Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Pharmacology ; 108(2): 176-187, 2023.
Article in English | MEDLINE | ID: mdl-36696888

ABSTRACT

INTRODUCTION: Neutrophils are a pivotal cell type in the K/BxN mouse model of rheumatoid arthritis and play an essential role in the progression of the arthritis. They are readily activated by immune complexes (ICs) via their FcγRs to release IL-1ß in addition to other cytokines, which are inducing cartilage destruction. Neutrophils also release neutrophil-active chemokines to recruit themselves in an autocrine manner to perpetuate tissue destruction. FcγR-expression on neutrophils is of crucial importance for the recognition of ICs. METHODS: In this study, due to its high avidity for binding to FcγRs, we investigated the potential anti-inflammatory effect of a recombinant IgG1 Fc hexamer (rFc-µTP-L309C) on neutrophils in the K/BxN mouse model of endogenously generated chronic arthritis. 200 mg/kg rFc-µTP-L309C and human serum albumin (HSA), used as controls, were administered subcutaneously every other day. Mouse ankle joints were monitored daily to generate a clinical score. Immunohistology was used to evaluate neutrophil infiltration and TUNEL to assess apoptosis. ELISA was used to measure IL-1ß. RESULTS: Treatment with rFc-µTP-L309C, but not HSA, was able to significantly ameliorate the arthritis in the K/BxN mice. Significant neutrophil infiltration into the ankle joint was found, but treatment with rFc-µTP-L309C resulted in significantly less neutrophil infiltration. There was no significant influence of rFc-µTP-L309C on neutrophil death or apoptosis. Less neutrophil infiltration could not be correlated to chemokine-mediated migration. Significantly less IL-1ß was measured in mice treated with rFc-µTP-L309C. CONCLUSION: In the endogenous K/BxN mouse model of rheumatoid arthritis, amelioration can be explained in part by inhibition of neutrophil infiltration into the joints as well as inhibition of IL-1ß production. Given the observed inhibitory properties on neutrophils, rFc-µTP-L309C may be a potential therapeutic candidate to treat autoimmune and inflammatory conditions in which neutrophils are the predominant cell type involved in pathogenesis.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Humans , Mice , Animals , Immunoglobulin G/metabolism , Neutrophils/metabolism , Neutrophils/pathology , Uridine Triphosphate/metabolism , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Immunologic Factors , Mice, Inbred C57BL
2.
EJHaem ; 2(4): 789-793, 2021 Nov.
Article in English | MEDLINE | ID: mdl-35845218

ABSTRACT

Recombinant Fc-µTP-L309C is more efficacious than intravenous immunoglobulin (IVIg) at ameliorating antibody-mediated autoimmune diseases through its effects on Fcγ receptors (FcγRs). Fc-µTP-L309C inhibited in-vitro FcγR-mediated phagocytosis 104/105-fold better than IVIg. Fc-µTP-L309C, given subcutaneously, recovered platelet counts in an immune thrombocytopenia (ITP) mouse model to a higher degree than IVIg at a 10-fold lower dose. We show, using confocal microscopy, that Fc-µTP-L309C binds to monocyte-macrophages and is rapidly internalized, whereas, IVIg remains on the cell surface. Western blotting showed that internalized FcγRIII is degraded through a lysosomal pathway, and this reduction of cell surface FcγRIII is likely responsible for the increased efficacy to ameliorate ITP.

3.
Pharmacology ; 105(11-12): 618-629, 2020.
Article in English | MEDLINE | ID: mdl-32570239

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation, swelling, and pain in the joints and involves systemic complications. Mouse models of RA have been extensively used to model the pathogenesis of RA and to develop effective therapies. Although many components of the immune system have been studied in these models, the role of crystallizable fragment (Fc) gamma receptors (FcγRs) in RA has been sorely neglected. The aim of this review was to introduce the different mouse models of RA and to describe the different drug development strategies that have been tested in these models to target FcγR function, with the focus being on drugs that have been made from the Fc of immunoglobulin G (IgG). SUMMARY: Evidence suggests that FcγRs play a major role in immune complex-induced inflammation in autoimmune diseases, such as RA. However, there is limited knowledge on the importance of FcγRs in the human disease even though there has been extensive work in mouse models of RA. Numerous mouse models of RA are available, with each model depicting certain aspects of the disease. Induced models of RA have nonspecific immune activation with cartilage-directed autoimmunity, whereas spontaneous models of RA develop without immunization, which results in a more chronic form of arthritis. These models have been used to test FcγR-targeting monoclonal antibodies, intravenous immunoglobulin (IVIg), subcutaneously administered IVIg, and recombinant Fcs for their ability to interact with and modify FcγR function. Recombinant Fcs avidly bind FcγRs and exhibit enhanced therapeutic efficacy in mouse models of RA. Key Message: The therapeutic utility of targeting FcγRs with recombinant Fcs is great and should be explored in human clinical trials for autoimmune diseases, such as RA.


Subject(s)
Arthritis, Experimental/etiology , Arthritis, Experimental/immunology , Biological Products/therapeutic use , Immunoglobulin Fc Fragments/therapeutic use , Receptors, Fc/immunology , Receptors, Fc/metabolism , Animals , Biological Products/pharmacology , Humans , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulins, Intravenous/pharmacology , Immunoglobulins, Intravenous/therapeutic use , Mice , Receptors, Fc/antagonists & inhibitors , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
4.
BMC Immunol ; 20(1): 44, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31801459

ABSTRACT

BACKGROUND: High-dose intravenous immunoglobulin (IVIg), and more recently, subcutaneously-delivered Ig (SCIg), are used to treat a variety of autoimmune diseases; however, there are challenges associated with product production, availability, access and efficacy. These challenges have provided incentives to develop a human recombinant Fc as a more potent alternative to IVIg and SCIg for the treatment of autoimmune diseases. Recently, a recombinant human IgG1 Fc hexamer (Fc-µTP-L309C) was shown to be more efficacious than IVIg in a variety of autoimmune mouse models. We have now examined its efficacy compared to IVIg and SCIg in the K/BxN mouse model of endogenous, chronic rheumatoid arthritis (RA). RESULT: Using the serum-transfer K/BxN model and the endogenous autoimmune model, amelioration of the arthritis was achieved. Effective treatment required high and frequent doses of IVIg, SCIg and Fc-µTP-L309C. However, Fc-µTP-L309C was efficacious at 10-fold lower doses that IVIg/SCIg. Also, arthritis could be prevented when Fc-µTP-L309C was given prior to onset of the arthritis in both the endogenous model and in the serum transfer model. CONCLUSIONS: Our results show that Fc-µTP-L309C is a powerful treatment for the prevention and amelioration of severe, chronic arthritis in a true autoimmune mouse model of RA. Thus, the K/BxN endogenous arthritis model should be useful for testing potential therapeutics for RA. Our findings provide rationale for further examination of the treatment efficacy of immunoglobulin-based therapeutics in rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulins, Intravenous/pharmacology , Immunologic Factors/pharmacology , Animals , Arthritis, Rheumatoid/pathology , Chronic Disease , Disease Models, Animal , Humans , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin G/chemistry , Immunoglobulins, Intravenous/administration & dosage , Immunologic Factors/administration & dosage , Mice , Treatment Outcome
5.
J Immunol ; 200(8): 2542-2553, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29531170

ABSTRACT

Activation of Fc receptors and complement by immune complexes is a common important pathogenic trigger in many autoimmune diseases and so blockade of these innate immune pathways may be an attractive target for treatment of immune complex-mediated pathomechanisms. High-dose IVIG is used to treat autoimmune and inflammatory diseases, and several studies demonstrate that the therapeutic effects of IVIG can be recapitulated with the Fc portion. Further, recent data indicate that recombinant multimerized Fc molecules exhibit potent anti-inflammatory properties. In this study, we investigated the biochemical and biological properties of an rFc hexamer (termed Fc-µTP-L309C) generated by fusion of the IgM µ-tailpiece to the C terminus of human IgG1 Fc. Fc-µTP-L309C bound FcγRs with high avidity and inhibited FcγR-mediated effector functions (Ab-dependent cell-mediated cytotoxicity, phagocytosis, respiratory burst) in vitro. In addition, Fc-µTP-L309C prevented full activation of the classical complement pathway by blocking C2 cleavage, avoiding generation of inflammatory downstream products (C5a or sC5b-9). In vivo, Fc-µTP-L309C suppressed inflammatory arthritis in mice when given therapeutically at approximately a 10-fold lower dose than IVIG, which was associated with reduced inflammatory cytokine production and complement activation. Likewise, administration of Fc-µTP-L309C restored platelet counts in a mouse model of immune thrombocytopenia. Our data demonstrate a potent anti-inflammatory effect of Fc-µTP-L309C in vitro and in vivo, likely mediated by blockade of FcγRs and its unique inhibition of complement activation.


Subject(s)
Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/immunology , Autoimmune Diseases/immunology , Complement System Proteins/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Receptors, Fc/immunology , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Line , Complement Activation/immunology , Humans , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Phagocytosis/immunology , Receptors, IgG/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...