracellular pathways. The inhibition of FcRn using engineered antibodies that bind to this receptor with increased affinity through their Fc region can be exploited to treat antibody mediated autoimmunity. The efficacy of this approach in mouse models of arthritis and multiple sclerosis has been demonstrated. Finally, the cross-species difference between mouse and man for FcRn-IgG interactions needs to be considered when engineering antibodies for improved activities in FcRn-mediated functions.
Collapse
Affiliation(s)
- E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| | - Ramraj Velmurugan
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Biomedical Engineering Graduate Program, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas TX 75390, USA.
| | - Raimund J Ober
- Department of Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Department of Electrical Engineering, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
18
|
Niknami M, Wang MX, Nguyen T, Pollard JD. Beneficial effect of a multimerized immunoglobulin Fc in an animal model of inflammatory neuropathy (experimental autoimmune neuritis). J Peripher Nerv Syst 2014; 18:141-52. [PMID: 23781961 DOI: 10.1111/jns5.12022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Intravenous immunoglobulin (IVIg) is one of the first-line therapies for inflammatory neuropathies. Clinical use of IVIg for these disorders is limited by expense and availability. Here, we investigated a synthetic product alternative to IVIg. The aim of this study was to test the therapeutic efficacy of a novel recombinant polyvalent murine IgG2a Fc compound (stradomer™) in experimental autoimmune neuritis (EAN). Seventy-four Lewis rats were immunized with myelin, randomized into three groups, and were treated with albumin, IVIg, or stradomer at 1% of IVIg dose. Rats were assessed clinically, electrophysiologically, and histologically. The clinical disease severity was evaluated by clinical grading and weight changes. The electrophysiological studies recorded motor conduction velocity (MCV), amplitudes, and latencies of the evoked compound muscle action potential (CMAP) and spinal somatosensory evoked potential. The treatment efficacy of the IVIg and stradomer groups was compared to the albumin (control) group. We demonstrate that stradomer has a similar therapeutic efficacy to human IVIg in EAN. Rats receiving stradomer or IVIg showed significantly lower clinical scores and less prominent weight loss compared with controls. A statistically significant improvement in both MCV and the amplitudes of distal and proximal evoked CMAP was observed in the stradomer and IVIg groups. Finally, treatment with both IVIg and stradomer resulted in statistically less inflammation and demyelinating changes in the sciatic nerve as evidenced by lower histological grade. These results reveal the potential of using fully recombinant multimerized immunoglobulin Fc instead of IVIg for treating inflammatory neuropathies.
Collapse
Affiliation(s)
- Marzieh Niknami
- Department of Neurology, Central Clinical School, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
19
|
Kyrmizi I, Ioannou M, Hatziapostolou M, Tsichlis PN, Boumpas DT, Tassiulas I. Tpl2 kinase regulates FcγR signaling and immune thrombocytopenia in mice. J Leukoc Biol 2013; 94:751-7. [PMID: 23898046 DOI: 10.1189/jlb.0113039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The MAPK3 Tpl2 controls innate and adaptive immunity by regulating TLR, TNF-α, and GPCR signaling in a variety of cell types. Its ablation gives rise to an anti-inflammatory phenotype characterized by resistance to LPS-induced endotoxin shock, DSS-induced colitis, and TNF-α-induced IBD. Here, we address the role of Tpl2 in autoimmunity. Our data show that the ablation and the pharmacological inhibition of Tpl2 protect mice from antiplatelet antibody-induced thrombocytopenia, a model of ITP. Thrombocytopenia in this model and in ITP is caused by phagocytosis of platelets opsonized with antiplatelet antibodies and depends on FcγR activation in splenic and hepatic myeloid cells. Further studies explained how Tpl2 inhibition protects from antibody-induced thrombocytopenia, by showing that Tpl2 is activated by FcγR signals in macrophages and that its activation by these signals is required for ERK activation, cytoplasmic Ca(2+) influx, the induction of cytokine and coreceptor gene expression, and phagocytosis.
Collapse
Affiliation(s)
- Irene Kyrmizi
- 2.Div. of Allergy, Clinical Immunology and Rheumatology, New York Medical College, 40 Sunshine Cottage Rd., Valhalla, NY 10595, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Ramakrishna C, Openshaw H, Cantin EM. The case for immunomodulatory approaches in treating HSV encephalitis. Future Virol 2013; 8:259-272. [PMID: 23956785 DOI: 10.2217/fvl.12.138] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
HSV encephalitis (HSE) is the most prevalent sporadic viral encephalitis. Although safe and effective antiviral therapies and greatly improved noninvasive diagnostic procedures have significantly improved outcomes, mortality (~20%) and debilitating neurological sequelae in survivors remain unacceptably high. An encouraging new development is that the focus is now shifting away from the virus exclusively, to include consideration of the host immune response to infection in the pathology underlying development of HSE. In this article, the authors discuss results from recent studies in experimental mouse models, as well as clinical reports that demonstrate a role for exaggerated host inflammatory responses in the brain in the development of HSE that is motivating researchers and clinicians to consider new therapeutic approaches for treating HSE. The authors also discuss results from a few studies that have shown that immunomodulatory drugs can be highly protective against HSE, which supports a role for deleterious host inflammatory responses in HSE. The impressive outcomes of some immunomodulatory approaches in mouse models of HSE emphasize the urgent need for clinical trials to rigorously evaluate combination antiviral and immunomodulatory therapy in comparison with standard antiviral therapy for treatment of HSE, and support for such an initiative is gaining momentum.
Collapse
Affiliation(s)
- Chandran Ramakrishna
- Department of Virology, Beckman Research Institute of City of Hope; Duarte, CA 91010-3000, USA
| | | | | |
Collapse
|
21
|
Kozakiewicz L, Phuah J, Flynn J, Chan J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:225-50. [PMID: 23468112 DOI: 10.1007/978-1-4614-6111-1_12] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tuberculosis (TB) remains a serious threat to public health, causing 2 million deaths annually world-wide. The control of TB has been hindered by the requirement of long duration of treatment involving multiple chemotherapeutic agents, the increased susceptibility to Mycobacterium tuberculosis infection in the HIV-infected population, and the development of multi-drug resistant and extensively resistant strains of tubercle bacilli. An efficacious and cost-efficient way to control TB is the development of effective anti-TB vaccines. This measure requires thorough understanding of the immune response to M. tuberculosis. While the role of cell-mediated immunity in the development of protective immune response to the tubercle bacillus has been well established, the role of B cells in this process is not clearly understood. Emerging evidence suggests that B cells and humoral immunity can modulate the immune response to various intracellular pathogens, including M. tuberculosis. These lymphocytes form conspicuous aggregates in the lungs of tuberculous humans, non-human primates, and mice, which display features of germinal center B cells. In murine TB, it has been shown that B cells can regulate the level of granulomatous reaction, cytokine production, and the T cell response. This chapter discusses the potential mechanisms by which specific functions of B cells and humoral immunity can shape the immune response to intracellular pathogens in general, and to M. tuberculosis in particular. Knowledge of the B cell-mediated immune response to M. tuberculosis may lead to the design of novel strategies, including the development of effective vaccines, to better control TB.
Collapse
Affiliation(s)
- Lee Kozakiewicz
- Department of Medicine and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | |
Collapse
|
22
|
Communication in the heart: the role of the innate immune system in coordinating cellular responses to ischemic injury. J Cardiovasc Transl Res 2012; 5:827-36. [PMID: 23054658 DOI: 10.1007/s12265-012-9410-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 09/05/2012] [Indexed: 12/14/2022]
Abstract
Ischemic cardiac injury is the leading cause of heart failure and mortality in the USA and is a major expense to health-care systems. Once the heart is injured, a highly dynamic and coordinated immune response is initiated, which is dependent on both resident and recruited leukocytes. The goal of the inflammatory response is to remove ischemic and necrotic material and to promote infarct healing. If this system is perturbed, the myocardium heals poorly, leading to significant left ventricular dysfunction. Understanding how inflammatory cells coordinate and interact with each other is required prior to designing therapeutic interventions that target pathological processes at play and leave untouched those processes that are protective. This review will discuss the intercellular cross talk between cells of the innate immune system following myocardial ischemic injury and how that response is coordinated over time.
Collapse
|
23
|
Intravenous immunoglobulin in the management of lupus nephritis. Autoimmune Dis 2012; 2012:589359. [PMID: 23056926 PMCID: PMC3465901 DOI: 10.1155/2012/589359] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 11/17/2022] Open
Abstract
The occurrence of nephritis in patients with systemic lupus erythematosus is associated with increased morbidity and mortality. The pathogenesis of lupus nephritis is complex, involving innate and adaptive cellular and humoral immune responses. Autoantibodies in particular have been shown to be critical in the initiation and progression of renal injury, via interactions with both Fc-receptors and complement. One approach in the management of patients with lupus nephritis has been the use of intravenous immunoglobulin. This therapy has shown benefit in the setting of many forms of autoantibody-mediated injury; however, the mechanisms of efficacy are not fully understood. In this paper, the data supporting the use of immunoglobulin therapy in lupus nephritis will be evaluated. In addition, the potential mechanisms of action will be discussed with respect to the known involvement of complement and Fc-receptors in the kidney parenchyma. Results are provocative and warrant additional clinical trials.
Collapse
|
24
|
Jain A, Olsen HS, Vyzasatya R, Burch E, Sakoda Y, Mérigeon EY, Cai L, Lu C, Tan M, Tamada K, Schulze D, Block DS, Strome SE. Fully recombinant IgG2a Fc multimers (stradomers) effectively treat collagen-induced arthritis and prevent idiopathic thrombocytopenic purpura in mice. Arthritis Res Ther 2012; 14:R192. [PMID: 22906120 PMCID: PMC3580588 DOI: 10.1186/ar4024] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 08/20/2012] [Indexed: 12/21/2022] Open
Abstract
Introduction Soluble immune aggregates bearing intact Fc fragments are effective treatment for a variety of autoimmune disorders in mice. The better to understand the mechanisms by which Fc-bearing immune complexes suppress autoimmunity, and to develop a platform for clinical translation, we created a series of fully recombinant forms of polyvalent IgG2a Fc, termed stradomers, and tested their efficacy in a therapeutic model of collagen-induced arthritis (CIA) and preventive models of both idiopathic thrombocytopenic purpura (ITP) and graft-versus-host disease (GVHD). Methods Stradomers were created by engineering either the human IgG2 hinge sequence (IgG2H) or the isoleucine zipper (ILZ) onto either the carboxy or amino termini of murine IgG2a Fc. Multimerization and binding to the canonical Fc receptors and the C-type lectin SIGN-RI were evaluated by using sodium dodecylsulfate-polymerase chain reaction (SDS-PAGE) and Biacore/Octet assays. The efficacy of stradomers in alleviating CIA and preventing ITP and GVHD was compared with "gold standard" therapies, including prednisolone and intravenous immune globulin (IVIG). Results Stradomers exist as both homodimeric and highly ordered sequential multimers. Higher-order multimers demonstrate increasingly stable associations with the canonic Fcγ receptors (FcγRs), and SIGN-R1, and are more effective than Fc homodimers in treating CIA. Furthermore, stradomers confer partial protection against platelet loss in a murine model ITP, but do not prevent GVHD. Conclusion These data suggest that fully human stradomers might serve as valuable tools for the treatment of selected autoimmune disorders and as reagents to study the function of Fc:FcR interactions in vivo.
Collapse
|
25
|
Cross-presentation of IgG-containing immune complexes. Cell Mol Life Sci 2012; 70:1319-34. [PMID: 22847331 DOI: 10.1007/s00018-012-1100-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 07/09/2012] [Accepted: 07/17/2012] [Indexed: 12/23/2022]
Abstract
IgG is a molecule that functionally combines facets of both innate and adaptive immunity and therefore bridges both arms of the immune system. On the one hand, IgG is created by adaptive immune cells, but can be generated by B cells independently of T cell help. On the other hand, once secreted, IgG can rapidly deliver antigens into intracellular processing pathways, which enable efficient priming of T cell responses towards epitopes from the cognate antigen initially bound by the IgG. While this process has long been known to participate in CD4(+) T cell activation, IgG-mediated delivery of exogenous antigens into a major histocompatibility complex (MHC) class I processing pathway has received less attention. The coordinated engagement of IgG with IgG receptors expressed on the cell-surface (FcγR) and within the endolysosomal system (FcRn) is a highly potent means to deliver antigen into processing pathways that promote cross-presentation of MHC class I and presentation of MHC class II-restricted epitopes within the same dendritic cell. This review focuses on the mechanisms by which IgG-containing immune complexes mediate such cross-presentation and the implications that this understanding has for manipulation of immune-mediated diseases that depend upon or are due to the activities of CD8(+) T cells.
Collapse
|
26
|
Tremblay T, Paré I, Bazin R. Immunoglobulin G dimers and immune complexes are dispensable for the therapeutic efficacy of intravenous immune globulin in murine immune thrombocytopenia. Transfusion 2012; 53:261-9. [DOI: 10.1111/j.1537-2995.2012.03725.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
27
|
IgG1 and IVIg induce inhibitory ITAM signaling through FcγRIII controlling inflammatory responses. Blood 2012; 119:3084-96. [PMID: 22337713 DOI: 10.1182/blood-2011-08-376046] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intravenous immunoglobulin (IVIg) has been used in the treatment of several autoimmune and inflammatory diseases. However, its mechanism of action remains incompletely understood. Here, we investigated the possibility that IVIg induces its anti-inflammatory effects through activating Fcγ receptors bearing an immunoreceptor tyrosine-based activation motif (ITAM) in the FcRγ signaling adaptor. Recently, the concept of inhibitory ITAM (ITAMi) has emerged as a new means to negatively control the immune response. We found that interaction of FcRγ-associated mouse or human FcγRIII with uncomplexed IgG1 or IVIg, or with bivalent anti-FcγRIII F(ab')(2) reduced calcium responses, reactive oxygen species production, endocytosis, and phagocytosis, induced by heterologous activating receptors on monocyte/macrophages and FcγRIII(+) transfectants. Inhibition required the ITAMi configuration of the FcγRIII-associated FcRγ subunit and SHP-1 recruitment involving formation of intracellular "inhibisome" clusters containing FcγRIII, and the targeted heterologous activating receptor. IVIg as well as anti-FcγRIII treatments controlled the development of nonimmune mediated inflammation in vivo independently of FcγRIIB. These results demonstrate that circulating immunoglobulins (Ig)Gs are not functionally inert but act through continuous interaction with FcγRIII-inducing ITAMi signaling to maintain immune homeostasis. These data support a new mechanism of action for IVIg and demonstrate the therapeutic potential of FcγRIIIA targeting in inflammation.
Collapse
|
28
|
The neonatal Fc receptor (FcRn) is not required for IVIg or anti-CD44 monoclonal antibody–mediated amelioration of murine immune thrombocytopenia. Blood 2011; 118:6403-6. [DOI: 10.1182/blood-2011-08-374223] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
To definitively determine whether the neonatal Fc receptor (FcRn) is required for the acute amelioration of immune thrombocytopenia (ITP) by IVIg, we used FcRn-deficient mice in a murine ITP model. Mice injected with antiplatelet antibody in the presence or absence of IVIg displayed no difference in platelet-associated IgG between FcRn deficient versus C57BL/6 mice. FcRn-deficient mice treated with high-dose (2 g/kg) IVIg or a low–dose (2 mg/kg) of an IVIg-mimetic CD44 antibody were, however, protected from thrombocytopenia to an equivalent extent as wild-type mice. To verify and substantiate the results found with FcRn-deficient mice, we used β2-microglobulin–deficient mice (which do not express functional FcRn) and found that IVIg or CD44 antibody also protected them from thrombocytopenia. These data suggest that for both high-dose IVIg as well as low-dose CD44 antibody treatment in an acute ITP model, FcRn expression is neither necessary nor required.
Collapse
|
29
|
Mekhaiel DNA, Daniel-Ribeiro CT, Cooper PJ, Pleass RJ. Do regulatory antibodies offer an alternative mechanism to explain the hygiene hypothesis? Trends Parasitol 2011; 27:523-9. [PMID: 21943801 DOI: 10.1016/j.pt.2011.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 08/17/2011] [Accepted: 08/18/2011] [Indexed: 12/24/2022]
Abstract
The 'hygiene hypothesis', or lack of microbial and parasite exposure during early life, is postulated as an explanation for the recent increase in autoimmune and allergic diseases in developed countries. The favored mechanism is that microbial and parasite-derived products interact directly with pathogen recognition receptors to subvert proinflammatory signaling via T regulatory cells, thereby inducing anti-inflammatory effects and control of autoimmune disease. Parasites, such as helminths, are considered to have a major role in the induction of immune regulatory mechanisms among children living in developing countries. Invoking Occam's razor, we believe we can select an alternative mechanism to explain the hygiene hypothesis, based on antibody-mediated inhibition of immune responses that may more simply explain the available evidence.
Collapse
Affiliation(s)
- David N A Mekhaiel
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | | | | | | |
Collapse
|
30
|
Wymann S, Zuercher AW, Schaub A, Bolli R, Stadler BM, Miescher SM. Monomeric and dimeric IgG fractions show differential reactivity against pathogen-derived antigens. Scand J Immunol 2011; 74:31-41. [PMID: 21338382 DOI: 10.1111/j.1365-3083.2011.02537.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polyvalent Ig preparations, derived from the pooled plasma of thousands of healthy donors, contain a complex mix of both 'acquired' and natural antibodies directed against pathogens as well as foreign and self/auto antigens (Ag). Depending on their formulation, donor pool size, etc., liquid Ig preparations contain monomeric and dimeric IgG. The dimeric IgG fraction is thought to represent mainly idiotype-antiidiotype Ab pairs. Treatment of all IgG fractions at pH 4 effectively monomerizes the IgG dimers resulting in separated idiotype-antiidiotype Ab pairs and thus in a comparable F(ab')(2) binding site availability of the different IgG fractions. Previously, we identified an increased anti-self-reactivity within the monomerized dimer fraction. This study addressed if, among the different IgG fractions, an analogous preferential reactivity was evident in the response against different pathogen-derived protein and carbohydrate antigens. Therefore, we assessed the activity of total unseparated IgG, the monomeric and dimeric IgG fractions against antigenic structures of bacterial and viral antigens/virulence factors. All fractions showed similar reactivity to protein antigens except for exotoxin A of Pseudomonas aeruginosa, where the dimeric fraction, especially when monomerized, showed a marked increase in reactivity. This suggests that the production of antiidiotypic IgG antibodies contributes to controlling the immune response to certain categories of pathogens. In contrast, the monomeric IgG fractions showed increased reactivity towards pathogen-associated polysaccharides, classically regarded as T-independent antigens. Taken together, the differential reactivity of the IgG fractions seems to indicate a preferential segregation of antibody reactivities according to the nature of the antigen.
Collapse
Affiliation(s)
- S Wymann
- CSL Behring AG, Wankdorfstrasse 10, Bern, Switzerland.
| | | | | | | | | | | |
Collapse
|
31
|
Schaub A, von Gunten S, Vogel M, Wymann S, Rüegsegger M, Stadler BM, Spycher M, Simon HU, Miescher S. Dimeric IVIG contains natural anti-Siglec-9 autoantibodies and their anti-idiotypes. Allergy 2011; 66:1030-7. [PMID: 21385183 DOI: 10.1111/j.1398-9995.2011.02579.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Intravenous immunoglobulin (IVIG) preparations are increasingly used for the treatment of autoimmune and chronic inflammatory diseases. Naturally occurring autoantibodies against Siglec-9 and Fas are thought to contribute to the anti-inflammatory effects of IVIG via cell death regulation of leukocytes and tissue cells. Dimeric IVIG fractions are suspected to contain idiotypic (Id)-anti-idiotypic complexes of antibodies, which might also include anti-Siglec-9 and anti-Fas autoantibodies. METHODS Dimeric IVIG fractions were separated from monomeric IVIG by size-exclusion chromatography and remonomerized by low pH treatment. Binding studies of total, monomeric, and dimeric IVIG were performed using surface plasmon resonance and flow cytometry on primary human neutrophils. RESULTS Anti-Siglec-9 and anti-Fas autoantibodies were contained in both monomeric and dimeric IVIG fractions, but anti-Siglec-9 antibodies were highly enriched in dimeric IVIG. The propensity to engage in dimer formation was paratope dependent. IVIG binding to Siglec-9 was specific and sialylation independent. Interestingly, we detected anti-idiotypic antibodies (anti-Ids) against anti-Siglec-9 autoantibodies in dimeric, but not in monomeric fractions of IVIG. CONCLUSIONS Our study supports the concept that idiotype-anti-idiotype (Id-anti-Id) interactions contribute to the dimer formation in IVIG preparations. To our knowledge, this is the first description of Id-anti-Id dimers of death receptor-specific antibodies in IVIG. Such Id-anti-Id interactions might determine the activity of immunomodulatory antibodies present both in IVIG and the patient.
Collapse
Affiliation(s)
- A Schaub
- University Institute of Immunology, Bern, Switzerland CSL Behring AG, Bern, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ramakrishna C, Newo ANS, Shen YW, Cantin E. Passively administered pooled human immunoglobulins exert IL-10 dependent anti-inflammatory effects that protect against fatal HSV encephalitis. PLoS Pathog 2011; 7:e1002071. [PMID: 21655109 PMCID: PMC3107211 DOI: 10.1371/journal.ppat.1002071] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 04/05/2011] [Indexed: 11/24/2022] Open
Abstract
HSV-1 is the leading cause of sporadic encephalitis in humans. HSV infection of susceptible 129S6 mice results in fatal encephalitis (HSE) caused by massive inflammatory brainstem lesions comprising monocytes and neutrophils. During infection with pathogenic microorganisms or autoimmune disease, IgGs induce proinflammatory responses and recruit innate effector cells. In contrast, high dose intravenous immunoglobulins (IVIG) are an effective treatment for various autoimmune and inflammatory diseases because of potent anti-inflammatory effects stemming in part from sialylated IgGs (sIgG) present at 1–3% in IVIG. We investigated the ability of IVIG to prevent fatal HSE when given 24 h post infection. We discovered a novel anti-inflammatory pathway mediated by low-dose IVIG that protected 129S6 mice from fatal HSE by modulating CNS inflammation independently of HSV specific antibodies or sIgG. IVIG suppressed CNS infiltration by pathogenic CD11b+ Ly6Chigh monocytes and inhibited their spontaneous degranulation in vitro. FcγRIIb expression was required for IVIG mediated suppression of CNS infiltration by CD45+ Ly6Clow monocytes but not for inhibiting development of Ly6Chigh monocytes. IVIG increased accumulation of T cells in the CNS, and the non-sIgG fraction induced a dramatic expansion of FoxP3+ CD4+ T regulatory cells (Tregs) and FoxP3− ICOS+ CD4+ T cells in peripheral lymphoid organs. Tregs purified from HSV infected IVIG treated, but not control, mice protected adoptively transferred mice from fatal HSE. IL-10, produced by the ICOS+ CD4+ T cells that accumulated in the CNS of IVIG treated, but not control mice, was essential for induction of protective anti-inflammatory responses. Our results significantly enhance understanding of IVIG's anti-inflammatory and immunomodulatory capabilities by revealing a novel sIgG independent anti-inflammatory pathway responsible for induction of regulatory T cells that secrete the immunosuppressive cytokine IL-10 and further reveal the therapeutic potential of IVIG for treating viral induced inflammatory diseases. We show that fatal HSV encephalitis (HSE) is caused by excessive brainstem inflammation. Once brainstem inflammation is initiated, antiviral drugs that inhibit only viral replication are ineffective in protecting against fatal HSE. Infusion of high doses of pooled human IgG (IVIG) is an effective anti-inflammatory treatment for various autoimmune diseases. One anti-inflammatory mechanism depends on sialylated IgGs (sIgG) present in limiting amounts (1–3%) in IVIG, hence the need for high doses of IVIG. We discovered a novel anti-inflammatory pathway mediated by low doses of IVIG independent of sIgG that prevented fatal HSE by suppressing CNS inflammation. The non-sIgG fraction of IVIG induced regulatory CD4+ T cells that produced the immunosuppressive cytokine IL-10 in the brainstem. Importantly, we show that IL-10 is critical for suppressing the generation of pathogenic inflammatory macrophages. Thus, IVIG has a remarkable ability to balance the host inflammatory responses to virus infection and thereby promotes virus clearance without bystander damage to the CNS, accounting for survival of all infected mice. Overall, our results provide important new insights in understanding IVIG's anti-inflammatory activity and further reveal its potential for use in treatment of viral inflammatory diseases.
Collapse
MESH Headings
- Animals
- Antigens, Ly/metabolism
- Blood-Brain Barrier/immunology
- Brain Stem/pathology
- CD4-Positive T-Lymphocytes/immunology
- Encephalitis, Herpes Simplex/immunology
- Encephalitis, Herpes Simplex/mortality
- Encephalitis, Herpes Simplex/prevention & control
- Encephalitis, Herpes Simplex/virology
- Flow Cytometry
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/pathogenicity
- Humans
- Immunoglobulins, Intravenous/administration & dosage
- Immunoglobulins, Intravenous/immunology
- Immunoglobulins, Intravenous/therapeutic use
- Immunologic Factors/therapeutic use
- Interleukin-10/administration & dosage
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Leukocytes/immunology
- Macrophages/immunology
- Mice
- Mice, Transgenic
- Monocytes/immunology
- Neutrophils/immunology
- T-Lymphocytes, Regulatory/immunology
- Time Factors
Collapse
Affiliation(s)
- Chandran Ramakrishna
- Division of Virology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Alain N. S. Newo
- Division of Virology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Yueh-Wei Shen
- Division of Virology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Edouard Cantin
- Division of Virology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
- Immunology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
- Neurology, Beckman Research Institute, City of Hope, Duarte, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
Effect of intravenous immunoglobulin with or without cytotoxic drugs on pemphigus intercellular antibodies. J Am Acad Dermatol 2011; 64:484-9. [DOI: 10.1016/j.jaad.2010.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 03/02/2010] [Accepted: 04/05/2010] [Indexed: 11/23/2022]
|
34
|
Abstract
Abstract
Intravenous immunoglobulin (IVIg) is an effective treatment against immune thrombocytopenia (ITP). Previous studies suggested that IVIg exerts this ameliorative role through 2 different leukocyte subsets. Dendritic cells (DCs) modulate the immunosuppression in an adoptive cell transfer model, and phagocytes up-regulate their inhibitory IgG Fc receptors (FcγR)IIB expression and thereby ameliorate the inflammatory response and platelet clearance. However, whether or not regulatory mechanisms exist among DCs, phagocytes, and platelets is still largely unknown. In this study we present findings that IVIg-primed splenic CD11c+ DCs (IVIg-DCs) primarily mediate their anti-inflammatory effects at the level of the platelet rather than the phagocyte. IVIg-DCs did not ameliorate ITP in Fcgr2b−/−, Fcgr3−/−, nor P-Selp−/− mice, implicating the potential involvement of these pathways in IVIg action. As platelets are a component of DC regulatory circuits, these findings may suggest an alternative perspective for the use of IVIg treatment.
Collapse
|
35
|
Smith SD, Dennington PM, Cooper A. The use of intravenous immunoglobulin for treatment of dermatological conditions in Australia: A review. Australas J Dermatol 2010; 51:227-37. [DOI: 10.1111/j.1440-0960.2009.00578.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
A novel role for the IgG Fc glycan: the anti-inflammatory activity of sialylated IgG Fcs. J Clin Immunol 2010; 30 Suppl 1:S9-14. [PMID: 20480216 DOI: 10.1007/s10875-010-9405-6] [Citation(s) in RCA: 226] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
IgG antibodies have long been recognized as proinflammatory mediators of the humoral immune response. Antibodies bind and neutralize antigens to promote antibody-dependent cytotoxicity, opsonization of antigens, and the initiation of phagocytosis. Whereas the antigen specificity of antibodies is determined by the antigen-binding Fab portion, the effector functions initiated by antibodies are triggered by the Fc (crystallizable) domain. These effector functions are heavily dependent on the single N-linked, biantennary glycan of the heavy chain, which resides just below the hinge region. This glycan is believed to maintain the two heavy chains of the Fc in an open confirmation required for interactions with activating Fcgamma receptors (FcgammaRs). However, the presence of specific sugar moieties on the glycan has profound implications on Fc effector functions. The addition of terminal sialic acid to the glycan reduces FcgammaR binding and converts IgG antibodies to anti-inflammatory mediators through the acquisition of novel binding activities. Studies from our laboratory demonstrated that these sialylated IgG Fcs are important for the in vivo activity of intravenous immunoglobulin. Instead of binding with FcgammaRs, sialylated Fcs initiate an anti-inflammatory cascade through the lectin receptor SIGN-R1 or DC-SIGN. This leads to upregulated surface expression of the inhibitory FcR, FcgammaRIIb, on inflammatory cells, thereby attenuating autoantibody-initiated inflammation.
Collapse
|
37
|
Abstract
Antibody-mediated rejection has become critical clinically because this form of rejection is usually unresponsive to conventional anti-rejection therapy, and therefore, it has been recognized as a major cause of allograft loss. Our group developed experimental animal models of vascularized organ transplantation to study pathogenesis of antibody- and complement-mediated endothelial cell injury leading to graft rejection. In this review, we discuss mechanisms of antibody-mediated graft rejection resulting from activation of complement by C1q- and MBL (mannose-binding lectin)-dependent pathways and interactions with a variety of effector cells, including macrophages and monocytes through Fcgamma receptors and complement receptors.
Collapse
|
38
|
Jordan SC, Reinsmoen N, Peng A, Lai CH, Cao K, Villicana R, Toyoda M, Kahwaji J, Vo AA. Advances in diagnosing and managing antibody-mediated rejection. Pediatr Nephrol 2010; 25:2035-45; quiz 2045-8. [PMID: 20077121 PMCID: PMC2923704 DOI: 10.1007/s00467-009-1386-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 10/05/2009] [Accepted: 11/02/2009] [Indexed: 02/07/2023]
Abstract
Antibody-mediated rejection (AMR) is a unique, significant, and often severe form of allograft rejection that is not amenable to treatment with standard immunosuppressive medications. Significant advances have occurred in our ability to predict patients at risk for, and to diagnose, AMR. These advances include the development of newer anti-human leukocyte antigen (HLA)-antibody detection techniques and assays for non-HLA antibodies associated with AMR. The pathophysiology of AMR suggests a prime role for antibodies, B cells and plasma cells, but other effector molecules, especially the complement system, point to potential targets that could modify the AMR process. An emerging and potentially larger problem is the development of chronic AMR (CAMR) resulting from de novo donor-specific anti-HLA antibodies (DSA) that emerge more than 100 days posttransplantation. Therapeutic options include: (1) High-dose intravenously administered immunoglobulin (IVIG), which has many potential benefits. (2) The use of IVIG+rituximab (anti-CD20, anti-B cell). (3) The combination of plasmapheresis (PP)+low-dose IVIG with or without rituximab. Data support the efficacy of all of the above approaches. Newer approaches to treating AMR include using the proteosome inhibitor (bortezomib), which induces apoptosis in plasma cells, and eculizumab (anti-C5, anticomplement monoclonal antibody).
Collapse
Affiliation(s)
- Stanley C Jordan
- The Transplant Immunotherapy Program, Comprehensive Transplant Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sharma KG, Radha R, Pao A, Amet N, Baden L, Jordan SC, Toyoda M. Mycophenolic acid and intravenous immunoglobulin exert an additive effect on cell proliferation and apoptosis in the mixed lymphocyte reaction. Transpl Immunol 2010; 23:117-20. [PMID: 20450974 DOI: 10.1016/j.trim.2010.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Revised: 04/20/2010] [Accepted: 04/26/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Intravenous immunoglobulin (IVIG) has known immunomodulatory effects in autoimmune diseases and transplantation and is commonly used in desensitization protocols and for treatment of antibody-mediated rejection (AMR). IVIG inhibits the MLR and induces apoptosis in immune cells. Mycophenolate mofetil inhibits immune cell proliferation and is an effective immunsuppressive agent. Here, we examined the possible synergistic effects of combined MMF and IVIG on cell proliferation and apoptosis induction in the MLR. METHODS Two-way MLRs were performed with mycophenolic acid (MPA), IVIG and both in combination. Cell proliferation and apoptosis were detected by 3H-thymidine incorporation and Annexin flow cytometry, respectively. RESULTS IVIG (1-10mg/ml) or MPA (0.01-0.25 microg/ml) alone inhibited cell proliferation in the MLR in a dose-dependent manner. MPA at 0.01-0.03 microg/ml showed minimal inhibition, but the addition of 5 and 10mg/ml IVIG increased inhibition significantly (p<0.05) to 43% and 64%, respectively. Annexin V positive cell number was significantly higher in IVIG (5mg/ml) treated CD19+ cells (68+/-13% vs. 43+/-12%, p=0.001) compared to untreated cells and to a lesser degree in CD3+ cells (29+/-7% vs. 25+/-10 %, p=0.02). MPA (0.25-10 microg/ml) alone neither induced nor inhibited apoptosis. Addition of MPA had no effect on apoptosis induced by IVIG. CONCLUSION 1) Combining low concentrations of IVIG (5-10 mg/ml) and MPA (0.01-0.03 microg/ml)has an additive effect on inhibition of cell proliferation in the MLR. 2) MPA alone neither induces nor inhibits apoptosis in T or B cells in the MLR, and has no effect on apoptosis induced by IVIG. These in vitro observations may have implications for modification of therapeutic approaches to protocols utilizing IVIG for desensitization and immune modulation.
Collapse
Affiliation(s)
- Kavita G Sharma
- Transplant Immunology Laboratory, Comprehensive Transplant Center, Cedars-Sinai Medical Center/UCLA School of Medicine, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Blank U, Launay P, Benhamou M, Monteiro RC. Inhibitory ITAMs as novel regulators of immunity. Immunol Rev 2009; 232:59-71. [DOI: 10.1111/j.1600-065x.2009.00832.x] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
41
|
Role of IgA and IgA fc receptors in inflammation. J Clin Immunol 2009; 30:1-9. [PMID: 19834792 DOI: 10.1007/s10875-009-9338-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Accepted: 09/24/2009] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Signals delivered by serum monomeric IgA (mIgA) are essential in controlling the immune system by preventing the development of autoimmunity and inflammation. However, IgA can also, when aggregated, be deleterious to the host, inducing inflammatory diseases. This Janus-like nature of IgA is mainly due to their heterogeneity in molecular forms and their interaction with IgA receptors. DISCUSSION While serum mIgA are mainly involved in FcalphaRI-mediated inhibition of immune responses, macromolecular serum IgA or circulating IgA immune complexes are often deleterious to the host by inducing sustained activation through IgA receptors including FcalphaRI and transferrin receptor. CONCLUSION FcalphaRI-mediated inhibitory function is able to suppress several inflammatory diseases in mice including asthma and glomerulonephritis. Intravenous mIgA (mIgAIV) and anti-FcalphaR monovalent antibodies represent thus promising tools for immunotherapy.
Collapse
|
42
|
Olovnikova NI, Ershler MA, Belkina EV, Nikolaeva TL, Miterev GY. Effect of producer cell line on functional activity of anti-D monoclonal antibodies destined for prevention of rhesus sensitization. Bull Exp Biol Med 2009; 147:448-52. [PMID: 19704946 DOI: 10.1007/s10517-009-0516-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The ability of anti-D antibodies to cause antigen-specific immunosuppression depends on their interaction with low-affinity Fcgamma-receptors. Human monoclonal antibodies to D antigen of the rhesus system were investigated by antibody-dependent cytotoxicity assay in order to estimate their ability to induce hemolysis mediated by low-affinity Fcgamma receptors. We demonstrate that affinity of monoclonal antibodies to receptors of this type does not depend on primary structure of Fc-fragment, but depends on the producer cell line which expresses the antibodies. Monoclonal IgG1 antibodies interacting with FcgammaRIIa and FcgammaRIII lost this property, if they were secreted by human-mouse heterohybridoma, but not by human B-cell line. On the opposite, monoclonal antibodies that could not activate low-affinity Fcgamma receptors were highly active after human cells fusion with rat myeloma YB2/0. Hemolytic activity of IgG3 remained unchanged after fusion of human cells with rodent cells.
Collapse
Affiliation(s)
- N I Olovnikova
- Hematological Research Center, Russian Academy of Medical Sciences, Moscow, Russia.
| | | | | | | | | |
Collapse
|
43
|
Abstract
Intravenous immunoglobulin (IVIG) products are derived from pooled human plasma and have been used for the treatment of primary immunodeficiency disorders for more than 25 years. IVIG products are also effective in the treatment of autoimmune and inflammatory disorders; however, the precise mechanism(s) of action is not known. Recent investigations suggest that IVIG has a much broader ability to regulate cellular immunity including innate and adaptive components. IVIG is also a recently recognized modifier of complement activation and injury. Here, we discuss these important advancements and how this knowledge applies to desensitization protocols and to the treatment of antibody-mediated rejection.
Collapse
|
44
|
Baker K, Qiao SW, Kuo T, Kobayashi K, Yoshida M, Lencer WI, Blumberg RS. Immune and non-immune functions of the (not so) neonatal Fc receptor, FcRn. Semin Immunopathol 2009; 31:223-36. [PMID: 19495758 PMCID: PMC3898171 DOI: 10.1007/s00281-009-0160-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Accepted: 05/14/2009] [Indexed: 02/06/2023]
Abstract
Careful regulation of the body's immunoglobulin-G (IgG) and albumin concentrations is necessitated by the importance of their respective functions. As such, the neonatal Fc receptor (FcRn) which, as a single receptor, is capable of regulating both of these molecules, has become an important focus of investigation. In addition to these essential protection functions, FcRn possesses a host of other functions that are equally as critical. During the very first stages of life, FcRn mediates the passive transfer of IgG from mother to offspring both before and after birth. In the adult, FcRn regulates the persistence of both IgG and albumin in the serum as well as the movement of IgG, and any bound cargo, between different compartments of the body. This shuttling allows for the movement not only of monomeric ligand but also of antigen/antibody complexes from one cell type to another in such a way as to facilitate the efficient initiation of immune responses towards opsonized pathogens. As such, FcRn continues to play the role of an immunological sensor throughout adult life, particularly in regions such as the gut which are exposed to a large number of infectious antigens. Increasing appreciation for the contributions of FcRn to both homeostatic and pathological states is generating an intense interest in the potential for therapeutic modulation of FcRn binding. A greater understanding of FcRn's pleiotropic roles is thus imperative for a variety of therapeutic purposes.
Collapse
Affiliation(s)
- Kristi Baker
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Shuo-Wang Qiao
- Rikshospitalet University Hospital, 0027 Oslo, Norway, University of Oslo, 0027 Oslo, Norway
| | - Timothy Kuo
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kanna Kobayashi
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Masaru Yoshida
- Department of Gastroenterology & The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medecine, Hyogo, Japan
| | - Wayne I. Lencer
- Harvard Digestive Diseases Center, Boston, MA 02115, USA, GI Cell Biology, Division of Pediatric Gastroenterology and Nutrition, Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Richard S. Blumberg
- Division of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA, Harvard Digestive Diseases Center, Boston, MA 02115, USA
| |
Collapse
|
45
|
Natural antibody contributes to host defense against an attenuated Brucella abortus virB mutant. Infect Immun 2009; 77:3004-13. [PMID: 19364836 DOI: 10.1128/iai.01114-08] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Brucella abortus is an intracellular pathogen that persists within phagocytic cells of the reticuloendothelial system. To identify in vivo interactions between B. abortus and the host that lead to persistent infection, we studied the persistence of B. abortus and an isogenic virB mutant deficient in the VirB type IV secretion system (T4SS) in knockout mice. In contrast to control mice, mice lacking B cells (Igh6(-/-)) were permissive for infection with the attenuated virB mutant. To determine the basis for this phenotype, we characterized immune functions of Igh6(-/-) mice in the context of B. abortus infection. Igh6(-/-) mice had greater numbers of extracellular bacteria in the spleen and increased early expression of proinflammatory cytokines during B. abortus infection. Further, a virB mutant, despite its wild-type level of survival, failed to elicit microgranuloma formation in the spleens of Igh6(-/-) mice, suggesting a requirement for the T4SS to elicit this pathological change. Passive transfer of immunoglobulin G from naïve mice restored the ability of Igh6(-/-) mice to control the persistence of the virB mutant by a complement-independent mechanism. Further, adoptive transfer of CD11b(+) cells from C57BL/6 mice to Igh6(-/-) mice restored the ability of the knockout mice to limit the replication of the virB mutant in the spleen, suggesting that the Igh6(-)(/)(-) mutation affects phagocyte function and that phagocyte function can be restored by natural antibody.
Collapse
|
46
|
Maglione PJ, Chan J. How B cells shape the immune response against Mycobacterium tuberculosis. Eur J Immunol 2009; 39:676-86. [PMID: 19283721 DOI: 10.1002/eji.200839148] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Extensive work illustrating the importance of cellular immune mechanisms for protection against Mycobacterium tuberculosis has largely relegated B-cell biology to an afterthought within the tuberculosis (TB) field. However, recent studies have illustrated that B lymphocytes, through a variety of interactions with the cellular immune response, play previously underappreciated roles in shaping host defense against non-viral intracellular pathogens, including M. tuberculosis. Work in our laboratory has recently shown that, by considering these lymphocytes more broadly within their variety of interactions with cellular immunity, B cells have a significant impact on the outcome of airborne challenge with M. tuberculosis as well as the resultant inflammatory response. In this review, we advocate for a revised view of TB immunology in which roles of cellular and humoral immunity are not mutually exclusive. In the context of our current understanding of host defense against non-viral intracellular infections, we review recent data supporting a more significant role of B cells during M. tuberculosis infection than previously thought.
Collapse
Affiliation(s)
- Paul J Maglione
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, NY, USA
| | | |
Collapse
|
47
|
Abstract
The benefit of immunoglobulin (IG) replacement in primary antibody deficiencies is unquestionable. Many of these congenital disorders present early in life and this therapy is often first implemented in the young. This article focuses on the indications of IG replacement in children, with an emphasis on the specific diagnostic problems encountered in this population. Also presented is an overview of the practical aspects of IG administration in the pediatric setting, including the recognition and management of adverse reactions. Finally, the advent of subcutaneous IG, a therapeutic IG modality with the potential to have a great impact on the quality of life of children with antibody deficiencies and their families, is discussed.
Collapse
|
48
|
Identification of a receptor required for the anti-inflammatory activity of IVIG. Proc Natl Acad Sci U S A 2008; 105:19571-8. [PMID: 19036920 DOI: 10.1073/pnas.0810163105] [Citation(s) in RCA: 420] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The anti-inflammatory activity of intravenous Ig (IVIG) results from a minor population of the pooled IgG molecules that contains terminal alpha2,6-sialic acid linkages on their Fc-linked glycans. These anti-inflammatory properties can be recapitulated with a fully recombinant preparation of appropriately sialylated IgG Fc fragments. We now demonstrate that these sialylated Fcs require a specific C-type lectin, SIGN-R1, (specific ICAM-3 grabbing non-integrin-related 1) expressed on macrophages in the splenic marginal zone. Splenectomy, loss of SIGN-R1(+) cells in the splenic marginal zone, blockade of the carbohydrate recognition domain (CRD) of SIGN-R1, or genetic deletion of SIGN-R1 abrogated the anti-inflammatory activity of IVIG or sialylated Fc fragments. Although SIGN-R1 has not previously been shown to bind to sialylated glycans, we demonstrate that it preferentially binds to 2,6-sialylated Fc compared with similarly sialylated, biantennary glycoproteins, thus suggesting that a specific binding site is created by the sialylation of IgG Fc. A human orthologue of SIGN-R1, DC-SIGN, displays a similar binding specificity to SIGN-R1 but differs in its cellular distribution, potentially accounting for some of the species differences observed in IVIG protection. These studies thus identify an antibody receptor specific for sialylated Fc, and present the initial step that is triggered by IVIG to suppress inflammation.
Collapse
|
49
|
Tha-In T, Bayry J, Metselaar HJ, Kaveri SV, Kwekkeboom J. Modulation of the cellular immune system by intravenous immunoglobulin. Trends Immunol 2008; 29:608-15. [PMID: 18926775 DOI: 10.1016/j.it.2008.08.004] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 08/21/2008] [Accepted: 08/25/2008] [Indexed: 01/20/2023]
Abstract
Intravenous immunoglobulin (IVIg) is therapeutically used in a variety of immune-mediated diseases. The beneficial effects of IVIg in auto-antibody-mediated diseases can be explained by neutralization, accelerated clearance and prevention of Fcgamma-receptor binding of auto-antibodies. However, the means by which IVIg exerts therapeutic effects in disorders mediated by cellular immunity have remained enigmatic. Clinical improvements, followed by IVIg treatment, often extend beyond the half-life of infused IgG, thereby indicating that IVIg modifies the cellular immune compartment for a prolonged period. Here, we discuss recent advances in the understanding of different, mutually non-exclusive mechanisms of action of IVIg on cells of the innate and adaptive immune system. These mechanisms might explain the beneficial effects of IVIg in certain autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Thanyalak Tha-In
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | | |
Collapse
|
50
|
Stangel M. New advances in the treatment of neurological diseases using high dose intravenous immunoglobulins. Ther Adv Neurol Disord 2008; 1:52-61. [PMID: 21180569 PMCID: PMC3002549 DOI: 10.1177/1756285608095747] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Since the incidental discovery in 1981 that intravenous immunoglobulins (IVIg) are immunomodulatory, they have been investigated in a large number of putative autoimmune diseases. This has led to licensing for idiopathic thrombocytopenic purpura, Kawasaki disease, and in neurological disorders for Guillain-Barré syndrome (GBS). Although not licensed, randomized controlled trials have also shown IVIg efficacy in other neuroimmunological diseases such as multifocal motor neuropathy (MMN), chronic inflammatory demyelinating neuropathy (CIDP), myasthenia gravis, dermatomyositis, and stiff-person syndrome. However, other indications are currently being explored including Alzheimer's disease, postpolio syndrome, and narcolepsy. There are even reports from experimental studies in stroke. The results of recently published clinical trials in both the classical neuroimmunological disorders as well as for new indications are reported and their role in clinical practice is discussed.
Collapse
Affiliation(s)
- Martin Stangel
- Department of Neurology Hannover Medical School Carl-Neuberg-Str. 1 30625 Hannover Germany.
| |
Collapse
|