1
|
Hematianlarki M, Nimmerjahn F. Immunomodulatory and anti-inflammatory properties of immunoglobulin G antibodies. Immunol Rev 2024; 328:372-386. [PMID: 39340138 DOI: 10.1111/imr.13404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2024]
Abstract
Antibodies provide an essential layer of protection from infection and reinfection with microbial pathogens. An impaired ability to produce antibodies results in immunodeficiency and necessitates the constant substitution with pooled serum antibodies from healthy donors. Among the five antibody isotypes in humans and mice, immunoglobulin G (IgG) antibodies are the most potent anti-microbial antibody isotype due to their long half-life, their ability to penetrate almost all tissues and due to their ability to trigger a wide variety of effector functions. Of note, individuals suffering from IgG deficiency frequently produce self-reactive antibodies, suggesting that a normal serum IgG level also may contribute to maintaining self-tolerance. Indeed, the substitution of immunodeficient patients with pooled serum IgG fractions from healthy donors, also referred to as intravenous immunoglobulin G (IVIg) therapy, not only protects the patient from infection but also diminishes autoantibody induced pathology, providing more direct evidence that IgG antibodies play an active role in maintaining tolerance during the steady state and during resolution of inflammation. The aim of this review is to discuss different conceptual models that may explain how serum IgG or IVIg can contribute to maintaining a balanced immune response. We will focus on pathways depending on the IgG fragment crystallizable (Fc) as pre-clinical data in various mouse model systems as well as human clinical data have demonstrated that the IgG Fc-domain recapitulates the ability of intact IVIg with respect to its ability to trigger resolution of inflammation. We will further discuss how the findings already have or are in the process of being translated to novel therapeutic approaches to substitute IVIg in treating autoimmune inflammation.
Collapse
Affiliation(s)
- Marjan Hematianlarki
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Boulard P, Azzopardi N, Levard R, Cornec JM, Lamamy J, Prieur B, Demattei MV, Watier H, Gatault P, Gouilleux-Gruart V. Albumin influences leucocyte FcRn expression in the early days of kidney transplantation. Clin Exp Immunol 2024; 216:307-317. [PMID: 38353127 PMCID: PMC11097912 DOI: 10.1093/cei/uxae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/28/2023] [Accepted: 02/08/2024] [Indexed: 05/18/2024] Open
Abstract
FcRn, a receptor originally known for its involvement in IgG and albumin transcytosis and recycling, is also important in the establishment of the innate and adaptive immune response. Dysregulation of the immune response has been associated with variations in FcRn expression, as observed in cancer. Recently, a link between autophagy and FcRn expression has been demonstrated. Knowing that autophagy is strongly involved in the development of reperfusion injury in kidney transplantation and that albuminemia is transiently decreased in the first 2 weeks after transplantation, we investigated variations in FcRn expression after kidney transplantation. We monitored FcRn levels by flow cytometry in leukocytes from 25 renal transplant patients and considered parameters such as albumin concentrations, estimated glomerular filtration rate, serum creatinine, serum IgG levels, and ischaemia/reperfusion time. Two groups of patients could be distinguished according to their increased or non-increased FcRn expression levels between days 2 and 6 (d2-d6) post-transplantation. Leukocyte FcRn expression at d2-d6 was correlated with albumin concentrations at d0-d2. These results suggest that albumin concentrations at d0-d2 influence FcRn expression at d2-d6, raising new questions about the mechanisms underlying these original observations.
Collapse
Affiliation(s)
- Pierre Boulard
- Centre d’Étude des Pathologies Respiratoires (CEPR) U1100 INSERM, Tours, France
- Laboratoire d’immunologie, CHU de Tours, Tours,France
| | | | - Romain Levard
- Laboratoire d’immunologie, CHU de Tours, Tours,France
| | | | - Juliette Lamamy
- EA7501 GICC, Faculté de Médecine, Université de Tours, Tours,France
| | | | | | - Hervé Watier
- Laboratoire d’immunologie, CHU de Tours, Tours,France
- EA7501 GICC, Faculté de Médecine, Université de Tours, Tours,France
| | - Philippe Gatault
- EA4245 T2I, Faculté de Médecine, Université de Tours, Tours,France
- Service de Néphrologie, CHU de Tours, Tours,France
| | - Valérie Gouilleux-Gruart
- Laboratoire d’immunologie, CHU de Tours, Tours,France
- EA7501 GICC, Faculté de Médecine, Université de Tours, Tours,France
| |
Collapse
|
3
|
Li Z, Xu Q, Huang J, Zhu Q, Yang X, Zhang M, Zhang S, Huang S, Yu G, Zheng P, Qin X, Feng J. Efgartigimod as rescue treatment in acute phase of neuromyelitis optica spectrum disorder: A Case Report. Heliyon 2024; 10:e30421. [PMID: 38720715 PMCID: PMC11076956 DOI: 10.1016/j.heliyon.2024.e30421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Neuromyelitis optica spectrum disorder (NMOSD) is a central nervous system demyelinating disease. Current therapy methods, however, have limited effect on acute attacks except for intravenous methylprednisolone (IVMP). Efgartigimod is a first-in-class novel human immunoglobulin G1 (IgG1) Fc fragment approved for the treatment of generalized myasthenia gravis. Its capacity to rapidly decrease serum IgG levels, including pathogenic autoantibodies, positions it as a potentially effective option for managing the acute phase of NMOSD. Case presentation We report the case of a 59-year-old female patient with acute NMOSD, presenting with vision loss and numbness in all four limbs. Despite an initial inadequate response to intravenous methylprednisolone (IVMP), the addition of Efgartigimod to her treatment regimen led to rapid improvement, notably including a significant reduction in serum aquaporin-4 antibody titers, total IgG levels, and inflammation cytokine levels. Furthermore, no adverse events were reported during a four-month follow-up period. Conclusion As an adjunct to glucocorticoid therapy, Efgartigimod has proven effective and safe for this patient. However, to ascertain its potential as a novel therapeutic option for acute NMOSD, larger-scale prospective clinical trials are required.
Collapse
Affiliation(s)
- Zhizhong Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiao Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jialu Huang
- 958th Hospital of the People's Liberation Army, Chongqing, 400038, China
| | - Qiyuan Zhu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaolin Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengjie Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shaoru Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Siyuan Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Yu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinzhou Feng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Nashaat HAH, Anani M, Attia FM. Convalescent plasma in COVID-19: renewed focus on the timing and effectiveness of an old therapy. Blood Res 2022; 57:6-12. [PMID: 35197369 PMCID: PMC8958377 DOI: 10.5045/br.2021.2021151] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/19/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19) pandemic that has strained health care systems worldwide and resulted in high mortality. The current COVID-19 treatment is based on supportive and symptomatic care. Therefore, convalescent plasma (CP), which provides passive immunization against many infectious diseases, has been studied for COVID-19 management. To date, a large number of randomized and non-randomized clinical trials as well as many systematic reviews have revealed conflicting results. This article summarizes the basic principles of passive immunization, particularly addressing CP in COVID-19. It also evaluates the effectiveness of CP as a therapy in patients with COVID-19, clinical trial reports and systematic reviews, regulatory considerations and different protocols that are authorized in different countries to use it safely and effectively. An advanced search was carried out in major databases (PubMed, Cochrane Library, and MEDLINE) and Google Scholar using the following key words: SARS-CoV-2, COVID-19, convalescent plasma, and the applied query was "convalescent plasma" AND "COVID-19 OR SARS-CoV-2". The results were filtered and duplicate data were removed. Collective evidence indicates that two cardinal players determine the effectiveness of CP use, time of infusion, and quality of CP. Early administration of CP with high neutralizing anti-spike IgG titer is hypothesized to be effective in improving clinical outcome, prevent progression, decrease the length of hospital stay, and reduce mortality. However, more reliable, high quality, well-controlled, double-blinded, randomized, international and multicenter collaborative trials are still needed.
Collapse
Affiliation(s)
- Hebat-Allah Hassan Nashaat
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Maha Anani
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Fadia M. Attia
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
5
|
Lamamy J, Boulard P, Brachet G, Tourlet S, Gouilleux-Gruart V, Ramdani Y. "Ways in which the neonatal Fc-receptor is involved in autoimmunity". J Transl Autoimmun 2021; 4:100122. [PMID: 34568803 PMCID: PMC8449123 DOI: 10.1016/j.jtauto.2021.100122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/06/2021] [Indexed: 11/18/2022] Open
Abstract
Since the neonatal IgG Fc receptor (FcRn) was discovered, its role has evolved from immunoglobulin recycling and biodistribution to antigen presentation and immune complex routing, bringing it to the center of both humoral and cellular immune responses. FcRn is thus involved in the pathophysiology of immune-related diseases such as cancer, infection, and autoimmune disorders. This review focuses on the role of FcRn in autoimmunity, based on the available data from both animal models and human studies. The knowledge concerning ways in which FcRn is involved in autoimmune response has led to the development of inhibitors for the treatment of autoimmune diseases, also described here. Up to date, the literature remains scarce, shedding light on the need for further studies to fully understand the various pathophysiological roles of this unique receptor. FcRn is an intracellular receptor with a key role in IgG and immune complex management. FcRn-targeting therapies are a promising way of treatment in antibodies mediated diseases.
Collapse
Affiliation(s)
- Juliette Lamamy
- EA7501, GICC, Université François Rabelais de Tours, F-37032, Tours, France
| | - Pierre Boulard
- Laboratoire d'immunologie, CHU Tours, F-37032, Tours, France
| | | | | | | | - Yanis Ramdani
- Service de Médecine Interne, CHU Tours, F-37032, Tours, France
| |
Collapse
|
6
|
Bhargava R, Maeda K, Tsokos MG, Pavlakis M, Stillman IE, Tsokos GC. N-glycosylated IgG in patients with kidney transplants increases calcium/calmodulin kinase IV in podocytes and causes injury. Am J Transplant 2021; 21:148-160. [PMID: 32531122 PMCID: PMC8188503 DOI: 10.1111/ajt.16140] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/29/2020] [Accepted: 06/06/2020] [Indexed: 01/25/2023]
Abstract
Transplant glomerulopathy (TG) is a major cause of late allograft loss. Increased urine podocin/creatinine ratio in TG signifies accelerated podocyte loss. The mechanisms that lead to podocyte injury in TG remain unclear. We report that IgG from kidney transplant recipients with TG, but not from those without TG, cause a reduction in the expression of nephrin, significant podocyte actin cytoskeleton, and motility changes. These changes are preceded by increased expression of calcium/calmodulin kinase IV (CAMK4). Mechanistically, we found that CAMK4 phosphorylates GSK3β (glycogen synthase kinase 3 beta), activates the Wnt pathway and stabilizes the nephrin transcriptional repressor SNAIL. Silencing neonatal Fc Receptor (FcRn) or CAMK4 prevented the podocyte-damaging effects of IgG from patients with TG. Furthermore, we show that removal of N-linked glycosyl residues from these IgG did not interfere with its entry into the podocytes but eliminated its ability to upregulate CAMK4 and cause podocyte injury. The translational value of these findings is signified by the fact that CAMK4 is increased in podocytes of patients with TG but not in those without TG despite other forms of renal dysfunction. Our results offer novel considerations to limit podocyte injury in patients with kidney transplants, which may lead to eventual glomerular destabilization and transplant glomerulopathy.
Collapse
Affiliation(s)
- Rhea Bhargava
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Kayaho Maeda
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Maria G. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Martha Pavlakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Isaac E. Stillman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - George C. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Natural Antibodies: from First-Line Defense Against Pathogens to Perpetual Immune Homeostasis. Clin Rev Allergy Immunol 2020; 58:213-228. [PMID: 31161341 DOI: 10.1007/s12016-019-08746-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Natural antibodies (nAbs) are most commonly defined as immunoglobulins present in the absence of pathological conditions or deliberate immunizations. Occurrence of nAbs in germ- and antigen-free mice suggest that their production is driven, at least in part, by self-antigens. Accordingly, nAbs are constituted of natural autoantibodies (nAAbs), and can belong to the IgM, IgG, or IgA subclasses. These nAbs provide immediate protection against infection while the adaptive arm of the immune system mounts a specific and long-term response. Beyond immediate protection from infection, nAbs have been shown to play various functional roles in the immune system, which include clearance of apoptotic debris, suppression of autoimmune and inflammatory responses, regulation of B cell responses, selection of the B cell repertoires, and regulation of B cell development. These various functions of nAbs are afforded by their reactivity, which is broad, cross-reactive, and shown to recognize evolutionarily fixed epitopes shared between foreign and self-antigens. Furthermore, nAbs have unique characteristics that also contribute to their functional roles and set them apart from antigen-specific antibodies. In further support for the role of nAbs in the protection against infections and in the maintenance of immune homeostasis, the therapeutic preparation of polyclonal immunoglobulins, intravenous immunoglobulin (IVIG), rich in nAbs is commonly used in the replacement therapy of primary and secondary immunodeficiencies and in the immunotherapy of a large number of autoimmune and inflammatory diseases. Here, we review several topics on nAbs features and functions, and therapeutic applications in human diseases.
Collapse
|
8
|
Rojas M, Rodríguez Y, Monsalve DM, Acosta-Ampudia Y, Camacho B, Gallo JE, Rojas-Villarraga A, Ramírez-Santana C, Díaz-Coronado JC, Manrique R, Mantilla RD, Shoenfeld Y, Anaya JM. Convalescent plasma in Covid-19: Possible mechanisms of action. Autoimmun Rev 2020. [PMID: 32380316 DOI: 10.1016/j.autrev.2020.102554.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible of the coronavirus disease 2019 (COVID-19) pandemic. Therapeutic options including antimalarials, antivirals, and vaccines are under study. Meanwhile the current pandemic has called attention over old therapeutic tools to treat infectious diseases. Convalescent plasma (CP) constitutes the first option in the current situation, since it has been successfully used in other coronaviruses outbreaks. Herein, we discuss the possible mechanisms of action of CP and their repercussion in COVID-19 pathogenesis, including direct neutralization of the virus, control of an overactive immune system (i.e., cytokine storm, Th1/Th17 ratio, complement activation) and immunomodulation of a hypercoagulable state. All these benefits of CP are expected to be better achieved if used in non-critically hospitalized patients, in the hope of reducing morbidity and mortality.
Collapse
Affiliation(s)
- Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Bernardo Camacho
- Instituto Distrital de Ciencia Biotecnología e Investigación en Salud, IDCBIS, Bogota, Colombia
| | | | | | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | | | - Rubén Manrique
- Epidemiology and Biostatistics Research Group, Universidad CES, Medellin, Colombia
| | - Ruben D Mantilla
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to Tel-Aviv University, Tel Aviv, Israel; Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia.
| |
Collapse
|
9
|
Rojas M, Rodríguez Y, Monsalve DM, Acosta-Ampudia Y, Camacho B, Gallo JE, Rojas-Villarraga A, Ramírez-Santana C, Díaz-Coronado JC, Manrique R, Mantilla RD, Shoenfeld Y, Anaya JM. Convalescent plasma in Covid-19: Possible mechanisms of action. Autoimmun Rev 2020; 19:102554. [PMID: 32380316 PMCID: PMC7198427 DOI: 10.1016/j.autrev.2020.102554] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 12/17/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible of the coronavirus disease 2019 (COVID-19) pandemic. Therapeutic options including antimalarials, antivirals, and vaccines are under study. Meanwhile the current pandemic has called attention over old therapeutic tools to treat infectious diseases. Convalescent plasma (CP) constitutes the first option in the current situation, since it has been successfully used in other coronaviruses outbreaks. Herein, we discuss the possible mechanisms of action of CP and their repercussion in COVID-19 pathogenesis, including direct neutralization of the virus, control of an overactive immune system (i.e., cytokine storm, Th1/Th17 ratio, complement activation) and immunomodulation of a hypercoagulable state. All these benefits of CP are expected to be better achieved if used in non-critically hospitalized patients, in the hope of reducing morbidity and mortality. Coronavirus disease 19 (COVID-19) is an emerging viral threat with major repercussions for public health. There is not specific treatment for COVID-19. Convalescent plasma (CP) emerges as the first option of management for hospitalized patients with COVID-19. Transference of neutralizing antibodies helps to control COVID-19 infection and modulates inflammatory response. Other plasma components may enhance the antiviral and anti-inflammatory properties of CP.
Collapse
Affiliation(s)
- Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Bernardo Camacho
- Instituto Distrital de Ciencia Biotecnología e Investigación en Salud, IDCBIS, Bogota, Colombia
| | | | | | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | | | - Rubén Manrique
- Epidemiology and Biostatistics Research Group, Universidad CES, Medellin, Colombia
| | - Ruben D Mantilla
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to Tel-Aviv University, Tel Aviv, Israel; Laboratory of the Mosaics of Autoimmunity, Saint Petersburg State University, Saint-Petersburg, Russian Federation
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia; Clínica del Occidente, Bogota, Colombia.
| |
Collapse
|
10
|
Fitzpatrick EA, Wang J, Strome SE. Engineering of Fc Multimers as a Protein Therapy for Autoimmune Disease. Front Immunol 2020; 11:496. [PMID: 32269572 PMCID: PMC7109252 DOI: 10.3389/fimmu.2020.00496] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/04/2020] [Indexed: 02/06/2023] Open
Abstract
The success of Intravenous Immunoglobulin in treating autoimmune and inflammatory processes such as immune thrombocytopenia purpura and Kawasaki disease has led to renewed interest in developing recombinant molecules capable of recapitulating these therapeutic effects. The anti-inflammatory properties of IVIG are, in part, due to the Fc region of the IgG molecule, which interacts with activating or inhibitory Fcγ receptors (FcγRs), the neonatal Fc Receptor, non-canonical FcRs expressed by immune cells and complement proteins. In most cases, Fc interactions with these cognate receptors are dependent upon avidity—avidity which naturally occurs when polyclonal antibodies recognize unique antigens on a given target. The functional consequences of these avid interactions include antibody dependent cell-mediated cytotoxicity, antibody dependent cell phagocytosis, degranulation, direct killing, and/or complement activation—all of which are associated with long-term immunomodulatory effects. Many of these immunologic effects can be recapitulated using recombinant or non-recombinant approaches to induce Fc multimerization, affording the potential to develop a new class of therapeutics. In this review, we discuss the history of tolerance induction by immune complexes that has led to the therapeutic development of artificial Fc bearing immune aggregates and recombinant Fc multimers. The contribution of structure, aggregation and N-glycosylation to human IgG: FcγR interactions and the functional effect(s) of these interactions are reviewed. Understanding the mechanisms by which Fc multimers induce tolerance and attempts to engineer Fc multimers to target specific FcγRs and/or specific effector functions in autoimmune disorders is explored in detail.
Collapse
Affiliation(s)
- Elizabeth A Fitzpatrick
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| | - Jin Wang
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - S E Strome
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center (UTHSC), Memphis, TN, United States
| |
Collapse
|
11
|
Blumberg LJ, Humphries JE, Jones SD, Pearce LB, Holgate R, Hearn A, Cheung J, Mahmood A, Del Tito B, Graydon JS, Stolz LE, Bitonti A, Purohit S, de Graaf D, Kacena K, Andersen JT, Christianson GJ, Roopenian DC, Hubbard JJ, Gandhi AK, Lasseter K, Pyzik M, Blumberg RS. Blocking FcRn in humans reduces circulating IgG levels and inhibits IgG immune complex-mediated immune responses. SCIENCE ADVANCES 2019; 5:eaax9586. [PMID: 31897428 PMCID: PMC6920022 DOI: 10.1126/sciadv.aax9586] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/06/2019] [Indexed: 06/10/2023]
Abstract
The neonatal crystallizable fragment receptor (FcRn) functions as an intracellular protection receptor for immunoglobulin G (IgG). Recently, several clinical studies have reported the lowering of circulating monomeric IgG levels through FcRn blockade for the potential treatment of autoimmune diseases. Many autoimmune diseases, however, are derived from the effects of IgG immune complexes (ICs). We generated, characterized, and assessed the effects of SYNT001, a FcRn-blocking monoclonal antibody, in mice, nonhuman primates (NHPs), and humans. SYNT001 decreased all IgG subtypes and IgG ICs in the circulation of humans, as we show in a first-in-human phase 1, single ascending dose study. In addition, IgG IC induction of inflammatory pathways was dependent on FcRn and inhibited by SYNT001. These studies expand the role of FcRn in humans by showing that it controls not only IgG protection from catabolism but also inflammatory pathways associated with IgG ICs involved in a variety of autoimmune diseases.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antigen-Antibody Complex/immunology
- Autoantibodies/drug effects
- Autoimmune Diseases/drug therapy
- Cohort Studies
- Double-Blind Method
- Female
- Healthy Volunteers
- Histocompatibility Antigens Class I
- Humans
- Immunity, Humoral/immunology
- Immunoglobulin G/blood
- Immunoglobulin G/immunology
- Macaca fascicularis
- Male
- Mice
- Protein Binding
- Receptors, Fc/antagonists & inhibitors
Collapse
Affiliation(s)
| | | | - S. D. Jones
- BioProcess Technology Consultants, Woburn, MA 01801, USA
| | | | - R. Holgate
- Abzena, Babraham, Cambridge, CB22 3AT, UK
| | - A. Hearn
- Abzena, Babraham, Cambridge, CB22 3AT, UK
| | - J. Cheung
- New York Structural Biology Center, New York, NY 10027, USA
| | - A. Mahmood
- New York Structural Biology Center, New York, NY 10027, USA
| | - B. Del Tito
- Biologics Consulting, Alexandria, VA 22314, USA
| | | | | | | | - S. Purohit
- BioProcess Technology Consultants, Woburn, MA 01801, USA
| | | | - K. Kacena
- BioBridges, Wellesley, MA 02481, USA
| | - J. T. Andersen
- Department of Immunology and Centre for Immune Regulation, Oslo University Hospital Rikshospitalet and University of Oslo, Oslo 0424, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo 0424, Norway
| | | | | | - J. J. Hubbard
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA
| | - A. K. Gandhi
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - K. Lasseter
- Clinical Pharmacology of Miami, Miami, FL 33014, USA
| | - M. Pyzik
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - R. S. Blumberg
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Tonsawan P, Dylewski J, Lewis L, Blaine J. Knockout of the neonatal Fc receptor in cultured podocytes alters IL-6 signaling and the actin cytoskeleton. Am J Physiol Cell Physiol 2019; 317:C1048-C1060. [PMID: 31553647 PMCID: PMC6879880 DOI: 10.1152/ajpcell.00235.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 12/20/2022]
Abstract
The neonatal Fc receptor (FcRn) has been shown to be required for antigen presentation in dendritic cells, and global knockout of FcRn attenuates immune-mediated kidney disease. Podocytes express interleukin-6 (IL-6) receptor and produce IL-6 under proinflammatory conditions. Here we examined the role of FcRn in the IL-6-mediated inflammatory response in podocytes. We examined IL-6 production by ELISA and expression by qPCR in wild type (WT) and FcRn knockout (KO) podocytes after treatment with proinflammatory stimuli as well as IL-6-mediated signaling via the JAK/STAT pathway. We also examined podocyte motility in cultured WT and KO podocytes after a proinflammatory challenge. We found that FcRn KO podocytes produced minimal amount of IL-6 after treatment with albumin, IgG, or immune complexes whereas WT podocytes had a robust response. FcRn KO podocytes also had minimal expression of IL-6 compared with WT. By Western blotting, there was significantly less phosphorylated STAT3 in KO podocytes after treatment with IFNγ or immune complexes. In a scratch assay, FcRn KO podocytes showed increased motility comparted KO, suggesting a defect in actin dynamics. Cultured FcRn KO podocytes also demonstrated abnormal stress fibers compared with WT and the defect could be rescued by IL-6 treatment. This study shows that in podocytes, FcRn modulates the IL-6 mediated response to proinflammatory stimuli and regulates podocytes actin structure, motility and synaptopodin expression.
Collapse
Affiliation(s)
- Pantipa Tonsawan
- Division of Renal Disease and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
- Division of Nephrology, Department of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - James Dylewski
- Division of Renal Disease and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
- Department of Nephrology, Denver Health Medical Center, Denver, Colorado
| | - Linda Lewis
- Division of Renal Disease and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| | - Judith Blaine
- Division of Renal Disease and Hypertension, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
13
|
Pyzik M, Sand KMK, Hubbard JJ, Andersen JT, Sandlie I, Blumberg RS. The Neonatal Fc Receptor (FcRn): A Misnomer? Front Immunol 2019; 10:1540. [PMID: 31354709 PMCID: PMC6636548 DOI: 10.3389/fimmu.2019.01540] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Antibodies are essential components of an adaptive immune response. Immunoglobulin G (IgG) is the most common type of antibody found in circulation and extracellular fluids. Although IgG alone can directly protect the body from infection through the activities of its antigen binding region, the majority of IgG immune functions are mediated via proteins and receptors expressed by specialized cell subsets that bind to the fragment crystallizable (Fc) region of IgG. Fc gamma (γ) receptors (FcγR) belong to a broad family of proteins that presently include classical membrane-bound surface receptors as well as atypical intracellular receptors and cytoplasmic glycoproteins. Among the atypical FcγRs, the neonatal Fc receptor (FcRn) has increasingly gained notoriety given its intimate influence on IgG biology and its ability to also bind to albumin. FcRn functions as a recycling or transcytosis receptor that is responsible for maintaining IgG and albumin in the circulation, and bidirectionally transporting these two ligands across polarized cellular barriers. More recently, it has been appreciated that FcRn acts as an immune receptor by interacting with and facilitating antigen presentation of peptides derived from IgG immune complexes (IC). Here we review FcRn biology and focus on newer advances including how emerging FcRn-targeted therapies may affect the immune responses to IgG and IgG IC.
Collapse
Affiliation(s)
- Michal Pyzik
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Kine M K Sand
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jonathan J Hubbard
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Inger Sandlie
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Harvard Digestive Diseases Center, Boston, MA, United States
| |
Collapse
|
14
|
Human cytomegalovirus evades antibody-mediated immunity through endoplasmic reticulum-associated degradation of the FcRn receptor. Nat Commun 2019; 10:3020. [PMID: 31289263 PMCID: PMC6617459 DOI: 10.1038/s41467-019-10865-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 06/05/2019] [Indexed: 01/09/2023] Open
Abstract
Human cytomegalovirus (HCMV) can persistently infect humans, but how HCMV avoids humoral immunity is not clear. The neonatal Fc receptor (FcRn) controls IgG transport from the mother to the fetus and prolongs IgG half-life. Here we show that US11 inhibits the assembly of FcRn with β2m and retains FcRn in the endoplasmic reticulum (ER), consequently blocking FcRn trafficking to the endosome. Furthermore, US11 recruits the ubiquitin enzymes Derlin-1, TMEM129 and UbE2J2 to engage FcRn, consequently initiating the dislocation of FcRn from the ER to the cytosol and facilitating its degradation. Importantly, US11 inhibits IgG-FcRn binding, resulting in a reduction of IgG transcytosis across intestinal or placental epithelial cells and IgG degradation in endothelial cells. Hence, these results identify the mechanism by which HCMV infection exploits an ER-associated degradation pathway through US11 to disable FcRn functions. These results have implications for vaccine development and immune surveillance. Human cytomegalovirus (HCMV) can persist for the life of a host in the face of robust immune responses owing to a wide range of immune evasion strategies. Here Liu and colleagues show that HCMV evades the IgG-mediated response by the endoplasmic reticulum-associated degradation of the neonatal Fc receptor for IgG.
Collapse
|
15
|
Wiatr M, Merle NS, Boudhabhay I, Poillerat V, Rossini S, Lecerf M, Kaveri SV, Lacroix-Desmazes S, Roumenina LT, Dimitrov JD. Anti-inflammatory activity of intravenous immunoglobulin through scavenging of heme. Mol Immunol 2019; 111:205-208. [PMID: 31078967 PMCID: PMC6560225 DOI: 10.1016/j.molimm.2019.04.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 04/13/2019] [Accepted: 04/24/2019] [Indexed: 12/22/2022]
Abstract
Therapeutic intravenous immunoglobulin preparations (IVIg) are used for treatment of wide range of autoimmune and inflammatory diseases. Versatile mechanisms have been reported to contribute to the immunomodulatory effects of IVIg. Here we demonstrate that IVIg has a strong potential to inhibit pro-inflammatory effect of extracellular heme. Indeed, the presence of immunoglobulins reduced the potential of heme to activate the complement system on the surface of human endothelial cells. Since extracellular heme is considered as one of the principal pathogenic factors in hemolytic disorders, its therapeutic scavenging by IVIg may have significant clinical repercussions.
Collapse
Affiliation(s)
- Marie Wiatr
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Nicolas S Merle
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Idris Boudhabhay
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Victoria Poillerat
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Sofia Rossini
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Maxime Lecerf
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Srini V Kaveri
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Sébastian Lacroix-Desmazes
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Lubka T Roumenina
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France
| | - Jordan D Dimitrov
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France.
| |
Collapse
|
16
|
Smith B, Kiessling A, Lledo-Garcia R, Dixon KL, Christodoulou L, Catley MC, Atherfold P, D'Hooghe LE, Finney H, Greenslade K, Hailu H, Kevorkian L, Lightwood D, Meier C, Munro R, Qureshi O, Sarkar K, Shaw SP, Tewari R, Turner A, Tyson K, West S, Shaw S, Brennan FR. Generation and characterization of a high affinity anti-human FcRn antibody, rozanolixizumab, and the effects of different molecular formats on the reduction of plasma IgG concentration. MAbs 2018; 10:1111-1130. [PMID: 30130439 PMCID: PMC6291300 DOI: 10.1080/19420862.2018.1505464] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Rozanolixizumab (UCB7665), a humanized high-affinity anti-human neonatal Fc receptor (FcRn) monoclonal antibody (IgG4P), has been developed to reduce pathogenic IgG in autoimmune and alloimmune diseases. We document the antibody isolation and compare rozanolixizumab with the same variable region expressed in various mono-, bi- and trivalent formats. We report activity data for rozanolixizumab and the different molecular formats in human cells, FcRn-transgenic mice, and cynomolgus monkeys. Rozanolixizumab, considered the most effective molecular format, dose-dependently and selectively reduced plasma IgG concentrations in an FcRn-transgenic mouse model (no effect on albumin). Intravenous (IV) rozanolixizumab dosing in cynomolgus monkeys demonstrated non-linear pharmacokinetics indicative of target-mediated drug disposition; single IV rozanolixizumab doses (30 mg/kg) in cynomolgus monkeys reduced plasma IgG concentration by 69% by Day 7 post-administration. Daily IV administration of rozanolixizumab (initial 30 mg/kg loading dose; 5 mg/kg daily thereafter) reduced plasma IgG concentrations in all cynomolgus monkeys, with low concentrations maintained throughout the treatment period (42 days). In a 13-week toxicology study in cynomolgus monkeys, supra-pharmacological subcutaneous and IV doses of rozanolixizumab (≤ 150 mg/kg every 3 days) were well tolerated, inducing sustained (but reversible) reductions in IgG concentrations by up to 85%, with no adverse events observed. We have demonstrated accelerated natural catabolism of IgG through inhibition of IgG:FcRn interactions in mice and cynomolgus monkeys. Inhibition of FcRn with rozanolixizumab may provide a novel therapeutic approach to reduce pathogenic IgG in human autoimmune disease. Rozanolixizumab is being investigated in patients with immune thrombocytopenia (NCT02718716) and myasthenia gravis (NCT03052751).
Collapse
|
17
|
Stöppler D, Macpherson A, Smith-Penzel S, Basse N, Lecomte F, Deboves H, Taylor RD, Norman T, Porter J, Waters LC, Westwood M, Cossins B, Cain K, White J, Griffin R, Prosser C, Kelm S, Sullivan AH, Fox D, Carr MD, Henry A, Taylor R, Meier BH, Oschkinat H, Lawson AD. Insight into small molecule binding to the neonatal Fc receptor by X-ray crystallography and 100 kHz magic-angle-spinning NMR. PLoS Biol 2018; 16:e2006192. [PMID: 29782488 PMCID: PMC5983862 DOI: 10.1371/journal.pbio.2006192] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/01/2018] [Accepted: 05/02/2018] [Indexed: 01/09/2023] Open
Abstract
Aiming at the design of an allosteric modulator of the neonatal Fc receptor (FcRn)-Immunoglobulin G (IgG) interaction, we developed a new methodology including NMR fragment screening, X-ray crystallography, and magic-angle-spinning (MAS) NMR at 100 kHz after sedimentation, exploiting very fast spinning of the nondeuterated soluble 42 kDa receptor construct to obtain resolved proton-detected 2D and 3D NMR spectra. FcRn plays a crucial role in regulation of IgG and serum albumin catabolism. It is a clinically validated drug target for the treatment of autoimmune diseases caused by pathogenic antibodies via the inhibition of its interaction with IgG. We herein present the discovery of a small molecule that binds into a conserved cavity of the heterodimeric, extracellular domain composed of an α-chain and β2-microglobulin (β2m) (FcRnECD, 373 residues). X-ray crystallography was used alongside NMR at 100 kHz MAS with sedimented soluble protein to explore possibilities for refining the compound as an allosteric modulator. Proton-detected MAS NMR experiments on fully protonated [13C,15N]-labeled FcRnECD yielded ligand-induced chemical-shift perturbations (CSPs) for residues in the binding pocket and allosteric changes close to the interface of the two receptor heterodimers present in the asymmetric unit as well as potentially in the albumin interaction site. X-ray structures with and without ligand suggest the need for an optimized ligand to displace the α-chain with respect to β2m, both of which participate in the FcRnECD-IgG interaction site. Our investigation establishes a method to characterize structurally small molecule binding to nondeuterated large proteins by NMR, even in their glycosylated form, which may prove highly valuable for structure-based drug discovery campaigns.
Collapse
Affiliation(s)
- Daniel Stöppler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | | | | | | | | | | | | | | | - Lorna C. Waters
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | | | | | | | | | | | | | | | - Amy H. Sullivan
- Beryllium Discovery, Bedford, Massachusetts, United States of America
| | - David Fox
- Beryllium Discovery, Bedford, Massachusetts, United States of America
| | - Mark D. Carr
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | | | | | - Beat H. Meier
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | - Hartmut Oschkinat
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- * E-mail: (HO); (ADL)
| | | |
Collapse
|
18
|
Seijsing J, Yu S, Frejd FY, Höiden-Guthenberg I, Gräslund T. In vivo depletion of serum IgG by an affibody molecule binding the neonatal Fc receptor. Sci Rep 2018; 8:5141. [PMID: 29572538 PMCID: PMC5865129 DOI: 10.1038/s41598-018-23481-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 03/12/2018] [Indexed: 12/28/2022] Open
Abstract
Lowering the total level of Immunoglobulin G (IgG) in circulation is a promising general treatment option for many autoimmune diseases driven by pathogenic autoantibodies. The half-life of IgG in circulation is unusually long as a consequence of its interaction with the neonatal Fc receptor (FcRn), which protects it from lysosomal degradation by cells in contact with blood. Blocking the IgG/FcRn interaction prevents FcRn-mediated rescue, which may lead to increased catabolism and a lowering of the total IgG level. Here, we find that an engineered alternative scaffold protein, an affibody molecule, interacting specifically with FcRn, is able to block the IgG/FcRn interaction in vitro. The affibody molecule (ZFcRn) was expressed alone or as a fusion to an albumin binding domain (ABD), to extend its half-life in circulation, in both cases with retained affinity and blocking potential. Repeated i.v. injections in mice of ZFcRn and ZFcRn-ABD were found to result in an up to 40% reduction of the IgG serum-level after 5 days. Potential applications of ZFcRn as a general treatment modality for autoimmune diseases are discussed.
Collapse
Affiliation(s)
- Johan Seijsing
- School of Biotechnology, KTH Royal Institute of Technology, Roslagstullsbacken 21, 11417, Stockholm, Sweden.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Shengze Yu
- School of Biotechnology, KTH Royal Institute of Technology, Roslagstullsbacken 21, 11417, Stockholm, Sweden
| | | | | | - Torbjörn Gräslund
- School of Biotechnology, KTH Royal Institute of Technology, Roslagstullsbacken 21, 11417, Stockholm, Sweden.
| |
Collapse
|
19
|
Priming the body to receive the therapeutic agent to redefine treatment benefit/risk profile. Sci Rep 2018; 8:4797. [PMID: 29556068 PMCID: PMC5859131 DOI: 10.1038/s41598-018-23140-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/06/2018] [Indexed: 12/16/2022] Open
Abstract
Many therapeutic agents offer a low useful dose (dose responsible for efficacy)/useless dose (dose eliminated or responsible for toxicity) ratio, mainly due to the fact that therapeutic agents must ensure in one single object all the functions required to deliver the treatment, which leads to compromises in their physico-chemical design. Here we introduce the concept of priming the body to receive the treatment by uncorrelating these functions into two distinct objects sequentially administered: a nanoprimer occupying transiently the main pathway responsible for therapeutic agent limited benefit/risk ratio followed by the therapeutic agent. The concept was evaluated for different nature of therapeutic agents: For nanomedicines we designed a liposomal nanoprimer presenting preferential hepatic accumulation without sign of acute toxicity. This nanoprimer was able to increase the blood bioavailability of nanomedicine correlated with a lower hepatic accumulation. Finally this nanoprimer markedly enhanced anti-tumor efficacy of irinotecan loaded liposomes in the HT-29 tumor model when compared to the nanomedicine alone. Then, for small molecules we demonstrated the ability of a cytochrome inhibitor loaded nanoprimer to increase efficacy of docetaxel treatment. These results shown that specific nanoprimers could be designed for each family of therapeutic agents to answer to their specific needs.
Collapse
|
20
|
Nandakumar KS. Targeting IgG in Arthritis: Disease Pathways and Therapeutic Avenues. Int J Mol Sci 2018; 19:E677. [PMID: 29495570 PMCID: PMC5877538 DOI: 10.3390/ijms19030677] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/25/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is a polygenic and multifactorial syndrome. Many complex immunological and genetic interactions are involved in the final outcome of the clinical disease. Autoantibodies (rheumatoid factors, anti-citrullinated peptide/protein antibodies) are present in RA patients' sera for a long time before the onset of clinical disease. Prior to arthritis onset, in the autoantibody response, epitope spreading, avidity maturation, and changes towards a pro-inflammatory Fc glycosylation phenotype occurs. Genetic association of epitope specific autoantibody responses and the induction of inflammation dependent and independent changes in the cartilage by pathogenic autoantibodies emphasize the crucial contribution of antibody-initiated inflammation in RA development. Targeting IgG by glyco-engineering, bacterial enzymes to specifically cleave IgG/alter N-linked Fc-glycans at Asn 297 or blocking the downstream effector pathways offers new avenues to develop novel therapeutics for arthritis treatment.
Collapse
Affiliation(s)
- Kutty Selva Nandakumar
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China.
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
21
|
Ahmed AR, Carrozzo M, Caux F, Cirillo N, Dmochowski M, Alonso AE, Gniadecki R, Hertl M, López-Zabalza MJ, Lotti R, Pincelli C, Pittelkow M, Schmidt E, Sinha AA, Sprecher E, Grando SA. Monopathogenic vs multipathogenic explanations of pemphigus pathophysiology. Exp Dermatol 2018; 25:839-846. [PMID: 27305362 DOI: 10.1111/exd.13106] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2016] [Indexed: 01/31/2023]
Abstract
This viewpoint highlights major, partly controversial concepts about the pathogenesis of pemphigus. The monopathogenic theory explains intra-epidermal blistering through the "desmoglein (Dsg) compensation" hypothesis, according to which an antibody-dependent disabling of Dsg 1- and/or Dsg 3-mediated cell-cell attachments of keratinocytes (KCs) is sufficient to disrupt epidermal integrity and cause blistering. The multipathogenic theory explains intra-epidermal blistering through the "multiple hit" hypothesis stating that a simultaneous and synchronized inactivation of the physiological mechanisms regulating and/or mediating intercellular adhesion of KCs is necessary to disrupt epidermal integrity. The major premise for a multipathogenic theory is that a single type of autoantibody induces only reversible changes, so that affected KCs can recover due to a self-repair. The damage, however, becomes irreversible when the salvage pathway and/or other cell functions are altered by a partnering autoantibody and/or other pathogenic factors. Future studies are needed to (i) corroborate these findings, (ii) characterize in detail patient populations with non-Dsg-specific autoantibodies, and (iii) determine the extent of the contribution of non-Dsg antibodies in disease pathophysiology.
Collapse
Affiliation(s)
- A Razzaque Ahmed
- Department of Dermatology of Tufts University and Center for Blistering Diseases, Boston, MA, USA
| | - Marco Carrozzo
- School of Dental Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Frédéric Caux
- Department of Dermatology, University Paris 13, Avicenne Hospital, APHP, Bobigny, France
| | - Nicola Cirillo
- Melbourne Dental School and Oral Health CRC, The University of Melbourne, Melbourne, Vic., Australia
| | - Marian Dmochowski
- Autoimmune Blistering Dermatoses Section, Department of Dermatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Agustín España Alonso
- Department of Dermatology, School of Medicine, University Clinic of Navarra, University of Navarra, Navarra, Spain
| | - Robert Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, AB, Canada
| | - Michael Hertl
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | | | - Roberta Lotti
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Pincelli
- Department of Dermatology, University of Modena and Reggio Emilia, Modena, Italy
| | - Mark Pittelkow
- Department of Dermatology, Mayo Clinic, Scottsdale, AZ, USA
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Animesh A Sinha
- Department of Dermatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Eli Sprecher
- Department of Dermatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Sergei A Grando
- Institute for Immunology and Departments of Dermatology and Biological Chemistry, University of California, Irvine, CA, USA.
| |
Collapse
|
22
|
Kenniston JA, Taylor BM, Conley GP, Cosic J, Kopacz KJ, Lindberg AP, Comeau SR, Atkins K, Bullen J, TenHoor C, Adelman BA, Sexton DJ, Edwards TE, Nixon AE. Structural basis for pH-insensitive inhibition of immunoglobulin G recycling by an anti-neonatal Fc receptor antibody. J Biol Chem 2017; 292:17449-17460. [PMID: 28878017 DOI: 10.1074/jbc.m117.807396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/17/2017] [Indexed: 11/06/2022] Open
Abstract
The neonatal Fc receptor FcRn plays a critical role in the trafficking of IgGs across tissue barriers and in retaining high circulating concentrations of both IgG and albumin. Although generally beneficial from an immunological perspective in maintaining IgG populations, FcRn can contribute to the pathogenesis of autoimmune disorders when an abnormal immune response targets normal biological components. We previously described a monoclonal antibody (DX-2507) that binds to FcRn with high affinity at both neutral and acidic pH, prevents the simultaneous binding of IgG, and reduces circulating IgG levels in preclinical animal models. Here, we report a 2.5 Å resolution X-ray crystal structure of an FcRn-DX-2507 Fab complex, revealing a nearly complete overlap of the IgG-Fc binding site in FcRn by complementarity-determining regions in DX-2507. This overlap explains how DX-2507 blocks IgG binding to FcRn and thereby shortens IgG half-life by preventing IgGs from recycling back into circulation. Moreover, the complex structure explains how the DX-2507 interaction is pH-insensitive unlike normal Fc interactions and how serum albumin levels are unaffected by DX-2507 binding. These structural studies could inform antibody-based therapeutic approaches for limiting the effects of IgG-mediated autoimmune disease.
Collapse
Affiliation(s)
- Jon A Kenniston
- From Shire, Lexington, Massachusetts 02421, .,Dyax Corp., Burlington, Massachusetts 01803
| | - Brandy M Taylor
- Beryllium Discovery Corp., Bainbridge Island, Washington 98110, and
| | | | | | | | | | | | - Kateri Atkins
- Beryllium Discovery Corp., Bainbridge Island, Washington 98110, and
| | - Jameson Bullen
- Beryllium Discovery Corp., Bainbridge Island, Washington 98110, and
| | | | | | | | - Thomas E Edwards
- Beryllium Discovery Corp., Bainbridge Island, Washington 98110, and
| | | |
Collapse
|
23
|
The neonatal Fc receptor, FcRn, as a target for drug delivery and therapy. Adv Drug Deliv Rev 2015; 91:109-24. [PMID: 25703189 DOI: 10.1016/j.addr.2015.02.005] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/10/2015] [Accepted: 02/11/2015] [Indexed: 12/22/2022]
Abstract
Immunoglobulin G (IgG)-based drugs are arguably the most successful class of protein therapeutics due in part to their remarkably long blood circulation. This arises from IgG interaction with the neonatal Fc receptor, FcRn. FcRn is the central regulator of IgG and albumin homeostasis throughout life and is increasingly being recognized as an important player in autoimmune disease, mucosal immunity, and tumor immune surveillance. Various engineering approaches that hijack or disrupt the FcRn-mediated transport pathway have been devised to develop long-lasting and non-invasive protein therapeutics, protein subunit vaccines, and therapeutics for treatment of autoimmune and infectious disease. In this review, we highlight the diverse biological functions of FcRn, emerging therapeutic opportunities, as well as the associated challenges of targeting FcRn for drug delivery and disease therapy.
Collapse
|
24
|
Mikulska JE. Analysis of Response Elements Involved in the Regulation of the Human Neonatal Fc Receptor Gene (FCGRT). PLoS One 2015; 10:e0135141. [PMID: 26252948 PMCID: PMC4529178 DOI: 10.1371/journal.pone.0135141] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 07/19/2015] [Indexed: 12/26/2022] Open
Abstract
Human epithelial, endothelial and PMA-differentiated THP-1 cell lines were used as model systems to study the transcriptional regulation of the human FCGRT gene encoding the alpha chain of hFcRn. The data obtained from site-directed mutagenesis in transient transfection experiments indicate that the Sp1 sites at positions -641, -635, and -313, CF1/YY1 elements at positions -586 and -357, and the AP-1 motif at -276 within the-660/-233 fragment of the human FCGRT promoter (hFCGRT) participate in the regulation of human FCGRT in all selected cell lines. However, their individual contribution to promoter activity is not equivalent. The Sp1 binding site at -313 and the AP-1 site at -276 are critical for the activity of the hFCGRT promoter in epithelial and endothelial cells. Moreover, the CF1/YY1 site at -586 in differentiated THP-1 cells, plays an essential role in the transcriptional activity of the promoter. In addition, the C/EBPbeta binding site at -497 of the hFCGRT promoter in epithelial and endothelial cells, and the C/EBPbeta motif located at -497 and -233 within the hFCGRT promoter in differentiated THP-1 cells may function as positive regulatory sequences in response to LPS or PMA stimulation. EMSA and supershift analyses showed that the functionally identified binding motifs in the hFCGRT promoter were able to specifically interact with their corresponding (Sp1, Sp2, Sp3, c-Fos, c-Jun, YY1, and C/EBPbeta or C/EBPdelta) transcription factors (TFs), suggesting their possible involvement in the regulation of the human FCGRT gene expression.
Collapse
Affiliation(s)
- Joanna E. Mikulska
- Department of Immunochemistry, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- * E-mail:
| |
Collapse
|
25
|
Proetzel G, Wiles MV, Roopenian DC. Genetically engineered humanized mouse models for preclinical antibody studies. BioDrugs 2015; 28:171-80. [PMID: 24150980 DOI: 10.1007/s40259-013-0071-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The use of genetic engineering has vastly improved our capabilities to create animal models relevant in preclinical research. With the recent advances in gene-editing technologies, it is now possible to very rapidly create highly tunable mouse models as needs arise. Here, we provide an overview of genetic engineering methods, as well as the development of humanized neonatal Fc receptor (FcRn) models and their use for monoclonal antibody in vivo studies.
Collapse
|
26
|
Nixon AE, Chen J, Sexton DJ, Muruganandam A, Bitonti AJ, Dumont J, Viswanathan M, Martik D, Wassaf D, Mezo A, Wood CR, Biedenkapp JC, TenHoor C. Fully human monoclonal antibody inhibitors of the neonatal fc receptor reduce circulating IgG in non-human primates. Front Immunol 2015; 6:176. [PMID: 25954273 PMCID: PMC4407741 DOI: 10.3389/fimmu.2015.00176] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 03/29/2015] [Indexed: 12/24/2022] Open
Abstract
The therapeutic management of antibody-mediated autoimmune disease typically involves immunosuppressant and immunomodulatory strategies. However, perturbing the fundamental role of the neonatal Fc receptor (FcRn) in salvaging IgG from lysosomal degradation provides a novel approach - depleting the body of pathogenic immunoglobulin by preventing IgG binding to FcRn and thereby increasing the rate of IgG catabolism. Herein, we describe the discovery and preclinical evaluation of fully human monoclonal IgG antibody inhibitors of FcRn. Using phage display, we identified several potent inhibitors of human-FcRn in which binding to FcRn is pH-independent, with over 1000-fold higher affinity for human-FcRn than human IgG-Fc at pH 7.4. FcRn antagonism in vivo using a human-FcRn knock-in transgenic mouse model caused enhanced catabolism of exogenously administered human IgG. In non-human primates, we observed reductions in endogenous circulating IgG of >60% with no changes in albumin, IgM, or IgA. FcRn antagonism did not disrupt the ability of non-human primates to mount IgM/IgG primary and secondary immune responses. Interestingly, the therapeutic anti-FcRn antibodies had a short serum half-life but caused a prolonged reduction in IgG levels. This may be explained by the high affinity of the antibodies to FcRn at both acidic and neutral pH. These results provide important preclinical proof of concept data in support of FcRn antagonism as a novel approach to the treatment of antibody-mediated autoimmune diseases.
Collapse
Affiliation(s)
| | - Jie Chen
- Dyax Corp. , Burlington, MA , USA
| | | | | | - Alan J Bitonti
- Syntonix Pharmaceuticals (a wholly-owned subsidiary of Biogen Idec.) , Waltham, MA , USA
| | | | | | | | | | - Adam Mezo
- Syntonix Pharmaceuticals (a wholly-owned subsidiary of Biogen Idec.) , Waltham, MA , USA
| | | | | | | |
Collapse
|
27
|
Rath T, Baker K, Pyzik M, Blumberg RS. Regulation of immune responses by the neonatal fc receptor and its therapeutic implications. Front Immunol 2015; 5:664. [PMID: 25601863 PMCID: PMC4283642 DOI: 10.3389/fimmu.2014.00664] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 12/10/2014] [Indexed: 11/13/2022] Open
Abstract
As a single receptor, the neonatal Fc receptor (FcRn) is critically involved in regulating albumin and IgG serum concentrations by protecting these two ligands from degradation. In addition to these essential homeostatic functions, FcRn possesses important functions in regulating immune responses that are equally as critical and are increasingly coming to attention. During the first stages of life, FcRn mediates the passive transfer of IgG across the maternal placenta or neonatal intestinal walls of mammals, thereby conferring passive immunity to the offspring before and after birth. In fact, FcRn is one of the very few molecules that are known to move from luminal to serosal membranes of polarized cells that form epithelial barriers of the lung and intestines. Together with FcRn's recently explored critical role in eliciting MHC II presentation and MHC class I cross-presentation of IgG-complexed antigen, this renders FcRn capable of exerting broad and potent functions in regulating immune responses and immunosurveillance at mucosal sites. Further, it is now clear that FcRn dependent mucosal absorption of therapeutic molecules is a clinically feasible and potent novel route of non-invasive drug delivery, and the interaction between FcRn and IgG has also been utilized for the acquisition of humoral immunity at mucosal sites. In this review, we begin by briefly summarizing the basic knowledge on FcRn expression and IgG binding, then describe more recent discoveries pertaining to the mechanisms by which FcRn orchestrates IgG related mucosal immune responses and immunosurveillance at host-environment interfaces within the adult organism. Finally, we outline how the knowledge of actions of FcRn at mucosal boundaries can be capitalized for the development and engineering of powerful mucosal vaccination strategies and novel routes for the non-invasive delivery of Fc-based therapeutics.
Collapse
Affiliation(s)
- Timo Rath
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Division of Gastroenterology, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nueremberg , Erlangen , Germany
| | - Kristi Baker
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Michal Pyzik
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Richard S Blumberg
- Department of Medicine, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA ; Harvard Digestive Diseases Center , Boston, MA , USA
| |
Collapse
|
28
|
Abstract
Mast cells are innate immune effector cells that reside in the healthy synovial sublining and expand in number with inflammation. These cells can play an important role in initiation of arthritis, but much about their biology and importance remains obscure. This chapter reviews the use of animal models for the study of mast cells in arthritis, with a particular focus on the K/BxN serum transfer model. We discuss tissue preparation and histological analysis for the assessment of joint inflammation, injury, and the presence and phenotype of synovial mast cells, as well as the use of bone marrow-derived mast cell (BMMC) engraftment into W/Wv mice as a tool to isolate the role of mast cells in joint inflammation and injury.
Collapse
Affiliation(s)
- Peter A Nigrovic
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Smith 516B, One Jimmy Fund Way, Boston, MA, 02115, USA,
| | | |
Collapse
|
29
|
Abstract
INTRODUCTION The Fc receptors (FcRs) and their interactions with immunoglobulin and innate immune opsonins, such as C-reactive protein, are key players in humoral and cellular immune responses. As the effector mechanism for some therapeutic monoclonal antibodies, and often a contributor to the pathogenesis and progression of autoimmunity, FcRs are promising targets for treating autoimmune diseases. AREAS COVERED This review discusses the nature of different FcRs and the various mechanisms of their involvement in initiating and modulating immunocyte functions and their biological consequences. It describes a range of current strategies in targeting FcRs and manipulating their interaction with specific ligands, while presenting the pros and cons of these approaches. This review also discusses potential new strategies including regulation of FcR expression and receptor crosstalk. EXPERT OPINION FcRs are appealing targets in the treatment of inflammatory autoimmune diseases. However, there are still knowledge limitations and technical challenges, the most important being a better understanding of the individual roles of each of the FcRs and enhancement of the specificity in targeting particular cell types and specific FcRs.
Collapse
Affiliation(s)
- Xinrui Li
- The University of Alabama , SHEL 272, 1825 University Blvd, Birmingham, AL 35294 , USA
| | | |
Collapse
|
30
|
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: 0.8] [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
|
31
|
Guilliams M, Bruhns P, Saeys Y, Hammad H, Lambrecht BN. The function of Fcγ receptors in dendritic cells and macrophages. Nat Rev Immunol 2014; 14:94-108. [PMID: 24445665 DOI: 10.1038/nri3582] [Citation(s) in RCA: 484] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) and macrophages use various receptors to recognize foreign antigens and to receive feedback control from adaptive immune cells. Although it was long believed that all immunoglobulin Fc receptors are universally expressed by phagocytes, recent findings indicate that only monocyte-derived DCs and macrophages express high levels of activating Fc receptors for IgG (FcγRs), whereas conventional and plasmacytoid DCs express the inhibitory FcγR. In this Review, we discuss how the uptake, processing and presentation of antigens by DCs and macrophages is influenced by FcγR recognition of immunoglobulins and immune complexes in the steady state and during inflammation.
Collapse
Affiliation(s)
- Martin Guilliams
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Pierre Bruhns
- 1] Institut Pasteur, Département d'Immunologie, Laboratoire Anticorps en Thérapie et Pathologie, 75015 Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U760, 75015 Paris, France
| | - Yvan Saeys
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Hamida Hammad
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- 1] Laboratory of Immunoregulation, VIB Inflammation Research Center, 9052 Ghent, Belgium. [2] Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium. [3] Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
32
|
Olaru F, Luo W, Suleiman H, St John PL, Ge L, Mezo AR, Shaw AS, Abrahamson DR, Miner JH, Borza DB. Neonatal Fc receptor promotes immune complex-mediated glomerular disease. J Am Soc Nephrol 2013; 25:918-25. [PMID: 24357670 DOI: 10.1681/asn.2013050498] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The neonatal Fc receptor (FcRn) is a major regulator of IgG and albumin homeostasis systemically and in the kidneys. We investigated the role of FcRn in the development of immune complex-mediated glomerular disease in mice. C57Bl/6 mice immunized with the noncollagenous domain of the α3 chain of type IV collagen (α3NC1) developed albuminuria associated with granular capillary loop deposition of exogenous antigen, mouse IgG, C3 and C5b-9, and podocyte injury. High-resolution imaging showed abundant IgG deposition in the expanded glomerular basement membrane, especially in regions corresponding to subepithelial electron dense deposits. FcRn-null and -humanized mice immunized with α3NC1 developed no albuminuria and had lower levels of serum IgG anti-α3NC1 antibodies and reduced glomerular deposition of IgG, antigen, and complement. Our results show that FcRn promotes the formation of subepithelial immune complexes and subsequent glomerular pathology leading to proteinuria, potentially by maintaining higher serum levels of pathogenic IgG antibodies. Therefore, reducing pathogenic IgG levels by pharmacologic inhibition of FcRn may provide a novel approach for the treatment of immune complex-mediated glomerular diseases. As proof of concept, we showed that a peptide inhibiting the interaction between human FcRn and human IgG accelerated the degradation of human IgG anti-α3NC1 autoantibodies injected into FCRN-humanized mice as effectively as genetic ablation of FcRn, thus preventing the glomerular deposition of immune complexes containing human IgG.
Collapse
Affiliation(s)
- Florina Olaru
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Wentian Luo
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Patricia L St John
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Linna Ge
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Adam R Mezo
- Biogen Idec Hemophilia, Waltham, Massachusetts; and
| | | | - Dale R Abrahamson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jeffrey H Miner
- Renal Division, Washington University School of Medicine, St. Louis, Missouri
| | - Dorin-Bogdan Borza
- Division of Nephrology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Microbiology and Immunology, Meharry Medical College, Nashville, Tennessee
| |
Collapse
|
33
|
Haasken S, Auger JL, Taylor JJ, Hobday PM, Goudy BD, Titcombe PJ, Mueller DL, Binstadt BA. Macrophage scavenger receptor 1 (Msr1, SR-A) influences B cell autoimmunity by regulating soluble autoantigen concentration. THE JOURNAL OF IMMUNOLOGY 2013; 191:1055-62. [PMID: 23794629 DOI: 10.4049/jimmunol.1201680] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The class A macrophage scavenger receptor Msr1 (SR-A, CD204) has been reported to participate in the maintenance of immunological tolerance. We investigated the role of Msr1 in a mouse model of autoantibody-dependent arthritis. Genetic deficiency of Msr1 in K/BxN TCR transgenic mice decreased the incidence and severity of arthritis because of decreased autoantibody production. Despite normal initial activation of autoreactive CD4(+) T cells, potentially autoreactive B cells in Msr1(-/-) K/BxN mice retained a naive phenotype and did not expand. This was not due to an intrinsic B cell defect. Rather, we found that macrophages lacking Msr1 were inefficient at taking up the key autoantigen glucose-6-phosphate isomerase and that Msr1-deficient mice had elevated serum concentrations of glucose-6-phosphate isomerase. Arthritis developed normally when bone marrow from Msr1(-/-) K/BxN mice was transplanted into hosts whose macrophages did express Msr1. Thus, Msr1 can regulate the concentration of a soluble autoantigen. In this model, the absence of Msr1 led to higher levels of soluble autoantigen and protected mice from developing pathogenic autoantibodies, likely because of altered cognate interactions of autoreactive T and B cells with impaired differentiation of follicular Th cells.
Collapse
Affiliation(s)
- Stefanie Haasken
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Rath T, Kuo TT, Baker K, Qiao SW, Kobayashi K, Yoshida M, Roopenian D, Fiebiger E, Lencer WI, Blumberg RS. The immunologic functions of the neonatal Fc receptor for IgG. J Clin Immunol 2013; 33 Suppl 1:S9-17. [PMID: 22948741 PMCID: PMC3548031 DOI: 10.1007/s10875-012-9768-y] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 08/09/2012] [Indexed: 01/05/2023]
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), as a single receptor, is capable of regulating both of these molecules and has become an important focus of investigation. In addition to these essential protection functions, FcRn possesses a number of other functions that are equally as critical and are increasingly coming to attention. 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 via transcytosis across polarized cells. FcRn is also expressed by hematopoietic cells; consistent with this, FcRn regulates MHC class II presentation and MHC class I cross-presentation by dendritic cells. As such, FcRn plays an important role in immune surveillance throughout adult life. The increasing appreciation for FcRn in both homeostatic and pathological conditions is generating an intense interest in the potential for therapeutic modulation of FcRn binding to IgG and albumin.
Collapse
Affiliation(s)
- Timo Rath
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, UDA.
| |
Collapse
|
36
|
Christianson GJ, Sun VZ, Akilesh S, Pesavento E, Proetzel G, Roopenian DC. Monoclonal antibodies directed against human FcRn and their applications. MAbs 2012; 4:208-16. [PMID: 22453095 DOI: 10.4161/mabs.4.2.19397] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The MHC class I-like Fc receptor (FcRn) is an intracellular trafficking Fc receptor that is uniquely responsible for the extended serum half-life of antibodies of the IgG subclass and their ability to transport across cellular barriers. By performing these functions, FcRn affects numerous facets of antibody biology and pathobiology. Its critical role in controlling IgG pharmacokinetics has been leveraged for the design of therapeutic antibodies and related biologics. FcRn also traffics serum albumin and is responsible for the enhanced pharmacokinetic properties of albumin-conjugated therapeutics. The understanding of FcRn and its therapeutic applications has been limited by a paucity of reliable serological reagents against human FcRn. Here, we describe the properties of a new panel of highly specific monoclonal antibodies (mAbs) directed against human FcRn with diverse epitope specificities. We show that this antibody panel can be used to study the tissue expression pattern of human FcRn, to selectively block IgG and serum albumin binding to human FcRn in vitro and to inhibit FcRn function in vivo. This mAb panel provides a powerful resource for probing the biology of human FcRn and for the evaluation of therapeutic FcRn blockade strategies.
Collapse
Affiliation(s)
| | | | - Shreeram Akilesh
- The Jackson Laboratory; Bar Harbor, ME USA; Current address: Barnes-Jewish Hospital; St. Louis, MO USA
| | - Emanuele Pesavento
- The Jackson Laboratory; Bar Harbor, ME USA; Current address: VIB Department of Molecular and Cellular Interactions; Laboratory for Cellular and Molecular Immunology; Vrije Universiteit; Brussels, Belgium
| | | | | |
Collapse
|
37
|
IgG placental transfer in healthy and pathological pregnancies. Clin Dev Immunol 2011; 2012:985646. [PMID: 22235228 PMCID: PMC3251916 DOI: 10.1155/2012/985646] [Citation(s) in RCA: 625] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 07/24/2011] [Indexed: 12/13/2022]
Abstract
Placental transfer of maternal IgG antibodies to the fetus is an important mechanism that provides protection to the infant while his/her humoral response is inefficient. IgG is the only antibody class that significantly crosses the human placenta. This crossing is mediated by FcRn expressed on syncytiotrophoblast cells. There is evidence that IgG transfer depends on the following: (i) maternal levels of total IgG and specific antibodies, (ii) gestational age, (iii) placental integrity, (iv) IgG subclass, and (v) nature of antigen, being more intense for thymus-dependent ones. These features represent the basis for maternal immunization strategies aimed at protecting newborns against neonatal and infantile infectious diseases. In some situations, such as mothers with primary immunodeficiencies, exogenous IgG acquired by intravenous immunoglobulin therapy crosses the placenta in similar patterns to endogenous immunoglobulins and may also protect the offspring from infections in early life. Inversely, harmful autoantibodies may cross the placenta and cause transitory autoimmune disease in the neonate.
Collapse
|
38
|
McPhee CG, Sproule TJ, Shin DM, Bubier JA, Schott WH, Steinbuck MP, Avenesyan L, Morse HC, Roopenian DC. MHC class I family proteins retard systemic lupus erythematosus autoimmunity and B cell lymphomagenesis. THE JOURNAL OF IMMUNOLOGY 2011; 187:4695-704. [PMID: 21964024 DOI: 10.4049/jimmunol.1101776] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Dysregulation of the T cell-dependent Ab response can lead to numerous immunological disorders, ranging from systemic lupus erythematosus to B cell lymphomas. Cellular processes governed by MHC class II proteins play a major role in this response and its dysregulation. The extent to which processes controlled by the diverse family of MHC class I proteins impact such autoimmune and neoplastic disorders, however, is less clear. In this study, we genetically dissect the contributions of individual MHC class I family members and the pathological processes under their control in the systemic lupus erythematosus-like disease of BXSB.Yaa mice and B cell lymphomagenesis of SJL mice. This study reveals a powerful repressive regulatory axis comprised of MHC class I-dependent CD8(+) T cells and NK cells. These results indicate that the predominant role of the MHC class I protein family in such immunological disorders is to protect from more aggressive diseases.
Collapse
|
39
|
Amrani YM, Gill J, Matevossian A, Alonzo ES, Yang C, Shieh JH, Moore MA, Park CY, Sant'Angelo DB, Denzin LK. The Paf oncogene is essential for hematopoietic stem cell function and development. ACTA ACUST UNITED AC 2011; 208:1757-65. [PMID: 21844206 PMCID: PMC3171089 DOI: 10.1084/jem.20102170] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The Paf oncogene is highly expressed in cycling hematopoietic stem cells (HSCs) and is required for the development of long-term HSCs. Hematopoietic stem cells (HSCs) self-renew to maintain the lifelong production of all blood populations. Here, we show that the proliferating cell nuclear antigen–associated factor (Paf) is highly expressed in cycling bone marrow HSCs and plays a critical role in hematopoiesis. Mice lacking Paf exhibited reduced bone marrow cellularity; reduced numbers of HSCs and committed progenitors; and leukopenia. These phenotypes are caused by a cell-intrinsic blockage in the development of long-term (LT)-HSCs into multipotent progenitors and preferential loss of lymphoid progenitors caused by markedly increased p53-mediated apoptosis. In addition, LT-HSCs from Paf−/− mice had increased levels of reactive oxygen species (ROS), failed to maintain quiescence, and were unable to support LT hematopoiesis. The loss of lymphoid progenitors was likely due the increased levels of ROS in LT-HSCs caused by treatment of Paf−/− mice with the anti-oxidant N-acetylcysteine restored lymphoid progenitor numbers to that of Paf+/+ mice. Collectively, our studies identify Paf as a novel and essential regulator of early hematopoiesis.
Collapse
Affiliation(s)
- Yacine M Amrani
- Immunology Program, Sloan-Kettering Institute, 3 Department of Pediatrics, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Haasken S, Auger JL, Binstadt BA. Absence of β2 integrins impairs regulatory T cells and exacerbates CD4+ T cell-dependent autoimmune carditis. THE JOURNAL OF IMMUNOLOGY 2011; 187:2702-10. [PMID: 21795599 DOI: 10.4049/jimmunol.1000967] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The immunopathogenic mechanisms mediating inflammation in multiorgan autoimmune diseases may vary between the different target tissues. We used the K/BxN TCR transgenic mouse model to investigate the contribution of CD4(+) T cells and β(2) integrins in the pathogenesis of autoimmune arthritis and endocarditis. Depletion of CD4(+) T cells following the onset of arthritis specifically prevented the development of cardiac valve inflammation. Genetic absence of β(2) integrins had no effect on the severity of arthritis and unexpectedly increased the extent of cardiovascular pathology. The exaggerated cardiac phenotype of the β(2) integrin-deficient K/BxN mice was accompanied by immune hyperactivation and was linked to a defect in regulatory T cells. These findings are consistent with a model in which the development of arthritis in K/BxN mice relies primarily on autoantibodies, whereas endocarditis depends on an additional contribution of effector T cells. Furthermore, strategies targeting β(2) integrins for the treatment of systemic autoimmune conditions need to consider not only the role of these molecules in leukocyte recruitment to sites of inflammation, but also their impact on the regulation of immunological tolerance.
Collapse
Affiliation(s)
- Stefanie Haasken
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA
| | | | | |
Collapse
|
41
|
Patel DA, Puig-Canto A, Challa DK, Perez Montoyo H, Ober RJ, Ward ES. Neonatal Fc receptor blockade by Fc engineering ameliorates arthritis in a murine model. THE JOURNAL OF IMMUNOLOGY 2011; 187:1015-22. [PMID: 21690327 DOI: 10.4049/jimmunol.1003780] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multiple autoimmune diseases are characterized by the involvement of autoreactive Abs in pathogenesis. Problems associated with existing therapeutics such as the delivery of intravenous immunoglobulin have led to interest in developing alternative approaches using recombinant or synthetic methods. Toward this aim, in the current study, we demonstrate that the use of Fc-engineered Abs (Abs that enhance IgG degradation [Abdegs]) to block neonatal FcR (FcRn) through high-affinity, Fc region binding is an effective strategy for the treatment of Ab-mediated disease. Specifically, Abdegs can be used at low, single doses to treat disease in the K/B×N serum transfer model of arthritis using BALB/c mice as recipients. Similar therapeutic effects are induced by 25- to 50-fold higher doses of i.v. Ig. Importantly, we show that FcRn blockade is a primary contributing factor toward the observed reduction in disease severity. The levels of albumin, which is also recycled by FcRn, are not affected by Abdeg delivery. Consequently, Abdegs do not alter FcRn expression levels or subcellular trafficking behavior. The engineering of Ab Fc regions to generate potent FcRn blockers therefore holds promise for the therapy of Ab-mediated autoimmunity.
Collapse
Affiliation(s)
- Dipesh A Patel
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9093, USA
| | | | | | | | | | | |
Collapse
|
42
|
Neonatal Fc receptor for IgG (FcRn) regulates cross-presentation of IgG immune complexes by CD8-CD11b+ dendritic cells. Proc Natl Acad Sci U S A 2011; 108:9927-32. [PMID: 21628593 DOI: 10.1073/pnas.1019037108] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cross-presentation of IgG-containing immune complexes (ICs) is an important means by which dendritic cells (DCs) activate CD8(+) T cells, yet it proceeds by an incompletely understood mechanism. We show that monocyte-derived CD8(-)CD11b(+) DCs require the neonatal Fc receptor for IgG (FcRn) to conduct cross-presentation of IgG ICs. Consequently, in the absence of FcRn, Fcγ receptor (FcγR)-mediated antigen uptake fails to initiate cross-presentation. FcRn is shown to regulate the intracellular sorting of IgG ICs to the proper destination for such cross-presentation to occur. We demonstrate that FcRn traps antigen and protects it from degradation within an acidic loading compartment in association with the rapid recruitment of key components of the phagosome-to-cytosol cross-presentation machinery. This unique mechanism thus enables cross-presentation to evolve from an atypically acidic loading compartment. FcRn-driven cross-presentation is further shown to control cross-priming of CD8(+) T-cell responses in vivo such that during chronic inflammation, FcRn deficiency results in inadequate induction of CD8(+) T cells. These studies thus demonstrate that cross-presentation in CD8(-)CD11b(+) DCs requires a two-step mechanism that involves FcγR-mediated internalization and FcRn-directed intracellular sorting of IgG ICs. Given the centrality of FcRn in controlling cross-presentation, these studies lay the foundation for a unique means to therapeutically manipulate CD8(+) T-cell responses.
Collapse
|
43
|
Freiberger T, Grodecká L, Ravčuková B, Kuřecová B, Postránecká V, Vlček J, Jarkovský J, Thon V, Litzman J. Association of FcRn expression with lung abnormalities and IVIG catabolism in patients with common variable immunodeficiency. Clin Immunol 2010; 136:419-25. [DOI: 10.1016/j.clim.2010.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 05/07/2010] [Accepted: 05/12/2010] [Indexed: 01/02/2023]
|
44
|
High-dose intravenous immunoglobulin (IVIG) therapy in autoimmune skin blistering diseases. Clin Rev Allergy Immunol 2010; 38:186-95. [PMID: 19557317 DOI: 10.1007/s12016-009-8153-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Treatment of autoimmune bullous skin diseases can often be challenging and primarily consists of systemic corticosteroids and a variety of immunosuppressants. Current treatment strategies are effective in most cases but hampered by the side effects of long-term immunosuppressive treatment. Intravenous immunoglobulin (IVIG) is one potential promising therapy for patients with autoimmune bullous skin diseases, and evidence of its effectiveness and safety is increasing. A number of autoimmune bullous skin diseases have been identified in which IVIG treatment may be beneficial. However, experience with IVIG in patients with autoimmune skin blistering disease is limited, where it is recommended for patients not responding to conventional therapy. The mode of action of IVIG in autoimmune diseases, including bullous diseases is far from being completely understood. We here summarize the clinical evidence supporting the notion, that IVIG is a promising therapeutic agent for the treatment of patients with autoimmune bullous skin disease. In addition, we review the proposed modes of action. In the future, randomized controlled trials are necessary to better determine the efficacy and adverse effects of IVIG in the treatment of autoimmune bullous skin diseases. In addition, insights into IVIG's mode of action might enable us to develop novel therapeutics to overcome the current shortage of IVIG.
Collapse
|
45
|
Abès R, Teillaud JL. Impact of Glycosylation on Effector Functions of Therapeutic IgG. Pharmaceuticals (Basel) 2010; 3:146-157. [PMID: 27713246 PMCID: PMC3991024 DOI: 10.3390/ph3010146] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 12/30/2009] [Accepted: 01/08/2010] [Indexed: 01/14/2023] Open
Abstract
Human IgG has only one conserved glycosylation site located in the Cγ2 domain of the Fc region that accounts for the presence of two sugar moieties per IgG. These IgG sugar cores play a critical role in a number of IgG effector functions. In the present review, we describe the main characteristics of IgG Fc glycosylation and some abnormalities of serum IgG glycosylation. We also discuss how glycosylation impacts on monoclonal antibodies (mAbs) and IVIg effector functions and how these molecules can be engineered. Several therapeutic antibodies have now been engineered to be no- or low-fucose antibodies and are currently tested in clinical trials. They exhibit an increased binding to activating FcγRIIIA and trigger a strong antibody-dependent cell cytotoxicity (ADCC) as compared to their highly-fucosylated counterparts. They represent a new generation of therapeutic antibodies that are likely to show a better clinical efficacy in patients, notably in cancer patients where cytotoxic antibodies are needed.
Collapse
Affiliation(s)
- Riad Abès
- INSERM UMRS 872, Paris, F-75006 France.
- Cordeliers Research Center, Université Pierre & Marie Curie, UMRS 872, Paris, F-75006, France.
- Université Paris-Descartes, UMRS 872, Paris, F-75006 France.
- Laboratoire français du Fractionnement et des Biotechnologies (LFB), Les Ulis, France.
| | - Jean-Luc Teillaud
- INSERM UMRS 872, Paris, F-75006 France.
- Cordeliers Research Center, Université Pierre & Marie Curie, UMRS 872, Paris, F-75006, France.
- Université Paris-Descartes, UMRS 872, Paris, F-75006 France.
| |
Collapse
|
46
|
Aschermann S, Lux A, Baerenwaldt A, Biburger M, Nimmerjahn F. The other side of immunoglobulin G: suppressor of inflammation. Clin Exp Immunol 2009; 160:161-7. [PMID: 20041883 DOI: 10.1111/j.1365-2249.2009.04081.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Immunoglobulin G (IgG) molecules can have two completely opposite functions. On one hand, they induce proinflammatory responses and recruit innate immune effector cells during infection with pathogenic microorganisms or autoimmune disease. On the other hand, intravenous infusion of high doses of pooled IgG molecules from thousands of donors [intravenous IG (IVIG) therapy] represents an efficient anti-inflammatory treatment for many autoimmune diseases. Whereas our understanding of the mechanism of the proinflammatory activity of IgG is quite advanced, we are only at the very beginning to comprehend how the anti-inflammatory activity comes about and what cellular and molecular players are involved in this activity. This review will summarize our current knowledge and focus upon the two major models of either IVIG-mediated competition for IgG-triggered effector functions or IVIG-mediated adjustment of cellular activation thresholds used to explain the mechanism of the anti-inflammatory activity.
Collapse
Affiliation(s)
- S Aschermann
- Laboratory of Experimental Immunology and Immunotherapy, Nikolaus-Fiebiger-Centre for Molecular Medicine, Medical Department III, University of Erlangen-Nuernberg, Erlangen, Germany
| | | | | | | | | |
Collapse
|
47
|
Masuda A, Yoshida M, Shiomi H, Morita Y, Kutsumi H, Inokuchi H, Mizuno S, Nakamura A, Takai T, Blumberg RS, Azuma T. Role of Fc Receptors as a therapeutic target. ACTA ACUST UNITED AC 2009; 8:80-6. [PMID: 19275696 DOI: 10.2174/187152809787582525] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It has been forty years since the discovery of Fc Receptors and their function. Fc Receptors include the IgG receptors (FcgammaR), high-affinity IgE receptor (FcepsilonRI), IgA and IgA/IgM receptors, and neonatal Fc receptor for IgG (FcRn). In particular, the FcgammaRs have been well known to play an important role in many biologic processes including those associated with the response to infection and cancer as well as in the pathogenesis of immune-mediated diseases. Both positive and negative regulatory function has ascribed to Fc receptors and FcgammaRs in particular which serve to establish a threshold for immune cell activation. In other cases, Fc receptors such as FcRn possess a novel structure and function by playing a major role in the transport of IgG across polarized epithelial barriers at mucosal surfaces and in the regulation of IgG half-life. These diverse functions highlight the potential effectiveness of targeting Fc receptors for therapeutic purposes. This review summarizes new information available in the therapeutic applications of this biology.
Collapse
Affiliation(s)
- Atsuhiro Masuda
- Department of Gastroenterology, Kobe University School of Medicine, Kobe, Hyogo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kobayashi K, Qiao SW, Yoshida M, Baker K, Lencer WI, Blumberg RS. An FcRn-dependent role for anti-flagellin immunoglobulin G in pathogenesis of colitis in mice. Gastroenterology 2009; 137:1746-56.e1. [PMID: 19664634 PMCID: PMC2787451 DOI: 10.1053/j.gastro.2009.07.059] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 06/25/2009] [Accepted: 07/22/2009] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS The neonatal Fc receptor for immunoglobulin (Ig)G (FcRn) protects monomeric IgG from catabolism in parenchymal and hematopoietic cells during adult life. In dendritic cells, FcRn also promotes presentation of antigens in association with IgG. Because IgGs with anti-bacterial specificity are a hallmark of inflammatory bowel disease, we sought to determine their significance and relationship to FcRn expression in antigen-presenting cells, focusing on IgGs specific for flagellin. METHODS Levels of circulating anti-flagellin IgG were induced in wild-type and FcRn(-/-) mice, followed by induction of colitis with dextran sodium sulfate (DSS). Bone marrow chimera models were used to localize the site of FcRn action. RESULTS Wild-type mice that received anti-flagellin IgG exhibited more severe colitis following administration of DSS, compared with mice that received control IgG. Wild-type mice immunized with flagellin exhibited significantly more severe colitis in response to DSS administration than that observed in similarly treated FcRn(-/-) mice. In chimera studies, FcRn(-/-) mice given wild-type bone marrow and immunized with flagellin exhibited significantly more colitis than wild-type mice given FcRn(-/-) bone marrow and immunized with flagellin. Serum anti-flagellin IgG levels were similar in both sets of chimeric mice, consistent with the equal participation of hematopoietic and nonhematopoeitic cells in FcRn-mediated IgG protection. CONCLUSIONS Anti-bacterial IgG antibodies are involved in the pathogenesis of colitis; this pathway requires FcRn in antigen presenting cells, the major subset of hematopoietic cells that express FcRn.
Collapse
Affiliation(s)
- Kanna Kobayashi
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | | | - Masaru Yoshida
- Gastrointestinal Center for Medical Research, Kobe University School of Medicine, Kobe, 650-0017, Japan
| | - Kristi Baker
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Wayne I. Lencer
- Gastroenterology Division, Children’s Hospital Boston, Harvard Medical School, Boston MA 02115
| | - Richard S. Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
49
|
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: 6.3] [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
|
50
|
Low SC, Mezo AR. Inhibitors of the FcRn:IgG protein-protein interaction. AAPS JOURNAL 2009; 11:432-4. [PMID: 19499344 DOI: 10.1208/s12248-009-9120-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 05/20/2009] [Indexed: 11/30/2022]
Abstract
The neonatal Fc receptor, FcRn, is responsible for controlling the half-life of IgG antibodies. As a result, inhibitors of FcRn have been investigated as a possible way to modulate IgG half-lives. Such inhibitors could have possible applications in reducing autoantibody levels in autoimmune disease states. To date, monoclonal antibodies, engineered Fc domains, and short peptides have been reported to inhibit FcRn function and modulate IgG half-lives in vivo.
Collapse
Affiliation(s)
- Susan C Low
- Syntonix Pharmaceuticals Inc, A subsidiary of Biogen Idec, Waltham, MA 02451, USA
| | | |
Collapse
|