Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Immunity ; 52(2): 404-416.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32049054

ABSTRACT

Mast cells are rare tissue-resident cells of importance to human allergies. To understand the structural basis of principle mast cell functions, we analyzed the proteome of primary human and mouse mast cells by quantitative mass spectrometry. We identified a mast-cell-specific proteome signature, indicative of a unique lineage, only distantly related to other immune cell types, including innate immune cells. Proteome comparison between human and mouse suggested evolutionary conservation of core mast cell functions. In addition to specific proteases and proteins associated with degranulation and proteoglycan biosynthesis, mast cells expressed proteins potentially involved in interactions with neurons and neurotransmitter metabolism, including cell adhesion molecules, ion channels, and G protein coupled receptors. Toward targeted cell ablation in severe allergic diseases, we used MRGPRX2 for mast cell depletion in human skin biopsies. These proteome analyses suggest a unique role of mast cells in the immune system, probably intertwined with the nervous system.


Subject(s)
Mast Cells/cytology , Mast Cells/immunology , Animals , Biomarkers/metabolism , Cell Degranulation , Cell Lineage , Cells, Cultured , Connective Tissue/immunology , Humans , Immunotherapy , Mast Cells/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Neuroimmunomodulation , Proteoglycans/biosynthesis , Proteome , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/immunology , Receptors, Neuropeptide/metabolism , Skin/immunology
2.
Nature ; 620(7974): 634-642, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37438525

ABSTRACT

The physiological functions of mast cells remain largely an enigma. In the context of barrier damage, mast cells are integrated in type 2 immunity and, together with immunoglobulin E (IgE), promote allergic diseases. Allergic symptoms may, however, facilitate expulsion of allergens, toxins and parasites and trigger future antigen avoidance1-3. Here, we show that antigen-specific avoidance behaviour in inbred mice4,5 is critically dependent on mast cells; hence, we identify the immunological sensor cell linking antigen recognition to avoidance behaviour. Avoidance prevented antigen-driven adaptive, innate and mucosal immune activation and inflammation in the stomach and small intestine. Avoidance was IgE dependent, promoted by Th2 cytokines in the immunization phase and by IgE in the execution phase. Mucosal mast cells lining the stomach and small intestine rapidly sensed antigen ingestion. We interrogated potential signalling routes between mast cells and the brain using mutant mice, pharmacological inhibition, neural activity recordings and vagotomy. Inhibition of leukotriene synthesis impaired avoidance, but overall no single pathway interruption completely abrogated avoidance, indicating complex regulation. Collectively, the stage for antigen avoidance is set when adaptive immunity equips mast cells with IgE as a telltale of past immune responses. On subsequent antigen ingestion, mast cells signal termination of antigen intake. Prevention of immunopathology-causing, continuous and futile responses against per se innocuous antigens or of repeated ingestion of toxins through mast-cell-mediated antigen-avoidance behaviour may be an important arm of immunity.


Subject(s)
Allergens , Avoidance Learning , Hypersensitivity , Mast Cells , Animals , Mice , Allergens/immunology , Avoidance Learning/physiology , Hypersensitivity/immunology , Immunoglobulin E/immunology , Mast Cells/immunology , Stomach/immunology , Vagotomy , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Th2 Cells/immunology , Cytokines/immunology , Leukotrienes/biosynthesis , Leukotrienes/immunology , Intestine, Small/immunology
3.
Nature ; 590(7844): 151-156, 2021 02.
Article in English | MEDLINE | ID: mdl-33442055

ABSTRACT

Up to 20% of people worldwide develop gastrointestinal symptoms following a meal1, leading to decreased quality of life, substantial morbidity and high medical costs. Although the interest of both the scientific and lay communities in this issue has increased markedly in recent years, with the worldwide introduction of gluten-free and other diets, the underlying mechanisms of food-induced abdominal complaints remain largely unknown. Here we show that a bacterial infection and bacterial toxins can trigger an immune response that leads to the production of dietary-antigen-specific IgE antibodies in mice, which are limited to the intestine. Following subsequent oral ingestion of the respective dietary antigen, an IgE- and mast-cell-dependent mechanism induced increased visceral pain. This aberrant pain signalling resulted from histamine receptor H1-mediated sensitization of visceral afferents. Moreover, injection of food antigens (gluten, wheat, soy and milk) into the rectosigmoid mucosa of patients with irritable bowel syndrome induced local oedema and mast cell activation. Our results identify and characterize a peripheral mechanism that underlies food-induced abdominal pain, thereby creating new possibilities for the treatment of irritable bowel syndrome and related abdominal pain disorders.


Subject(s)
Abdominal Pain/immunology , Abdominal Pain/pathology , Allergens/immunology , Food Hypersensitivity/immunology , Food/adverse effects , Intestines/immunology , Irritable Bowel Syndrome/immunology , Abdominal Pain/etiology , Abdominal Pain/microbiology , Adult , Animals , Citrobacter rodentium/immunology , Diarrhea/immunology , Diarrhea/microbiology , Diarrhea/pathology , Enterobacteriaceae Infections/complications , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Female , Food Hypersensitivity/complications , Food Hypersensitivity/microbiology , Food Hypersensitivity/pathology , Glutens/immunology , Humans , Immunoglobulin E/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/microbiology , Intestines/pathology , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/pathology , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Middle Aged , Milk/immunology , Ovalbumin/immunology , Quality of Life , Receptors, Histamine H1/metabolism , Soybean Proteins/immunology , Triticum/immunology
4.
Nature ; 548(7668): 456-460, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28813413

ABSTRACT

Developmental deconvolution of complex organs and tissues at the level of individual cells remains challenging. Non-invasive genetic fate mapping has been widely used, but the low number of distinct fluorescent marker proteins limits its resolution. Much higher numbers of cell markers have been generated using viral integration sites, viral barcodes, and strategies based on transposons and CRISPR-Cas9 genome editing; however, temporal and tissue-specific induction of barcodes in situ has not been achieved. Here we report the development of an artificial DNA recombination locus (termed Polylox) that enables broadly applicable endogenous barcoding based on the Cre-loxP recombination system. Polylox recombination in situ reaches a practical diversity of several hundred thousand barcodes, allowing tagging of single cells. We have used this experimental system, combined with fate mapping, to assess haematopoietic stem cell (HSC) fates in vivo. Classical models of haematopoietic lineage specification assume a tree with few major branches. More recently, driven in part by the development of more efficient single-cell assays and improved transplantation efficiencies, different models have been proposed, in which unilineage priming may occur in mice and humans at the level of HSCs. We have introduced barcodes into HSC progenitors in embryonic mice, and found that the adult HSC compartment is a mosaic of embryo-derived HSC clones, some of which are unexpectedly large. Most HSC clones gave rise to multilineage or oligolineage fates, arguing against unilineage priming, and suggesting coherent usage of the potential of cells in a clone. The spreading of barcodes, both after induction in embryos and in adult mice, revealed a basic split between common myeloid-erythroid development and common lymphocyte development, supporting the long-held but contested view of a tree-like haematopoietic structure.


Subject(s)
Attachment Sites, Microbiological/genetics , Cell Lineage/genetics , Cell Tracking/methods , DNA Barcoding, Taxonomic/methods , Hematopoietic Stem Cells/cytology , Recombination, Genetic/genetics , Single-Cell Analysis/methods , Animals , Clone Cells/cytology , Clone Cells/metabolism , Embryo, Mammalian/cytology , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Mice , Mosaicism , Myeloid Cells/cytology , Myeloid Cells/metabolism
5.
Allergy ; 76(7): 2057-2069, 2021 07.
Article in English | MEDLINE | ID: mdl-33486786

ABSTRACT

BACKGROUND: Airway hyperresponsiveness (AHR) is a feature of asthma in which airways are hyperreactive to stimuli causing extensive airway narrowing. Methacholine provocations assess AHR in asthma patients mainly by direct stimulation of smooth muscle cells. Using in vivo mouse models, mast cells have been implicated in AHR, but the mechanism behind has remained unknown. METHODS: Cpa3Cre/+ mice, which lack mast cells, were used to assess the role of mast cells in house dust mite (HDM)-induced experimental asthma. Effects of methacholine in presence or absence of ketanserin were assessed on lung function and in lung mast cells in vitro. Airway inflammation, mast cell accumulation and activation, smooth muscle proliferation, and HDM-induced bronchoconstriction were evaluated. RESULTS: Repeated intranasal HDM sensitization induced allergic airway inflammation associated with accumulation and activation of lung mast cells. Lack of mast cells, absence of activating Fc-receptors, or antagonizing serotonin (5-HT)2A receptors abolished HDM-induced trachea contractions. HDM-sensitized mice lacking mast cells had diminished lung-associated 5-HT levels, reduced AHR and methacholine-induced airway contraction, while blocking 5-HT2A receptors in wild types eliminated AHR, implying that mast cells contribute to AHR by releasing 5-HT. Primary mouse and human lung mast cells express muscarinic M3 receptors. Mouse lung mast cells store 5-HT intracellularly, and methacholine induces release of 5-HT from lung-derived mouse mast cells and Ca2+ flux in human LAD-2 mast cells. CONCLUSIONS: Methacholine activates mast cells to release 5-HT, which by acting on 5-HT2A receptors enhances bronchoconstriction and AHR. Thus, M3-directed asthma treatments like tiotropium may also act by targeting mast cells.


Subject(s)
Asthma , Mast Cells , Animals , Asthma/diagnosis , Asthma/etiology , Disease Models, Animal , Humans , Lung , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Pyroglyphidae , Serotonin
6.
Immunity ; 37(1): 13-24, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22840840

ABSTRACT

Immunological functions of mast cells are currently considered to be much broader than the original role of mast cells in IgE-driven allergic disease. The spectrum of proposed mast cell functions includes areas as diverse as the regulation of innate and adaptive immune responses, protective immunity against viral, microbial, and parasitic pathogens, autoimmunity, tolerance to graft rejection, promotion of or protection from cancer, wound healing, angiogenesis, cardiovascular diseases, diabetes, obesity, and others. The vast majority of in vivo mast cell data have been based on mast cell-deficient Kit mutant mice. However, work in new mouse mutants with unperturbed Kit function, which have a surprisingly normal immune system, has failed to corroborate some key immunological aspects, formerly attributed to mast cells. Here, we consider the implications of these recent developments for the state of the field as well as for future work, aiming at deciphering the physiological functions of mast cells.


Subject(s)
Mast Cells/immunology , Animals , Humans , Immunity/physiology , Mast Cells/metabolism , Mice , Mutation , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism
7.
Immunity ; 35(5): 832-44, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22101159

ABSTRACT

Immunological functions of mast cells remain poorly understood. Studies in Kit mutant mice suggest key roles for mast cells in certain antibody- and T cell-mediated autoimmune diseases. However, Kit mutations affect multiple cell types of both immune and nonimmune origin. Here, we show that targeted insertion of Cre-recombinase into the mast cell carboxypeptidase A3 locus deleted mast cells in connective and mucosal tissues by a genotoxic Trp53-dependent mechanism. Cre-mediated mast cell eradication (Cre-Master) mice had, with the exception of a lack of mast cells and reduced basophils, a normal immune system. Cre-Master mice were refractory to IgE-mediated anaphylaxis, and this defect was rescued by mast cell reconstitution. This mast cell-deficient strain was fully susceptible to antibody-induced autoimmune arthritis and to experimental autoimmune encephalomyelitis. Differences comparing Kit mutant mast cell deficiency models to selectively mast cell-deficient mice call for a systematic re-evaluation of immunological functions of mast cells beyond allergy.


Subject(s)
Autoantibodies/immunology , Autoimmunity/immunology , Integrases/metabolism , Mast Cells/immunology , T-Lymphocytes/immunology , Anaphylaxis/genetics , Anaphylaxis/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Carboxypeptidases A/genetics , Carboxypeptidases A/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Profiling , Gene Targeting , Genetic Predisposition to Disease , Immunoglobulin E/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Stem Cell Factor/deficiency , T-Lymphocytes/metabolism , Th2 Cells/immunology , Tumor Suppressor Protein p53/metabolism
8.
Immunity ; 30(1): 67-79, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19110448

ABSTRACT

Notch1 signaling is required for T cell development and has been implicated in fate decisions in the thymus. We showed that Notch1 deletion in progenitor T cells (pro-T cells) revealed their latent developmental potential toward becoming conventional and plasmacytoid dendritic cells. In addition, Notch1 deletion in pro-T cells resulted in large numbers of thymic B cells, previously explained by T-to-B cell fate conversion. Single-cell genotyping showed, however, that the majority of these thymic B cells arose from Notch1-sufficient cells by a cell-extrinsic pathway. Fate switching nevertheless exists for a subset of thymic B cells originating from Notch1-deleted pro-T cells. Chimeric mice lacking the Notch ligand delta-like 4 (Dll4) in thymus epithelium revealed an essential role for Dll4 in T cell development. Thus, Notch1-Dll4 signaling fortifies T cell commitment by suppressing non-T cell lineage potential in pro-T cells, and normal Notch1-driven T cell development repels excessive B cells in the thymus.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Gene Deletion , Receptor, Notch1/genetics , T-Lymphocytes/immunology , Thymus Gland/cytology , Animals , Cell Lineage , Flow Cytometry , Mice , Mice, Inbred C57BL , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
Eur J Immunol ; 46(1): 114-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26449668

ABSTRACT

The genus leishmania comprises different protozoan parasites which are causative agents of muco-cutaneous and systemic, potentially lethal diseases. After infection with the species Leishmania major, resistant mice expand Th1 cells which stimulate macrophages for Leishmania destruction. In contrast, susceptible mice generate Th2 cells which deactivate macrophages, leading to systemic spread of the pathogens. Th-cell differentiation is determined within the first days, and Th2 cell differentiation requires IL-4, whereby the initial IL-4 source is often unknown. Mast cells are potential sources of IL-4, and hence their role in murine leishmaniasis has previously been studied in mast cell-deficient Kit mutant mice, although these mice display immunological phenotypes beyond mast cell deficiency. We therefore readdressed this question by infecting Kit-independent mast cell-deficient mice that are Th1 (C57BL/6 Cpa(Cre) ) or Th2 (BALB/c Cpa(Cre) ) prone with L. major. Using different parasite doses and intra- or subcutaneous infection routes, the results demonstrate no role of mast cells on lesion size development, parasite load, immune cell phenotypes expanding in draining lymph nodes, and cytokine production during murine cutaneous leishmaniasis. Thus, other cell types such as ILCs or T cells have to be considered as primary source of Th2-driving IL-4.


Subject(s)
Leishmaniasis, Cutaneous/immunology , Mast Cells/immunology , Th2 Cells/immunology , Animals , Cell Differentiation/immunology , Disease Models, Animal , Disease Susceptibility , Leishmania major , Leishmaniasis, Cutaneous/parasitology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Parasite Load
11.
PLoS Pathog ; 10(2): e1003913, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516385

ABSTRACT

Accumulating evidence suggests that IL-9-mediated immunity plays a fundamental role in control of intestinal nematode infection. Here we report a different impact of Foxp3⁺ regulatory T cells (Treg) in nematode-induced evasion of IL-9-mediated immunity in BALB/c and C57BL/6 mice. Infection with Strongyloides ratti induced Treg expansion with similar kinetics and phenotype in both strains. Strikingly, Treg depletion reduced parasite burden selectively in BALB/c but not in C57BL/6 mice. Treg function was apparent in both strains as Treg depletion increased nematode-specific humoral and cellular Th2 response in BALB/c and C57BL/6 mice to the same extent. Improved resistance in Treg-depleted BALB/c mice was accompanied by increased production of IL-9 and accelerated degranulation of mast cells. In contrast, IL-9 production was not significantly elevated and kinetics of mast cell degranulation were unaffected by Treg depletion in C57BL/6 mice. By in vivo neutralization, we demonstrate that increased IL-9 production during the first days of infection caused accelerated mast cell degranulation and rapid expulsion of S. ratti adults from the small intestine of Treg-depleted BALB/c mice. In genetically mast cell-deficient (Cpa3-Cre) BALB/c mice, Treg depletion still resulted in increased IL-9 production but resistance to S. ratti infection was lost, suggesting that IL-9-driven mast cell activation mediated accelerated expulsion of S. ratti in Treg-depleted BALB/c mice. This IL-9-driven mast cell degranulation is a central mechanism of S. ratti expulsion in both, BALB/c and C57BL/6 mice, because IL-9 injection reduced and IL-9 neutralization increased parasite burden in the presence of Treg in both strains. Therefore our results suggest that Foxp3⁺ Treg suppress sufficient IL-9 production for subsequent mast cell degranulation during S. ratti infection in a non-redundant manner in BALB/c mice, whereas additional regulatory pathways are functional in Treg-depleted C57BL/6 mice.


Subject(s)
Forkhead Transcription Factors/immunology , Interleukin-9/immunology , Mast Cells/immunology , Strongyloidiasis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Degranulation/immunology , Enzyme-Linked Immunosorbent Assay , Interleukin-9/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Strongyloides ratti/immunology , Strongyloidiasis/metabolism , T-Lymphocyte Subsets/immunology
12.
Am J Pathol ; 185(6): 1575-87, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25843682

ABSTRACT

Chronic inflammatory skin diseases, such as atopic dermatitis, affect a large percentage of the population, but the role of different immune cells in the pathogenesis of these disorders is largely unknown. Recently, we found that mice lacking fibroblast growth factor receptor 1 (Fgfr1) and Fgfr2 (K5-R1/R2 mice) in the epidermis have a severe impairment in the epidermal barrier, which leads to the development of a chronic inflammatory skin disease that shares many features with human atopic dermatitis. Using Fgfr1-/Fgfr2-deficient mice, we analyzed the consequences of the loss of mast cells. Mast cells accumulated and degranulated in the skin of young Fgfr1-/Fgfr2-deficient mice, most likely as a consequence of increased expression of the mast cell chemokine Ccl2. The increase in mast cells occurred before the development of histological abnormalities, indicating a functional role of these cells in the inflammatory skin phenotype. To test this hypothesis, we mated the Fgfr1-/Fgfr2-deficient mice with mast cell-deficient CreMaster mice. Surprisingly, loss of mast cells did not or only mildly affect keratinocyte proliferation, epidermal thickness, epidermal barrier function, accumulation and activation of different immune cells, or expression of different proinflammatory cytokines in the skin. These results reveal that mast cells are dispensable for the development of chronic inflammation in response to a defect in the epidermal barrier.


Subject(s)
Dermatitis/pathology , Mast Cells/pathology , Skin/pathology , Animals , Cell Proliferation , Chemokine CCL2/metabolism , Dermatitis/genetics , Dermatitis/immunology , Disease Models, Animal , Keratinocytes/immunology , Keratinocytes/pathology , Mast Cells/metabolism , Mice , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Skin/immunology , Skin/metabolism
13.
J Immunol ; 191(12): 6147-55, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227781

ABSTRACT

The growth and differentiation factor activin A is a key regulator of tissue repair, inflammation, fibrosis, and tumorigenesis. However, the cellular targets, which mediate the different activin functions, are still largely unknown. In this study, we show that activin increases the number of mature mast cells in mouse skin in vivo. To determine the relevance of this finding for wound healing and skin carcinogenesis, we mated activin transgenic mice with CreMaster mice, which are characterized by Cre recombinase-mediated mast cell eradication. Using single- and double-mutant mice, we show that loss of mast cells neither affected the stimulatory effect of overexpressed activin on granulation tissue formation and reepithelialization of skin wounds nor its protumorigenic activity in a model of chemically induced skin carcinogenesis. Furthermore, mast cell deficiency did not alter wounding-induced inflammation and new tissue formation or chemically induced angiogenesis and tumorigenesis in mice with normal activin levels. These findings reveal that mast cells are not major targets of activin during wound healing and skin cancer development and also argue against nonredundant functions of mast cells in wound healing and skin carcinogenesis in general.


Subject(s)
Activins/pharmacology , Carcinoma, Squamous Cell/pathology , Mast Cells/physiology , Papilloma/pathology , Skin Neoplasms/pathology , Wound Healing/drug effects , 9,10-Dimethyl-1,2-benzanthracene , Activins/administration & dosage , Activins/deficiency , Animals , Carcinogens , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/chemically induced , Chemotaxis/drug effects , Female , Humans , Injections, Intralesional , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Pathologic/pathology , Neutrophil Infiltration , Papilloma/blood supply , Papilloma/chemically induced , Proto-Oncogene Proteins c-kit/deficiency , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Skin/injuries , Skin/pathology , Skin Neoplasms/blood supply , Skin Neoplasms/chemically induced , Tetradecanoylphorbol Acetate
14.
J Immunol ; 190(11): 5534-44, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23636054

ABSTRACT

Mast cell-deficient Kit(W-sh) "sash" mice are widely used to investigate mast cell functions. However, mutations of c-Kit also affect additional cells of hematopoietic and nonimmune origin. In this study, we demonstrate that Kit(W-sh) causes aberrant extramedullary myelopoiesis characterized by the expansion of immature lineage-negative cells, common myeloid progenitors, and granulocyte/macrophage progenitors in the spleen. A consistent feature shared by these cell types is the reduced expression of c-Kit. Populations expressing intermediate and high levels of Ly6G, a component of the myeloid differentiation Ag Gr-1, are also highly expanded in the spleen of sash mice. These cells are able to suppress T cell responses in vitro and phenotypically and functionally resemble myeloid-derived suppressor cells (MDSC). MDSC typically accumulate in tumor-bearing hosts and are able to dampen immune responses. Consequently, transfer of MDSC from naive sash mice into line 1 alveolar cell carcinoma tumor-bearing wild-type littermates leads to enhanced tumor progression. However, although it can also be observed in sash mice, accelerated growth of transplanted line 1 alveolar cell carcinoma tumors is a mast cell-independent phenomenon. Thus, the Kit(W-sh) mutation broadly affects key steps in myelopoiesis that may have an impact on mast cell research.


Subject(s)
Mast Cells/immunology , Mutation , Myeloid Cells/immunology , Myelopoiesis/genetics , Myelopoiesis/immunology , Proto-Oncogene Proteins c-kit/genetics , Spleen/cytology , Adoptive Transfer , Animals , Antigens, Ly/metabolism , CD11b Antigen/metabolism , Female , Hematopoiesis, Extramedullary/genetics , Hematopoiesis, Extramedullary/immunology , Immunophenotyping , Mast Cells/metabolism , Mice , Mice, Knockout , Myeloid Cells/metabolism , Neoplasm Transplantation , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins c-kit/deficiency , Spleen/immunology , Spleen/metabolism
15.
PLoS One ; 19(4): e0300668, 2024.
Article in English | MEDLINE | ID: mdl-38578780

ABSTRACT

Mast cells are effector cells known to contribute to allergic airway disease. When activated, mast cells release a broad spectrum of inflammatory mediators, including the mast cell-specific protease carboxypeptidase A3 (CPA3). The expression of CPA3 in the airway epithelium and lumen of asthma patients has been associated with a Th2-driven airway inflammation. However, the role of CPA3 in asthma is unclear and therefore, the aim of this study was to investigate the impact of CPA3 for the development and severity of allergic airway inflammation using knockout mice with a deletion in the Cpa3 gene. We used the ovalbumin (OVA)- and house-dust mite (HDM) induced murine asthma models, and monitored development of allergic airway inflammation. In the OVA model, mice were sensitized with OVA intraperitoneally at seven time points and challenged intranasally (i.n.) with OVA three times. HDM-treated mice were challenged i.n. twice weekly for three weeks. Both asthma protocols resulted in elevated airway hyperresponsiveness, increased number of eosinophils in bronchoalveolar lavage fluid, increased peribronchial mast cell degranulation, goblet cell hyperplasia, thickening of airway smooth muscle layer, increased expression of IL-33 and increased production of allergen-specific IgE in allergen-exposed mice as compared to mocktreated mice. However, increased number of peribronchial mast cells was only seen in the HDM asthma model. The asthma-like responses in Cpa3-/- mice were similar as in wild type mice, regardless of the asthma protocol used. Our results demonstrated that the absence of a functional Cpa3 gene had no effect on several symptoms of asthma in two different mouse models. This suggest that CPA3 is dispensable for development of allergic airway inflammation in acute models of asthma in mice.


Subject(s)
Asthma , Mast Cells , Animals , Mice , Allergens/metabolism , Bronchoalveolar Lavage Fluid , Carboxypeptidases/metabolism , Disease Models, Animal , Inflammation/genetics , Inflammation/metabolism , Lung/metabolism , Mast Cells/metabolism , Mice, Inbred BALB C , Ovalbumin/metabolism
16.
Blood ; 117(4): 1329-39, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21063022

ABSTRACT

Bruton tyrosine kinase (Btk) is essential for B cell development and function and also appears to be important for myeloid cells. The bone marrow of Btk-deficient mice shows enhanced granulopoiesis compared with that of wild-type mice. In purified granulocyte-monocyte-progenitors (GMP) from Btk-deficient mice, the development of granulocytes is favored at the expense of monocytes. However, Btk-deficient neutrophils are impaired in maturation and function. Using bone marrow chimeras, we show that this defect is cell-intrinsic to neutrophils. In GMP and neutrophils, Btk plays a role in GM-CSF- and Toll-like receptor-induced differentiation. Molecular analyses revealed that expression of the lineage-determining transcription factors C/EBPα, C/EBPß, and PU.1, depends on Btk. In addition, expression of several granule proteins, including myeloperoxidase, neutrophilic granule protein, gelatinase and neutrophil elastase, is Btk-dependent. In the Arthus reaction, an acute inflammatory response, neutrophil migration into tissues, edema formation, and hemorrhage are significantly reduced in Btk-deficient animals. Together, our findings implicate Btk as an important regulator of neutrophilic granulocyte maturation and function in vivo.


Subject(s)
Neutrophils/physiology , Protein-Tyrosine Kinases/physiology , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Agammaglobulinemia/metabolism , Agammaglobulinemia/pathology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Disease Models, Animal , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Neutrophils/immunology , Neutrophils/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Toll-Like Receptors/physiology
17.
Cell Stem Cell ; 30(2): 207-218.e7, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36652946

ABSTRACT

In response to infections and stress, hematopoiesis rapidly enhances blood and immune cell production. The stage within the hematopoietic hierarchy that accounts for this regeneration is unclear under natural conditions in vivo. We analyzed by differentiation tracing, using inducible Tie2- or Flt3-driven Cre recombinase, the roles of mouse hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs). During polymicrobial sepsis, HSCs responded transcriptionally and increased their proliferation and cell death, yet HSC differentiation rates remained at steady-state levels. HSC differentiation was also independent from the ablation of various cellular compartments-bleeding, the antibody-mediated ablation of granulocytes or B lymphocytes, and genetic lymphocyte deficiency. By marked contrast, the fate mapping of MPPs in polymicrobial sepsis identified these cells as a major source for accelerated myeloid cell production. The regulation of blood and immune cell homeostasis by progenitors rather than stem cells may ensure a rapid response while preserving the integrity of the HSC population.


Subject(s)
Hematopoietic Stem Cells , Sepsis , Animals , Mice , Cell Differentiation/genetics , Cell Lineage , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Multipotent Stem Cells , Sepsis/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Receptor, TIE-2/metabolism
18.
Leukemia ; 37(7): 1474-1484, 2023 07.
Article in English | MEDLINE | ID: mdl-37161070

ABSTRACT

The persistence of leukemic stem cells (LSCs) represents a problem in the therapy of chronic myeloid leukemia (CML). Hence, it is of utmost importance to explore the underlying mechanisms to develop new therapeutic approaches to cure CML. Using the genetically engineered ScltTA/TRE-BCR::ABL1 mouse model for chronic phase CML, we previously demonstrated that the loss of the docking protein GAB2 counteracts the infiltration of mast cells (MCs) in the bone marrow (BM) of BCR::ABL1 positive mice. Here, we show for the first time that BCR::ABL1 drives the cytokine independent expansion of BM derived MCs and sensitizes them for FcεRI triggered degranulation. Importantly, we demonstrate that genetic mast cell deficiency conferred by the Cpa3Cre allele prevents BCR::ABL1 induced splenomegaly and impairs the production of pro-inflammatory cytokines. Furthermore, we show in CML patients that splenomegaly is associated with high BM MC counts and that upregulation of pro-inflammatory cytokines in patient serum samples correlates with tryptase levels. Finally, MC-associated transcripts were elevated in human CML BM samples. Thus, our study identifies MCs as essential contributors to disease progression and suggests considering them as an additional target in CML therapy. Mast cells play a key role in the pro-inflammatory tumor microenvironment of the bone marrow. Shown is a cartoon summarizing our results from the mouse model. BCR::ABL1 transformed MCs, as part of the malignant clone, are essential for the elevation of pro-inflammatory cytokines, known to be important in disease initiation and progression.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid , Humans , Mice , Animals , Mast Cells/metabolism , Splenomegaly/etiology , Splenomegaly/prevention & control , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Cytokines , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Disease Models, Animal , Protein Kinase Inhibitors/therapeutic use , Tumor Microenvironment
19.
J Biol Chem ; 286(7): 5423-33, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21123167

ABSTRACT

Mast cell secretory granules (secretory lysosomes) contain large amounts of fully active proteases bound to serglycin proteoglycan. Damage to the granule membrane will thus lead to the release of serglycin and serglycin-bound proteases into the cytosol, which potentially could lead to proteolytic activation of cytosolic pro-apoptotic compounds. We therefore hypothesized that mast cells are susceptible to apoptosis induced by permeabilization of the granule membrane and that this process is serglycin-dependent. Indeed, we show that wild-type mast cells are highly sensitive to apoptosis induced by granule permeabilization, whereas serglycin-deficient cells are largely resistant. The reduced sensitivity of serglycin(-/-) cells to apoptosis was accompanied by reduced granule damage, reduced release of proteases into the cytosol, and defective caspase-3 activation. Mechanistically, the apoptosis-promoting effect of serglycin involved serglycin-dependent proteases, as indicated by reduced sensitivity to apoptosis and reduced caspase-3 activation in cells lacking individual mast cell-specific proteases. Together, these findings implicate serglycin proteoglycan as a novel player in mast cell apoptosis.


Subject(s)
Apoptosis/physiology , Intracellular Membranes/metabolism , Mast Cells/metabolism , Proteoglycans/metabolism , Secretory Vesicles/metabolism , Vesicular Transport Proteins/metabolism , Animals , Caspase 3/genetics , Caspase 3/metabolism , Enzyme Activation/physiology , Mast Cells/cytology , Mice , Mice, Knockout , Permeability , Proteoglycans/genetics , Secretory Vesicles/genetics , Vesicular Transport Proteins/genetics
20.
Biol Chem ; 393(12): 1555-67, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23667909

ABSTRACT

Mast cell (MC) granules contain large amounts of proteases of the chymase, tryptase and carboxypeptidase A (MC-CPA) type that are stored in complex with serglycin,a proteoglycan with heparin side chains. Hence, serglycinprotease complexes are released upon MC degranulation and may influence local inflammation. Here we explored the possibility that a serglycin-protease axis may regulate levels of IL-13, a cytokine involved in allergic asthma. Indeed, we found that wild-type MCs efficiently degraded exogenous or endogenously produced IL-13 upon degranulation,whereas serglycin −/− MCs completely lacked this ability.Moreover, MC-mediated IL-13 degradation was blocked both by a serine protease inhibitor and by a heparin antagonist,which suggests that IL-13 degradation is catalyzed by serglycin-dependent serine proteases and that optimal IL-13 degradation is dependent on both the serglycin and the protease component of the serglycin-protease complex.Moreover, IL-13 degradation was abrogated in MC-CPA −/−MC cultures, but was normal in cultures of MCs with an inactivating mutation of MC-CPA, which suggests that the IL-13-degrading serine proteases rely on MC-CPA protein.Together, our data implicate a serglycin-serine protease axis in the regulation of extracellular levels of IL-13. Reduction of IL-13 levels through this mechanism possibly can provide a protective function in the context of allergic inflammation.


Subject(s)
Interleukin-13/metabolism , Mast Cells/physiology , Proteoglycans/metabolism , Serine Proteases/metabolism , Vesicular Transport Proteins/metabolism , Animals , Carboxypeptidases A/genetics , Carboxypeptidases A/immunology , Carboxypeptidases A/metabolism , Cell Degranulation , Cells, Cultured , Gene Deletion , Heparin/immunology , Heparin/metabolism , Heparin Antagonists/pharmacology , Hypersensitivity/immunology , Hypersensitivity/metabolism , Interleukin-13/immunology , Mice , Peritoneum/cytology , Proteoglycans/genetics , Proteoglycans/immunology , Proteolysis , Serine Proteases/immunology , Serine Proteinase Inhibitors/pharmacology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL