Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Nat Immunol ; 23(6): 947-959, 2022 06.
Article in English | MEDLINE | ID: mdl-35552540

ABSTRACT

Inflammation is an important component of fibrosis but immune processes that orchestrate kidney fibrosis are not well understood. Here we apply single-cell sequencing to a mouse model of kidney fibrosis. We identify a subset of kidney tubule cells with a profibrotic-inflammatory phenotype characterized by the expression of cytokines and chemokines associated with immune cell recruitment. Receptor-ligand interaction analysis and experimental validation indicate that CXCL1 secreted by profibrotic tubules recruits CXCR2+ basophils. In mice, these basophils are an important source of interleukin-6 and recruitment of the TH17 subset of helper T cells. Genetic deletion or antibody-based depletion of basophils results in reduced renal fibrosis. Human kidney single-cell, bulk gene expression and immunostaining validate a function for basophils in patients with kidney fibrosis. Collectively, these studies identify basophils as contributors to the development of renal fibrosis and suggest that targeting these cells might be a useful clinical strategy to manage chronic kidney disease.


Subject(s)
Basophils , Renal Insufficiency, Chronic , Animals , Fibrosis , Humans , Kidney/metabolism , Kidney Tubules , Mice , Renal Insufficiency, Chronic/metabolism , Single-Cell Analysis
2.
Nat Immunol ; 21(10): 1181-1193, 2020 10.
Article in English | MEDLINE | ID: mdl-32807943

ABSTRACT

Type 2 cytokine responses promote parasitic immunity and initiate tissue repair; however, they can also result in immunopathologies when not properly restricted. Although basophilia is recognized as a common feature of type 2 inflammation, the roles basophils play in regulating these responses are unknown. Here, we demonstrate that helminth-induced group 2 innate lymphoid cell (ILC2) responses are exaggerated in the absence of basophils, resulting in increased inflammation and diminished lung function. Additionally, we show that ILC2s from basophil-depleted mice express reduced amounts of the receptor for the neuropeptide neuromedin B (NMB). Critically, NMB stimulation inhibited ILC2 responses from control but not basophil-depleted mice, and basophils were sufficient to directly enhance NMB receptor expression on ILC2s. These studies suggest that basophils prime ILC2s to respond to neuron-derived signals necessary to maintain tissue integrity. Further, these data provide mechanistic insight into the functions of basophils and identify NMB as a potent inhibitor of type 2 inflammation.


Subject(s)
Basophils/immunology , Lung/metabolism , Lymphocytes/immunology , Nippostrongylus/physiology , Strongylida Infections/immunology , Animals , Cell Communication , Cells, Cultured , Cytokines/metabolism , Immunity, Innate , Lung/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurokinin B/analogs & derivatives , Neurokinin B/metabolism , Th2 Cells/immunology , Tryptases/genetics
3.
Nat Immunol ; 17(4): 356-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27002843

ABSTRACT

Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.


Subject(s)
Adaptive Immunity/immunology , Cytokines/immunology , Immunity, Innate/immunology , Infections/immunology , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Animals , Basophils/immunology , Eosinophils/immunology , Humans , Macrophages/immunology , Mast Cells/immunology , Monocytes/immunology , Neutrophils/immunology
4.
Immunity ; 49(4): 709-724.e8, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291028

ABSTRACT

B cells thwart antigenic aggressions by releasing immunoglobulin M (IgM), IgG, IgA, and IgE, which deploy well-understood effector functions. In contrast, the role of secreted IgD remains mysterious. We found that some B cells generated IgD-secreting plasma cells following early exposure to external soluble antigens such as food proteins. Secreted IgD targeted basophils by interacting with the CD44-binding protein galectin-9. When engaged by antigen, basophil-bound IgD increased basophil secretion of interleukin-4 (IL-4), IL-5, and IL-13, which facilitated the generation of T follicular helper type 2 cells expressing IL-4. These germinal center T cells enhanced IgG1 and IgE but not IgG2a and IgG2b responses to the antigen initially recognized by basophil-bound IgD. In addition, IgD ligation by antigen attenuated allergic basophil degranulation induced by IgE co-ligation. Thus, IgD may link B cells with basophils to optimize humoral T helper type 2-mediated immunity against common environmental soluble antigens.


Subject(s)
Basophils/immunology , Galectins/immunology , Hyaluronan Receptors/immunology , Immunoglobulin D/immunology , Th2 Cells/immunology , Animals , Basophils/metabolism , Cell Line, Tumor , Cells, Cultured , Galectins/genetics , Galectins/metabolism , Gene Expression Profiling/methods , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Immunoglobulin D/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-4/metabolism , Mice, Inbred BALB C , Protein Binding , Th2 Cells/metabolism
5.
Semin Immunol ; 53: 101529, 2021 03.
Article in English | MEDLINE | ID: mdl-34815162

ABSTRACT

It has been appreciated that basophilia is a common feature of helminth infections for approximately 50 years. The ability of basophils to secrete IL-4 and other type 2 cytokines has supported the prevailing notion that basophils contribute to antihelminth immunity by promoting optimal type 2 T helper (Th2) cell responses. While this appears to be the case in several helminth infections, emerging studies are also revealing that the effector functions of basophils are extremely diverse and parasite-specific. Further, new reports now suggest that basophils can restrict type 2 inflammation in a manner that preserves the integrity of helminth-affected tissue. Finally, exciting data has also demonstrated that basophils can regulate inflammation by participating in neuro-immune interactions. This article will review the current state of basophil biology and describe how recent studies are transforming our understanding of the role basophils play in the context of helminth infections.


Subject(s)
Basophils , Helminths , Animals , Cytokines/metabolism , Helminths/metabolism , Humans , Inflammation , Th2 Cells
6.
Proc Natl Acad Sci U S A ; 119(37): e2201645119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36070344

ABSTRACT

Neuroimmune interactions are crucial for regulating immunity and inflammation. Recent studies have revealed that the central nervous system (CNS) senses peripheral inflammation and responds by releasing molecules that limit immune cell activation, thereby promoting tolerance and tissue integrity. However, the extent to which this is a bidirectional process, and whether peripheral immune cells also promote tolerance mechanisms in the CNS remains poorly defined. Here we report that helminth-induced type 2 inflammation promotes monocyte responses in the brain that are required to inhibit excessive microglial activation and host death. Mechanistically, infection-induced monocytes express YM1 that is sufficient to inhibit tumor necrosis factor production from activated microglia. Importantly, neuroprotective monocytes persist in the brain, and infected mice are protected from subsequent lipopolysaccharide-induced neuroinflammation months after infection-induced inflammation has resolved. These studies demonstrate that infiltrating monocytes promote CNS homeostasis in response to inflammation in the periphery and demonstrate that a peripheral infection can alter the immunologic landscape of the host brain.


Subject(s)
Brain , Encephalitis , Homeostasis , Monocytes , Neuroimmunomodulation , Trichinella spiralis , Trichinellosis , Animals , Brain/immunology , Brain/parasitology , Encephalitis/immunology , Encephalitis/parasitology , Homeostasis/immunology , Lectins/metabolism , Mice , Microglia/immunology , Monocytes/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Trichinellosis/pathology , beta-N-Acetylhexosaminidases/metabolism
7.
J Immunol ; 209(10): 1827-1831, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36216513

ABSTRACT

Neutrophils are critical for the direct eradication of Aspergillus fumigatus conidia, but whether they mediate antifungal defense beyond their role as effectors is unclear. In this study, we demonstrate that neutrophil depletion impairs the activation of protective antifungal CCR2+ inflammatory monocytes. In the absence of neutrophils, monocytes displayed limited differentiation into monocyte-derived dendritic cells, reduced formation of reactive oxygen species, and diminished conidiacidal activity. Upstream regulator analysis of the transcriptional response in monocytes predicted a loss of STAT1-dependent signals as the potential basis for the dysfunction seen in neutrophil-depleted mice. We find that conditional removal of STAT1 on CCR2+ cells results in diminished antifungal monocyte responses, whereas exogenous administration of IFN-γ to neutrophil-depleted mice restores monocyte-derived dendritic cell maturation and reactive oxygen species production. Altogether, our findings support a critical role for neutrophils in antifungal immunity not only as effectors but also as important contributors to antifungal monocyte activation, in part by regulating STAT1-dependent functions.


Subject(s)
Monocytes , Neutrophils , Mice , Animals , Antifungal Agents , Reactive Oxygen Species , Aspergillus fumigatus
8.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Article in English | MEDLINE | ID: mdl-32421753

ABSTRACT

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Subject(s)
Erythroid Precursor Cells/immunology , Erythropoiesis/immunology , Mast Cells/immunology , Mastocytosis/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Carbonic Anhydrase I/genetics , Carbonic Anhydrase I/immunology , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/pathology , Female , Mast Cells/parasitology , Mast Cells/pathology , Mastocytosis/genetics , Mastocytosis/pathology , Mice , Mice, Transgenic , Trichinellosis/genetics , Trichinellosis/pathology
9.
Immunity ; 39(6): 1158-70, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24332033

ABSTRACT

Extramedullary hematopoiesis (EMH) refers to the differentiation of hematopoietic stem cells (HSCs) into effector cells that occurs in compartments outside of the bone marrow. Previous studies linked pattern-recognition receptor (PRR)-expressing HSCs, EMH, and immune responses to microbial stimuli. However, whether EMH operates in broader immune contexts remains unknown. Here, we demonstrate a previously unrecognized role for thymic stromal lymphopoietin (TSLP) in promoting the population expansion of progenitor cells in the periphery and identify that TSLP-elicited progenitors differentiated into effector cells including macrophages, dendritic cells, and granulocytes and that these cells contributed to type 2 cytokine responses. The frequency of circulating progenitor cells was also increased in allergic patients with a gain-of-function polymorphism in TSLP, suggesting the TSLP-EMH pathway might operate in human disease. These data identify that TSLP-induced EMH contributes to the development of allergic inflammation and indicate that EMH is a conserved mechanism of innate immunity.


Subject(s)
Cytokines/metabolism , Hematopoiesis, Extramedullary/immunology , Hypersensitivity/immunology , Inflammation , Spleen/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Flow Cytometry , Gene Expression Profiling , Humans , Mice , Mice, Inbred C57BL , Polymorphism, Genetic , Precursor Cells, B-Lymphoid/cytology , Spleen/cytology , Trichinellosis/immunology , Thymic Stromal Lymphopoietin
10.
Nat Immunol ; 10(7): 697-705, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19465906

ABSTRACT

Dendritic cells can prime naive CD4+ T cells; however, here we demonstrate that dendritic cell-mediated priming was insufficient for the development of T helper type 2 cell-dependent immunity. We identify basophils as a dominant cell population that coexpressed major histocompatibility complex class II and interleukin 4 message after helminth infection. Basophilia was promoted by thymic stromal lymphopoietin, and depletion of basophils impaired immunity to helminth infection. Basophils promoted antigen-specific CD4+ T cell proliferation and interleukin 4 production in vitro, and transfer of basophils augmented the population expansion of helminth-responsive CD4+ T cells in vivo. Collectively, our studies suggest that major histocompatibility complex class II-dependent interactions between basophils and CD4+ T cells promote T helper type 2 cytokine responses and immunity to helminth infection.


Subject(s)
Basophils/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Histocompatibility Antigens Class II/immunology , Immunity/immunology , Animals , Basophils/cytology , Basophils/metabolism , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cell Proliferation , Cytokines/metabolism , Female , Flow Cytometry , Gene Expression , Histocompatibility Antigens Class II/genetics , Immunoblotting , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Th2 Cells/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , Trichuriasis/immunology , Trichuriasis/parasitology
11.
J Immunol ; 203(5): 1230-1241, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31366712

ABSTRACT

Basophils are innate immune cells associated with type 2 immunity, allergic reactions, and host defense against parasite infections. In this study, we show that the transcription factor PLZF, which is known for its essential role in the function and development of several innate lymphocyte subsets, is also important for the myeloid-derived basophil lineage. PLZF-deficient mice had decreased numbers of basophil progenitors in the bone marrow and mature basophils in multiple peripheral tissues. Functionally, PLZF-deficient basophils were less responsive to IgE activation and produced reduced amounts of IL-4. The altered function of basophils resulted in a blunted Th2 T cell response to a protein allergen. Additionally, PLZF-deficient basophils had reduced expression of the IL-18 receptor, which impacted migration to lungs. PLZF, therefore, is a major player in controlling type 2 immune responses mediated not only by innate lymphocytes but also by myeloid-derived cells.


Subject(s)
Basophils/immunology , Promyelocytic Leukemia Zinc Finger Protein/immunology , Transcription Factors/immunology , Allergens/immunology , Animals , Immunity, Innate/immunology , Immunoglobulin E/immunology , Interleukin-4/immunology , Interleukin-8/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Th2 Cells/immunology
12.
J Autoimmun ; 79: 39-52, 2017 May.
Article in English | MEDLINE | ID: mdl-28126203

ABSTRACT

Regulatory T cells (Tregs) are a subset of CD4+ T cells with suppressive function and are critical for limiting inappropriate activation of T cells. Hence, the expansion of Tregs is an attractive strategy for the treatment of autoimmune diseases. Here, we demonstrate that the skin possesses the remarkable capacity to systemically expand Treg numbers by producing thymic stromal lymphopoietin (TSLP) in response to vitamin D receptor stimulation. An ∼2-fold increase in the proportion and absolute number of Tregs was observed in mice treated topically but not systemically with the Vitamin D3 analog MC903. This expansion of Tregs was dependent on TSLP receptor signaling but not on VDR signaling in hematopoietic cells. However, TSLP receptor expression by Tregs was not required for their proliferation. Rather, skin-derived TSLP promoted Treg expansion through dendritic cells. Importantly, treatment of skin with MC903 significantly lowered the incidence of autoimmune diabetes in non-obese diabetic mice and attenuated disease score in experimental autoimmune encephalomyelitis. Together, these data demonstrate that the skin has the remarkable potential to control systemic immune responses and that Vitamin D-mediated stimulation of skin could serve as a novel strategy to therapeutically modulate the systemic immune system for the treatment of autoimmunity.


Subject(s)
Cytokines/metabolism , Skin/immunology , Skin/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Biomarkers , Cholecalciferol/analogs & derivatives , Cholecalciferol/pharmacology , Cytokines/pharmacology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Thymic Stromal Lymphopoietin
13.
Nature ; 477(7363): 229-33, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841801

ABSTRACT

CD4(+) T-helper type 2 (T(H)2) cells, characterized by their expression of interleukin (IL)-4, IL-5, IL-9 and IL-13, are required for immunity to helminth parasites and promote the pathological inflammation associated with asthma and allergic diseases. Polymorphisms in the gene encoding the cytokine thymic stromal lymphopoietin (TSLP) are associated with the development of multiple allergic disorders in humans, indicating that TSLP is a critical regulator of T(H)2 cytokine-associated inflammatory diseases. In support of genetic analyses, exaggerated TSLP production is associated with asthma, atopic dermatitis and food allergies in patients, and studies in murine systems demonstrated that TSLP promotes T(H)2 cytokine-mediated immunity and inflammation. However, the mechanisms through which TSLP induces T(H)2 cytokine responses remain poorly defined. Here we demonstrate that TSLP promotes systemic basophilia, that disruption of TSLP-TSLPR interactions results in defective basophil responses, and that TSLPR-sufficient basophils can restore T(H)2-cell-dependent immunity in vivo. TSLP acted directly on bone-marrow-resident progenitors to promote basophil responses selectively. Critically, TSLP could elicit basophil responses in both IL-3-IL-3R-sufficient and -deficient environments, and genome-wide transcriptional profiling and functional analyses identified heterogeneity between TSLP-elicited versus IL-3-elicited basophils. Furthermore, activated human basophils expressed TSLPR, and basophils isolated from eosinophilic oesophagitis patients were distinct from classical basophils. Collectively, these studies identify previously unrecognized heterogeneity within the basophil cell lineage and indicate that expression of TSLP may influence susceptibility to multiple allergic diseases by regulating basophil haematopoiesis and eliciting a population of functionally distinct basophils that promote T(H)2 cytokine-mediated inflammation.


Subject(s)
Basophils/cytology , Cytokines/metabolism , Hematopoiesis , Hypersensitivity, Immediate/immunology , Inflammation/immunology , Inflammation/metabolism , Interleukin-3 , Animals , Asthma/immunology , Basophils/metabolism , Cytokines/genetics , Cytokines/immunology , Dermatitis, Atopic/immunology , Food Hypersensitivity/immunology , Humans , Interleukin-3/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , Receptors, Cytokine/metabolism , Receptors, Interleukin-3/deficiency , Receptors, Interleukin-3/genetics , Receptors, Interleukin-3/metabolism , Th2 Cells/immunology , Thymic Stromal Lymphopoietin
14.
J Immunol ; 193(7): 3717-25, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25156365

ABSTRACT

Type 2 inflammation underlies allergic diseases such as atopic dermatitis, which is characterized by the accumulation of basophils and group 2 innate lymphoid cells (ILC2s) in inflamed skin lesions. Although murine studies have demonstrated that cutaneous basophil and ILC2 responses are dependent on thymic stromal lymphopoietin, whether these cell populations interact to regulate the development of cutaneous type 2 inflammation is poorly defined. In this study, we identify that basophils and ILC2s significantly accumulate in inflamed human and murine skin and form clusters not observed in control skin. We demonstrate that murine basophil responses precede ILC2 responses and that basophils are the dominant IL-4-enhanced GFP-expressing cell type in inflamed skin. Furthermore, basophils and IL-4 were necessary for the optimal accumulation of ILC2s and induction of atopic dermatitis-like disease. We show that ILC2s express IL-4Rα and proliferate in an IL-4-dependent manner. Additionally, basophil-derived IL-4 was required for cutaneous ILC2 responses in vivo and directly regulated ILC2 proliferation ex vivo. Collectively, these data reveal a previously unrecognized role for basophil-derived IL-4 in promoting ILC2 responses during cutaneous inflammation.


Subject(s)
Basophils/immunology , Dermatitis, Atopic/immunology , Immunity, Innate , Lymphocytes/immunology , Skin/immunology , Animals , Basophils/pathology , Cell Proliferation , Cytokines/genetics , Cytokines/immunology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Female , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocytes/pathology , Male , Mice , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Skin/pathology , Thymic Stromal Lymphopoietin
15.
Nature ; 464(7293): 1362-6, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20200520

ABSTRACT

CD4(+) T helper 2 (T(H)2) cells secrete interleukin (IL)4, IL5 and IL13, and are required for immunity to gastrointestinal helminth infections. However, T(H)2 cells also promote chronic inflammation associated with asthma and allergic disorders. The non-haematopoietic-cell-derived cytokines thymic stromal lymphopoietin, IL33 and IL25 (also known as IL17E) have been implicated in inducing T(H)2 cell-dependent inflammation at mucosal sites, but how these cytokines influence innate immune responses remains poorly defined. Here we show that IL25, a member of the IL17 cytokine family, promotes the accumulation of a lineage-negative (Lin(-)) multipotent progenitor (MPP) cell population in the gut-associated lymphoid tissue that promotes T(H)2 cytokine responses. The IL25-elicited cell population, termed MPP(type2) cells, was defined by the expression of Sca-1 (also known as Ly6a) and intermediate expression of c-Kit (c-Kit(int)), and exhibited multipotent capacity, giving rise to cells of monocyte/macrophage and granulocyte lineages both in vitro and in vivo. Progeny of MPP(type2) cells were competent antigen presenting cells, and adoptive transfer of MPP(type2) cells could promote T(H)2 cytokine responses and confer protective immunity to helminth infection in normally susceptible Il25(-/-) mice. The ability of IL25 to induce the emergence of an MPP(type2) cell population identifies a link between the IL17 cytokine family and extramedullary haematopoiesis, and suggests a previously unrecognized innate immune pathway that promotes T(H)2 cytokine responses at mucosal sites.


Subject(s)
Cell Differentiation , Interleukins/immunology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Antigens, Ly/metabolism , Cell Lineage , Granulocytes/cytology , Granulocytes/immunology , Granulocytes/metabolism , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Interleukins/biosynthesis , Interleukins/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Nippostrongylus/immunology , Proto-Oncogene Proteins c-kit/metabolism , Strongylida Infections/immunology , Th2 Cells/cytology , Trichuriasis/immunology , Trichuris/immunology
16.
J Allergy Clin Immunol ; 133(5): 1390-9, 1399.e1-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24560412

ABSTRACT

BACKGROUND: Exposure to food allergens through a disrupted skin barrier has been recognized as a potential factor in the increasing prevalence of food allergy. OBJECTIVE: We sought to test the immunologic mechanisms by which epicutaneous sensitization to food allergens predisposes to intestinal food allergy. METHODS: Mice were epicutaneously sensitized with ovalbumin or peanut on an atopic dermatitis-like skin lesion, followed by intragastric antigen challenge. Antigen-specific serum IgE levels and T(H)2 cytokine responses were measured by ELISA. Expression of type 2 cytokines and mast cell proteases in the intestine were measured by using real-time PCR. Accumulation of basophils in the skin and mast cells in the intestine was examined by using flow cytometry. In vivo basophil depletion was achieved by using diphtheria toxin treatment of Baso-DTR mice. For cell-transfer studies, the basophil population was expanded in vivo by means of hydrodynamic tail vein injection of thymic stromal lymphopoietin (TSLP) cDNA plasmid. RESULTS: Sensitization to food allergens through an atopic dermatitis-like skin lesion is associated with an expansion of TSLP-elicited basophils in the skin that promote antigen-specific T(H)2 cytokine responses, increased antigen-specific serum IgE levels, and accumulation of mast cells in the intestine, promoting the development of intestinal food allergy. Critically, disruption of TSLP responses or depletion of basophils reduced the susceptibility to intestinal food allergy, whereas transfer of TSLP-elicited basophils into intact skin promoted disease. CONCLUSION: Epicutaneous sensitization on a disrupted skin barrier is associated with accumulation of TSLP-elicited basophils, which are necessary and sufficient to promote antigen-induced intestinal food allergy.


Subject(s)
Allergens/immunology , Basophils/immunology , Cytokines/immunology , Dermatitis, Atopic/immunology , Food Hypersensitivity/immunology , Intestines/immunology , Animals , Basophils/pathology , Cytokines/genetics , Dermatitis, Atopic/complications , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Food Hypersensitivity/etiology , Food Hypersensitivity/genetics , Food Hypersensitivity/pathology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Intestinal Mucosa/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Skin/immunology , Skin/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Thymic Stromal Lymphopoietin
17.
J Immunol ; 189(9): 4371-8, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024277

ABSTRACT

CD4(+) Th2 cytokine responses promote the development of allergic inflammation and are critical for immunity to parasitic helminth infection. Recent studies highlighted that basophils can promote Th2 cytokine-mediated inflammation and that phenotypic and functional heterogeneity exists between classical IL-3-elicited basophils and thymic stromal lymphopoietin (TSLP)-elicited basophils. However, whether distinct basophil populations develop after helminth infection and their relative contributions to anti-helminth immune responses remain to be defined. After Trichinella spiralis infection of mice, we show that basophil responses are rapidly induced in multiple tissue compartments, including intestinal-draining lymph nodes. Trichinella-induced basophil responses were IL-3-IL-3R independent but critically dependent on TSLP-TSLPR interactions. Selective depletion of basophils after Trichinella infection impaired infection-induced CD4(+) Th2 cytokine responses, suggesting that TSLP-dependent basophils augment Th2 cytokine responses after helminth infection. The identification and functional classification of TSLP-dependent basophils in a helminth infection model, coupled with their recently described role in promoting atopic dermatitis, suggests that these cells may be a critical population in promoting Th2 cytokine-associated inflammation in a variety of inflammatory or infectious settings. Collectively, these data suggest that the TSLP-basophil pathway may represent a new target in the design of therapeutic intervention strategies to promote or limit Th2 cytokine-dependent immunity and inflammation.


Subject(s)
Basophils/immunology , Cytokines/biosynthesis , Intestinal Mucosa/immunology , Th2 Cells/immunology , Thymus Gland/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Basophils/parasitology , Basophils/pathology , Cytokines/physiology , Inflammation/immunology , Inflammation/metabolism , Inflammation/parasitology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Stromal Cells/immunology , Stromal Cells/parasitology , Stromal Cells/pathology , Th2 Cells/parasitology , Th2 Cells/pathology , Thymus Gland/parasitology , Thymus Gland/pathology , Trichinellosis/metabolism , Trichinellosis/pathology , Thymic Stromal Lymphopoietin
18.
J Allergy Clin Immunol ; 132(4): 789-801; quiz 788, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24075190

ABSTRACT

Basophils were discovered by Paul Ehrlich in 1879 and represent the least abundant granulocyte population in mammals. The relative rarity of basophils and their phenotypic similarities with mast cells resulted in this cell lineage being historically overlooked, both clinically and experimentally. However, recent studies in human subjects and murine systems have shown that basophils perform nonredundant effector functions and significantly contribute to the development and progression of TH2 cytokine-mediated inflammation. Although the potential functions of murine and human basophils have provoked some controversy, recent genetic approaches indicate that basophils can migrate into lymphoid tissues and, in some circumstances, cooperate with other immune cells to promote optimal TH2 cytokine responses in vivo. This article provides a brief historical perspective on basophil-related research and discusses recent studies that have identified previously unappreciated molecules and pathways that regulate basophil development, activation, and function in the context of allergic inflammation. Furthermore, we highlight the unique effector functions of basophils and discuss their contributions to the development and pathogenesis of allergic inflammation in human disease. Finally, we discuss the therapeutic potential of targeting basophils in preventing or alleviating the development and progression of allergic inflammation.


Subject(s)
Basophils/immunology , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/physiopathology , Inflammation/immunology , Inflammation/physiopathology , Animals , Basophils/cytology , Cytokines/metabolism , Humans , Hypersensitivity, Immediate/metabolism , Inflammation/metabolism , Mice , Th2 Cells/immunology , Th2 Cells/metabolism
19.
Front Immunol ; 13: 995432, 2022.
Article in English | MEDLINE | ID: mdl-36225918

ABSTRACT

Parasitic helminth infections remain a significant global health issue and are responsible for devastating morbidity and economic hardships. During infection, helminths migrate through different host organs, which results in substantial tissue damage and the release of diverse effector molecules by both hematopoietic and non-hematopoietic cells. Thus, host protective responses to helminths must initiate mechanisms that help to promote worm clearance while simultaneously mitigating tissue injury. The specialized immunity that promotes these responses is termed type 2 inflammation and is initiated by the recruitment and activation of hematopoietic stem/progenitor cells, mast cells, basophils, eosinophils, dendritic cells, neutrophils, macrophages, myeloid-derived suppressor cells, and group 2 innate lymphoid cells. Recent work has also revealed the importance of neuron-derived signals in regulating type 2 inflammation and antihelminth immunity. These studies suggest that multiple body systems coordinate to promote optimal outcomes post-infection. In this review, we will describe the innate immune events that direct the scope and intensity of antihelminth immunity. Further, we will highlight the recent progress made in our understanding of the neuro-immune interactions that regulate these pathways and discuss the conceptual advances they promote.


Subject(s)
Helminthiasis , Helminths , Animals , Immunity, Innate , Inflammation , Lymphocytes
20.
Cell Rep ; 38(2): 110215, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021079

ABSTRACT

Macrophages are known to mediate anti-helminth responses, but it remains uncertain which subsets are involved or how macrophages actually kill helminths. Here, we show rapid monocyte recruitment to the lung after infection with the nematode parasite Nippostrongylus brasiliensis. In this inflamed tissue microenvironment, these monocytes differentiate into an alveolar macrophage (AM)-like phenotype, expressing both SiglecF and CD11c, surround invading parasitic larvae, and preferentially kill parasites in vitro. Monocyte-derived AMs (Mo-AMs) express type 2-associated markers and show a distinct remodeling of the chromatin landscape relative to tissue-derived AMs (TD-AMs). In particular, they express high amounts of arginase-1 (Arg1), which we demonstrate mediates helminth killing through L-arginine depletion. These studies indicate that recruited monocytes are selectively programmed in the pulmonary environment to express AM markers and an anti-helminth phenotype.


Subject(s)
Lung/immunology , Macrophages, Alveolar/immunology , Strongylida Infections/immunology , Animals , Arginase/metabolism , Cell Differentiation , Cytokines , Female , Lung/parasitology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Nippostrongylus , Strongylida Infections/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL