125 results on '"Lazarus AH"'
Search Results
2. Induction of a secondary human anti-HLA alloimmune response in severe combined immunodeficient mice engrafted with human lymphocytes
- Author
-
Lazarus, AH, primary, Crow, AR, additional, Semple, JW, additional, Cosgrave, D, additional, Kalovsky, EJ, additional, Hannach, B, additional, Blanchette, V, additional, and Freedman, J, additional
- Published
- 1997
- Full Text
- View/download PDF
3. Mechanisms of anti-D action in the prevention of hemolytic disease of the fetus and newborn: what can we learn from rodent models?
- Author
-
Brinc D, Denomme GA, and Lazarus AH
- Published
- 2009
- Full Text
- View/download PDF
4. Analysis of transmembrane signalling and T cell defects associated with idiopathic thrombocytopenic purpura (ITP).
- Author
-
Lazarus, AH, Joy, T, and Crow, AR
- Subjects
- *
THROMBOCYTOPENIA , *IMMUNOTHERAPY , *AUTOIMMUNITY , *PATHOLOGICAL physiology , *TECHNOLOGICAL innovations , *DIAGNOSIS - Abstract
Adult chronic idiopathic thrombocytopenic purpura (ITP) is an autoimmune disease characterized by production of autoreactive antibodies to platelet antigens. It is now becoming clear that autoantibody production, in general, is regulated by T helper (Th) cells. Several recent studies have examined potential defects in T cell function in this disease and have demonstrated that patients with ITP possess abnormal lymphocyte activation and Th[sub 1]/Th[sub 2]-mediated cytokine production. Although the underlying cause(s) of aberrant T cell function in this disease are not known, studies from other models of autoimmune disease indicate that defects in T cell transmembrane signalling can be causally linked to abnormal T cell activation and cytokine production. This review will present some of the major T cell signalling pathways and discuss how altered T cell signalling may be linked to autoimmunity with an emphasis on ITP. Recent preliminary findings of a potential defect in the signal transduction apparatus in lymphocytes from three patients with ITP will also be presented. [ABSTRACT FROM AUTHOR]
- Published
- 1998
- Full Text
- View/download PDF
5. Antagonism of the Platelet-Activating Factor Pathway Mitigates Inflammatory Adverse Events Driven by Anti-erythrocyte Antibody Therapy in Mice.
- Author
-
Won KD, Gil Gonzalez L, Cruz-Leal Y, Pavon Oro A, and Lazarus AH
- Subjects
- Animals, Mice, Chemokine CCL5 immunology, Chemokine CXCL9 immunology, Receptors, G-Protein-Coupled immunology, Signal Transduction immunology, Mice, Inbred C57BL, Autoantibodies immunology, Disease Models, Animal, Platelet Activating Factor immunology, Purpura, Thrombocytopenic, Idiopathic immunology, Purpura, Thrombocytopenic, Idiopathic drug therapy, Erythrocytes immunology, Inflammation immunology, Platelet Membrane Glycoproteins antagonists & inhibitors, Platelet Membrane Glycoproteins immunology, Chemokine CCL2 immunology
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by low platelet counts primarily due to antiplatelet autoantibodies. Anti-D is a donor-derived polyclonal Ab against the rhesus D Ag on erythrocytes used to treat ITP. Unfortunately, adverse inflammatory/hypersensitivity reactions and a Food and Drug Administration-issued black box warning have limited its clinical use. This underscores the imperative to understand the inflammatory pathway associated with anti-erythrocyte Ab-based therapies. TER119 is an erythrocyte-specific Ab with anti-D-like therapeutic activity in murine ITP, while also exhibiting a distinct inflammatory signature involving production of CCL2, CCL5, and CXCL9 but not IFN-γ. Therefore, TER119 has been used to elucidate the potential mechanism underlying the adverse inflammatory activity associated with anti-erythrocyte Ab therapy in murine ITP. Prior work has demonstrated that TER119 administration is associated with a dramatic decrease in body temperature and inflammatory cytokine/chemokine production. The work presented in the current study demonstrates that inhibiting the highly inflammatory platelet-activating factor (PAF) pathway with PAF receptor antagonists prevents TER119-driven changes in body temperature and inhibits the production of the CCL2, CCL5, and CXCL9 inflammatory cytokines in CD-1 mice. Phagocytic cells and a functional TER119 Fc region were found to be necessary for TER119-induced body temperature changes and increases in CXCL9 and CCL2. Taken together, this work reveals the novel requirement of the PAF pathway in causing adverse inflammatory activity associated with anti-erythrocyte Ab therapy in a murine model and provides a strategy of mitigating these potential reactions without altering therapeutic activity., (Copyright © 2024 by The American Association of Immunologists, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
6. Human Fc gamma receptor IIIA blockade inhibits platelet destruction in a humanized murine model of ITP.
- Author
-
Gil Gonzalez L, Won KD, Tawhidi Z, Cummins E, Cruz-Leal Y, Tundidor Cabado Y, Sachs UJ, Norris PAA, Shan Y, Bhakta V, Li J, Samudio I, Silva-Moreno B, Cerna-Portillo L, Pavon Oro A, Bergqvist P, Chan P, Moorehead A, Sholzberg M, Sheffield WP, and Lazarus AH
- Subjects
- Humans, Mice, Animals, Receptors, IgG metabolism, Disease Models, Animal, Immunoglobulin G therapeutic use, Albumins therapeutic use, Purpura, Thrombocytopenic, Idiopathic drug therapy, Thrombocytopenia
- Abstract
Abstract: Fc gamma receptor (FcγR) IIIA is an important receptor for immunoglobulin G (IgG) and is involved in immune defense mechanisms as well as tissue destruction in some autoimmune diseases including immune thrombocytopenia (ITP). FcγRIIIA on macrophages can trigger phagocytosis of IgG-sensitized platelets, and prior pilot studies observed blockade of FcγRIIIA increased platelet counts in patients with ITP. Unfortunately, although blockade of FcγRIIIA in patients with ITP increased platelet counts, its engagement by the blocking antibody drove serious adverse inflammatory reactions. These adverse events were postulated to originate from the antibody's Fc and/or bivalent nature. The blockade of human FcγRIIIA in vivo with a monovalent construct lacking an active Fc region has not yet been achieved. To effectively block FcγRIIIA in vivo, we developed a high affinity monovalent single-chain variable fragment (scFv) that can bind and block human FcγRIIIA. This scFv (17C02) was expressed in 3 formats: a monovalent fusion protein with albumin, a 1-armed human IgG1 antibody, and a standard bivalent mouse (IgG2a) antibody. Both monovalent formats were effective in preventing phagocytosis of ITP serum-sensitized human platelets. In vivo studies using FcγR-humanized mice demonstrated that both monovalent therapeutics were also able to increase platelet counts. The monovalent albumin fusion protein did not have adverse event activity as assessed by changes in body temperature, whereas the 1-armed antibody induced some changes in body temperature even though the Fc region function was impaired by the Leu234Ala and Leu235Ala mutations. These data demonstrate that monovalent blockade of human FcγRIIIA in vivo can potentially be a therapeutic strategy for patients with ITP., (© 2024 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Trogocytosis drives red blood cell antigen loss in association with antibody-mediated immune suppression.
- Author
-
Cruz-Leal Y, Norris PAA, Gil Gonzalez L, Marjoram D, Wabnitz H, Shan Y, and Lazarus AH
- Subjects
- Pregnancy, Infant, Newborn, Female, Mice, Humans, Animals, Antibodies, Erythrocytes metabolism, Immunosuppression Therapy, Isoantibodies, Trogocytosis, Erythroblastosis, Fetal
- Abstract
Abstract: Red blood cell (RBC) alloimmunization to paternal antigens during pregnancy can cause hemolytic disease of the fetus and newborn (HDFN). This severe and potentially fatal neonatal disorder can be prevented by the administration of polyclonal anti-D through a mechanism referred to as antibody-mediated immune suppression (AMIS). Although anti-D prophylaxis effectively prevents HDFN, a lack of mechanistic clarity has hampered its replacement with recombinant agents. The major theories behind AMIS induction in the hematologic literature have classically centered around RBC clearance; however, antigen modulation/loss has recently been proposed as a potential mechanism of AMIS. To explore the primary mechanisms of AMIS, we studied the ability of 11 different antibodies to induce AMIS, RBC clearance, antigen loss, and RBC membrane loss in the HOD (hen egg lysozyme-ovalbumin-human Duffy) murine model. Antibodies targeting different portions of the HOD molecule could induce AMIS independent of their ability to clear RBCs; however, all antibodies capable of inducing a strong AMIS effect also caused significant in vivo loss of the HOD antigen in conjunction with RBC membrane loss. In vitro studies of AMIS-inducing antibodies demonstrated simultaneous RBC antigen and membrane loss, which was mediated by macrophages. Confocal live-cell microscopy revealed that AMIS-inducing antibodies triggered RBC membrane transfer to macrophages, consistent with trogocytosis. Furthermore, anti-D itself can induce trogocytosis even at low concentrations, when phagocytosis is minimal or absent. In view of these findings, we propose trogocytosis as a mechanism of AMIS induction., (© 2024 American Society of Hematology. Published by Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.)
- Published
- 2024
- Full Text
- View/download PDF
8. B cells and antibodies in refractory immune thrombocytopenia.
- Author
-
Roeser A, Lazarus AH, and Mahévas M
- Subjects
- Humans, Rituximab therapeutic use, Rituximab pharmacology, B-Lymphocytes, Plasma Cells pathology, Autoantibodies, Purpura, Thrombocytopenic, Idiopathic therapy, Purpura, Thrombocytopenic, Idiopathic pathology
- Abstract
Immune thrombocytopenia (ITP) is an acquired bleeding disorder mediated by pathogenic autoantibodies secreted by plasma cells (PCs) in many patients. In refractory ITP patients, the persistence of splenic and bone marrow autoreactive long-lived PCs (LLPCs) may explain primary failure of rituximab and splenectomy respectively. The reactivation of autoreactive memory B cells generating new autoreactive PCs contributes to relapses after initial response to rituximab. Emerging strategies targeting B cells and PCs aim to prevent the settlement of splenic LLPCs with the combination of anti-BAFF and rituximab, to deplete autoreactive PCs with anti-CD38 antibodies, and to induce deeper B-cell depletion in tissues with novel anti-CD20 monoclonal antibodies and anti-CD19 therapies. Alternative strategies, focused on controlling autoantibody mediated effects, have also been developed, including SYK and BTK inhibitors, complement inhibitors, FcRn blockers and inhibitors of platelet desialylation., (© 2023 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
9. Serum from half of patients with immune thrombocytopenia trigger macrophage phagocytosis of platelets.
- Author
-
Norris PAA, Tawhidi Z, Sachs UJ, Cserti-Gazdewich CM, Lin Y, Callum J, Gil Gonzalez L, Shan Y, Branch DR, and Lazarus AH
- Subjects
- Humans, Blood Platelets metabolism, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Immunoglobulin G, Phagocytosis, Macrophages metabolism, Autoantibodies, Purpura, Thrombocytopenic, Idiopathic diagnosis, Thrombocytopenia metabolism
- Abstract
Humoral antiplatelet factors, such as autoantibodies, are thought to primarily clear platelets by triggering macrophage phagocytosis in immune thrombocytopenia (ITP). However, there are few studies characterizing the capacity and mechanisms of humoral factor-triggered macrophage phagocytosis of platelets using specimens from patients with ITP. Here, we assessed sera from a cohort of 24 patients with ITP for the capacity to trigger macrophage phagocytosis of normal donor platelets and characterized the contribution of humoral factors to phagocytosis. Sera that produced a phagocytosis magnitude greater than a normal human serum mean + 2 standard deviations were considered phagocytosis-positive. Overall, 42% (8/19) of MHC I alloantibody-negative ITP sera were phagocytosis-positive. The indirect monoclonal antibody immobilization of platelet antigens assay was used to detect immunoglobulin G (IgG) autoantibodies to glycoproteins (GP)IIb/IIIa, GPIb/IX, and GPIa/IIa. Autoantibody-positive sera triggered a higher mean magnitude of phagocytosis than autoantibody-negative sera. Phagocytosis correlated inversely with platelet counts among autoantibody-positive patients but not among autoantibody-negative patients. Select phagocytosis-positive sera were separated into IgG-purified and -depleted fractions via protein G and reassessed for phagocytosis. Phagocytosis was largely retained in the purified IgG fractions. In addition, we assessed serum concentrations of C-reactive protein, serum amyloid P, and pentraxin 3 as potential phagocytosis modulators. Pentraxin 3 concentrations correlated inversely with platelet counts among patients positive for autoantibodies. Taken together, sera from approximately half of the patients with ITP studied triggered macrophage phagocytosis of platelets beyond a normal level. An important role for antiplatelet autoantibodies in phagocytosis is supported; a role for pentraxins such as pentraxin 3 may be suggested., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. TEMRA: the CD8 subset in chronic ITP?
- Author
-
Lazarus AH and Semple JW
- Subjects
- Humans, CD8-Positive T-Lymphocytes immunology, Clone Cells, Purpura, Thrombocytopenic, Idiopathic immunology, Thrombocytopenia
- Published
- 2023
- Full Text
- View/download PDF
11. Antigen copy number and antibody dose can determine the outcome of erythrocyte alloimmunization inducing either antibody-mediated immune suppression or enhancement in a murine model.
- Author
-
Wabnitz H, Cruz-Leal Y, and Lazarus AH
- Subjects
- Animals, Mice, Disease Models, Animal, Erythrocytes metabolism, Immunoglobulin G, Immunoglobulin M, DNA Copy Number Variations
- Abstract
Background: The administration of anti-D for the prevention of hemolytic disease of the fetus and newborn is one of the most successful clinical uses of the phenomenon of antibody-mediated immune suppression (AMIS). However, despite adequate prophylaxis, failures can still occur in the clinic and are poorly understood. Recently, the copy number of red blood cell (RBC) antigens has been shown to influence immunogenicity in the context of RBC alloimmunization; however, its influence on AMIS remains unexplored., Study Design and Methods: RBCs expressing approximately 3,600 and approximately 12,400 copy numbers of surface-bound hen egg lysozyme (HEL), named respectively HEL
med -RBCs and HELhi -RBCs, and selected doses of a polyclonal HEL-specific IgG were transfused into mice. Recipient HEL-specific IgM, IgG, and IgG subclass responses were evaluated by ELISA., Results: Antigen copy number affected the antibody dose required for AMIS induction with higher antigen copy numbers requiring larger doses of antibody. For instance, 5 μg of antibody caused AMIS for HELmed -RBCs but not HELhi -RBCs, while 20 μg induced significant suppression for both HEL-RBCs. Overall, increasing amounts of the AMIS-inducing antibody were associated with a more complete AMIS effect. In contrast, the lowest tested doses of the AMIS-inducing IgG led to evidence of enhancement at the IgM and IgG levels., Discussion: The results demonstrate that the relationship between antigen copy number and antibody dose can influence the outcome of AMIS. Further, this work suggests that the same antibody preparation can induce both AMIS and enhancement but that the outcome may depend on the quantitative interrelationship of antigen-antibody binding., (© 2023 The Authors. Transfusion published by Wiley Periodicals LLC on behalf of AABB.)- Published
- 2023
- Full Text
- View/download PDF
12. Antigen-specific IgG subclass composition in recipient mice can indicate the degree of red blood cell alloimmunization as well as discern between primary and secondary immunization.
- Author
-
Wabnitz H, Cruz-Leal Y, and Lazarus AH
- Subjects
- Mice, Animals, Immunization, Secondary, Antigens, Immunoglobulin G, Isoantibodies, Erythrocyte Transfusion, Erythrocytes
- Abstract
Background: Despite the vast antigen disparity between donor and recipient red blood cells (RBCs), only 2%-6% of transfusion patients mount an alloantibody response. Recently, RBC antigen density has been proposed as one of the factors that can influence alloimmunization, however, there has been no characterization of the role of antigen density along with RBC dose in primary and secondary immunization., Study Design and Methods: To generate RBCs that express distinct antigen copy numbers, different quantities of hen egg lysozyme (HEL) were coupled to murine RBCs. The HEL-RBCs were subsequently transfused into recipient mice at different RBC doses and their HEL-specific IgM, IgG, and IgG subclass response was evaluated., Results: Productive immune responses could be generated through a high copy number antigen transfused at low RBC doses or a low copy number transfused at high RBC doses. Further, primary but submaximal humoral immunization predominantly induced the IgG2b and IgG3 subclasses. In contrast, a maximal primary immunization or a secondary immunization induced all four IgG subclasses., Discussion: Our results confirm the existence of an antigen threshold for productive immune responses but indicate that a high antigen copy number alone might not be enough to induce a response, but rather a combination of both antigen copy number and cell dosage may determine the outcome of immunization. Further, this study provides a proof of concept that the IgG subclass composition can be an indicator of the level of RBC alloimmunization as well as discern between primary and secondary immunization at least in this murine model., (© 2023 AABB.)
- Published
- 2023
- Full Text
- View/download PDF
13. THP-1 cells transduced with CD16A utilize Fcγ receptor I and III in the phagocytosis of IgG-sensitized human erythrocytes and platelets.
- Author
-
Gil Gonzalez L, Fernandez-Marrero Y, Norris PAA, Tawhidi Z, Shan Y, Cruz-Leal Y, Won KD, Frias-Boligan K, Branch DR, and Lazarus AH
- Subjects
- Humans, THP-1 Cells, Phagocytosis, Monocytes metabolism, Erythrocytes metabolism, Receptors, IgG metabolism, Immunoglobulin G
- Abstract
Fc gamma receptors (FcγRs) are critical effector receptors for immunoglobulin G (IgG) antibodies. On macrophages, FcγRs mediate multiple effector functions, including phagocytosis, but the individual contribution of specific FcγRs to phagocytosis has not been fully characterized. Primary human macrophage populations, such as splenic macrophages, can express FcγRI, FcγRIIA, and FcγRIIIA. However, there is currently no widely available monocyte or macrophage cell line expressing all these receptors. Common sources of monocytes for differentiation into macrophages, such as human peripheral blood monocytes and the monocytic leukemia cell line THP-1, generally lack the expression of FcγRIIIA (CD16A). Here, we utilized a lentiviral system to generate THP-1 cells stably expressing human FcγRIIIA (CD16F158). THP-1-CD16A cells treated with phorbol 12-myristate 13-acetate for 24 hours phagocytosed anti-D-opsonized human red blood cells primarily utilizing FcγRI with a lesser but significant contribution of IIIA while phagocytosis of antibody-opsonized human platelets equally utilized FcγRI and Fcγ IIIA. Despite the well-known ability of FcγRIIA to bind IgG in cell free systems, this receptor did not appear to be involved in either RBC or platelet phagocytosis. These transgenic cells may constitute a valuable tool for studying macrophage FcγR utilization and function., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2022 Gil Gonzalez et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2022
- Full Text
- View/download PDF
14. Anti-inflammatory activity of CD44 antibodies in murine immune thrombocytopenia is mediated by Fcγ receptor inhibition.
- Author
-
Norris PAA, Kaur G, Khan R, Zhu G, Ni H, and Lazarus AH
- Subjects
- Animals, Blood Platelets immunology, Female, Humans, Inflammation immunology, Mice, Mice, Inbred C57BL, Phagocytosis, RAW 264.7 Cells, Hyaluronan Receptors immunology, Immunoglobulin G immunology, Purpura, Thrombocytopenic, Idiopathic immunology, Receptors, IgG immunology
- Abstract
Monoclonal immunoglobulin G (IgG) antibodies to CD44 (anti-CD44) are anti-inflammatory in numerous murine autoimmune models, but the mechanisms are poorly understood. Anti-CD44 anti-inflammatory activity shows complete therapeutic concordance with IV immunoglobulin (IVIg) in treating autoimmune disease models, making anti-CD44 a potential IVIg alternative. In murine immune thrombocytopenia (ITP), there is no mechanistic explanation for anti-CD44 activity, although anti-CD44 ameliorates disease similarly to IVIg. Here, we demonstrate a novel anti-inflammatory mechanism of anti-CD44 that explains disease amelioration by anti-CD44 in murine ITP. Macrophages treated with anti-CD44 in vitro had dramatically suppressed phagocytosis through FcγRs in 2 separate systems of IgG-opsonized platelets and erythrocytes. Phagocytosis inhibition by anti-CD44 was mediated by blockade of the FcγR IgG binding site without changing surface FcγR expression. Anti-CD44 of different subclasses revealed that FcγR blockade was specific to receptors that could be engaged by the respective anti-CD44 subclass, and Fc-deactivated anti-CD44 variants lost all FcγR-inhibiting activity. In vivo, anti-CD44 functioned analogously in the murine passive ITP model and protected mice from ITP when thrombocytopenia was induced through an FcγR that could be engaged by the CD44 antibody's subclass. Consistent with FcγR blockade, Fc-deactivated variants of anti-CD44 were completely unable to ameliorate ITP. Together, anti-CD44 inhibits macrophage FcγR function and ameliorates ITP consistent with an FcγR blockade mechanism. Anti-CD44 is a potential IVIg alternative and may be of particular benefit in ITP because of the significant role that FcγRs play in human ITP pathophysiology., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
15. Could antigen loss be a potential mechanism to explain antibody-mediated immune suppression?
- Author
-
Cruz-Leal Y and Lazarus AH
- Subjects
- Antigen-Antibody Reactions immunology, Antigen-Antibody Reactions physiology, Erythrocytes enzymology, Erythrocytes immunology, Fetus immunology, Humans, Immunosuppression Therapy adverse effects, Infant, Newborn, Phagocytosis immunology, Antibody Formation immunology, Antigens immunology, Erythroblastosis, Fetal immunology, Immune Tolerance immunology
- Published
- 2021
- Full Text
- View/download PDF
16. FcγRI and FcγRIII on splenic macrophages mediate phagocytosis of anti-glycoprotein IIb/IIIa autoantibody-opsonized platelets in immune thrombocytopenia.
- Author
-
Norris PAA, Segel GB, Burack WR, Sachs UJ, Lissenberg-Thunnissen SN, Vidarsson G, Bayat B, Cserti-Gazdewich CM, Callum J, Lin Y, Branch D, Kapur R, Semple JW, and Lazarus AH
- Subjects
- Blood Platelets, Humans, Macrophages, Phagocytosis, Platelet Glycoprotein GPIIb-IIIa Complex, Platelet Membrane Glycoproteins, Purpura, Thrombocytopenic, Idiopathic
- Published
- 2021
- Full Text
- View/download PDF
17. New insights into IVIg mechanisms and alternatives in autoimmune and inflammatory diseases.
- Author
-
Norris PAA, Kaur G, and Lazarus AH
- Subjects
- Animals, Autoimmune Diseases immunology, Complement Activation drug effects, Histocompatibility Antigens Class I immunology, Humans, Inflammation immunology, Receptors, Fc antagonists & inhibitors, Receptors, Fc immunology, T-Lymphocytes drug effects, T-Lymphocytes immunology, Autoimmune Diseases therapy, Immunoglobulins, Intravenous pharmacology, Immunoglobulins, Intravenous therapeutic use, Inflammation therapy
- Abstract
Purpose of Review: Intravenous immunoglobulin (IVIg) is an effective treatment for an increasing number of autoimmune and inflammatory conditions. However, IVIg continues to be limited by problems of potential shortages and cost. A number of mechanisms have been described for IVIg, which have been captured in newly emergent IVIg mimetic and IVIg alternative therapies. This review discusses the recent developments in IVIg mimetics and alternatives., Recent Findings: Newly emergent IVIg mimetics and alternatives capture major proposed mechanisms of IVIg, including FcγR blockade, FcRn inhibition, complement inhibition, immune complex mimetics and sialylated IgG. Many of these emergent therapies have promising preclinical and clinical trial results., Summary: Significant research has been undertaken into the mechanism of IVIg in the treatment of autoimmune and inflammatory disease. Understanding the major IVIg mechanisms has allowed for rational development of IVIg mimetics and alternatives for several IVIg-treatable diseases.
- Published
- 2020
- Full Text
- View/download PDF
18. Inhibition of platelet phagocytosis as an in vitro predictor for therapeutic potential of RBC antibodies in murine ITP.
- Author
-
Khan R, Menard M, Jen CC, Chen X, Norris PAA, and Lazarus AH
- Subjects
- Anemia etiology, Animals, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal immunology, Antibodies, Monoclonal therapeutic use, Female, Immunoglobulin G adverse effects, Immunoglobulin G immunology, In Vitro Techniques, Mice, Mice, Inbred C57BL, RAW 264.7 Cells, Rats, Recombinant Proteins adverse effects, Recombinant Proteins immunology, Recombinant Proteins therapeutic use, Blood Platelets, Erythrocytes immunology, Immunoglobulin G therapeutic use, Phagocytosis immunology, Purpura, Thrombocytopenic, Idiopathic blood, Purpura, Thrombocytopenic, Idiopathic therapy
- Abstract
Polyclonal anti-D is a first-line therapy for immune thrombocytopenia (ITP). Monoclonal antibodies are desirable alternatives, but none have yet proven successful despite their ability to opsonize erythrocytes (or red blood cells, RBCs) and cause anemia. Here, we examined 12 murine erythrocyte-specific antibodies of different specificity and subtypes and found that 8 of these antibodies could induce anemia in antigen-positive mice. Of these 8 antibodies, only 5 ameliorated ITP. All antibodies were examined for their in vitro ability to support macrophage-mediated phagocytosis of erythrocytes. Antibodies which supported erythrocyte phagocytosis in vitro successfully ameliorated ITP in vivo. To examine the ability of each antibody to inhibit phagocytosis of platelets, the antibodies were used to sensitize erythrocytes in vitro and these were added to a platelet phagocytosis assay. Antibodies that inhibited platelet phagocytosis in vitro also all ameliorated ITP in vivo. We conclude that inducing anemia is not a sufficient condition for amelioration of ITP but that the antibody's ability to prevent platelet phagocytosis in vitro predicted its ability to ameliorate ITP. We suggest that inhibition of in vitro platelet phagocytosis may prove to be a valuable tool for determining which erythrocyte antibodies would likely be candidates for clinical use in ITP., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
19. The use of IVIg in fetal and neonatal alloimmune thrombocytopenia- Principles and mechanisms.
- Author
-
Wabnitz H, Khan R, and Lazarus AH
- Subjects
- Female, Fetus, Humans, Infant, Newborn, Pregnancy, Immunoglobulins, Intravenous therapeutic use, Thrombocytopenia, Neonatal Alloimmune immunology
- Abstract
Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a rare neonatal disorder that is caused by alloimmunization against platelet antigens during pregnancy. Although rare, affecting only 1 in 1000 live births, it can cause intracranial hemorrhage and other bleeding complications that can lead to miscarriage, stillbirth and life-long neurological complications. One of the gold-standard therapies for at risk pregnancies is the administration of IVIg. Although IVIg has been used in a variety of different disorders for over 40 years, its exact mechanism of action is still unknown. In FNAIT, the majority of its therapeutic effect is thought the be mediated through the neonatal Fc receptor, however other mechanisms cannot be excluded. Due to safety, supply and other concerns that are associated with IVIg use, alternative therapies that could replace IVIg are additionally being investigated. This includes the possibility of a prophylaxis regimen for FNAIT, similarly to what has been successfully used in hemolytic disease of the fetus and newborn for over 50 years., Competing Interests: Declaration of Competing Interest None., (Copyright © 2019. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
20. Treating murine inflammatory diseases with an anti-erythrocyte antibody.
- Author
-
Crow AR, Kapur R, Koernig S, Campbell IK, Jen CC, Mott PJ, Marjoram D, Khan R, Kim M, Brasseit J, Cruz-Leal Y, Amash A, Kahlon S, Yougbare I, Ni H, Zuercher AW, Käsermann F, Semple JW, and Lazarus AH
- Subjects
- Acute Lung Injury blood, Acute Lung Injury complications, Anemia blood, Anemia complications, Animals, Arthritis blood, Arthritis complications, Arthritis, Experimental blood, Arthritis, Experimental complications, Arthritis, Experimental immunology, Blood Transfusion, Cell Movement, Chemokines metabolism, Disease Models, Animal, Disease Progression, Glycosylation, Immunoglobulin G metabolism, Inflammation blood, Inflammation complications, Inflammation Mediators metabolism, Mice, Inbred C57BL, Mice, SCID, Monocytes metabolism, Purpura, Thrombocytopenic, Idiopathic blood, Purpura, Thrombocytopenic, Idiopathic complications, Purpura, Thrombocytopenic, Idiopathic pathology, Receptors, IgG metabolism, Antibodies, Monoclonal therapeutic use, Erythrocytes immunology, Inflammation drug therapy
- Abstract
Treatment of autoimmune and inflammatory diseases typically involves immune suppression. In an opposite strategy, we show that administration of the highly inflammatory erythrocyte-specific antibody Ter119 into mice remodels the monocyte cellular landscape, leading to resolution of inflammatory disease. Ter119 with intact Fc function was unexpectedly therapeutic in the K/BxN serum transfer model of arthritis. Similarly, it rapidly reversed clinical disease progression in collagen antibody-induced arthritis (CAIA) and collagen-induced arthritis and completely corrected CAIA-induced increase in monocyte Fcγ receptor II/III expression. Ter119 dose-dependently induced plasma chemokines CCL2, CCL5, CXCL9, CXCL10, and CCL11 with corresponding alterations in monocyte percentages in the blood and liver within 24 hours. Ter119 attenuated chemokine production from the synovial fluid and prevented the accumulation of inflammatory cells and complement components in the synovium. Ter119 could also accelerate the resolution of hypothermia and pulmonary edema in an acute lung injury model. We conclude that this inflammatory anti-erythrocyte antibody simultaneously triggers a highly efficient anti-inflammatory effect with broad therapeutic potential., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2019
- Full Text
- View/download PDF
21. Glycoprotein V is a relevant immune target in patients with immune thrombocytopenia.
- Author
-
Vollenberg R, Jouni R, Norris PAA, Burg-Roderfeld M, Cooper N, Rummel MJ, Bein G, Marini I, Bayat B, Burack R, Lazarus AH, Bakchoul T, and Sachs UJ
- Subjects
- Animals, Blood Platelets immunology, Blood Platelets metabolism, Disease Models, Animal, Female, Humans, Male, Mice, Mice, Inbred NOD, Mice, SCID, Phagocytosis, Prevalence, Protein Binding immunology, Purpura, Thrombocytopenic, Idiopathic epidemiology, Autoantibodies immunology, Autoantigens immunology, Disease Susceptibility immunology, Platelet Membrane Glycoproteins immunology, Purpura, Thrombocytopenic, Idiopathic immunology
- Abstract
Platelet autoantibody-induced platelet clearance represents a major pathomechanism in immune thrombocytopenia (ITP). There is growing evidence for clinical differences between anti-glycoprotein IIb/IIIa and anti-glycoprotein Ib/IX mediated ITP. Glycoprotein V is a well characterized target antigen in Varicella-associated and drug-induced thrombocytopenia. We conducted a systematic study assessing the prevalence and functional capacity of autoantibodies against glycoprotein V. A total of 1140 patients were included. In one-third of patients, platelet-bound autoantibodies against glycoproteins Ib/IX, IIb/IIIa, or V were detected in a monoclonal antibody immobilization of platelet antigen assay; platelet-bound autoantiglycoprotein V was present in the majority of samples (222 out of 343, 64.7%). Investigation of patient sera revealed the presence of free autoantibodies against glycoprotein V in 13.5% of these patients by an indirect monoclonal antibody immobilization of platelet antigen assay, but in 39.6% by surface plasmon resonance technology. These antibodies showed significantly lower avidity (association/dissociation ratio 0.32±0.13 vs 0.73±0.14; P <0.001). High- and low-avidity antibodies induced comparable amounts of platelet uptake in a phagocytosis assay using CD14
+ positively-selected human macrophages [mean phagocytic index, 6.81 (range, 4.75-9.86) vs 6.01 (range, 5.00-6.98); P =0.954]. In a NOD/SCID mouse model, IgG prepared from both types of anti-glycoprotein V autoantibodies eliminated human platelets with no detectable difference between the groups from the murine circulation [mean platelet survival at 300 minutes, 40% (range, 27-55) vs 35% (16-46); P =0.025]. Our data establish glycoprotein V as a relevant immune target in immune thrombocytopenia. We would suggest that further studies including glycoprotein V will be required before ITP treatment can be tailored according to platelet autoantibody specificity., (Copyright© 2019 Ferrata Storti Foundation.)- Published
- 2019
- Full Text
- View/download PDF
22. GPIbα is required for platelet-mediated hepatic thrombopoietin generation.
- Author
-
Xu M, Li J, Neves MAD, Zhu G, Carrim N, Yu R, Gupta S, Marshall J, Rotstein O, Peng J, Hou M, Kunishima S, Ware J, Branch DR, Lazarus AH, Ruggeri ZM, Freedman J, and Ni H
- Subjects
- Animals, Bernard-Soulier Syndrome genetics, Cells, Cultured, Glycosylation, Hepatocytes metabolism, Homeostasis, Humans, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, N-Acetylneuraminic Acid metabolism, Platelet Transfusion, Protein Domains, Protein Processing, Post-Translational, Recombinant Proteins metabolism, Thrombopoietin blood, Bernard-Soulier Syndrome blood, Blood Platelets physiology, Liver metabolism, Platelet Glycoprotein GPIb-IX Complex physiology, Thrombopoietin biosynthesis
- Abstract
Thrombopoietin (TPO), a hematopoietic growth factor produced predominantly by the liver, is essential for thrombopoiesis. Prevailing theory posits that circulating TPO levels are maintained through its clearance by platelets and megakaryocytes via surface c-Mpl receptor internalization. Interestingly, we found a two- to threefold decrease in circulating TPO in GPIbα
-/- mice compared with wild-type (WT) controls, which was consistent in GPIbα-deficient human Bernard-Soulier syndrome (BSS) patients. We showed that lower TPO levels in GPIbα-deficient conditions were not due to increased TPO clearance by GPIbα-/- platelets but rather to decreased hepatic TPO mRNA transcription and production. We found that WT, but not GPIbα-/- , platelet transfusions rescued hepatic TPO mRNA and circulating TPO levels in GPIbα-/- mice. In vitro hepatocyte cocultures with platelets or GPIbα-coupled beads further confirm the disruption of platelet-mediated hepatic TPO generation in the absence of GPIbα. Treatment of GPIbα-/- platelets with neuraminidase caused significant desialylation; however, strikingly, desialylated GPIbα-/- platelets could not rescue impaired hepatic TPO production in vivo or in vitro, suggesting that GPIbα, independent of platelet desialylation, is a prerequisite for hepatic TPO generation. Additionally, impaired hepatic TPO production was recapitulated in interleukin-4/GPIbα-transgenic mice, as well as with antibodies targeting the extracellular portion of GPIbα, demonstrating that the N terminus of GPIbα is required for platelet-mediated hepatic TPO generation. These findings reveal a novel nonredundant regulatory role for platelets in hepatic TPO homeostasis, which improves our understanding of constitutive TPO regulation and has important implications in diseases related to GPIbα, such as BSS and auto- and alloimmune-mediated thrombocytopenias., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
23. Antiplatelet antibody-induced thrombocytopenia does not correlate with megakaryocyte abnormalities in murine immune thrombocytopenia.
- Author
-
Guo L, Kapur R, Aslam R, Hunt K, Hou Y, Zufferey A, Speck ER, Rondina MT, Lazarus AH, Ni H, and Semple JW
- Subjects
- Animals, Bone Marrow Cells pathology, Mice, Mice, Inbred BALB C, Autoantibodies immunology, Blood Platelets immunology, Megakaryocytes pathology, Purpura, Thrombocytopenic, Idiopathic immunology, Purpura, Thrombocytopenic, Idiopathic pathology
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by increased peripheral immune platelet destruction and megakaryocyte defects in the bone marrow. Although ITP was originally thought to be primarily due to antibody-mediated autoimmunity, it is now clear that T cells also play a significant role in the disease. However, the exact interplay between platelet destruction, megakaryocyte dysfunction and the elements of both humoral and cell-mediated immunity in ITP remains incompletely defined. While most studies have focused on immune platelet destruction in the spleen, an additional possibility is that the antiplatelet antibodies can also destroy bone marrow megakaryocytes. To address this, we negated the effects of T cells by utilizing an in vivo passive ITP model where BALB/c mice were administered various anti-αIIb, anti-β3 or anti-GPIb antibodies or antisera and platelet counts and bone marrow megakaryocytes were enumerated. Our results show that after 24 hours, all the different antiplatelet antibodies/sera induced variable degrees of thrombocytopenia in recipient mice. Compared with naïve control mice, however, histological examination of the bone marrow revealed that only 2 antibody preparations (mouse-anti-mouse β3 sera and an anti- αIIb monoclonal antibody (MWReg30) could affect bone marrow megakaryocyte counts. Our study shows that while most antiplatelet antibodies induce acute thrombocytopenia, the majority of them do not affect the number of megakaryocytes in the bone marrow. This suggests that other mechanisms may be responsible for megakaryocyte abnormalities seen during immune thrombocytopenia., (© 2018 The Foundation for the Scandinavian Journal of Immunology.)
- Published
- 2018
- Full Text
- View/download PDF
24. Red blood cell antibody-induced anemia causes differential degrees of tissue hypoxia in kidney and brain.
- Author
-
Mistry N, Mazer CD, Sled JG, Lazarus AH, Cahill LS, Solish M, Zhou YQ, Romanova N, Hare AGM, Doctor A, Fisher JA, Brunt KR, Simpson JA, and Hare GMT
- Subjects
- Acute Kidney Injury immunology, Acute Kidney Injury pathology, Acute Kidney Injury physiopathology, Anemia immunology, Anemia pathology, Anemia physiopathology, Animals, Brain metabolism, Brain pathology, Cerebrovascular Circulation, Disease Models, Animal, Erythrocytes immunology, Erythrocytes pathology, Erythropoietin genetics, Erythropoietin metabolism, Glycophorins blood, Glycophorins immunology, Hemolysis, Hypoxia, Brain immunology, Hypoxia, Brain pathology, Hypoxia, Brain physiopathology, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kidney metabolism, Kidney pathology, Male, Mice, Transgenic, Renal Circulation, Severity of Illness Index, Spleen metabolism, Spleen pathology, Up-Regulation, Acute Kidney Injury blood, Anemia blood, Antibodies, Monoclonal, Brain blood supply, Erythrocytes metabolism, Hypoxia, Brain blood, Kidney blood supply, Oxygen blood
- Abstract
Moderate anemia is associated with increased mortality and morbidity, including acute kidney injury (AKI), in surgical patients. A red blood cell (RBC)-specific antibody model was utilized to determine whether moderate subacute anemia could result in tissue hypoxia as a potential mechanism of injury. Cardiovascular and hypoxic cellular responses were measured in transgenic mice capable of expressing hypoxia-inducible factor-1α (HIF-1α)/luciferase activity in vivo. Antibody-mediated anemia was associated with mild intravascular hemolysis (6 h) and splenic RBC sequestration ( day 4), resulting in a nadir hemoglobin concentration of 89 ± 13 g/l on day 4. At this time point, renal tissue oxygen tension (P
t O2 ) was decreased in anemic mice relative to controls (13.1 ± 4.3 vs. 20.8 ± 3.7 mmHg, P < 0.001). Renal tissue hypoxia was associated with an increase in HIF/luciferase expression in vivo ( P = 0.04) and a 20-fold relative increase in renal erythropoietin mRNA transcription ( P < 0.001) but no increase in renal blood flow ( P = 0.67). By contrast, brain Pt O2 was maintained in anemic mice relative to controls (22.7 ± 5.2 vs. 23.4 ± 9.8 mmHg, P = 0.59) in part because of an increase in internal carotid artery blood flow (80%, P < 0.001) and preserved cerebrovascular reactivity. Despite these adaptive changes, an increase in brain HIF-dependent mRNA levels was observed (erythropoietin: P < 0.001; heme oxygenase-1: P = 0.01), providing evidence for subtle cerebral tissue hypoxia in anemic mice. These data demonstrate that moderate subacute anemia causes significant renal tissue hypoxia, whereas adaptive cerebrovascular responses limit the degree of cerebral tissue hypoxia. Further studies are required to assess whether hypoxia is a mechanism for acute kidney injury associated with anemia.- Published
- 2018
- Full Text
- View/download PDF
25. Erythrocyte Saturation with IgG Is Required for Inducing Antibody-Mediated Immune Suppression and Impacts Both Erythrocyte Clearance and Antigen-Modulation Mechanisms.
- Author
-
Cruz-Leal Y, Marjoram D, and Lazarus AH
- Subjects
- Animals, Humans, Mice, Mice, Transgenic, Muramidase immunology, Antigenic Modulation immunology, Erythrocytes immunology, Immunoglobulin G immunology, Immunosuppression Therapy methods
- Abstract
Anti-D prevents hemolytic disease of the fetus and newborn, and this mechanism has been referred to as Ab-mediated immune suppression (AMIS). Anti-D, as well as other polyclonal AMIS-inducing Abs, most often induce both epitope masking and erythrocyte clearance mechanisms. We have previously observed that some Abs that successfully induce AMIS effects could be split into those that mediate epitope masking versus those that induce erythrocyte clearance, allowing the ability to analyze these mechanisms separately. In addition, AMIS-inducing activity has recently been shown to induce Ag modulation (Ag loss from the erythrocyte surface). To assess these mechanisms, we immunized mice with transgenic murine RBCs expressing a single Ag protein comprising a recombinant Ag composed of hen egg lysozyme, OVA sequences comprising aa 251-349, and the human Duffy transmembrane protein (HOD-Ag) with serial doses of polyclonal anti-OVA IgG as the AMIS-inducing Ab. The anti-OVA Ab induced AMIS in the absence of apparent epitope masking. AMIS occurred only when the erythrocytes appeared saturated with IgG. This Ab was capable of inducing HOD-RBC clearance, as well as loss of the OVA epitope at doses of Ab that caused AMIS effects. HOD-RBCs also lost reactivity with Abs specific for the hen egg lysozyme and Duffy portions of the Ag consistent with the initiation of Ag modulation and/or trogocytosis mechanisms. These data support the concept that an AMIS-inducing Ab that does not cause epitope masking can induce AMIS effects in a manner consistent with RBC clearance and/or Ag modulation., (Copyright © 2018 by The American Association of Immunologists, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
26. Anti-RhD reduces levels of detectable RhD antigen following anti-RhD infusion.
- Author
-
Sullivan HC, Arthur CM, Thompson L, Patel SR, Stowell SR, Hendrickson JE, and Lazarus AH
- Subjects
- Humans, Infant, Male, Immunoglobulin G administration & dosage, Isoantibodies administration & dosage, Purpura, Thrombocytopenic, Idiopathic blood, Purpura, Thrombocytopenic, Idiopathic drug therapy, Rh-Hr Blood-Group System blood
- Published
- 2018
- Full Text
- View/download PDF
27. Immunoglobulin G Fc glycans are not essential for antibody-mediated immune suppression to murine erythrocytes.
- Author
-
Marjoram D, Cruz-Leal Y, Bernardo L, Le NPL, Crispin M, Yu X, Uchikawa M, and Lazarus AH
- Subjects
- Animals, Glycosylation, Mice, Polysaccharides immunology, Erythrocytes immunology, Immunoglobulin Fc Fragments immunology, Immunosuppression Therapy methods
- Published
- 2017
- Full Text
- View/download PDF
28. Prevention of hemolytic disease of the fetus and newborn: what have we learned from animal models?
- Author
-
Cruz-Leal Y, Marjoram D, and Lazarus AH
- Subjects
- Animals, Humans, Infant, Newborn, Mice, Disease Models, Animal, Erythroblastosis, Fetal metabolism, Erythroblastosis, Fetal pathology, Erythroblastosis, Fetal physiopathology, Erythroblastosis, Fetal prevention & control
- Abstract
Purpose of Review: This review aims to highlight recent advances in our understanding of how anti-red blood cell (RBC) antibodies prevent erythrocyte immunization with an emphasis on new murine models., Recent Findings: New murine models with clinically relevant human erythrocyte antigens have been used to understand the alloimmunization process and its inhibition. The search to elucidate the mechanism of action of IgG-mediated inhibition of erythrocyte alloimmunization has provided new evidence in support of a potential role for epitope masking, immune deviation and/or antigen modulation in this process. In addition, recent evidence suggests that blends of monoclonal antibodies targeting nonoverlapping epitopes on the RBC surface can improve the efficacy of monoclonal antibodies approaching that of polyclonal IgG., Summary: Animal models with defined alloantigens have helped to identify important mechanistic components that lead to alloimmunization and its inhibition by IgG. A better understanding of the underlying mechanisms leading to hemolytic disease of the fetus and newborn is required to develop the most effective prevention strategies for future patients.
- Published
- 2017
- Full Text
- View/download PDF
29. Antibody-mediated immune suppression is improved when blends of anti-RBC monoclonal antibodies are used in mice.
- Author
-
Bernardo L, Amash A, Marjoram D, and Lazarus AH
- Subjects
- Animals, Chickens, Erythrocytes immunology, Humans, Mice, Inbred C57BL, Muramidase immunology, Antibodies, Monoclonal immunology, Immunosuppression Therapy
- Abstract
Although the prevention of hemolytic disease of the fetus and newborn is highly effective using polyclonal anti-D, a recombinant alternative is long overdue. Unfortunately, anti-D monoclonal antibodies have been, at best, disappointing. To determine the primary attribute defining an optimal antibody, we assessed suppression of murine red blood cell (RBC) immunization by single-monoclonal antibodies vs defined blends of subtype-matched antibodies. Allogeneic RBCs expressing the HOD antigen (hen egg lysozyme [HEL]-ovalbumin-human transmembrane Duffy(b)) were transfused into naïve mice alone or together with selected combinations of HEL-specific antibodies, and the resulting suppressive effect was assessed by evaluating the antibody response. Polyclonal HEL antibodies dramatically inhibited the antibody response to the HOD antigen, whereas single-monoclonal HEL antibodies were less effective despite the use of saturating doses. A blend of monoclonal HEL-specific antibodies reactive with different HEL epitopes significantly increased the suppressive effect, whereas a blend of monoclonal antibodies that block each other's binding to the HEL protein did not increase suppression. In conclusion, these data show that polyclonal antibodies are superior to monoclonal antibodies at suppressing the immune response to the HOD cells, a feature that can be completely recapitulated using monoclonal antibodies to different epitopes., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
30. Targeting FcγRs to treat antibody-dependent autoimmunity.
- Author
-
Yu X and Lazarus AH
- Subjects
- Autoimmunity drug effects, Humans, Antibodies, Monoclonal immunology, Autoantibodies immunology, Autoimmunity immunology, Receptors, IgG chemistry
- Abstract
Self-reactive antibodies represent a significant force in autoimmune disease induction. In antibody-dependent autoimmune syndromes such as immune thrombocytopenia (ITP), systemic lupus erythematosus (SLE), myasthenia gravis and rheumatoid arthritis (RA), autoantibodies exert their inflammatory effect through FcγRs, a well-established class of cell surface receptors that interact with the Fc domain of IgG. Down-regulating FcγR functionality presents an attractive strategy to treat antibody-dependent autoimmune diseases. Various approaches, including nonspecific blocking of the IgG binding site as well as specific targeting using antagonistic monoclonal antibodies, have been explored to modulate the interaction between the Fc portion of IgG and FcγRs. The exquisite specificity and favorable pharmacokinetics of IgG make monoclonal antibodies a preferred choice. Indeed, the first antagonistic monoclonal antibody against the human FcγRIIIA had shown efficacy in refractory ITP patients; however, the practicality of using anti-FcγRIII antibody as a therapeutic was hindered by its associated adverse events, a phenomenon recapitulated in animal models. In this review, we discuss the role of FcγRs in autoimmune diseases, and focus on a novel monovalent approach to target FcγRs to resolve antibody-mediated autoimmunity., (Crown Copyright © 2016. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
31. CD44 Antibody Inhibition of Macrophage Phagocytosis Targets Fcγ Receptor- and Complement Receptor 3-Dependent Mechanisms.
- Author
-
Amash A, Wang L, Wang Y, Bhakta V, Fairn GD, Hou M, Peng J, Sheffield WP, and Lazarus AH
- Subjects
- Animals, Anti-Inflammatory Agents immunology, Anti-Inflammatory Agents pharmacology, Antibodies, Blocking immunology, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacology, Cell Line, Erythrocytes immunology, Immunoglobulin G immunology, Inflammation immunology, Inflammation prevention & control, Lectins, C-Type immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, Zymosan immunology, Antibodies, Blocking pharmacology, Hyaluronan Receptors immunology, Macrophages immunology, Phagocytosis immunology, Receptors, Complement immunology, Receptors, IgG immunology
- Abstract
Targeting CD44, a major leukocyte adhesion molecule, using specific Abs has been shown beneficial in several models of autoimmune and inflammatory diseases. The mechanisms contributing to the anti-inflammatory effects of CD44 Abs, however, remain poorly understood. Phagocytosis is a key component of immune system function and can play a pivotal role in autoimmune states where CD44 Abs have shown to be effective. In this study, we show that the well-known anti-inflammatory CD44 Ab IM7 can inhibit murine macrophage phagocytosis of RBCs. We assessed three selected macrophage phagocytic receptor systems: Fcγ receptors (FcγRs), complement receptor 3 (CR3), and dectin-1. Treatment of macrophages with IM7 resulted in significant inhibition of FcγR-mediated phagocytosis of IgG-opsonized RBCs. The inhibition of FcγR-mediated phagocytosis was at an early stage in the phagocytic process involving both inhibition of the binding of the target RBC to the macrophages and postbinding events. This CD44 Ab also inhibited CR3-mediated phagocytosis of C3bi-opsonized RBCs, but it did not affect the phagocytosis of zymosan particles, known to be mediated by the C-type lectin dectin-1. Other CD44 Abs known to have less broad anti-inflammatory activity, including KM114, KM81, and KM201, did not inhibit FcγR-mediated phagocytosis of RBCs. Taken together, these findings demonstrate selective inhibition of FcγR and CR3-mediated phagocytosis by IM7 and suggest that this broadly anti-inflammatory CD44 Ab inhibits these selected macrophage phagocytic pathways. The understanding of the immune-regulatory effects of CD44 Abs is important in the development and optimization of therapeutic strategies for the potential treatment of autoimmune conditions., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
32. Mechanistic properties of intravenous immunoglobulin in murine immune thrombocytopenia: support for FcγRIIB falls by the wayside.
- Author
-
Crow AR and Lazarus AH
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Immunoglobulins, Intravenous therapeutic use, Purpura, Thrombocytopenic, Idiopathic drug therapy, Receptors, IgG metabolism
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterised by platelet clearance resulting from the production of platelet-reactive autoantibodies. Platelet clearance appears to occur mainly via phagocytosis in the mononuclear phagocytic system, although T-cell-mediated platelet destruction, platelet apoptosis and dysregulation of platelet production can also play a role in disease pathogenesis. One of the most successful treatments for ITP is intravenous immunoglobulin (IVIg), and while it has been used in ITP for over 30 years, its mechanism(s) of action still remain unclear. Animal models of ITP have proven useful in understanding IVIg's immunomodulatory properties, providing a valuable tool to test new mechanistic theories as well as further explore the soundness of older ones. This model has also provided the key evidence that IVIg exerts its effects via activating receptors for IgG Fc, specifically FcγRIII, via formation of IgG dimers or immune complexes. Here, we discuss the validity of one prominent theory of IVIg function, anti-inflammatory activity mediated through the inhibitory Fcγ receptor FcγRIIB, and review evidence to suggest that this theory is not likely valid in the practical sense., (Crown Copyright © 2016. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. CD20+ B-cell depletion therapy suppresses murine CD8+ T-cell-mediated immune thrombocytopenia.
- Author
-
Guo L, Kapur R, Aslam R, Speck ER, Zufferey A, Zhao Y, Kim M, Lazarus AH, Ni H, and Semple JW
- Subjects
- Animals, B-Lymphocytes pathology, Integrin beta3 genetics, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, SCID, Platelet Count, Purpura, Thrombocytopenic, Idiopathic immunology, Purpura, Thrombocytopenic, Idiopathic pathology, Antigens, CD20 metabolism, B-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, Lymphocyte Depletion methods, Purpura, Thrombocytopenic, Idiopathic therapy
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder with a complex pathogenesis, which includes both antibody- and T-cell-mediated effector mechanisms. Rituximab (an anti-human CD20 monoclonal antibody [mAb]) is one of the treatments for ITP and is known to deplete B cells but may also work by affecting the T-cell compartments. Here, we investigated the outcome of B-cell depletion (Bdep) therapy on CD8(+) T-cell-mediated ITP using a murine model. CD61 knockout (KO) mice were immunized with CD61(+) platelets, and T-cell-mediated ITP was initiated by transfer of their splenocytes into severe combined immunodeficiency (SCID) mice. The CD61 KO mice were administrated an anti-mouse CD20 mAb either before or after CD61(+) platelet immunization. This resulted in efficient Bdep in vivo, accompanied by significant increases in splenic and lymph node CD4(+) and CD8(+) T cells and proportional increases of FOXP3(+) in CD4(+)and CD8(+) T cells. Moreover, Bdep therapy resulted in significantly decreased splenic CD8(+) T-cell proliferation in vitro that could be rescued by interleukin-2. This correlated with normalization of in vivo platelet counts in the transferred SCID mice suggesting that anti-CD20 therapy significantly reduces the ability of CD8(+) T cells to activate and mediate ITP., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
34. Monovalent Fc receptor blockade by an anti-Fcγ receptor/albumin fusion protein ameliorates murine ITP with abrogated toxicity.
- Author
-
Yu X, Menard M, Prechl J, Bhakta V, Sheffield WP, and Lazarus AH
- Subjects
- Albumins metabolism, Animals, Antibody-Dependent Cell Cytotoxicity immunology, Cells, Cultured, Female, Humans, Macrophages immunology, Macrophages metabolism, Macrophages pathology, Mice, Purpura, Thrombocytopenic, Idiopathic immunology, Receptors, IgG metabolism, Albumins immunology, Antibodies, Monoclonal pharmacology, Immunoglobulin Fc Fragments immunology, Purpura, Thrombocytopenic, Idiopathic therapy, Receptors, IgG antagonists & inhibitors, Receptors, IgG immunology, Recombinant Fusion Proteins immunology
- Abstract
Patients with immune thrombocytopenia (ITP) commonly have antiplatelet antibodies that cause thrombocytopenia through Fcγ receptors (FcγRs). Antibodies specific for FcγRs, designed to inhibit antibody-FcγR interaction, had been shown to improve ITP in refractory human patients. However, the development of such FcγR-specific antibodies has stalled because of adverse events, a phenomenon recapitulated in mouse models. One hypothesis behind these adverse events involved the function of the Fc region of the antibody, which engages FcγRs, leading to inflammatory responses. Unfortunately, inhibition of Fc function by deglycosylation failed to prevent this inflammatory response. In this work, we hypothesize that the bivalent antigen-binding fragment regions of immunoglobulin G are sufficient to trigger adverse events and have reasoned that designing a monovalent targeting strategy could circumvent the inflammatory response. To this end, we generated a fusion protein comprising a monovalent human FcγRIIIA-specific antibody linked in tandem to human serum albumin, which retained FcγR-binding activity in vitro. To evaluate clinically relevant in vivo FcγR-blocking function and inflammatory effects, we generated a murine version targeting the murine FcγRIII linked to murine albumin in a passive murine ITP model. Monovalent blocking of FcγR function dramatically inhibited antibody-dependent murine ITP and successfully circumvented the inflammatory response as assessed by changes in body temperature, basophil activation, and basophil depletion. Consistent with our hypothesis, in vivo cross-linking of the fusion protein induced these inflammatory effects, recapitulating the adverse events of the parent antibody. Thus, monovalent blocking of FcγR function demonstrates a proof of concept to successfully treat FcγR-mediated autoimmune diseases., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
35. IgG-Mediated Immune Suppression to Erythrocytes by Polyclonal Antibodies Can Occur in the Absence of Activating or Inhibitory Fcγ Receptors in a Full Mouse Model.
- Author
-
Bernardo L, Yu H, Amash A, Zimring JC, and Lazarus AH
- Subjects
- Animals, Duffy Blood-Group System immunology, Enzyme-Linked Immunosorbent Assay, Erythrocytes metabolism, Humans, Immune Tolerance genetics, Immunoglobulin M immunology, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Models, Immunological, Muramidase genetics, Muramidase immunology, Muramidase metabolism, Ovalbumin immunology, Receptors, IgG genetics, Receptors, IgG metabolism, Rh-Hr Blood-Group System immunology, Rho(D) Immune Globulin immunology, Antibodies, Monoclonal immunology, Erythrocytes immunology, Immune Tolerance immunology, Immunoglobulin G immunology, Receptors, IgG immunology
- Abstract
Polyclonal anti-D has been used to prevent RhD-negative mothers from becoming immunized against RhD positive fetal erythrocytes, and this mechanism has been referred as Ab or IgG-mediated immune suppression (AMIS). Although anti-D has been highly successful, the inhibitory mechanisms remain poorly understood. Two major theories behind AMIS involve the binding of IgG to activating or inhibitory FcγR, which can induce either erythrocyte clearance or immune inhibition, respectively. In this work, we explored the absolute role of activating and inhibitory FcγR in the AMIS mechanism using the HOD mouse model of RBC immunization. HOD mice contain a RBC-specific recombinant protein composed of hen egg lysozyme (HEL), OVA and human transmembrane Duffy Ag, and erythrocytes from HOD mice can stimulate an immune response to HEL. To assess the contribution of activating and inhibitory FcγR to AMIS, C57BL/6 versus FcRγ-chain(-/-) or FcγRIIB(-/-) mice were used as recipients of HOD-RBC alone or together with anti-HEL Abs (i.e., AMIS) and the resulting immune response to HEL evaluated. We show that anti-HEL polyclonal Abs induce the same degree of AMIS effect in mice lacking these IgG binding receptors as compared with wild-type mice. In agreement with this, F(ab')2 fragments of the AMIS Ab also significantly reduced the Ab response to the HOD cells. In conclusion, successful inhibition of in vivo Ab responses to HOD-RBC by polyclonal IgG can occur independently of activating or inhibitory FcγR involvement. These results may have implications for the understanding of RhD prophylaxis., (Copyright © 2015 by The American Association of Immunologists, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
36. CD8+ T cells are predominantly protective and required for effective steroid therapy in murine models of immune thrombocytopenia.
- Author
-
Ma L, Simpson E, Li J, Xuan M, Xu M, Baker L, Shi Y, Yougbaré I, Wang X, Zhu G, Chen P, Prud'homme GJ, Lazarus AH, Freedman J, and Ni H
- Subjects
- Animals, Blood Platelets immunology, CD8-Positive T-Lymphocytes transplantation, Combined Modality Therapy, Disease Models, Animal, Immunotherapy, Adoptive, Lymphocyte Depletion, Mice, Mice, Inbred BALB C, Mice, Knockout, Platelet Glycoprotein GPIIb-IIIa Complex genetics, Platelet Glycoprotein GPIIb-IIIa Complex immunology, Purpura, Thrombocytopenic, Idiopathic therapy, T-Lymphocytes, Cytotoxic, Treatment Outcome, CD8-Positive T-Lymphocytes physiology, Dexamethasone therapeutic use, Purpura, Thrombocytopenic, Idiopathic drug therapy, Purpura, Thrombocytopenic, Idiopathic immunology
- Abstract
Immune thrombocytopenia (ITP) is a common autoimmune bleeding disorder characterized by autoantibodies targeting platelet surface proteins, most commonly GPIIbIIIa (αIIbβ3 integrin), leading to platelet destruction. Recently, CD8(+) cytotoxic T-lymphocytes (CTLs) targeting platelets and megakaryocytes have also been implicated in thrombocytopenia. Because steroids are the most commonly administered therapy for ITP worldwide, we established both active (immunized splenocyte engraftment) and passive (antibody injection) murine models of steroid treatment. Surprisingly, we found that, in both models, CD8(+) T cells limited the severity of the thrombocytopenia and were required for an efficacious response to steroid therapy. Conversely, CD8(+) T-cell depletion led to more severe thrombocytopenia, whereas CD8(+) T-cell transfusion ameliorated thrombocytopenia. CD8(+) T-regulatory cell (Treg) subsets were detected, and interestingly, dexamethasone (DEX) treatment selectively expanded CD8(+) Tregs while decreasing CTLs. In vitro coculture studies revealed CD8(+) Tregs suppressed CD4(+) and CD19(+) proliferation, platelet-associated immunoglobulin G generation, CTL cytotoxicity, platelet apoptosis, and clearance. Furthermore, we found increased production of anti-inflammatory interleukin-10 in coculture studies and in vivo after steroid treatment. Thus, we uncovered subsets of CD8(+) Tregs and demonstrated their potent immunosuppressive and protective roles in experimentally induced thrombocytopenia. The data further elucidate mechanisms of steroid treatment and suggest therapeutic potential for CD8(+) Tregs in immune thrombocytopenia., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
37. A monoclonal antibody with anti-D-like activity in murine immune thrombocytopenia requires Fc domain function for immune thrombocytopenia ameliorative effects.
- Author
-
Yu X, Menard M, Seabright G, Crispin M, and Lazarus AH
- Subjects
- Animals, Animals, Outbred Strains, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal therapeutic use, Cells, Cultured, Disease Models, Animal, Female, Glycosylation, Immunoglobulin Fab Fragments chemistry, Immunoglobulin Fab Fragments metabolism, Immunoglobulin Fab Fragments pharmacology, Immunoglobulin Fab Fragments therapeutic use, Immunoglobulin Fc Fragments chemistry, Immunoglobulin Fc Fragments metabolism, Immunoglobulin Fc Fragments therapeutic use, Isoantibodies chemistry, Mice, Mice, Inbred C57BL, Protein Structure, Tertiary physiology, Purpura, Thrombocytopenic, Idiopathic immunology, Purpura, Thrombocytopenic, Idiopathic pathology, Rho(D) Immune Globulin, Antibodies, Monoclonal pharmacology, Immunoglobulin Fc Fragments pharmacology, Isoantibodies pharmacology, Purpura, Thrombocytopenic, Idiopathic therapy
- Abstract
Background: The mechanism of action of anti-D in ameliorating immune thrombocytopenia (ITP) remains unclear. The monoclonal antibody (MoAb) Ter119, which targets murine red blood cells (RBCs), has been shown to mimic the effect of anti-D in improving antibody-mediated murine ITP. The mechanism of Ter119-mediated ITP amelioration, especially the role of the antigen-binding and Fc domains, remains untested. A functional Fc domain is crucial for many therapeutic MoAb activity; therefore, the requirement of Ter119 Fc domain in ITP amelioration is investigated using outbred CD-1 mice., Study Design and Methods: Ter119 variants, including Ter119 F(ab')2 fragments, deglycosylated Ter119, and afucosylated Ter119, were generated to test their effect in ameliorating antibody-induced murine ITP. In vivo inhibition of FcγRIII and FcγRIIB was achieved using the Fab fragment of the FcγRIII/FcγRIIB-specific MoAb 2.4G2., Results: Ter119 F(ab')2 fragments and deglycosylated Ter119 were unable to ameliorate murine ITP or mediate phagocytosis of RBCs by RAW264.7 macrophages in vitro. Inhibition of FcγRIII and FcγRIIB, as well as Ter119 defucosylation, do not affect Ter119-mediated ITP amelioration., Conclusion: The Fc domain of Ter119, as well as its Fc glycosylation, is required for Ter119-mediated ITP amelioration. Moreover, both Fc and Fc glycosylation are required for Ter119-mediated phagocytosis in vitro. These findings demonstrate the importance of the Fc domain in a therapeutic MoAb with anti-D-like activity., (© 2015 AABB.)
- Published
- 2015
- Full Text
- View/download PDF
38. CD44 antibody-mediated amelioration of murine immune thrombocytopenia (ITP): mouse background determines the effect of FcγRIIb genetic disruption.
- Author
-
Crow AR, Amash A, and Lazarus AH
- Subjects
- Animals, Disease Models, Animal, Gene Targeting, Immunoglobulins, Intravenous therapeutic use, Immunotherapy, Mice, Mice, Inbred BALB C, Mice, Inbred C3H, Mice, Inbred C57BL, Mice, Knockout, Mice, SCID, Purpura, Thrombocytopenic, Idiopathic immunology, Species Specificity, Antibodies therapeutic use, Hyaluronan Receptors immunology, Purpura, Thrombocytopenic, Idiopathic genetics, Purpura, Thrombocytopenic, Idiopathic therapy, Receptors, IgG genetics
- Abstract
Background: Several monoclonal antibodies to CD44 can successfully ameliorate murine immune thrombocytopenia (ITP). As these antibodies may be a potential replacement for intravenous immune globulin (IVIG) in the treatment of ITP and other autoimmune diseases, an understanding of their mechanisms of action is important. The role of the inhibitory Fc receptor (FcγRIIb) in the mechanism of action of IVIG and therapeutic CD44 antibodies remains uncertain. To assess if FcγRIIb in splenic macrophages plays a critical role in the action of these two therapeutics, splenectomized mice and mice genetically deficient in FcγRIIb on different backgrounds were evaluated., Study Design and Methods: Thrombocytopenia was induced in FcγRIIb-deficient mice on B6;129S, C57BL/6, and BALB/C backgrounds, as well as splenectomized mice and control mice by platelet (PLT) antibody. PLT counts were enumerated before and after treatment with anti-CD44, red blood cell antibodies, or IVIG., Results: Anti-CD44 is ineffective at inhibiting thrombocytopenia in B6;129S FcγRIIb-deficient mice but, like IVIG, is effective in splenectomized mice and FcγRIIb-deficient mice on the BALB/C and C57BL/6 background., Conclusion: These data suggest that 1) the B6;129S background itself is unlikely to be the sole reason for anti-CD44's inability to function in B6;129S FcγRIIb-deficient mice, 2) the simple loss of macrophage FcγRIIb expression alone is insufficient to explain anti-CD44 ameliorative function, and 3) a combination of mouse background genes in addition to FcγRIIb genetic disruption may affect the ability of anti-CD44 to function therapeutically. Similarities between IVIG and anti-CD44 mechanisms suggest that patients responsive to IVIG may also potentially respond to anti-CD44 treatment., (© 2014 AABB.)
- Published
- 2015
- Full Text
- View/download PDF
39. Antibody-mediated immune suppression of erythrocyte alloimmunization can occur independently from red cell clearance or epitope masking in a murine model.
- Author
-
Yu H, Stowell SR, Bernardo L, Hendrickson JE, Zimring JC, Amash A, Uchikawa M, and Lazarus AH
- Subjects
- Animals, Antibodies, Monoclonal immunology, Duffy Blood-Group System biosynthesis, Duffy Blood-Group System genetics, Duffy Blood-Group System immunology, Erythrocyte Transfusion, Immune Tolerance immunology, Immunoglobulin M immunology, Isoantibodies immunology, Mice, Mice, Inbred C57BL, Muramidase biosynthesis, Muramidase genetics, Muramidase immunology, Ovalbumin biosynthesis, Ovalbumin genetics, Ovalbumin immunology, Receptors, Cell Surface biosynthesis, Receptors, Cell Surface genetics, Receptors, Cell Surface immunology, Allografts immunology, Epitopes immunology, Erythrocytes immunology, Immunoglobulin G immunology, Transplantation Tolerance immunology
- Abstract
Anti-D can prevent immunization to the RhD Ag on RBCs, a phenomenon commonly termed Ab-mediated immune suppression (AMIS). The most accepted theory to explain this effect has been the rapid clearance of RBCs. In mouse models using SRBC, these xenogeneic cells are always rapidly cleared even without Ab, and involvement of epitope masking of the SRBC Ags by the AMIS-inducing Ab (anti-SRBC) has been suggested. To address these hypotheses, we immunized mice with murine transgenic RBCs expressing the HOD Ag (hen egg lysozyme [HEL], in sequence with ovalbumin, and the human Duffy transmembrane protein) in the presence of polyclonal Abs or mAbs to the HOD molecule. The isotype, specificity, and ability to induce AMIS of these Abs were compared with accelerated clearance as well as steric hindrance of the HOD Ag. Mice made IgM and IgG reactive with the HEL portion of the molecule only. All six of the mAbs could inhibit the response. The HEL-specific Abs (4B7, IgG1; GD7, IgG2b; 2F4, IgG1) did not accelerate clearance of the HOD-RBCs and displayed partial epitope masking. The Duffy-specific Abs (MIMA 29, IgG2a; CBC-512, IgG1; K6, IgG1) all caused rapid clearance of HOD RBCs without steric hindrance. To our knowledge, this is the first demonstration of AMIS to erythrocytes in an all-murine model and shows that AMIS can occur in the absence of RBC clearance or epitope masking. The AMIS effect was also independent of IgG isotype and epitope specificity of the AMIS-inducing Ab., (Copyright © 2014 by The American Association of Immunologists, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
40. Therapeutic effect of IVIG on inflammatory arthritis in mice is dependent on the Fc portion and independent of sialylation or basophils.
- Author
-
Campbell IK, Miescher S, Branch DR, Mott PJ, Lazarus AH, Han D, Maraskovsky E, Zuercher AW, Neschadim A, Leontyev D, McKenzie BS, and Käsermann F
- Subjects
- Animals, Arthritis pathology, Basophils pathology, Disease Models, Animal, Immunoglobulins, Intravenous immunology, Immunologic Factors immunology, Male, Mice, Mice, Inbred NOD, Arthritis immunology, Arthritis prevention & control, Basophils immunology, Immunoglobulin Fc Fragments immunology, Immunoglobulins, Intravenous pharmacology, Immunologic Factors pharmacology, N-Acetylneuraminic Acid immunology
- Abstract
High-dose i.v. Ig (IVIG) is used to treat various autoimmune and inflammatory diseases; however, the mechanism of action remains unclear. Based on the K/BxN serum transfer arthritis model in mice, IVIG suppression of inflammation has been attributed to a mechanism involving basophils and the binding of highly sialylated IgG Fc to DC-SIGN-expressing myeloid cells. The requirement for sialylation was examined in the collagen Ab-induced arthritis (CAbIA) and K/BxN serum transfer arthritis models in mice. High-dose IVIG (1-2 g/kg body weight) suppressed inflammatory arthritis when given prophylactically. The same doses were also effective in the CAbIA model when given subsequent to disease induction. In this therapeutic CAbIA model, the anti-inflammatory effect of IVIG was dependent on IgG Fc but not F(ab')2 fragments. Removal of sialic acid residues by neuraminidase had no impact on the anti-inflammatory activity of IVIG or Fc fragments. Treatment of mice with basophil-depleting mAbs did not abrogate the suppression of either CAbIA or K/BxN arthritis by IVIG. Our data confirm the therapeutic benefit of IVIG and IgG Fc in Ab-induced arthritis but fail to support the significance of sialylation and basophil involvement in the mechanism of action of IVIG therapy., (Copyright © 2014 by The American Association of Immunologists, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
41. Antibody specific for the glycophorin A complex mediates intravenous immune globulin-resistant anemia in a murine model.
- Author
-
Chen X, Ghaffar H, Jen CC, and Lazarus AH
- Subjects
- Animals, Female, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Anemia drug therapy, Anemia immunology, Antibodies immunology, Glycophorins immunology, Immunoglobulins, Intravenous therapeutic use
- Abstract
Background: Therapy for patients with autoimmune hemolytic anemia (AHA) remains a major challenge. Patients with glycophorin A (GPA)-specific immunoglobulin G antibodies can have severe hemolysis, which may occur by mechanisms independent from traditional macrophage-dependent Fcγ receptor (FcγR)-mediated extravascular hemolysis. As intravenous immune globulin (IVIG) is known to display its beneficial effects in FcγR-mediated cytopenias, and IVIG responses in AHA are inconsistent at best, we sought to gain insight into the mechanism of anemia by a GPA complex-specific monoclonal antibody (TER119) in a mouse model of immune hemolytic anemia and evaluate the therapeutic effect of IVIG., Study Design and Methods: The anemic effect of the TER119 antibody was studied in vitro by incubation of mouse RBC with the antibody and in vivo by infusing the antibody into normal mice versus mice genetically deficient for the Fc receptor γ chain (Fcγ), complement C3, mice naturally deficient in complement C5, and splenectomized mice. IVIG efficacy in anemia was determined by treating mice with an intensive IVIG dosing regimen., Results: The TER119-mediated anemia was independent of classical FcγR-, C3-, and C5-dependent mechanisms, but occurred by a mechanism consistent with RBC agglutination. In accordance with agglutination, the presence of the spleen accelerated the anemia observed but anemia could still occur in splenectomized mice. IVIG did not significantly affect the induction of anemia by TER119., Conclusion: The mechanism of anemia induced by AHA-causing antibodies may be an important factor to consider in the response to therapy with IVIG., (© 2013 American Association of Blood Banks.)
- Published
- 2014
- Full Text
- View/download PDF
42. Allogeneic platelet transfusions prevent murine T-cell-mediated immune thrombocytopenia.
- Author
-
Guo L, Yang L, Speck ER, Aslam R, Kim M, McKenzie CG, Lazarus AH, Ni H, Hou M, Freedman J, and Semple JW
- Subjects
- Animals, Bone Marrow Cells cytology, Disease Models, Animal, Female, Histocompatibility Antigens Class I metabolism, Immunoglobulin G immunology, Integrin beta3 metabolism, Megakaryocytes cytology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Mice, SCID, Spleen cytology, Thrombocytopenia immunology, Time Factors, Platelet Transfusion methods, T-Lymphocytes immunology, Thrombocytopenia genetics, Thrombocytopenia prevention & control
- Abstract
Platelet transfusions are life-saving treatments for many patients with thrombocytopenia; however, their use is generally discouraged in the autoimmune disorder known as immune thrombocytopenia (ITP). We examined whether allogeneic platelet major histocompatibility complex (MHC) class I transfusions affected antiplatelet CD61-induced ITP. BALB/c CD61 knockout mice (CD61(-)/H-2(d)) were immunized against platelets from wild-type syngeneic BALB/c (CD61(+)/H-2(d)), allogeneic C57BL/6 (CD61(+)/H-2(b)), or C57BL/6 CD61 KO (CD61(-)/H-2(b)) mice, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice to induce ITP. When nondepleted splenocytes were transferred to induce antibody-mediated ITP, both CD61(+) platelet immunizations generated immunity that caused thrombocytopenia independently of allogeneic MHC molecules. In contrast, when B-cell-depleted splenocytes were transferred to induce T-cell-mediated ITP, transfer of allogeneic MHC-immunized splenocytes completely prevented CD61-induced ITP development. In addition, allogeneic platelet transfusions into SCID mice with established CD61-induced ITP rescued the thrombocytopenia. Compared with thrombocytopenic mice, bone marrow histology in the rescued mice showed normalized megakaryocyte morphology, and in vitro CD61-specific T-cell cytotoxicity was significantly suppressed. These results indicate that antibody-mediated ITP is resistant to allogeneic platelet transfusions, while the T-cell-mediated form of the disease is susceptible, suggesting that transfusion therapy may be beneficial in antibody-negative ITP.
- Published
- 2014
- Full Text
- View/download PDF
43. RhD Specific Antibodies Are Not Detectable in HLA-DRB1(*)1501 Mice Challenged with Human RhD Positive Erythrocytes.
- Author
-
Bernardo L, Denomme GA, Shah K, and Lazarus AH
- Abstract
The ability to study the immune response to the RhD antigen in the prevention of hemolytic disease of the fetus and newborn has been hampered by the lack of a mouse model of RhD immunization. However, the ability of transgenic mice expressing human HLA DRB1(*)1501 to respond to immunization with purified RhD has allowed this question to be revisited. In this work we aimed at inducing anti-RhD antibodies by administering human RhD(+) RBCs to mice transgenic for the human HLA DRB1(*)1501 as well as to several standard inbred and outbred laboratory strains including C57BL/6, DBA1/J, CFW(SW), CD1(ICR), and NSA(CF-1). DRB1(*)1501 mice were additionally immunized with putative extracellular immunogenic RhD peptides. DRB1(*)1501 mice immunized with RhD(+) erythrocytes developed an erythrocyte-reactive antibody response. Antibodies specific for RhD could not however be detected by flow cytometry. Despite this, DRB1(*)1501 mice were capable of recognizing immunogenic sequences of Rh as injection with Rh peptides induced antibodies reactive with RhD sequences, consistent with the presence of B cell repertoires capable of recognizing RhD. We conclude that while HLA DRB1(*)1501 transgenic mice may have the capability of responding to immunogenic sequences within RhD, an immune response to human RBC expressing RhD is not directly observed.
- Published
- 2014
- Full Text
- View/download PDF
44. Monoclonal versus polyclonal anti-D in the treatment of ITP.
- Author
-
Lazarus AH
- Subjects
- Animals, Humans, Mice, Purpura, Thrombocytopenic, Idiopathic immunology, Antibodies, Monoclonal therapeutic use, Isoantibodies therapeutic use, Purpura, Thrombocytopenic, Idiopathic drug therapy, Rho(D) Immune Globulin immunology
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder caused by low numbers of platelets generally due to the production of anti-platelet antibodies. One effective treatment for ITP patients who express the RhD antigen on their red blood cells has been the use of blood donor-derived pooled polyclonal anti-D. Although anti-D has served us well, it needs to be replaced with a recombinant product. While the mechanism of action of anti-D in ITP remains highly speculative, this has not thwarted attempts to replace anti-D with a monoclonal product. Although a single attempt at a monoclonal antibody was not successful in the 1990s for the treatment of ITP, more recent efforts in mouse models of ITP and ITP patients now show that monoclonal antibodies can be successful in ITP. These studies also finally help substantiate the concept that it is unlikely that contaminants in the original donor-derived preparations mediate the major ameliorative activity of anti-D in ITP.
- Published
- 2013
- Full Text
- View/download PDF
45. Amelioration of murine passive immune thrombocytopenia by IVIg and a therapeutic monoclonal CD44 antibody does not require the Myd88 signaling pathway.
- Author
-
Crow AR, Yu H, Han D, and Lazarus AH
- Subjects
- Animals, Female, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction genetics, Signal Transduction immunology, Thrombocytopenia genetics, Thrombocytopenia immunology, Toll-Like Receptor 4 physiology, Antibodies, Monoclonal administration & dosage, Hyaluronan Receptors immunology, Immunization, Passive methods, Immunoglobulins, Intravenous administration & dosage, Myeloid Differentiation Factor 88 physiology, Thrombocytopenia therapy
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by a low platelet count and the production of anti-platelet antibodies. The majority of ITP patients have antibodies to platelet integrin α(IIb)β₃ (GPIIbIIIa) which can direct platelet phagocytosis by macrophages. One effective treatment for patients with ITP is intravenous immunoglobulin (IVIg) which rapidly reverses thrombocytopenia. The exact mechanism of IVIg action in human patients is unclear, although in mouse models of passive ITP, IVIg can rapidly increase platelet counts in the absence of adaptive immunity. Another antibody therapeutic that can similarly increase platelet counts independent of adaptive immunity are CD44 antibodies. Toll-like receptors (TLRs) are pattern recognition receptors which play a central role in helping direct the innate immune system. Dendritic cells, which are notable for their expression of TLRs, have been directly implicated in IVIg function as an initiator cell, while CD44 can associate with TLR2 and TLR4. We therefore questioned whether IVIg, or the therapeutic CD44 antibody KM114, mediate their ameliorative effects in a manner dependent upon normal TLR function. Here, we demonstrate that the TLR4 agonist LPS does not inhibit IVIg or KM114 amelioration of antibody-induced thrombocytopenia, and that these therapeutics do not ameliorate LPS-induced thrombocytopenia. IVIg was able to significantly ameliorate murine ITP in C3H/HeJ mice which have defective TLR4. All known murine TLRs except TLR3 utilize the Myd88 adapter protein to drive TLR signaling. Employing Myd88 deficient mice, we found that both IVIg and KM114 ameliorate murine ITP in Myd88 deficient mice to the same extent as normal mice. Thus both IVIg and anti-CD44 antibody can mediate their ameliorative effects in murine passive ITP independent of the Myd88 signaling pathway. These data help shed light on the mechanism of action of IVIg and KM114 in the amelioration of murine ITP.
- Published
- 2013
- Full Text
- View/download PDF
46. CD44 antibodies and immune thrombocytopenia in the amelioration of murine inflammatory arthritis.
- Author
-
Mott PJ and Lazarus AH
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Arthritis complications, Arthritis drug therapy, Blood Platelets immunology, Disease Models, Animal, Female, Mice, Platelet Count, Purpura, Thrombocytopenic, Idiopathic complications, Thrombocytosis immunology, Antibodies, Monoclonal immunology, Arthritis immunology, Hyaluronan Receptors immunology, Purpura, Thrombocytopenic, Idiopathic immunology
- Abstract
Antibodies to CD44 have been used to successfully ameliorate murine models of autoimmune disease. The most often studied disease model has been murine inflammatory arthritis, where a clear mechanism for the efficacy of CD44 antibodies has not been established. We have recently shown in a murine passive-model of the autoimmune disease immune thrombocytopenia (ITP) that some CD44 antibodies themselves can induce thrombocytopenia in mice, and the CD44 antibody causing the most severe thrombocytopenia (IM7), also is known to be highly effective in ameliorating murine models of arthritis. Recent work in the K/BxN serum-induced model of arthritis demonstrated that antibody-induced thrombocytopenia reduced arthritis, causing us to question whether CD44 antibodies might primarily ameliorate arthritis through their thrombocytopenic effect. We evaluated IM7, IRAWB14.4, 5035-41.1D, KM201, KM114, and KM81, and found that while all could induce thrombocytopenia, the degree of protection against serum-induced arthritis was not closely related to the length or severity of the thrombocytopenia. CD44 antibody treatment was also able to reverse established inflammation, while thrombocytopenia induced by an anti-platelet antibody targeting the GPIIbIIIa platelet antigen, could not mediate this effect. While CD44 antibody-induced thrombocytopenia may contribute to some of its therapeutic effect against the initiation of arthritis, for established disease there are likely other mechanisms contributing to its efficacy. Humans are not known to express CD44 on platelets, and are therefore unlikely to develop thrombocytopenia after CD44 antibody treatment. An understanding of the relationship between arthritis, thrombocytopenia, and CD44 antibody treatment remains critical for continued development of CD44 antibody therapeutics.
- Published
- 2013
- Full Text
- View/download PDF
47. Innate and adaptive immunity in immune thrombocytopenia.
- Author
-
Lazarus AH, Semple JW, and Cines DB
- Subjects
- Adaptive Immunity immunology, Animals, Humans, Immunity, Innate immunology, Thrombocytopenia immunology
- Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by both accelerated clearance of autoantibody-sensitized platelets and suboptimal platelet production. A number of studies have provided evidence of disturbed innate and adaptive immune responses in patients with ITP. This brief review will highlight some of the more recent work in this field and highlight other findings that provide a potential link between ITP, systemic lupus erythematosus (SLE), and autoimmune hemolytic anemia (AHA)., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
48. Intravenous immunoglobulin prevents murine antibody-mediated acute lung injury at the level of neutrophil reactive oxygen species (ROS) production.
- Author
-
Semple JW, Kim M, Hou J, McVey M, Lee YJ, Tabuchi A, Kuebler WM, Chai ZW, and Lazarus AH
- Subjects
- Acids, Acute Lung Injury complications, Acute Lung Injury pathology, Animals, Chemokine CXCL2 metabolism, Edema complications, Humans, Hypothermia complications, Hypothermia pathology, Immunoglobulins, Intravenous pharmacology, Lung drug effects, Lung metabolism, Lung pathology, Male, Mice, Mice, Inbred BALB C, Mice, SCID, Neutrophils drug effects, Survival Analysis, Transfusion Reaction, Acute Lung Injury drug therapy, Acute Lung Injury prevention & control, Immunoglobulins, Intravenous therapeutic use, Neutrophils metabolism, Reactive Oxygen Species metabolism
- Abstract
Transfusion-related acute lung injury (TRALI) is a leading cause of transfusion-associated mortality that can occur with any type of transfusion and is thought to be primarily due to donor antibodies activating pulmonary neutrophils in recipients. Recently, a large prospective case controlled clinical study of cardiac surgery patients demonstrated that despite implementation of male donors, a high incidence of TRALI still occurred and suggested a need for additional interventions in susceptible patient populations. To examine if intravenous immunoglobulin (IVIg) may be effective, a murine model of antibody-mediated acute lung injury that approximates human TRALI was examined. When BALB/c mice were injected with the anti-major histocompatibility complex class I antibody 34-1-2s, mild shock (reduced rectal temperature) and respiratory distress (dyspnea) were observed and pre-treatment of the mice with 2 g/kg IVIg completely prevented these symptoms. To determine IVIg's usefulness to affect severe lung damage, SCID mice, previously shown to be hypersensitive to 34-1-2s were used. SCID mice treated with 34-1-2s underwent severe shock, lung damage (increased wet/dry ratios) and 40% mortality within 2 hours. Treatment with 2 g/kg IVIg 18 hours before 34-1-2s administration completely protected the mice from all adverse events. Treatment with IVIg after symptoms began also reduced lung damage and mortality. While the prophylactic IVIg administration did not affect 34-1-2s-induced pulmonary neutrophil accumulation, bone marrow-derived neutrophils from the IVIg-treated mice displayed no spontaneous ROS production nor could they be stimulated in vitro with fMLP or 34-1-2s. These results suggest that IVIg prevents murine antibody-mediated acute lung injury at the level of neutrophil ROS production and thus, alleviating tissue damage.
- Published
- 2012
- Full Text
- View/download PDF
49. The neonatal Fc receptor (FcRn) is not required for IVIg or anti-CD44 monoclonal antibody-mediated amelioration of murine immune thrombocytopenia.
- Author
-
Crow AR, Suppa SJ, Chen X, Mott PJ, and Lazarus AH
- Subjects
- Animals, Disease Models, Animal, Histocompatibility Antigens Class I genetics, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Platelet Count, Purpura, Thrombocytopenic, Idiopathic blood, Purpura, Thrombocytopenic, Idiopathic immunology, Receptors, Fc genetics, beta 2-Microglobulin genetics, beta 2-Microglobulin physiology, Antibodies, Monoclonal therapeutic use, Autoantibodies therapeutic use, Histocompatibility Antigens Class I physiology, Hyaluronan Receptors chemistry, Immunoglobulins, Intravenous therapeutic use, Purpura, Thrombocytopenic, Idiopathic prevention & control, Receptors, Fc physiology
- Abstract
To definitively determine whether the neonatal Fc receptor (FcRn) is required for the acute amelioration of immune thrombocytopenia (ITP) by IVIg, we used FcRn-deficient mice in a murine ITP model. Mice injected with antiplatelet antibody in the presence or absence of IVIg displayed no difference in platelet-associated IgG between FcRn deficient versus C57BL/6 mice. FcRn-deficient mice treated with high-dose (2 g/kg) IVIg or a low-dose (2 mg/kg) of an IVIg-mimetic CD44 antibody were, however, protected from thrombocytopenia to an equivalent extent as wild-type mice. To verify and substantiate the results found with FcRn-deficient mice, we used β(2)-microglobulin-deficient mice (which do not express functional FcRn) and found that IVIg or CD44 antibody also protected them from thrombocytopenia. These data suggest that for both high-dose IVIg as well as low-dose CD44 antibody treatment in an acute ITP model, FcRn expression is neither necessary nor required.
- Published
- 2011
- Full Text
- View/download PDF
50. Immunity against a therapeutic xenoprotein/Fc construct delivered by gene transfer is reduced through binding to the inhibitory receptor FcγRIIb.
- Author
-
Liang Y, Qiu H, Glinka Y, Lazarus AH, Ni H, Prud'homme GJ, and Wang Q
- Subjects
- Animals, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology, B-Lymphocytes immunology, B-Lymphocytes metabolism, Diabetes Mellitus, Type 2 therapy, Exenatide, Gene Expression Regulation, Gene Transfer Techniques, Immunoglobulin Fc Fragments metabolism, Injections, Intramuscular, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Plasmids administration & dosage, Plasmids genetics, Protein Binding, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins immunology, Recombinant Fusion Proteins metabolism, Transduction, Genetic, Genetic Therapy, Hypoglycemic Agents immunology, Immunoglobulin Fc Fragments genetics, Peptides genetics, Peptides immunology, Receptors, IgG metabolism, Venoms genetics, Venoms immunology
- Abstract
Background: Therapeutic xenoproteins are immunogenic and can induce neutralizing antibodies. When delivered by intramuscular injection of a plasmid vector, this mimics classical DNA vaccination. To demonstrate this, we chose Exendin-4 (Ex4), which is a glucagon-like peptide-1 mimetic xenoprotein in clinical use for treating type 2 diabetes. We constructed an Ex4 and mouse immunoglobulin (Ig)G1-Fc fusion fragment (Ex4/Fc), and hypothesized that it would have minimal immunogenicity as a result of its capacity to bind the inhibitory Fc receptor FcγRIIb expressed by B lymphocytes., Methods: Plasmid vectors encoding Ex4/Fc constructs, with wild-type or mutant Fc, were injected intramuscularly into mice, and local electroporation was applied to enhance gene transfer. Gene transfer was performed in both wild-type and FcγRIIb knockout mice. Antibody production was detected in serum by an enzyme-linked immunosorbent assay., Results: Recombinant Ex4/Fc bound only to B cells expressing FcγRIIb. This binding was dependent on a motif in the Fc region, which we mutated to abolish binding (Ex4/Fcmut). Ex4 antibody was detected in mice treated with Ex4, as well as Ex4/Fcmut, but not in those treated with Ex4/Fc. Thus, wild-type Fc was associated with reduced immunogenicity. To confirm this was related to the presence of inhibitory Fc receptors, we also performed experiments in FcγRIIb-null mice. Mice lacking this receptor produced antibodies against all Ex4 constructs, including the wild-type Fc (Ex4/Fc)., Conclusions: The present study shows that inhibitory FcγRIIb receptors interacting with the wild-type IgG1-Fc reduce immunity against Ex4/Fc, suggesting an approach for reducing the immunogenicity of therapeutic proteins in the context of gene therapy., (Copyright © 2011 John Wiley & Sons, Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.