Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Nat Immunol ; 19(3): 302-314, 2018 03.
Article in English | MEDLINE | ID: mdl-29476184

ABSTRACT

The quantification and characterization of circulating immune cells provide key indicators of human health and disease. To identify the relative effects of environmental and genetic factors on variation in the parameters of innate and adaptive immune cells in homeostatic conditions, we combined standardized flow cytometry of blood leukocytes and genome-wide DNA genotyping of 1,000 healthy, unrelated people of Western European ancestry. We found that smoking, together with age, sex and latent infection with cytomegalovirus, were the main non-genetic factors that affected variation in parameters of human immune cells. Genome-wide association studies of 166 immunophenotypes identified 15 loci that showed enrichment for disease-associated variants. Finally, we demonstrated that the parameters of innate cells were more strongly controlled by genetic variation than were those of adaptive cells, which were driven by mainly environmental exposure. Our data establish a resource that will generate new hypotheses in immunology and highlight the role of innate immunity in susceptibility to common autoimmune diseases.


Subject(s)
Genetic Variation/immunology , Immunity, Innate/genetics , Adaptive Immunity/genetics , Adult , Aged , Female , Genome-Wide Association Study , Humans , Immunophenotyping , Male , Middle Aged , Young Adult
3.
Eur J Immunol ; 53(12): e2350454, 2023 12.
Article in English | MEDLINE | ID: mdl-37621208

ABSTRACT

Human immune system (HIS) mice provide a model to study human immune responses in vivo. Currently available HIS mouse models may harbor mouse Fc Receptor (FcR)-expressing cells that exert potent effector functions following administration of human Ig. Previous studies showed that the ablation of the murine FcR gamma chain (FcR-γ) results in loss of antibody-dependent cellular cytotoxicity and antibody-dependent cellular phagocytosis in vivo. We created a new FcR-γ-deficient HIS mouse model to compare host (mouse) versus graft (human) effects underlying antibody-mediated immune responses in vivo. FcR-γ-deficient HIS recipients lack expression and function of mouse activating FcRs and can be stably and robustly reconstituted with human immune cells. By screening blood B-cell depletion by rituximab Ig variants, we found that human FcγRs-mediated IgG1 effects, whereas mouse activating FcγRs were dominant in IgG4 effects. Complement played a role as an IgG1 variant (IgG1 K322A) lacking complement binding activity was largely ineffective. Finally, we provide evidence that FcγRIIIA on human NK cells could mediate complement-independent B-cell depletion by IgG1 K322A. We anticipate that our FcR-γ-deficient HIS model will help clarify mechanisms of action of exogenous administered human antibodies in vivo.


Subject(s)
Receptors, Fc , Receptors, IgG , Humans , Mice , Animals , Receptors, IgG/genetics , Immunoglobulin G , Antibody-Dependent Cell Cytotoxicity , Macrophages , Complement System Proteins , Adaptive Immunity
4.
Cytometry A ; 105(2): 124-138, 2024 02.
Article in English | MEDLINE | ID: mdl-37751141

ABSTRACT

Flow cytometry is the method of choice for immunophenotyping in the context of clinical, translational, and systems immunology studies. Among the latter, the Milieu Intérieur (MI) project aims at defining the boundaries of a healthy immune response to identify determinants of immune response variation. MI used immunophenotyping of a 1000 healthy donor cohort by flow cytometry as a principal outcome for immune variance at steady state. New generation spectral cytometers now enable high-dimensional immune cell characterization from small sample volumes. Therefore, for the MI 10-year follow up study, we have developed two high-dimensional spectral flow cytometry panels for deep characterization of innate and adaptive whole blood immune cells (35 and 34 fluorescent markers, respectively). We have standardized the protocol for sample handling, staining, acquisition, and data analysis. This approach enables the reproducible quantification of over 182 immune cell phenotypes at a single site. We have applied the protocol to discern minor differences between healthy and patient samples and validated its value for application in immunomonitoring studies. Our protocol is currently used for characterization of the impact of age and environmental factors on peripheral blood immune phenotypes of >400 donors from the initial MI cohort.


Subject(s)
Follow-Up Studies , Humans , Immunophenotyping , Phenotype , Flow Cytometry/methods
5.
J Immunol ; 209(7): 1243-1251, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36165182

ABSTRACT

Mouse models of active systemic anaphylaxis rely predominantly on IgG Abs forming IgG-allergen immune complexes that induce IgG receptor-expressing neutrophils and monocytes/macrophages to release potent mediators, leading to systemic effects. Whether anaphylaxis initiates locally or systemically remains unknown. In this study, we aimed at identifying the anatomical location of IgG-allergen immune complexes during anaphylaxis. Active systemic anaphylaxis was induced following immunization with BSA and i.v. challenge with fluorescently labeled BSA. Ag retention across different organs was examined using whole-body fluorescence imaging, comparing immunized and naive animals. Various mouse models and in vivo deletion strategies were employed to determine the contribution of IgG receptors, complement component C1q, myeloid cell types, and anaphylaxis mediators. We found that following challenge, Ag diffused systemically, but specifically accumulated in the lungs of mice sensitized to that Ag, where it formed large Ab-dependent aggregates in the vasculature. Ag retention in the lungs did not rely on IgG receptors, C1q, neutrophils, or macrophages. IgG2a-mediated, but neither IgG1- nor IgG2b-mediated, passive systemic anaphylaxis led to Ag retention in the lung. Neutrophils and monocytes significantly accumulated in the lungs after challenge and captured high amounts of Ag, which led to downmodulation of surface IgG receptors and triggered their activation. Thus, within minutes of systemic injection in sensitized mice, Ag formed aggregates in the lung and liver vasculature, but accumulated specifically and dose-dependently in the lung. Neutrophils and monocytes recruited to the lung captured Ag and became activated. However, Ag aggregation in the lung vasculature was not necessary for anaphylaxis induction.


Subject(s)
Anaphylaxis , Allergens , Animals , Antigen-Antibody Complex , Complement C1q , Disease Models, Animal , Immunoglobulin G , Lung , Mice , Mice, Inbred C57BL , Receptors, Complement , Receptors, IgG
6.
Eur J Immunol ; 52(5): 753-759, 2022 05.
Article in English | MEDLINE | ID: mdl-35133670

ABSTRACT

IgG is the predominant antibody class generated during infections and used for the generation of therapeutic antibodies. Antibodies are mainly characterized in or generated from animal models that support particular infections, respond to particular antigens or allow the generation of hybridomas. Due to the availability of numerous transgenic mouse models and the ease of performing bioassays with human blood cells in vitro, most antibodies from species other than mice and humans are tested in vitro using human cells and/or in vivo using mice. In this process, it is expected, but not yet systematically documented, that IgG from these species interact with human or mouse IgG receptors (FcγRs). In this study, we undertook a systematic assessment of binding specificities of IgG from various species to the families of mouse and human FcγRs including their polymorphic variants. Our results document the specific binding patterns for each of these IgG (sub)classes, reveal possible caveats of antibody-based immunoassays, and will be a useful reference for the transition from one animal model to preclinical mouse models or human cell-based bioassays.


Subject(s)
Immunoglobulin G , Receptors, IgG , Animals , Carrier Proteins/metabolism , Humans , Hybridomas , Mice , Mice, Transgenic
7.
J Cell Sci ; 133(5)2020 03 06.
Article in English | MEDLINE | ID: mdl-31974112

ABSTRACT

Actin dynamics is essential for T-cell development. We show here that cofilin1 is the key molecule for controlling actin filament turnover in this process. Mice with specific depletion of cofilin1 in thymocytes showed increased steady-state levels of actin filaments, and associated alterations in the pattern of thymocyte migration and adhesion. Our data suggest that cofilin1 is controlling oscillatory F-actin changes, a parameter that influences the migration pattern in a 3-D environment. In a collagen matrix, cofilin1 controls the speed and resting intervals of migrating thymocytes. Cofilin1 was not involved in thymocyte proliferation, cell survival, apoptosis or surface receptor trafficking. However, in cofilin1 mutant mice, impaired adhesion and migration resulted in a specific block of thymocyte differentiation from CD4/CD8 double-positive thymocytes towards CD4 and CD8 single-positive cells. Our data suggest that tuning of the dwelling time of thymocytes in the thymic niches is tightly controlled by cofilin1 and essential for positive selection during T-cell differentiation. We describe a novel role of cofilin1 in the physiological context of migration-dependent cell differentiation.


Subject(s)
Actins , Thymocytes , Actins/genetics , Animals , CD8-Positive T-Lymphocytes , Cell Differentiation , Cell Movement , Cofilin 1 , Mice
8.
Immunol Rev ; 268(1): 25-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26497511

ABSTRACT

Mouse and human FcRs have been a major focus of attention not only of the scientific community, through the cloning and characterization of novel receptors, and of the medical community, through the identification of polymorphisms and linkage to disease but also of the pharmaceutical community, through the identification of FcRs as targets for therapy or engineering of Fc domains for the generation of enhanced therapeutic antibodies. The availability of knockout mouse lines for every single mouse FcR, of multiple or cell-specific--'à la carte'--FcR knockouts and the increasing generation of hFcR transgenics enable powerful in vivo approaches for the study of mouse and human FcR biology. This review will present the landscape of the current FcR family, their effector functions and the in vivo models at hand to study them. These in vivo models were recently instrumental in re-defining the properties and effector functions of FcRs that had been overlooked or discarded from previous analyses. A particular focus will be made on the (mis)concepts on the role of high-affinity IgG receptors in vivo and on results from antibody engineering to enhance or abrogate antibody effector functions mediated by FcRs.


Subject(s)
Receptors, Fc/metabolism , Animals , Animals, Genetically Modified , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mice , Mice, Knockout , Mutation , Protein Binding , Receptors, Fc/chemistry , Receptors, Fc/genetics , Receptors, IgG/genetics , Receptors, IgG/metabolism
11.
J Allergy Clin Immunol ; 139(4): 1253-1265.e14, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27568081

ABSTRACT

BACKGROUND: Anaphylaxis can proceed through distinct IgE- or IgG-dependent pathways, which have been investigated in various mouse models. We developed a novel mouse strain in which the human low-affinity IgG receptor locus, comprising both activating (hFcγRIIA, hFcγRIIIA, and hFcγRIIIB) and inhibitory (hFcγRIIB) hFcγR genes, has been inserted into the equivalent murine locus, corresponding to a locus swap. OBJECTIVE: We sought to determine the capabilities of hFcγRs to induce systemic anaphylaxis and identify the cell types and mediators involved. METHODS: hFcγR expression on mouse and human cells was compared to validate the model. Passive systemic anaphylaxis was induced by injection of heat-aggregated human intravenous immunoglobulin and active systemic anaphylaxis after immunization and challenge. Anaphylaxis severity was evaluated based on hypothermia and mortality. The contribution of receptors, mediators, or cell types was assessed based on receptor blockade or depletion. RESULTS: The human-to-mouse low-affinity FcγR locus swap engendered hFcγRIIA/IIB/IIIA/IIIB expression in mice comparable with that seen in human subjects. Knock-in mice were susceptible to passive and active anaphylaxis, accompanied by downregulation of both activating and inhibitory hFcγR expression on specific myeloid cells. The contribution of hFcγRIIA was predominant. Depletion of neutrophils protected against hypothermia and mortality. Basophils contributed to a lesser extent. Anaphylaxis was inhibited by platelet-activating factor receptor or histamine receptor 1 blockade. CONCLUSION: Low-affinity FcγR locus-switched mice represent an unprecedented model of cognate hFcγR expression. Importantly, IgG-related anaphylaxis proceeds within a native context of activating and inhibitory hFcγRs, indicating that, despite robust hFcγRIIB expression, activating signals can dominate to initiate a severe anaphylactic reaction.


Subject(s)
Anaphylaxis/immunology , Receptors, IgG/immunology , Animals , Disease Models, Animal , GPI-Linked Proteins/immunology , Gene Knock-In Techniques , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL
12.
J Allergy Clin Immunol ; 139(1): 269-280.e7, 2017 01.
Article in English | MEDLINE | ID: mdl-27246523

ABSTRACT

BACKGROUND: Animal models have demonstrated that allergen-specific IgG confers sensitivity to systemic anaphylaxis that relies on IgG Fc receptors (FcγRs). Mouse IgG2a and IgG2b bind activating FcγRI, FcγRIII, and FcγRIV and inhibitory FcγRIIB; mouse IgG1 binds only FcγRIII and FcγRIIB. Although these interactions are of strikingly different affinities, these 3 IgG subclasses have been shown to enable induction of systemic anaphylaxis. OBJECTIVE: We sought to determine which pathways control the induction of IgG1-, IgG2a-, and IgG2b-dependent passive systemic anaphylaxis. METHODS: Mice were sensitized with IgG1, IgG2a, or IgG2b anti-trinitrophenyl mAbs and challenged with trinitrophenyl-BSA intravenously to induce systemic anaphylaxis that was monitored by using rectal temperature. Anaphylaxis was evaluated in mice deficient for FcγRs injected with mediator antagonists or in which basophils, monocytes/macrophages, or neutrophils had been depleted. FcγR expression was evaluated on these cells before and after anaphylaxis. RESULTS: Activating FcγRIII is the receptor primarily responsible for all 3 models of anaphylaxis, and subsequent downregulation of this receptor was observed. These models differentially relied on histamine release and the contribution of mast cells, basophils, macrophages, and neutrophils. Strikingly, basophil contribution and histamine predominance in mice with IgG1- and IgG2b-induced anaphylaxis correlated with the ability of inhibitory FcγRIIB to negatively regulate these models of anaphylaxis. CONCLUSION: We propose that the differential expression of inhibitory FcγRIIB on myeloid cells and its differential binding of IgG subclasses controls the contributions of mast cells, basophils, neutrophils, and macrophages to IgG subclass-dependent anaphylaxis. Collectively, our results unravel novel complexities in the involvement and regulation of cell populations in IgG-dependent reactions in vivo.


Subject(s)
Anaphylaxis/immunology , Immunoglobulin G/immunology , Protein Subunits/immunology , Animals , Antibodies, Monoclonal/immunology , Female , Haptens/immunology , Histamine/immunology , Immunoglobulin E/immunology , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/immunology , Receptors, IgG/genetics , Receptors, IgG/immunology , Serum Albumin, Bovine/immunology
13.
J Autoimmun ; 80: 95-102, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27745779

ABSTRACT

Two activating mouse IgG receptors (FcγRs) have the ability to bind monomeric IgG, the high-affinity mouse FcγRI and FcγRIV. Despite high circulating levels of IgG, reports using FcγRI-/- or FcγRIV-/- mice or FcγRIV-blocking antibodies implicate these receptors in IgG-induced disease severity or therapeutic Ab efficacy. From these studies, however, one cannot conclude on the effector capabilities of a given receptor, because different activating FcγRs possess redundant properties in vivo, and cooperation between FcγRs may occur, or priming phenomena. To help resolve these uncertainties, we used mice expressing only FcγRI to determine its intrinsic properties in vivo. FcγRIonly mice were sensitive to IgG-induced autoimmune thrombocytopenia and anti-CD20 and anti-tumour immunotherapy, but resistant to IgG-induced autoimmune arthritis, anaphylaxis and airway inflammation. Our results show that the in vivo roles of FcγRI are more restricted than initially reported using FcγRI-/- mice, but confirm effector capabilities for this high-affinity IgG receptor in vivo.


Subject(s)
Antibodies, Blocking/therapeutic use , B-Lymphocytes/immunology , Immunotherapy/methods , Purpura, Thrombocytopenic, Idiopathic/immunology , Receptors, IgG/metabolism , Animals , Antibody Affinity , Disease Models, Animal , Hepatectomy , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Purpura, Thrombocytopenic, Idiopathic/therapy , Receptors, IgG/genetics , Splenectomy
15.
Blood ; 121(9): 1563-73, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23293080

ABSTRACT

Receptors for the Fc portion of IgG (FcγRs) are mandatory for the induction of various IgG-dependent models of autoimmunity, inflammation, anaphylaxis, and cancer immunotherapy. A few FcγRs have the ability to bind monomeric IgG: high-affinity mouse mFcγRI, mFcγRIV, and human hFcγRI. All others bind IgG only when aggregated in complexes or bound to cells or surfaces: low-affinity mouse mFcγRIIB and mFcγRIII and human hFcγRIIA/B/C and hFcγRIIIA/B. Although it has been proposed that high-affinity FcγRs are occupied by circulating IgG, multiple roles for mFcγRI and mFcγRIV have been reported in vivo. However, the potential roles of hFcγRI that is expressed on monocytes, macrophages, and neutrophils have not been reported. In the present study, we therefore investigated the role of hFcγRI in antibody-mediated models of disease and therapy by generating hFcγRI-transgenic mice deficient for multiple endogenous FcRs. hFcγRI was sufficient to trigger autoimmune arthritis and thrombocytopenia, immune complex-induced airway inflammation, and active and passive systemic anaphylaxis. We found monocyte/macrophages to be responsible for thrombocytopenia, neutrophils to be responsible for systemic anaphylaxis, and both cell types to be responsible for arthritis induction. Finally, hFcγRI was capable of mediating antibody-induced immunotherapy of metastatic melanoma. Our results unravel novel capabilities of human FcγRI that confirm the role of high-affinity IgG receptors in vivo.


Subject(s)
Anaphylaxis/genetics , Immunoglobulin G/physiology , Immunotherapy , Inflammation/genetics , Neoplasms/therapy , Receptors, IgG/physiology , Anaphylaxis/immunology , Animals , Antineoplastic Agents/pharmacology , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Immunoglobulin G/pharmacology , Immunotherapy/methods , Inflammation/chemically induced , Inflammation/immunology , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Receptors, IgG/genetics , Xenograft Model Antitumor Assays
16.
Blood ; 122(18): 3160-4, 2013 Oct 31.
Article in English | MEDLINE | ID: mdl-23980063

ABSTRACT

Tumor engraftment followed by monoclonal antibody (mAb) therapy targeting tumor antigens represents a gold standard for assessing the efficiency of mAbs to eliminate tumor cells. Mouse models have demonstrated that receptors for the Fc portion of immunoglobulin G (FcγRs) are critical determinants of mAb therapeutic efficacy, but the FcγR-expressing cell populations responsible remain elusive. We show that neutrophils are responsible for mAb-induced therapy of both subcutaneous syngeneic melanoma and human breast cancer xenografts. mAb-induced tumor reduction, abolished in neutropenic mice, could be restored in FcγR-deficient hosts upon transfer of FcγR+ neutrophils or upon human FcγRIIA/CD32A transgenic expression. Finally, conditional knockout mice unable to perform FcγR-mediated activation and phagocytosis specifically in neutrophils were resistant to mAb-induced therapy. Our work suggests that neutrophils are necessary and sufficient for mAb-induced therapy of subcutaneous tumors, and represent a new and critical focal point for optimizing mAb-induced immunotherapies that will impact on human cancer treatment.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Melanoma, Experimental/immunology , Neutrophils/immunology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Humans , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, IgG/genetics , Receptors, IgG/immunology , Receptors, IgG/metabolism , Rituximab , Trastuzumab , Tumor Burden/drug effects , Tumor Burden/immunology
17.
Blood ; 119(11): 2533-44, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22138510

ABSTRACT

IgE and IgE receptors (FcεRI) are well-known inducers of allergy. We recently found in mice that active systemic anaphylaxis depends on IgG and IgG receptors (FcγRIIIA and FcγRIV) expressed by neutrophils, rather than on IgE and FcεRI expressed by mast cells and basophils. In humans, neutrophils, mast cells, basophils, and eosinophils do not express FcγRIIIA or FcγRIV, but FcγRIIA. We therefore investigated the possible role of FcγRIIA in allergy by generating novel FcγRIIA-transgenic mice, in which various models of allergic reactions induced by IgG could be studied. In mice, FcγRIIA was sufficient to trigger active and passive anaphylaxis, and airway inflammation in vivo. Blocking FcγRIIA in vivo abolished these reactions. We identified mast cells to be responsible for FcγRIIA-dependent passive cutaneous anaphylaxis, and monocytes/macrophages and neutrophils to be responsible for FcγRIIA-dependent passive systemic anaphylaxis. Supporting these findings, human mast cells, monocytes and neutrophils produced anaphylactogenic mediators after FcγRIIA engagement. IgG and FcγRIIA may therefore contribute to allergic and anaphylactic reactions in humans.


Subject(s)
Hypersensitivity/etiology , Hypersensitivity/pathology , Inflammation/etiology , Inflammation/pathology , Passive Cutaneous Anaphylaxis , Receptors, IgG/physiology , Respiratory System/pathology , Animals , Cells, Cultured , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Humans , Hypersensitivity/metabolism , Inflammation/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Respiratory System/metabolism
18.
J Immunol ; 189(6): 2995-3006, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22908332

ABSTRACT

Besides high-affinity IgE receptors (FcεRI), human basophils express activating (FcγRIIA) and inhibitory (FcγRIIB) low-affinity IgG receptors. IgG receptors (FcγR) were also found on mouse basophils, but not identified. We investigated in this study FcγR and the biological consequences of their engagement in basophils of the two species. We found the following: (1) that mouse basophils also express activating (FcγRIIIA) and inhibitory (FcγRIIB) low-affinity FcγR; (2) that activating FcγR can activate both human and mouse basophils, albeit with different efficacies; (3) that negative signals triggered by inhibitory FcγR are dominant over positive signals triggered by activating FcγR, thus preventing both human and mouse basophils from being activated by IgG immune complexes; (4) that the coengagement of FcεRI with inhibitory and activating FcγR results in a FcγRIIB-dependent inhibition of IgE-induced responses of both human and mouse basophils; (5) that FcγRIIB has a similar dominant inhibitory effect in basophils from virtually all normal donors; and (6) that IL-3 upregulates the expression of both activating and inhibitory FcγR on human basophils from normal donors, but further enhances FcγRIIB-dependent inhibition. FcγR therefore function as a regulatory module, made of two subunits with antagonistic properties, that prevents IgG-induced and controls IgE-induced basophil activation in both mice and humans.


Subject(s)
Basophils/immunology , Basophils/metabolism , Down-Regulation/immunology , Receptors, IgG/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , GPI-Linked Proteins/physiology , Humans , Immunoglobulin E/biosynthesis , Immunoglobulin E/blood , Immunoglobulin G/blood , Immunoglobulin G/physiology , Mice , Mice, Transgenic , Receptors, IgG/biosynthesis , Receptors, IgG/blood
19.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798354

ABSTRACT

Platelets are highly reactive fragments of megakaryocytes that play a fundamental role in thrombosis and hemostasis. Predictably, all conventional anti-platelet therapies elicit bleeding, raising the question whether the thrombotic activity of platelets can be targeted separately. In this study, we describe a novel approach of inhibiting platelet activation through the use of bispecific single-chain variable fragments (bi-scFvs), termed cis-acting platelet receptor inhibitors (CAPRIs) that harness the immunoreceptor tyrosine-based inhibition motif (ITIM)-containing co-inhibitory receptor G6b-B (G6B) to suppress immunoreceptor tyrosine-based (ITAM)-containing receptor-mediated platelet activation. CAPRI-mediated hetero-clustering of G6B with either the ITAM-containing GPVI-FcR γ-chain complex or FcγRIIA (CD32A) inhibited collagen- or immune complex-induced platelet aggregation. G6B-GPVI CAPRIs strongly and specifically inhibited thrombus formation on collagen under arterial shear, whereas G6B-CD32A CAPRI strongly and specifically inhibited thrombus formation to heparin-induced thrombocytopenia, vaccine-induced thrombotic thrombocytopenia and antiphospholipid syndrome complexes on Von Willebrand Factor-coated surfaces and photochemical-injured endothelial cells under arterial shear. Our findings provide proof-of-concept that CAPRIs are highly effective at inhibiting ITAM receptor-mediated platelet activation, laying the foundation for a novel family of anti-thrombotic therapeutics with potentially improved efficacy and fewer bleeding outcomes compared with current anti-platelet therapies. .

20.
Eur J Immunol ; 42(9): 2395-408, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684987

ABSTRACT

Using N-ethyl-N-nitrosourea-induced mutagenesis, we established a mouse model with a novel form of neutropenia resulting from a point mutation in the transcriptional repressor Growth Factor Independence 1 (Gfi1). These mice, called Genista, had normal viability and no weight loss, in contrast to mice expressing null alleles of the Gfi1 gene. Furthermore, the Genista mutation had a very limited impact on lymphopoiesis or on T- and B-cell function. Within the bone marrow (BM), the Genista mutation resulted in a slight increase of monopoiesis and in a block of terminal granulopoiesis. This block occurred just after the metamyelocytic stage and resulted in the generation of small numbers of atypical CD11b(+) Ly-6G(int) neutrophils, the nuclear morphology of which resembled that of mature WT neutrophils. Unexpectedly, once released from the BM, these atypical neutrophils contributed to induce mild forms of autoantibody-induced arthritis and of immune complex-mediated lung alveolitis. They additionally failed to provide resistance to acute bacterial infection. Our study demonstrates that a hypomorphic mutation in the Gfi1 transcriptional repressor results in a novel form of neutropenia characterized by a split pattern of functional responses, reflecting the distinct thresholds required for eliciting neutrophil-mediated inflammatory and anti-infectious responses.


Subject(s)
DNA-Binding Proteins/genetics , Neutropenia/genetics , Point Mutation , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Arthritis/genetics , Arthritis/metabolism , B-Lymphocytes/metabolism , Bone Marrow/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , DNA-Binding Proteins/metabolism , Ethylnitrosourea , Female , Inflammation/genetics , Inflammation/metabolism , Lymphocytes/metabolism , Lymphopoiesis/genetics , Male , Mice , Mice, Inbred C57BL , Neutropenia/chemically induced , Neutrophils/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL