Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Cell Rep ; 38(5): 110303, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108544

ABSTRACT

Across the animal kingdom, multivalency discriminates antibodies from all other immunoglobulin superfamily members. The evolutionary forces conserving multivalency above other structural hallmarks of antibodies remain, however, incompletely defined. Here, we engineer monovalent either Fc-competent or -deficient antibody formats to investigate mechanisms of protection of neutralizing antibodies (nAbs) and non-neutralizing antibodies (nnAbs) in virus-infected mice. Antibody bivalency enables the tethering of virions to the infected cell surface, inhibits the release of virions in cell culture, and suppresses viral loads in vivo independently of Fc gamma receptor (FcγR) interactions. In return, monovalent antibody formats either do not inhibit virion release and fail to protect in vivo or their protective efficacy is largely FcγR dependent. Protection in mice correlates with virus-release-inhibiting activity of nAb and nnAb rather than with their neutralizing capacity. These observations provide mechanistic insights into the evolutionary conservation of antibody bivalency and help refining correlates of nnAb protection for vaccine development.


Subject(s)
Antibodies, Viral/pharmacology , Antiviral Agents/pharmacology , HIV Antibodies/pharmacology , Receptors, Fc/drug effects , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Epitopes/drug effects , Epitopes/immunology , HIV Antibodies/immunology , Immunoglobulin G/drug effects , Immunoglobulin G/immunology , Mice, Inbred C57BL , Receptors, IgG/drug effects , Receptors, IgG/immunology
2.
Int J Mol Sci ; 22(16)2021 Aug 21.
Article in English | MEDLINE | ID: mdl-34445732

ABSTRACT

Infection with viruses, such as the lactate dehydrogenase-elevating virus (LDV), is known to trigger the onset of autoimmune anemia through the enhancement of the phagocytosis of autoantibody-opsonized erythrocytes by activated macrophages. Type I interferon receptor-deficient mice show enhanced anemia, which suggests a protective effect of these cytokines, partly through the control of type II interferon production. The development of anemia requires the expression of Fcγ receptors (FcγR) I, III, and IV. Whereas LDV infection decreases FcγR III expression, it enhances FcγR I and IV expression in wild-type animals. The LDV-associated increase in the expression of FcγR I and IV is largely reduced in type I interferon receptor-deficient mice, through both type II interferon-dependent and -independent mechanisms. Thus, the regulation of the expression of FcγR I and IV, but not III, by interferons may partly explain the exacerbating effect of LDV infection on anemia that results from the enhanced phagocytosis of IgG autoantibody-opsonized erythrocytes.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Arterivirus Infections/immunology , Interferons/metabolism , Lactate dehydrogenase-elevating virus/immunology , Receptors, IgG/metabolism , Anemia, Hemolytic, Autoimmune/virology , Animals , Arterivirus Infections/virology , Host-Pathogen Interactions , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis
3.
Cancers (Basel) ; 13(10)2021 May 14.
Article in English | MEDLINE | ID: mdl-34069226

ABSTRACT

Promising strategies for maximizing IgG effector functions rely on the introduction of natural and non-immunogenic modifications. The Fc domain of IgG antibodies contains an N-linked oligosaccharide at position 297. Human IgG antibodies lacking the core fucose in this glycan have enhanced binding to human (FcγR) IIIa/b, resulting in enhanced antibody dependent cell cytotoxicity and phagocytosis through these receptors. However, it is not yet clear if glycan-enhancing modifications of human IgG translate into more effective treatment in mouse models. We generated humanized hIgG1-TA99 antibodies with and without core-fucose. C57Bl/6 mice that were injected intraperitoneally with B16F10-gp75 mouse melanoma developed significantly less metastasis outgrowth after treatment with afucosylated hIgG1-TA99 compared to mice treated with wildtype hhIgG1-TA99. Afucosylated human IgG1 showed stronger interaction with the murine FcγRIV, the mouse orthologue of human FcγRIIIa, indicating that this glycan change is functionally conserved between the species. In agreement with this, no significant differences were observed in tumor outgrowth in FcγRIV-/- mice treated with human hIgG1-TA99 with or without the core fucose. These results confirm the potential of using afucosylated therapeutic IgG to increase their efficacy. Moreover, we show that afucosylated human IgG1 antibodies act across species, supporting that mouse models can be suitable to test afucosylated antibodies.

4.
Cancer Cell ; 37(6): 850-866.e7, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32442402

ABSTRACT

Anti-CD40 monoclonal antibodies (mAbs) comprise agonists and antagonists, which display promising therapeutic activities in cancer and autoimmunity, respectively. We previously showed that epitope and isotype interact to deliver optimal agonistic anti-CD40 mAbs. The impact of Fc engineering on antagonists, however, remains largely unexplored. Here, we show that clinically relevant antagonists used for treating autoimmune conditions can be converted into potent FcγR-independent agonists with remarkable antitumor activity by isotype switching to hIgG2. One antagonist is converted to a super-agonist with greater potency than previously reported highly agonistic anti-CD40 mAbs. Such conversion is dependent on the unique disulfide bonding properties of the hIgG2 hinge. This investigation highlights the transformative capacity of the hIgG2 isotype for converting antagonists to agonists to treat cancer.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD40 Antigens/immunology , CD40 Ligand/immunology , Dendritic Cells/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin G/immunology , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dendritic Cells/drug effects , Immunoglobulin Class Switching/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Receptors, IgE/physiology , Receptors, IgG/physiology , Thymus Neoplasms/drug therapy , Thymus Neoplasms/immunology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
5.
Sci Rep ; 10(1): 3933, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32127568

ABSTRACT

The use of Trastuzumab (Herceptin), a monoclonal antibody (mAb) targeting HER2/neu, results in an increased median survival in Her2+ breast cancer patients. The tumour mutational burden and the presence of tumour infiltrating lymphocytes (TILs) clearly correlate with response to trastuzumab. Here, we investigated if the immunogenicity of the transplantable rat-neu+ tumour cell line (TUBO) derived from a BALB/c-NeuT primary tumour is associated with the response to anti-neu mAb therapy. We compared the TUBO tumour outgrowth and tumour infiltrating T cells in isogenic (BALB/c-NeuT) and non-isogenic (WT BALB/c) recipient mice. Furthermore, therapeutic efficacy of anti-neu mAb and the contribution of T cells were examined in both mouse strains. The outgrowth of untreated tumours was significantly better in BALB/c-NeuT than WT BALB/c mice. Moreover, tumour infiltrating T cells were more abundantly present in WT BALB/c than BALB/c-NeuT mice, showing that the TUBO tumour was more immunogenic in WT BALB/c mice. In TUBO tumour bearing WT BALB/c mice, anti-neu mAb therapy resulted in an increase of tumour infiltrating T cells and long-term survival. When T cells were depleted, this strong anti-tumour effect was reduced to an outgrowth delay. In contrast, in TUBO tumour bearing BALB/c-NeuT mice, treatment with anti-neu mAb resulted only in tumour outgrowth delay, both in the presence and absence of T cells. We concluded that in immunogenic tumours the response to anti-neu mAb therapy is enhanced by additional T cell involvement compared to the response to anti-neu mAb in non-immunogenic tumours.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , T-Lymphocytes/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Female , Flow Cytometry , Lapatinib/therapeutic use , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred BALB C , Quinolines/therapeutic use , Rats , Receptor, ErbB-2/genetics , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Trastuzumab/therapeutic use
6.
Gut ; 69(6): 1053-1063, 2020 06.
Article in English | MEDLINE | ID: mdl-31506328

ABSTRACT

OBJECTIVE: Macrophage interleukin (IL)-10 signalling plays a critical role in the maintenance of a regulatory phenotype that prevents the development of IBD. We have previously found that anti-tumour necrosis factor (TNF) monoclonal antibodies act through Fcγ-receptor (FcγR) signalling to promote repolarisation of proinflammatory intestinal macrophages to a CD206+ regulatory phenotype. The role of IL-10 in anti-TNF-induced macrophage repolarisation has not been examined. DESIGN: We used human peripheral blood monocytes and mouse bone marrow-derived macrophages to study IL-10 production and CD206+ regulatory macrophage differentiation. To determine whether the efficacy of anti-TNF was dependent on IL-10 signalling in vivo and in which cell type, we used the CD4+CD45Rbhigh T-cell transfer model in combination with several genetic mouse models. RESULTS: Anti-TNF therapy increased macrophage IL-10 production in an FcγR-dependent manner, which caused differentiation of macrophages to a more regulatory CD206+ phenotype in vitro. Pharmacological blockade of IL-10 signalling prevented the induction of these CD206+ regulatory macrophages and diminished the therapeutic efficacy of anti-TNF therapy in the CD4+CD45Rbhigh T-cell transfer model of IBD. Using cell type-specific IL-10 receptor mutant mice, we found that IL-10 signalling in macrophages but not T cells was critical for the induction of CD206+ regulatory macrophages and therapeutic response to anti-TNF. CONCLUSION: The therapeutic efficacy of anti-TNF in resolving intestinal inflammation is critically dependent on IL-10 signalling in macrophages.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Interleukin-10/metabolism , Macrophages/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Animals , Antibodies, Monoclonal , Crohn Disease/drug therapy , Crohn Disease/metabolism , Disease Models, Animal , Female , Humans , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Macrophages/drug effects , Male , Mice , Mice, Knockout , Middle Aged , Signal Transduction/drug effects , Young Adult
7.
Front Immunol ; 10: 2061, 2019.
Article in English | MEDLINE | ID: mdl-31681256

ABSTRACT

FcγRIIb is the only inhibitory Fc receptor and controls many aspects of immune and inflammatory responses. The observation 19 years ago that FcγRIIb-/- mice generated by gene targeting in 129 derived ES cells developed severe lupus like disease when backcrossed more than 7 generations into C57BL/6 background initiated extensive research on the functional understanding of this strong autoimmune phenotype. The genomic region in the distal part of Chr1 both in human and mice in which the FcγR gene cluster is located shows a high level of complexity in relation to the susceptibility to SLE. Specific haplotypes of closely linked genes including the FcγRIIb and Slamf genes are associated with increased susceptibility to SLE both in mice and human. Using forward and reverse genetic approaches including in human GWAS and in mice congenic strains, KO mice (germline and cell type specific, on different genetic background), knockin mice, overexpressing transgenic mice combined with immunological models such as adoptive transfer of B cells from Ig transgenic mice the involved genes and the causal mutations and their associated functional alterations were analyzed. In this review the results of this 19 years extensive research are discussed with a focus on (genetically modified) mouse models.


Subject(s)
Genetic Predisposition to Disease , Lupus Erythematosus, Systemic , Models, Immunological , Receptors, IgG , Animals , Disease Models, Animal , Genome-Wide Association Study , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Knockout , Multigene Family , Receptors, IgG/genetics , Receptors, IgG/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology
8.
Int Immunol ; 31(11): 687-696, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31063541

ABSTRACT

AbstractMonocytes are evolutionally conserved innate immune cells that play essential roles for the protection of the host against pathogens and also produce several inflammatory cytokines. Thus, the aberrant functioning of monocytes may affect not only host defense but also the development of inflammatory diseases. Monocytes are a heterogeneous population with phenotypical and functional differences. Most recent studies have shown that monocytes are divided into three subsets, namely classical, intermediate and non-classical subsets, both in humans and mice. Accumulating evidence showed that monocyte activation is associated with the disease progression in autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). However, it remains to be determined how monocytes contribute to the disease process and which subset is involved. In this review, we discuss the pathogenic role of monocyte subsets in SLE and RA on the basis of current studies by ourselves and others to shed light on the suitability of monocyte-targeted therapies in these diseases.


Subject(s)
Arthritis, Rheumatoid/immunology , Lupus Erythematosus, Systemic/immunology , Monocytes/immunology , Animals , Humans
9.
Viruses ; 11(2)2019 02 08.
Article in English | MEDLINE | ID: mdl-30744065

ABSTRACT

Dendritic cells (DCs) express Fcγ receptors (FcγRs) for the binding immune complexes (ICs) consisting of IgG and antigens (Ags). IC⁻FcγR interactions have been demonstrated to enhance activation and antigen-presenting functions of DCs. Utilizing Friend virus (FV), an oncogenic mouse retrovirus, we investigated the effect of IgG-opsonization of retroviral particles on the infection of DCs and the subsequent presentation of viral antigens by DCs to virus-specific CD8 T cells. We found that opsonization by virus-specific non-neutralizing IgG abrogated DC infection and as a consequence significantly reduced the capacity of DCs to activate virus-specific CD8 T cells. Effects of IgG-opsonization were mediated by the high-affinity FcγR type I, CD64, expressed on DCs. Our results suggest that different opsonization patterns on the retroviral surface modulate infection and antigen-presenting functions of DCs, whereby, in contrast to complement, IgG reduces the capacity of DCs to activate cytotoxic T cell (CTL) responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Friend murine leukemia virus/immunology , Lymphocyte Activation , Receptors, IgG/immunology , Animals , Antigen Presentation , Antigen-Antibody Complex/immunology , Dendritic Cells/virology , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Receptors, IgG/genetics
10.
Eur J Immunol ; 49(4): 626-637, 2019 04.
Article in English | MEDLINE | ID: mdl-30636035

ABSTRACT

Infection of C57BL/6 mice with lymphocytic choriomeningitis virus (LCMV) strain Armstrong (Arm) induces an acute infection with rapid virus clearance by CD8+ T cells independently of CD4+ T cell help. Residual viral antigen may, however, persist for a prolonged time. Here, we demonstrate that mice that had been transiently depleted of CD4+ T cells during acute LCMV Arm infection generated high levels of virus-specific IgG antibodies (Ab) after viral clearance. Robust induction of LCMV-specific IgG after transient CD4+ T cell depletion was dependent on Fcγ receptors but not on the complement receptors CD21/CD35. In contrast to the potent production of LCMV-specific IgG, the generation of LCMV-specific isotype-switched memory B cells after transient CD4+ T cell depletion was considerably reduced. Moreover, mice depleted of CD4+ T cells during acute infection were strongly impaired in generating a secondary LCMV-specific B cell response upon LCMV rechallenge. In conclusion, our data indicate that LCMV antigen depots after viral clearance were capable of inducing high levels of virus-specific IgG. They failed, however, to induce robust virus-specific B cell memory revealing a previously unappreciated dichotomy of specific Ab production and memory cell formation after priming with residual antigen.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Biomarkers , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunologic Memory , Immunophenotyping , Lymphocyte Depletion , Lymphocytic Choriomeningitis/virology , Mice , Mice, Knockout , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocytes, Cytotoxic/immunology
11.
Mol Cancer Ther ; 18(2): 312-322, 2019 02.
Article in English | MEDLINE | ID: mdl-30381448

ABSTRACT

Immunotherapy of cancer with CD3-targeting bispecific antibodies (CD3 bsAb) is a fast developing field, and multiple tumor-associated antigens (TAA) are evaluated for hematologic and solid malignancies. The efficacy of these CD3 bsAb is usually examined in xenograft mouse tumor models with human T cells or in genetically engineered mouse models, where human TAA are introduced. These models often fail to fully recapitulate the natural tumor environment, especially for solid cancers, because of interspecies differences. Here, we investigated the systemic and intratumoral effects of a mouse CD3 bsAb in a fully immune-competent mouse melanoma model. Systemic administration of 0.5 mg/kg antibody induced a brief overall T-cell activation that was selectively sustained in the tumor microenvironment for several days. A fast subsequent influx of inflammatory macrophages into the tumor microenvironment was observed, followed by an increase in the number of CD4+ and CD8+ T cells. Although the capacity to directly kill melanoma cells in vitro was very modest, optimal tumor elimination was observed in vivo, even in the absence of CD8+ T cells, implying a redundancy in T-cell subsets for therapeutic efficacy. Finally, we took advantage of the full immune competence of our mouse model and tested immune memory induction. Despite a strong initial immunity against melanoma, treatment with the CD3 bsAb did not install protective memory responses. The observed mechanisms of action revealed in this immune-competent mouse model might form a rational basis for combinatorial approaches.


Subject(s)
Antibodies, Bispecific/administration & dosage , CD3 Complex/antagonists & inhibitors , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Melanoma/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Female , Humans , Immunologic Memory , Lymphocyte Activation , Melanoma/immunology , Mice , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
12.
Int J Cancer ; 144(2): 345-354, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30259976

ABSTRACT

Immunomodulatory antibodies blocking interactions of coinhibitory receptors to their ligands such as CTLA-4, PD1 and PD-L1 on immune cells have shown impressive therapeutic efficacy in clinical studies. The therapeutic effect of these antibodies is mainly mediated by reactivating antitumor T cell immune responses. Detailed analysis of anti-CTLA4 antibody therapy revealed that an optimal therapeutic efficacy also requires binding to Fc receptors for IgG, FcγR, mediating depletion of intratumoral regulatory T cells. Here, we investigated the role of Fc binding in anti-PD-L1 antibody therapy in the MC38 C57BL/6 and CT26 BALB/c colon adenocarcinoma tumor models. In the MC38 tumor model, all IgG subclasses anti-PD-L1 showed similar therapeutic efficacy when compared to each other in either wild-type mice or in mice deficient for all FcγR. In contrast, in the CT26 tumor model, anti-PD-L1 mIgG2a, the IgG subclass with the highest affinity for activating FcγR, showed stronger therapeutic efficacy than other IgG subclasses. This was associated with a reduction of a myeloid cell subset with high expression of PD-L1 in the tumor microenvironment. This subclass preference for mIgG2a was lost in C57BL/6 × BALB/c F1 mice, indicating that the genetic background of the host may determine the additional clinical benefit of the high affinity antibody subclasses. Based on these data, we conclude that FcγR are not crucial for anti-PD-L1 antibody therapy but might play a role in some tumor models.


Subject(s)
Adenocarcinoma , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Colonic Neoplasms , Receptors, IgG , Animals , Antibodies, Monoclonal , Disease Models, Animal , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
13.
J Immunol ; 201(12): 3741-3749, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30397036

ABSTRACT

Therapy with tumor-specific Abs is common in the clinic but has limited success against solid malignancies. We aimed at improving the efficacy of this therapy by combining a tumor-specific Ab with immune-activating compounds. In this study, we demonstrate in the aggressive B16F10 mouse melanoma model that concomitant application of the anti-TRP1 Ab (clone TA99) with TLR3-7/8 or -9 ligands, and IL-2 strongly enhanced tumor control in a therapeutic setting. Depletion of NK cells, macrophages, or CD8+ T cells all mitigated the therapeutic response, showing a coordinated immune rejection by innate and adaptive immune cells. FcγRs were essential for the therapeutic effect, with a dominant role for FcγRI and a minor role for FcγRIII and FcγRIV. FcγR expression on NK cells and granulocytes was dispensable, indicating that other tumoricidal functions of NK cells were involved and implicating that FcγRI, -III, and -IV exerted their activity on macrophages. Indeed, F4/80+Ly-6C+ inflammatory macrophages in the tumor microenvironment displayed high levels of these receptors. Whereas administration of the anti-TRP1 Ab alone reduced the frequency of these macrophages, the combination with a TLR agonist retained these cells in the tumor microenvironment. Thus, the addition of innate stimulatory compounds, such as TLR ligands, to tumor-specific Ab therapy could greatly enhance its efficacy in solid cancers via optimal exploitation of FcγRs.


Subject(s)
Antibodies/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy/methods , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Macrophages/immunology , Melanoma/therapy , Receptors, IgG/metabolism , Animals , Antigens, Neoplasm/immunology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Immunization , Male , Melanoma/immunology , Melanoma, Experimental , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases/immunology , Receptors, IgG/genetics , Toll-Like Receptors/agonists
14.
Immunity ; 49(5): 958-970.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30446386

ABSTRACT

The costimulatory receptor 4-1BB is expressed on activated immune cells, including activated T cells. Antibodies targeting 4-1BB enhance the proliferation and survival of antigen-stimulated T cells in vitro and promote CD8 T cell-dependent anti-tumor immunity in pre-clinical cancer models. We found that T regulatory (Treg) cells infiltrating human or murine tumors expressed high amounts of 4-1BB. Intra-tumoral Treg cells were preferentially depleted by anti-4-1BB mAbs in vivo. Anti-4-1BB mAbs also promoted effector T cell agonism to promote tumor rejection. These distinct mechanisms were competitive and dependent on antibody isotype and FcγR availability. Administration of anti-4-1BB IgG2a, which preferentially depletes Treg cells, followed by either agonistic anti-4-1BB IgG1 or anti-PD-1 mAb augmented anti-tumor responses in multiple solid tumor models. An antibody engineered to optimize both FcγR-dependent Treg cell depleting capacity and FcγR-independent agonism delivered enhanced anti-tumor therapy. These insights into the effector mechanisms of anti-4-1BB mAbs lay the groundwork for translation into the clinic.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunomodulation/drug effects , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors , Animals , Gene Expression , Humans , Immunoglobulin G/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Knockout , Neoplasms/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
15.
J Immunol ; 201(11): 3199-3210, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30373853

ABSTRACT

C57BL/6 (B6).FcγRIIb-/- Yaa mice spontaneously develop lethal lupus nephritis. To define the cell type-specific role of FcγRIIb in Yaa-associated lupus, we established B cell- (CD19Cre Yaa), myeloid cell- (C/EBPαCre Yaa), and dendritic cell- (DC) (CD11cCre Yaa) specific FcγRIIb-deficient B6.Yaa mouse strains. CD19Cre Yaa mice developed milder lupus than B6.FcγRIIb-/- Yaa mice, indicating that FcγRIIb deficiency on B cells is not sufficient for the development of severe disease. Surprisingly, C/EBPαCre Yaa mice also showed autoantibody production and mild lupus similar to that in CD19Cre Yaa mice, whereas CD11cCre Yaa mice stayed disease free. These observations indicate that FcγRIIb deficiency in B cells and myeloid cells, but not DCs, contributes to the severe disease in B6.FcγRIIb-/- Yaa mice. Flow cytometric analysis showed that the frequency of peripheral Gr-1- but not Gr-1+ monocyte was increased in B6.FcγRIIb-/- Yaa and C/EBPαCre Yaa but not CD19Cre Yaa mice, suggesting a link between FcγRIIb deficiency on myeloid cells and the high frequency of Gr-1- monocytes. RNA sequencing revealed that compared with Gr-1+ monocytes, Gr-1- monocytes expressed higher levels of the B cell-stimulating cytokines BSF-3, IL-10, and IL-1ß, the DC markers CD11c, CD83, and Adamdec1, and the antiapoptotic factors Bcl2 and Bcl6. In conclusion, in Yaa-associated lupus nephritis, FcγRIIb on B cells and myeloid cells modulates B cell activation via different but synergistic pathways. Gr-1- monocytes are the most likely candidate myeloid cells involved.


Subject(s)
B-Lymphocytes/physiology , Dendritic Cells/physiology , Lupus Nephritis/immunology , Myeloid Cells/physiology , Receptors, IgG/metabolism , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Autoantibodies/metabolism , Cells, Cultured , Disease Susceptibility , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, IgG/genetics
16.
Arthritis Res Ther ; 20(1): 80, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720243

ABSTRACT

BACKGROUND: Osteoclast-mediated bone erosion is a central feature of rheumatoid arthritis (RA). Immune complexes, present in a large percentage of patients, bind to Fcγ receptors (FcγRs), thereby modulating the activity of immune cells. In this study, we investigated the contribution of FcγRs, and FcγRIV in particular, during antigen-induced arthritis (AIA). METHODS: AIA was induced in knee joints of wild-type (WT), FcγRI,II,III-/-, and FcγRI,II,III,IV-/- mice. Bone destruction, numbers of tartrate-resistant acid phosphatase-positive (TRAP+) osteoclasts, and inflammation were evaluated using histology; expression of the macrophage marker F4/80, neutrophil marker NIMPR14, and alarmin S100A8 was evaluated using immunohistochemistry. The percentage of osteoclast precursors in the bone marrow was determined using flow cytometry. In vitro osteoclastogenesis was evaluated with TRAP staining, and gene expression was assessed using real-time PCR. RESULTS: FcγRI,II,III,IV-/- mice showed decreased bone erosion compared with WT mice during AIA, whereas both the humoral and cellular immune responses against methylated bovine serum albumin were not impaired in FcγRI,II,III,IV-/- mice. The percentage of osteoclast precursors in the bone marrow of arthritic mice and their ability to differentiate into osteoclasts in vitro were comparable between FcγRI,II,III,IV-/- and WT mice. In line with these observations, numbers of TRAP+ osteoclasts on the bone surface during AIA were comparable between the two groups. Inflammation, a process that strongly activates osteoclast activity, was reduced in FcγRI,II,III,IV-/- mice, and of note, mainly decreased numbers of neutrophils were present in the joint. In contrast to FcγRI,II,III,IV-/- mice, AIA induction in knee joints of FcγRI,II,III-/- mice resulted in increased bone erosion, inflammation, and numbers of neutrophils, suggesting a crucial role for FcγRIV in the joint pathology by the recruitment of neutrophils. Finally, significant correlations were found between bone erosion and the number of neutrophils present in the joint as well as between bone erosion and the number of S100A8-positive cells, with S100A8 being an alarmin strongly produced by neutrophils that stimulates osteoclast resorbing activity. CONCLUSIONS: FcγRs play a crucial role in the development of bone erosion during AIA by inducing inflammation. In particular, FcγRIV mediates bone erosion in AIA by inducing the influx of S100A8/A9-producing neutrophils into the arthritic joint.


Subject(s)
Arthritis, Experimental/immunology , Bone and Bones/immunology , Calgranulin A/immunology , Calgranulin B/immunology , Neutrophils/immunology , Receptors, IgG/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology , Calgranulin A/metabolism , Calgranulin B/metabolism , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Osteoclasts/immunology , Osteoclasts/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Tartrate-Resistant Acid Phosphatase/immunology , Tartrate-Resistant Acid Phosphatase/metabolism
17.
J Immunol ; 200(8): 2615-2626, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29523656

ABSTRACT

By their interaction with IgG immune complexes, FcγR and complement link innate and adaptive immunity, showing functional redundancy. In complement-deficient mice, IgG downstream effector functions are often impaired, as well as adaptive immunity. Based on a variety of model systems using FcγR-knockout mice, it has been concluded that FcγRs are also key regulators of innate and adaptive immunity; however, several of the model systems underpinning these conclusions suffer from flawed experimental design. To address this issue, we generated a novel mouse model deficient for all FcγRs (FcγRI/II/III/IV-/- mice). These mice displayed normal development and lymphoid and myeloid ontogeny. Although IgG effector pathways were impaired, adaptive immune responses to a variety of challenges, including bacterial infection and IgG immune complexes, were not. Like FcγRIIb-deficient mice, FcγRI/II/III/IV-/- mice developed higher Ab titers but no autoantibodies. These observations indicate a redundant role for activating FcγRs in the modulation of the adaptive immune response in vivo. We conclude that FcγRs are downstream IgG effector molecules with a restricted role in the ontogeny and maintenance of the immune system, as well as the regulation of adaptive immunity.

18.
Front Immunol ; 9: 3097, 2018.
Article in English | MEDLINE | ID: mdl-30671062

ABSTRACT

Non-invasive imaging technologies to visualize the location and functionality of T cells are of great value in immunology. Here, we describe the design and generation of a transgenic mouse in which all T cells constitutively express green-emitting click-beetle luciferase (CBG99) while expression of the red-emitting firefly luciferase (PpyRE9) is induced by Nuclear Factor of Activated T cells (NFAT) such as during T cell activation, which allows multicolor bioluminescence imaging of T cell location and function. This dual-luciferase mouse, which we named TbiLuc, showed high constitutive luciferase expression in lymphoid organs such as lymph nodes and the spleen. Ex vivo purified CD8+ and CD4+ T cells both constitutively expressed luciferase, whereas B cells showed no detectable signal. We cross-bred TbiLuc mice to T cell receptor-transgenic OT-I mice to obtain luciferase-expressing naïve CD8+ T cells with defined antigen-specificity. TbiLuc*OT-I T cells showed a fully antigen-specific induction of the T cell activation-dependent luciferase. In vaccinated mice, we visualized T cell localization and activation in vaccine-draining lymph nodes with high sensitivity using two distinct luciferase substrates, D-luciferin and CycLuc1, of which the latter specifically reacts with the PpyRE9 enzyme. This dual-luciferase T cell reporter mouse can be applied in many experimental models studying the location and functional state of T cells.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Luciferases, Firefly/metabolism , Luminescent Measurements/methods , Animals , Benzothiazoles/metabolism , Cell Line , Coleoptera/enzymology , Dendritic Cells/immunology , Epitopes , Firefly Luciferin/chemical synthesis , Firefly Luciferin/metabolism , Lymph Nodes/cytology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , NFATC Transcription Factors/metabolism , Ovalbumin/immunology , Spleen/cytology , Vaccination
19.
Gastroenterology ; 153(5): 1351-1362.e4, 2017 11.
Article in English | MEDLINE | ID: mdl-28756234

ABSTRACT

BACKGROUND & AIMS: Although tumor necrosis factor (TNF) antagonists reduce many clinical features of inflammatory bowel disease, complete mucosal healing occurs in fewer than 50% of patients. The Fc-region of monoclonal antibodies against TNF has immunosuppressive properties via effects on macrophage polarization. We examined the interaction between the anti-TNF Fc-region and Fcγ receptors (FcγR), and whether the absence of the Fc core fucose (which increases binding to FcγRIIIa) increases the efficacy of anti-TNF in mice with colitis. METHODS: We generated Rag1-/- mice that lack all activating FcγRs (FcγRI, FcγRIII, and FcγRIV; called FcγR-/-Rag1-/- mice). We produced hypo-fucosylated antibodies against mouse and human TNF (adalimumab). Colitis was induced in mice by transfer of CD4+CD45RBhi to FcγR-/-Rag1-/- or Rag1-/- littermates; mice were given different antibodies against TNF or isotype (control) antibodies and disease activity index scores were determined. Colon tissues were collected and analyzed by histology. Human peripheral blood mononuclear cells (PBMCs) were isolated from blood of healthy donors. T-cell proliferation and proportions of CD206+ (immune regulatory) macrophages were measured in mixed lymphocyte reactions. Human PBMCs were genotyped for FCGR3A158 (the FcγRIIIa-158F allotype displays a lower Fc binding affinity) using the TaqMan single nucleotide polymorphism genotype assay. RESULTS: Rag1-/- mice with colitis given anti-TNF had near complete mucosal healing and Rag1-/- mice given an isotype control antibody developed severe colitis. In contrast, FcγR-/-Rag1-/- mice were refractory to the effects of anti-TNF: their histological colitis scores were as severe as those from FcγR-/-Rag1-/- mice given a control antibody. Colons from Rag1-/- mice that received anti-TNF had an increased number of CD206+ macrophages compared with Rag1-/- mice given control antibody; in FcγR-/-Rag1-/- mice given anti-TNF these numbers were as low as FcγR-/-Rag1-/- given the control antibody. In human PBMCs, anti-TNF increased the number of CD206+ macrophages: this required expression of FcγRIIIa; numbers of these cells were reduced in PBMCs with the low-affinity FcγRIIIa-158F genotype. A hypo-fucosylated form of adalimumab bound human FcγRIIIa with a higher affinity than control adalimumab. When hypo-fucosylated adalimumab was added to PBMCs, a larger number of CD206+ macrophages formed and T-cell proliferation was reduced, compared with addition of a control adalimumab. Hypo-fucosylated adalimumab increased the number of CD206+ macrophages in PMBCs that expressed the low-affinity FcγRIIIa. In mice with colitis, hypo-fucosylated anti-TNF significantly increased the number of CD206+ macrophages in the colon compared with control anti-TNF and was more effective in reducing colitis severity as measured by histology. CONCLUSIONS: In a study of the in vitro and in vivo mechanisms of anti-TNF, we found FcγR engagement by anti-TNF to be required for reduction of colitis in mice and development of CD206+ macrophages. A hypo-fucosylated form of anti-TNF binds FcγRIIIa with higher affinity and induces development of CD206+ macrophages in human PBMCs, especially PBMCs that express low-affinity FcγRIIIa. Hypo-fucosylated anti-TNF might be more effective in patients with inflammatory bowel disease.


Subject(s)
Adalimumab/pharmacology , Antibodies, Monoclonal/pharmacology , Colitis/drug therapy , Colon/drug effects , Immunosuppressive Agents/pharmacology , Intestinal Mucosa/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adoptive Transfer , Animals , Cell Proliferation/drug effects , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, IgG/deficiency , Receptors, IgG/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Time Factors , Tumor Necrosis Factor-alpha/immunology , Wound Healing/drug effects
20.
J Immunol ; 198(11): 4235-4243, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28432146

ABSTRACT

Dendritic cells (DCs) are specialized in Ag engulfment via a wide variety of uptake receptors on their cell surface. In the present study we investigated Ag uptake and presentation of in vivo-formed Ag-Ab complexes by i.v. injecting mice with Ag-specific Abs followed by the cognate Ag. We show by this natural Ab-mediated Ag targeting system that uptake by splenic APC subsets is severely hampered in mice lacking complement factor C1q (C1qa-/-). Moreover, no detectable Ag cross-presentation by CD8α+ DCs from C1qa-/- mice was found. On the contrary, Ag uptake was not hampered by APCs in FcγRI/II/III/IV-deficient (FcγR quadruple-/-) mice, and the cross-presentation ability of CD8α+ DCs was not affected. In conclusion, we show that C1q rather than FcγRs controls the Ab-mediated Ag uptake and its presentation by spleen APC subsets to T cells.


Subject(s)
Antigen Presentation , Antigen-Antibody Complex/immunology , Complement C1q/immunology , Dendritic Cells/immunology , Adaptive Immunity , Animals , CD8 Antigens/immunology , Complement C1q/deficiency , Complement C1q/genetics , Cross-Priming , Mice , Mice, Inbred C57BL , Receptors, IgG/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...