Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.536
Filter
Add more filters

Publication year range
1.
Cell ; 184(8): 2151-2166.e16, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33765440

ABSTRACT

Cutaneous mast cells mediate numerous skin inflammatory processes and have anatomical and functional associations with sensory afferent neurons. We reveal that epidermal nerve endings from a subset of sensory nonpeptidergic neurons expressing MrgprD are reduced by the absence of Langerhans cells. Loss of epidermal innervation or ablation of MrgprD-expressing neurons increased expression of a mast cell gene module, including the activating receptor, Mrgprb2, resulting in increased mast cell degranulation and cutaneous inflammation in multiple disease models. Agonism of MrgprD-expressing neurons reduced expression of module genes and suppressed mast cell responses. MrgprD-expressing neurons released glutamate which was increased by MrgprD agonism. Inhibiting glutamate release or glutamate receptor binding yielded hyperresponsive mast cells with a genomic state similar to that in mice lacking MrgprD-expressing neurons. These data demonstrate that MrgprD-expressing neurons suppress mast cell hyperresponsiveness and skin inflammation via glutamate release, thereby revealing an unexpected neuroimmune mechanism maintaining cutaneous immune homeostasis.


Subject(s)
Glutamic Acid/metabolism , Mast Cells/metabolism , Neurons/metabolism , Skin/metabolism , Animals , Cells, Cultured , Dermatitis/metabolism , Dermatitis/pathology , Diphtheria Toxin/pharmacology , Disease Models, Animal , Female , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Langerhans Cells/cytology , Langerhans Cells/drug effects , Langerhans Cells/metabolism , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Skin/pathology , beta-Alanine/chemistry , beta-Alanine/metabolism , beta-Alanine/pharmacology
2.
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
3.
Immunity ; 48(6): 1160-1171.e5, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29858009

ABSTRACT

Hematopoiesis occurs in distinct waves. "Definitive" hematopoietic stem cells (HSCs) with the potential for all blood lineages emerge in the aorta-gonado-mesonephros, while "primitive" progenitors, whose potential is thought to be limited to erythrocytes, megakaryocytes, and macrophages, arise earlier in the yolk sac (YS). Here, we questioned whether other YS lineages exist that have not been identified, partially owing to limitations of current lineage tracing models. We established the use of Cdh5-CreERT2 for hematopoietic fate mapping, which revealed the YS origin of mast cells (MCs). YS-derived MCs were replaced by definitive MCs, which maintained themselves independently from the bone marrow in the adult. Replacement occurred with tissue-specific kinetics. MCs in the embryonic skin, but not other organs, remained largely YS derived prenatally and were phenotypically and transcriptomically distinct from definite adult MCs. We conclude that within myeloid lineages, dual hematopoietic origin is shared between macrophages and MCs.


Subject(s)
Cell Lineage/immunology , Hematopoiesis/physiology , Mast Cells/cytology , Animals , Hemangioblasts/cytology , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Macrophages/immunology , Mast Cells/immunology , Mice , Skin/cytology , Skin/immunology , Yolk Sac/cytology , Yolk Sac/embryology
4.
Immunity ; 49(4): 640-653.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332630

ABSTRACT

Tissue-resident mast cells are associated with many inflammatory and physiological processes. Although mast cells arise from the yolk sac, the exact ontogeny of adult mast cells remains unclear. Here we have investigated the hematopoietic origin of mast cells using fate-mapping systems. We have shown that early erythro-myeloid progenitors (EMPs), late EMPs, and definitive hematopoietic stem cells (HSCs) each gave rise to mast cells in succession via an intermediate integrin ß7+ progenitor. From late embryogenesis to adult, early EMP-derived mast cells were largely replaced by late EMP-derived cells in most connective tissues except adipose and pleural cavity. Thus, mast cells with distinct origin displayed tissue-location preferences: early EMP-derived cells were limited to adipose and pleural cavity and late EMP-derived cells dominated most connective tissues, while HSC-derived cells were a main group in mucosa. Therefore, embryonic origin shapes the heterogeneity of adult mast cells, with diverse functions in immunity and development.


Subject(s)
Erythroid Cells/immunology , Mast Cells/immunology , Myeloid Progenitor Cells/immunology , Animals , Cell Lineage/immunology , Cells, Cultured , Connective Tissue/immunology , Connective Tissue/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/immunology , Erythroid Cells/cytology , Erythroid Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Integrin beta Chains/immunology , Integrin beta Chains/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Mice, Transgenic , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism
5.
Cell ; 149(1): 159-72, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22464328

ABSTRACT

Specialized niche environments specify and maintain stem and progenitor cells, but little is known about the identities and functional interactions of niche components in vivo. Here, we describe a modular system for the generation of artificial thymopoietic environments in the mouse embryo. Thymic epithelium that lacks hematopoietic function but is physiologically accessible for hematopoietic progenitor cells is functionalized by individual and combinatorial expression of four factors, the chemokines Ccl25 and Cxcl12, the cytokine Scf, and the Notch ligand DLL4. The distinct phenotypes and variable numbers of hematopoietic cells in the resulting epithelial environments reveal synergistic, context-dependent, and hierarchical interactions among effector molecules. The surprisingly simple rules determining hematopoietic properties enable the in vivo engineering of artificial environments conducive to the presence of distinct myeloid or T or B lymphoid lineage precursors; moreover, synthetic environments facilitate the procurement of physiological progenitor cell types for analytical purposes and future therapeutic applications.


Subject(s)
Cellular Microenvironment , Hematopoietic Stem Cells/metabolism , Thymus Gland/cytology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Proliferation , Epithelium/metabolism , Forkhead Transcription Factors/metabolism , Humans , Mast Cells/cytology , Mice , Mice, Transgenic , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymus Gland/metabolism
6.
Immunity ; 46(5): 863-874.e4, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514691

ABSTRACT

Mast cells are important for eradication of intestinal nematodes; however, their precise mechanisms of action have remained elusive, especially in the early phase of infection. We found that Spi-B-deficient mice had increased numbers of mast cells and rapidly expelled the Heligmosomoides polygyrus (Hp) nematode. This was accompanied by induction of interleukin-13 (IL-13)-producing group 2 innate lymphoid cells (ILC2) and goblet cell hyperplasia. Immediately after Hp infection, mast cells were rapidly activated to produce IL-33 in response to ATP released from apoptotic intestinal epithelial cells. In vivo inhibition of the P2X7 ATP receptor rendered the Spi-B-deficient mice susceptible to Hp, concomitant with elimination of mast cell activation and IL-13-producing ILC2 induction. These results uncover a previously unknown role for mast cells in innate immunity in that activation of mast cells by ATP orchestrates the development of a protective type 2 immune response, in part by producing IL-33, which contributes to ILC2 activation.


Subject(s)
Helminthiasis/immunology , Helminthiasis/parasitology , Helminths/immunology , Immunity, Innate , Lymphocyte Subsets/immunology , Mast Cells/immunology , Adenosine Triphosphate/metabolism , Animals , Cell Communication , Cell Differentiation , Disease Models, Animal , Disease Resistance/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression , Helminthiasis/genetics , Immunophenotyping , Interleukin-33/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Male , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Knockout , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Purinergic P2X7/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Cell Physiol Biochem ; 58(3): 212-225, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38852193

ABSTRACT

BACKGROUND/AIMS: Adrenaline quickly inhibits the release of histamine from mast cells. Besides ß2-adrenergic receptors, several in vitro studies also indicate the involvement of α-adrenergic receptors in the process of exocytosis. Since exocytosis in mast cells can be detected electrophysiologically by the changes in the membrane capacitance (Cm), its continuous monitoring in the presence of drugs would determine their mast cell-stabilizing properties. METHODS: Employing the whole-cell patch-clamp technique in rat peritoneal mast cells, we examined the effects of adrenaline on the degranulation of mast cells and the increase in the Cm during exocytosis. We also examined the degranulation of mast cells in the presence or absence of α-adrenergic receptor agonists or antagonists. RESULTS: Adrenaline dose-dependently suppressed the GTP-γ-S-induced increase in the Cm and inhibited the degranulation from mast cells, which was almost completely erased in the presence of butoxamine, a ß2-adrenergic receptor antagonist. Among α-adrenergic receptor agonists or antagonists, high dose prazosin, a selective α1-adrenergic receptor antagonist, significantly reduced the ratio of degranulating mast cells and suppressed the increase in the Cm. Additionally, prazosin augmented the inhibitory effects of adrenaline on the degranulation of mast cells. CONCLUSION: This study provided electrophysiological evidence for the first time that adrenaline dose-dependently inhibited the process of exocytosis, confirming its usefulness as a potent mast cell-stabilizer. The pharmacological blockade of α1-adrenergic receptor by prazosin synergistically potentiated such mast cell-stabilizing property of adrenaline, which is primarily mediated by ß2-adrenergic receptors.


Subject(s)
Cell Degranulation , Epinephrine , Exocytosis , Mast Cells , Prazosin , Animals , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/cytology , Epinephrine/pharmacology , Rats , Prazosin/pharmacology , Cell Degranulation/drug effects , Male , Exocytosis/drug effects , Patch-Clamp Techniques , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Rats, Wistar
8.
Immunity ; 42(2): 279-293, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25692702

ABSTRACT

Crosslinking of the immunoglobulin receptor FcεRI activates basophils and mast cells to induce immediate and chronic allergic inflammation. However, it remains unclear how the chronic allergic inflammation is regulated. Here, we showed that ecto-nucleotide pyrophosphatase-phosphodiesterase 3 (E-NPP3), also known as CD203c, rapidly induced by FcεRI crosslinking, negatively regulated chronic allergic inflammation. Basophil and mast cell numbers increased in Enpp3(-/-) mice with augmented serum ATP concentrations. Enpp3(-/-) mice were highly sensitive to chronic allergic pathologies, which was reduced by ATP blockade. FcεRI crosslinking induced ATP secretion from basophils and mast cells, and ATP activated both cells. ATP clearance was impaired in Enpp3(-/-) cells. Enpp3(-/-)P2rx7(-/-) mice showed decreased responses to FcεRI crosslinking. Thus, ATP released by FcεRI crosslinking stimulates basophils and mast cells for further activation causing allergic inflammation. E-NPP3 decreases ATP concentration and suppresses basophil and mast cell activity.


Subject(s)
Adenosine Triphosphate/metabolism , Asthma/immunology , Basophils/immunology , Mast Cells/immunology , Phosphoric Diester Hydrolases/immunology , Pyrophosphatases/immunology , Receptors, IgE/immunology , Adenosine Triphosphate/pharmacology , Animals , Basophils/cytology , Dermatitis, Contact/immunology , Diarrhea/immunology , Diarrhea/pathology , Immunoglobulin E/immunology , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Mice, Knockout , Passive Cutaneous Anaphylaxis/immunology , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/genetics , RNA Interference , RNA, Small Interfering , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/immunology , Trinitrobenzenes/immunology
9.
Nature ; 555(7694): 54-60, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29466336

ABSTRACT

The formation of red blood cells begins with the differentiation of multipotent haematopoietic progenitors. Reconstructing the steps of this differentiation represents a general challenge in stem-cell biology. Here we used single-cell transcriptomics, fate assays and a theory that allows the prediction of cell fates from population snapshots to demonstrate that mouse haematopoietic progenitors differentiate through a continuous, hierarchical structure into seven blood lineages. We uncovered coupling between the erythroid and the basophil or mast cell fates, a global haematopoietic response to erythroid stress and novel growth factor receptors that regulate erythropoiesis. We defined a flow cytometry sorting strategy to purify early stages of erythroid differentiation, completely isolating classically defined burst-forming and colony-forming progenitors. We also found that the cell cycle is progressively remodelled during erythroid development and during a sharp transcriptional switch that ends the colony-forming progenitor stage and activates terminal differentiation. Our work showcases the utility of linking transcriptomic data to predictive fate models, and provides insights into lineage development in vivo.


Subject(s)
Erythrocytes/cytology , Erythroid Precursor Cells/cytology , Erythropoiesis , Animals , Basophils/cytology , Cell Cycle/genetics , Cell Cycle/physiology , Cell Lineage/drug effects , Cell Lineage/genetics , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Female , Flow Cytometry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mast Cells/cytology , Mice , Proto-Oncogene Proteins c-kit/metabolism , RNA, Small Cytoplasmic/analysis , RNA, Small Cytoplasmic/genetics , Single-Cell Analysis , Transcriptome
10.
Nat Immunol ; 12(11): 1035-44, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-22012443

ABSTRACT

Hematopoietic cells, including lymphoid and myeloid cells, can develop into phenotypically distinct 'subpopulations' with different functions. However, evidence indicates that some of these subpopulations can manifest substantial plasticity (that is, undergo changes in their phenotype and function). Here we focus on the occurrence of phenotypically distinct subpopulations in three lineages of myeloid cells with important roles in innate and acquired immunity: macrophages, mast cells and neutrophils. Cytokine signals, epigenetic modifications and other microenvironmental factors can substantially and, in some cases, rapidly and reversibly alter the phenotype of these cells and influence their function. This suggests that regulation of the phenotype and function of differentiated hematopoietic cells by microenvironmental factors, including those generated during immune responses, represents a common mechanism for modulating innate or adaptive immunity.


Subject(s)
Cytokines/immunology , Immunity, Innate , Macrophages/metabolism , Mast Cells/metabolism , Neutrophils/metabolism , Adaptive Immunity , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Epigenesis, Genetic/immunology , Gene Expression Regulation/immunology , Humans , Macrophages/cytology , Macrophages/immunology , Mast Cells/cytology , Mast Cells/immunology , Myeloid Progenitor Cells/cytology , Neutrophils/cytology , Neutrophils/immunology , Signal Transduction/immunology , Stem Cell Niche/immunology
11.
Cell ; 134(3): 485-95, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18692471

ABSTRACT

Mast cells are known to play a pivotal role in allergic diseases. Cross-linking of the high-affinity receptor for IgE (FcepsilonRI) leads to degranulation and allergic inflammation; however, the regulatory mechanisms of IgE-dependent exocytosis remain unknown. We show here that IkappaB kinase (IKK) 2 in mast cells plays critical roles in IgE-mediated anaphylaxis in vivo, and IgE-mediated degranulation in vitro, in an NF-kB-independent manner. Upon FcvarepsilonRI stimulation, IKK2 phosphorylates SNAP-23, the target membrane soluble N-ethylmaleimide-sensitive fusion factor attachment protein receptor (SNARE), and ectopic expression of a phospho-mimetic mutant of SNAP-23 partially rescued the impaired IgE-mediated degranulation in IKK2-deficient mast cells. These results suggest that IKK2 phosphorylation of SNAP-23 leads to degranulation and anaphylactic reactions. While this reaction is NF-kB-independent, we additionally show that IKK2 also regulates late-phase allergic reactions promoted by the release of proinflammatory cytokines in an NF-kB-dependent manner. The findings suggest that IKK2 is a central player in allergic reactions.


Subject(s)
Cell Degranulation , I-kappa B Kinase/metabolism , Mast Cells/cytology , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , Anaphylaxis/immunology , Animals , Immunoglobulin E/immunology , Mast Cells/immunology , Mice , Phosphorylation
12.
Br J Haematol ; 196(2): 304-315, 2022 01.
Article in English | MEDLINE | ID: mdl-34374078

ABSTRACT

Historically, understanding of the human mast cell (MC) compartment has lagged behind the appreciation of other cell lineages. MCs exist in vascularised tissues but do not under normal circumstances circulate in blood, and there has been no pharmacological agent identified that totally and selectively inhibits human MC function. There are no substantiated accounts of an apparently healthy individual who is severely lacking in MCs. Thus, some of the approaches employed to understand the function of a specific immune cell are not available to the MC biologist. The disease categories that have provided the greatest insight into MC biology have been monoclonal and IgE-mediated MC disorders. This has led to the categorisation of MC diseases as intrinsic or extrinsic to the MC compartment and to the recognition of the role of mediators in MC activation disorders. Mastocytosis as a clonal disorder not only impacts the MC compartment through changes intrinsic to the MC, but also by the effects of episodes of significant release of MC mediators. The availability of newer therapeutic approaches developed to treat monoclonal MC disorders offer insights into how to more selectively approach management of MC centric diseases.


Subject(s)
Mast Cells/immunology , Mast Cells/metabolism , Age Factors , Biomarkers , Biopsy , Combined Modality Therapy , Diagnosis, Differential , Disease Management , Disease Susceptibility/immunology , Genetic Predisposition to Disease , Homeostasis , Humans , Immunophenotyping , Mast Cells/cytology , Mastocytosis/diagnosis , Mastocytosis/etiology , Mastocytosis/metabolism , Mastocytosis/therapy , Molecular Diagnostic Techniques , Phenotype , Prognosis , Treatment Outcome
13.
Immunity ; 39(1): 97-110, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23871207

ABSTRACT

It remains unclear whether basophils and mast cells are derived from a common progenitor. Furthermore, how basophil versus mast cell fate is specified has not been investigated. Here, we have identified a population of granulocyte-macrophage progenitors (GMPs) that were highly enriched in the capacity to differentiate into basophils and mast cells while retaining a limited capacity to differentiate into myeloid cells. We have designated these progenitor cells "pre-basophil and mast cell progenitors" (pre-BMPs). STAT5 signaling was required for the differentiation of pre-BMPs into both basophils and mast cells and was critical for inducing two downstream molecules: C/EBPα and MITF. We have identified C/EBPα as the critical basophil transcription factor for specifying basophil cell fate and MITF as the crucial transcription factor for specifying mast cell fate. C/EBPα and MITF silenced each other's transcription in a directly antagonistic fashion. Our study reveals how basophil and mast cell fate is specified.


Subject(s)
Basophils/immunology , CCAAT-Enhancer-Binding Protein-alpha/immunology , Mast Cells/immunology , Microphthalmia-Associated Transcription Factor/immunology , Animals , Basophils/cytology , Basophils/metabolism , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/metabolism , HEK293 Cells , Humans , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Stem Cells/immunology , Stem Cells/metabolism
14.
J Immunol ; 205(12): 3311-3318, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33188077

ABSTRACT

IL-13 plays a critical role in mediating many biological processes responsible for allergic inflammation. Mast cells express Il13 mRNA and produce IL-13 protein in response to antigenic stimulation. Enhancers are essential in promoting gene transcription and are thought to activate transcription by delivering essential accessory cofactors to the promoter to potentiate gene transcription. However, enhancers mediating Il13 have not been identified. Furthermore, which Il13 enhancers detect signals triggered by antigenic stimulation have not yet been defined. In this study, we identified potential mouse Il13 enhancers using histone modification monomethylation at lysine residue 4 on histone 3 (H3K4me1) chromatin immunoprecipitation sequencing and acetylation at lysine residue 27 on histone 3 (H3K27ac) chromatin immunoprecipitation sequencing. We used Omni-assay for transposase-accessible chromatin sequencing to determine which accessible regions within the potential Il13 enhancers that responded to IgE receptor crosslinking. We also demonstrated that the transcription factor cluster consisting of the NFATC2, STAT5, GATA2, AP1, and RUNX1 binding sites at the proximal Il13 enhancer and the transcription factor cluster consisting of the EGR2 binding site at the distal Il13 E+6.5 enhancer are critical in sensing the signals triggered by antigenic stimulation. Those enhancers, which are responsive to antigenic stimulation and are constitutively active, cooperate to generate greater transcriptional outputs. Our study reveals a novel mechanism underlying how antigenic stimulation induces robust Il13 mRNA expression in mouse mast cells.


Subject(s)
Antigens/immunology , Core Binding Factor Alpha 2 Subunit/immunology , Early Growth Response Protein 2/immunology , GATA2 Transcription Factor/immunology , Interleukin-13/immunology , Mast Cells/immunology , NFATC Transcription Factors/immunology , Response Elements/immunology , STAT5 Transcription Factor/immunology , Transcription Factor AP-1/immunology , Transcription, Genetic/immunology , Animals , Cell Line , Mast Cells/cytology , Mice
15.
Proc Natl Acad Sci U S A ; 116(41): 20500-20504, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548430

ABSTRACT

Skin wound infections are a significant health problem, and antibiotic resistance is on the rise. Mast cells (MCs) have been shown to contribute to host-defense responses in certain bacterial infections, but their role in skin wound superinfection is unknown. We subjected 2 MC-deficient mouse strains to Pseudomonas aeruginosa skin wound infection and found significantly delayed wound closure in infected skin wounds. This delay was associated with impaired bacterial clearance in the absence of MCs. Engraftment of MCs restored both bacterial clearance and wound closure. Bacterial killing was dependent on IL-6 released from MCs, and engraftment with IL-6-deficient MCs failed to control wound infection. Treatment with recombinant IL-6 enhanced bacterial killing and resulted in the control of wound infection and normal wound healing in vivo. Taken together, our results demonstrate a defense mechanism for boosting host innate immune responses, namely effects of MC-derived IL-6 on antimicrobial functions of keratinocytes.


Subject(s)
Keratinocytes/immunology , Mast Cells/immunology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Skin/immunology , Wound Healing/immunology , Wound Infection/prevention & control , Animals , Anti-Bacterial Agents/pharmacology , Cells, Cultured , Humans , Interleukin-6/pharmacology , Keratinocytes/drug effects , Mast Cells/cytology , Mice , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Skin/drug effects , Wound Healing/drug effects , Wound Infection/immunology , Wound Infection/microbiology
16.
Int J Mol Sci ; 23(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35008966

ABSTRACT

INTRODUCTION: Osteophytes are a prominent feature of osteoarthritis (OA) joints and one of the clinical hallmarks of the disease progression. Research on osteophytes is fragmentary and modes of its contribution to OA pathology are obscure. AIM: To elucidate the role of osteophytes in OA pathology from a perspective of molecular and cellular events. METHODS: RNA-seq of fully grown osteophytes, collected from tibial plateau of six OA patients revealed patterns corresponding to active extracellular matrix re-modulation and prominent participation of mast cells. Presence of mast cells was further confirmed by immunohistochemistry, performed on the sections of the osteophytes using anti-tryptase alpha/beta-1 and anti-FC epsilon RI antibodies and the related key up-regulated genes were validated by qRT-PCR. To test the role of OA synovial fluid (SF) in mast cell maturation as proposed by the authors, hematopoietic stem cells (HSCs) and ThP1 cells were cultured in a media supplemented with 10% SF samples, obtained from various grades of OA patients and were monitored using specific cell surface markers by flow cytometry. Proteomics analysis of SF samples was performed to detect additional markers specific to mast cells and inflammation that drive the cell differentiation and maturation. RESULTS: Transcriptomics of osteophytes revealed a significant upregulation of mast cells specific genes such as chymase 1 (CMA1; 5-fold) carboxypeptidase A3 (CPA3; 4-fold), MS4A2/FCERI (FCERI; 4.2-fold) and interleukin 1 receptor-like 1 (IL1RL1; 2.5-fold) indicating their prominent involvement. (In IHC, anti-tryptase alpha/beta-1 and anti- FC epsilon RI-stained active mast cells were seen populated in cartilage, subchondral bone, and trabecular bone.) Based on these outcomes and previous learnings, the authors claim a possibility of mast cells invasion into osteophytes is mediated by SF and present in vitro cell differentiation assay results, wherein ThP1 and HSCs showed differentiation into HLA-DR+/CD206+ and FCERI+ phenotype, respectively, after exposing them to medium containing 10% SF for 9 days. Proteomics analysis of these SF samples showed an accumulation of mast cell-specific inflammatory proteins. CONCLUSIONS: RNA-seq analysis followed by IHC study on osteophyte samples showed a population of mast cells resident in them and may further accentuate inflammatory pathology of OA. Besides subchondral bone, the authors propose an alternative passage of mast cells invasion in osteophytes, wherein OA SF was found to be necessary and sufficient for maturation of mast cell precursor into effector cells.


Subject(s)
Cell Differentiation , Mast Cells/cytology , Mast Cells/metabolism , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteophyte/metabolism , Synovial Fluid/metabolism , Biomarkers , Computational Biology/methods , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Immunohistochemistry , Molecular Sequence Annotation , Osteoarthritis/pathology , Osteophyte/pathology
17.
Immunol Rev ; 282(1): 47-57, 2018 03.
Article in English | MEDLINE | ID: mdl-29431208

ABSTRACT

Since their establishment in 1981, RBL-2H3 cells have been widely used as a mast cell (MC) model. Their ability to be easily grown in culture in large amounts, their responsiveness to FcεRI-mediated triggers and the fact that they can be genetically manipulated, have provided advantages over primary MCs, in particular for molecular studies relying on genetic screening. Furthermore, the ability to generate clones that stably express proteins of interest, for example, a human receptor, have marked the RBL cells as an attractive MC model for drug screening. Indeed, 3 RBL reporter cell lines (RS-ATL8, NFAT-DsRed, and NPY-mRFP) have been generated providing useful models for drug and allergen screening. Similarly, RBL cells stably expressing the human MrgprX2 receptor provide a unique paradigm for analyzing ligand interactions and signaling pathways of the unique human receptor. Finally, transient co-transfections of RBL cells allow functional genomic analyses of MC secretion by combining library screening with simultaneous expression of a reporter for exocytosis. RBL cells thus comprise powerful tools for the study of intracellular membrane trafficking and exocytosis and the detection of allergens, vaccine safety studies and diagnosis of allergic sensitization. Their recent uses as an investigative tool are reviewed here.


Subject(s)
Basophils/physiology , Hypersensitivity/diagnosis , Mast Cells/physiology , Allergens/immunology , Animals , Basophils/cytology , Cell Degranulation , Cell Line , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Mast Cells/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, IgE/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Signal Transduction
18.
Eur J Immunol ; 50(1): 110-118, 2020 01.
Article in English | MEDLINE | ID: mdl-31706262

ABSTRACT

Mast cells are generally classified into two phenotypically distinct populations: mucosal-type mast cells (MMCs) and connective tissue-type mast cells (CTMCs). However, the molecular basis determining the different characteristics of the mast cell subclasses still remains unclear. Unfortunately, the number of mast cells that can be obtained from tissues is limited, which makes it difficult to study the function of each mast cell subclass. Here, we report the generation and characterization of MMCs and CTMCs derived from mouse BM mast cells (BMMCs). We found that the expression of galanin receptor 3 was elevated in MMCs when compared to the expression in CTMCs. Moreover, intraperitoneal injection of a galanin antagonist reduced MMCs and inhibited the inflammation of dextran sodium sulfate-induced colitis in mice. Therefore, these results suggest that galanin promotes MMC differentiation in vivo, and provide important insights into the molecular mechanisms underlying the differentiation of mast cell subclasses.


Subject(s)
Cell Differentiation/immunology , Galanin/metabolism , Mast Cells/cytology , Mucous Membrane/cytology , Mucous Membrane/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL
19.
Biochem Biophys Res Commun ; 551: 127-132, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33725574

ABSTRACT

Mast cell-deficient mice are helpful for understanding the roles of mast cells in vivo. To date, a dozen mouse models for mast cell deficiency have been reported. However, mice with a specific depletion of all populations of mast cells have not been reported. We generated knock-in mice, termed Mcpt5/Cma1DTR mice, expressing human diphtheria toxin A (DT) receptor under the endogenous promoter of Mcpt5 (also known as Cma1), which encodes mouse mast cell protease-5. Flow cytometry and histological analysis showed that intraperitoneal injection of DT induced almost complete depletion of mast cells in heterozygote Mcpt5/Cma1DTR/+ mice. The deletion rates of mast cells in peritoneal cavity, mesentery, abdominal skin, ear skin, and glandular stomach were 99.9%, 100%, 98.7%, 97.7%, and 100%, respectively. Passive cutaneous anaphylaxis reaction also revealed mast cell deficiency in ear skin after DT treatment. Other than mast cells, a small percentage of marginal zone B cells in Mcpt5/Cma1DTR/+ mice were killed by DT treatment. In conclusion, the Mcpt5/Cma1DTR/+ mouse model is valuable for achieving conditional depletion of all populations of mast cells without inducing a marked reduction in other cells.


Subject(s)
Cell Separation/methods , Chymases/genetics , Mast Cells/cytology , Models, Animal , Animals , Connective Tissue Cells/cytology , Female , Humans , Injections, Intraperitoneal , Mice , Mucous Membrane/cytology , Promoter Regions, Genetic/genetics
20.
Biochem Biophys Res Commun ; 561: 101-105, 2021 07 05.
Article in English | MEDLINE | ID: mdl-34020140

ABSTRACT

Mast cells (MCs) are present in various organs including the skin, peritoneal cavity, lung, and intestine and involved in the development of allergic diseases and host defense against infection. However, the regulatory mechanism of mast cell activation remains incompletely understood. We found in a database that Clec12b encoding a C-type lectin receptor Clec12b is preferentially expressed in skin MCs in mice. However, neither MCs in other tissues such as trachea, tongue, esophagus, or peritoneal cavity nor most lymphocytes and myeloid cells express Clec12b. To analyze the protein expression of Clec12b, we newly generated a monoclonal antibody (named TX109), which recognizes both mouse and human Clec12b. Consistent with the gene expression profile, flow cytometry analysis demonstrated that Clec12b is expressed only on MCs in the skin, but not on any other immune cell types in various tissues, in mice. Similarly, Clec12b is also expressed on skin MCs, but not on circulating lymphocytes and myeloid cells, in humans. Our results suggest that Clec12b plays an important role in the regulation of MCs activation in the skin.


Subject(s)
Antibodies, Monoclonal/immunology , Lectins, C-Type/metabolism , Mast Cells/metabolism , Receptors, Mitogen/metabolism , Skin/metabolism , Animals , Cell Differentiation , Cells, Cultured , Flow Cytometry/methods , Humans , Lectins, C-Type/immunology , Mast Cells/cytology , Mast Cells/immunology , Mice , Receptors, Mitogen/immunology , Skin/cytology , Skin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL