Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Mucosal Immunol ; 16(4): 408-421, 2023 08.
Article in English | MEDLINE | ID: mdl-37121384

ABSTRACT

Early life is characterized by extraordinary challenges, including rapid tissue growth and immune adaptation to foreign antigens after birth. During this developmental stage, infants have an increased risk of immune-mediated diseases. Here, we demonstrate that tissue-resident, interleukin (IL)-13- and IL-4-producing group 2 innate lymphoid cells (ILC2s) are enriched in human infant intestines compared to adult intestines. Organoid systems were employed to assess the role of infant intestinal ILC2s in intestinal development and showed that IL-13 and IL-4 increased epithelial cell proliferation and skewed cell differentiation toward secretory cells. IL-13 furthermore upregulated the production of mediators of type-2 immunity by infant intestinal epithelial cells, including vascular endothelial growth factor-A and IL-26, a chemoattractant for eosinophils. In line with these in vitro findings increased numbers of eosinophils were detected in vivo in infant intestines. Taken together, ILC2s are enriched in infant intestines and can support intestinal development while inducing an epithelial secretory response associated with type 2 immune-mediated diseases.


Subject(s)
Immunity, Innate , Interleukin-13 , Adult , Humans , Infant , Lymphocytes , Vascular Endothelial Growth Factor A , Interleukin-4 , Intestines , Interleukin-33 , Cytokines/metabolism
2.
Curr Opin Immunol ; 68: 28-33, 2021 02.
Article in English | MEDLINE | ID: mdl-32971468

ABSTRACT

Five subsets of ILCs are extensively described, Lymphoid Tissue inducer (LTi) cells, cytotoxic NK cells and non-cytotoxic helper ILC1s, ILC2s and ILC3s. So far, the main focus has been on the potent cytokine production by helper ILCs and their plastic nature that allows them to switch function and phenotype upon environmental changes. Recent advances in the field indicate the presence of cytotoxic helper ILCs that are distinct from conventional NK cells. In humans, these cytotoxic ILCs can develop from conventional helper ILCs whereas in mice this remains to be elucidated. In this review we discuss the identification, development and function of cytotoxic helper ILCs subsets in humans and mice.


Subject(s)
Immunity, Innate/immunology , Lymphocytes/immunology , Animals , Humans , Killer Cells, Natural/immunology
3.
J Pathol ; 253(4): 374-383, 2021 04.
Article in English | MEDLINE | ID: mdl-33305354

ABSTRACT

Pneumonia represents a major health care burden and Gram-negative bacteria provide an increasing therapeutic challenge at least in part through the emergence of multidrug-resistant strains. IL-33 is a multifunctional cytokine belonging to the IL-1 family that can affect many different cell types. We sought here to determine the effect of recombinant IL-33 on the host response during murine pneumonia caused by the common Gram-negative pathogen Klebsiella pneumoniae. IL-33 pretreatment prolonged survival for more than 1 day during lethal airway infection and decreased bacterial loads at the primary site of infection and distant organs. Postponed treatment with IL-33 (3 h) also reduced bacterial growth and dissemination. IL-33-mediated protection was not observed in mice deficient for the IL-33 receptor component IL-1 receptor-like 1. IL-33 induced a brisk type 2 response, characterized by recruitment of type 2 innate lymphoid cells to the lungs and enhanced release of IL-5 and IL-13. However, neither absence of innate lymphoid cells or IL-13, nor blocking of IL-5 impacted on IL-33 effects in mice infected with Klebsiella. Likewise, IL-33 remained effective in reducing bacterial loads in mice lacking B, T, and natural killer T cells. Experiments using antibody-mediated cell depletion indicated that neutrophils and inflammatory monocytes were of importance for antibacterial defense. The capacity of IL-33 to restrict bacterial growth in the lungs was strongly reduced in mice depleted of both neutrophils and inflammatory monocytes, but not in mice selectively depleted of either one of these cell types. These results suggest that IL-33 boosts host defense during bacterial pneumonia by a combined effect on neutrophils and inflammatory monocytes. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Interleukin-33/immunology , Klebsiella Infections/immunology , Monocytes/immunology , Neutrophils/immunology , Pneumonia/immunology , Sepsis/immunology , Animals , Interleukin-33/pharmacology , Klebsiella Infections/complications , Klebsiella pneumoniae , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sepsis/etiology
4.
Eur J Immunol ; 51(4): 811-823, 2021 04.
Article in English | MEDLINE | ID: mdl-33300130

ABSTRACT

Human ILCs are classically categorized into five subsets; cytotoxic CD127- CD94+ NK cells and non-cytotoxic CD127+ CD94- , ILC1s, ILC2s, ILC3s, and LTi cells. Here, we identify a previously unrecognized subset within the CD127+ ILC population, characterized by the expression of the cytotoxic marker CD94. These CD94+ ILCs resemble conventional ILC3s in terms of phenotype, transcriptome, and cytokine production, but are highly cytotoxic. IL-15 was unable to induce differentiation of CD94+ ILCs toward mature NK cells. Instead, CD94+ ILCs retained RORγt, CD127 and CD200R1 expression and produced IL-22 in response to IL-15. Culturing non-cytotoxic ILC3s with IL-12 induced upregulation of CD94 and cytotoxic activity, effects that were not observed with IL-15 stimulation. Thus, human helper ILCs can acquire a cytotoxic program without differentiating into NK cells.


Subject(s)
Cell Differentiation/immunology , Gene Expression Profiling/methods , Gene Expression Regulation/immunology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Animals , Cell Differentiation/genetics , Cell Line , Cytotoxicity, Immunologic/genetics , Cytotoxicity, Immunologic/immunology , Gene Expression Regulation/drug effects , Humans , Immunity, Innate/immunology , Interleukin-15/pharmacology , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
6.
Nat Immunol ; 20(8): 992-1003, 2019 08.
Article in English | MEDLINE | ID: mdl-31263279

ABSTRACT

Here we identify a group 2 innate lymphoid cell (ILC2) subpopulation that can convert into interleukin-17 (IL-17)-producing NKp44- ILC3-like cells. c-Kit and CCR6 define this ILC2 subpopulation that exhibits ILC3 features, including RORγt, enabling the conversion into IL-17-producing cells in response to IL-1ß and IL-23. We also report a role for transforming growth factor-ß in promoting the conversion of c-Kit- ILC2s into RORγt-expressing cells by inducing the upregulation of IL23R, CCR6 and KIT messenger RNA in these cells. This switch was dependent on RORγt and the downregulation of GATA-3. IL-4 was able to reverse this event, supporting a role for this cytokine in maintaining ILC2 identity. Notably, this plasticity has physiological relevance because a subset of RORγt+ ILC2s express the skin-homing receptor CCR10, and the frequencies of IL-17-producing ILC3s are increased at the expense of ILC2s within the lesional skin of patients with psoriasis.


Subject(s)
Interleukin-17/immunology , Lymphocytes/immunology , Psoriasis/pathology , Skin/pathology , Cells, Cultured , Humans , Interleukin-1beta/immunology , Interleukin-23 Subunit p19/immunology , Interleukin-4/immunology , Lymphocytes/cytology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Psoriasis/immunology , Receptors, CCR10/metabolism , Skin/immunology , Transforming Growth Factor beta/metabolism
7.
Arthritis Rheumatol ; 71(3): 392-402, 2019 03.
Article in English | MEDLINE | ID: mdl-30260078

ABSTRACT

OBJECTIVE: Clinical trials of the anti-interleukin-17A (anti-IL-17A) antibody secukinumab have demonstrated a crucial role of the cytokine IL-17A in the pathogenesis of spondyloarthritis (SpA); however, its cellular source in this condition remains a matter of controversy. Group 3 innate lymphoid cells (ILC3s) have been recently identified as potent producers of proinflammatory cytokines, including IL-17A and IL-22, in a number of different tissues. This study was undertaken to characterize the presence and composition of ILCs, and investigate whether these cells are an important source of IL-17A, in the synovial tissue (ST) of patients with SpA. METHODS: Matched ST, synovial fluid, and peripheral blood (PB) samples were obtained from SpA patients with actively inflamed knee joints. ILC subsets were characterized by flow cytometry. Gene expression analysis at the single-cell level was performed directly ex vivo and after in vitro activation. An IL-17A enzyme-linked immunospot assay was used to detect IL-17A-secreting cells. RESULTS: ILCs, and particularly NKp44+ ILC3s, were expanded in inflamed arthritic joints. Single-cell expression analysis demonstrated that ST ILCs were clearly distinguishable from ST T cells and from their PB counterparts. Expression of the Th17 signature transcripts RORC, AHR, and IL23R was detected in a large proportion of ST ILC3s. These cells were capable of inducing expression of IL22 and CSF2, but not IL17A, in response to in vitro restimulation. CONCLUSION: Our findings demonstrate that absolute and relative numbers of ILC3s are enriched in the synovial joints of patients with SpA. However, these cells are not a significant source of IL-17A in this disease.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunity, Innate/immunology , Interleukin-17/immunology , Interleukins/immunology , Lymphocytes/immunology , Spondylarthritis/immunology , Adult , Arthritis, Rheumatoid/immunology , Case-Control Studies , Female , Humans , Male , Middle Aged , Spondylarthritis/pathology , Synovial Fluid/immunology , Interleukin-22
8.
J Immunol ; 201(4): 1165-1173, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29980610

ABSTRACT

Infusion of mesenchymal stromal cells (MSCs) is a promising and increasingly applied therapy for patients who suffer from a variety of inflammatory diseases, including graft-versus-host disease (GvHD), a common and life-threatening complication after allogeneic hematopoietic stem cell transplantation. The therapeutic effect of MSCs is mainly ascribed to their ability to suppress T cells and to support tissue repair. However, clinical response rates in patients with GvHD are limited to 50%, and the determinants for MSC responsiveness are unknown. We recently reported that high frequencies of activated group 3 innate lymphoid cells (ILC3s) before and after allogeneic hematopoietic stem cell transplantation were associated with a lower risk of GvHD. This may be related to IL-22 production by ILC3s, a cytokine important for intestinal epithelial cell homeostasis. In this study, we investigated whether ILC3s may contribute to the therapeutic effect of MSCs by studying the interaction between MSCs and ILC3s in vitro. ILC3s isolated from human tonsils were cocultured with human bone marrow-derived MSCs for 5 d in the presence of IL-2. Coculture with MSCs enhanced the proliferation and IL-22 production of ILC3s. Reciprocally, ILC3s promoted ICAM-1 and VCAM-1 expression on MSCs. For both directions, the activation was mainly mediated by cell-cell contact and by MSC-derived IL-7 and likely by aryl hydrocarbon receptor ligands. Thus, in addition to inhibiting the proliferation of alloreactive T cells, MSCs also promote the expansion and IL-22 production of ILC3s, which may contribute to healthy homeostasis and wound repair in the treatment of various inflammatory conditions in the intestine, including GvHD.


Subject(s)
Graft vs Host Disease/therapy , Interleukins/metabolism , Lymphocytes/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Homeostasis , Humans , Immunity, Innate , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation , Transplantation, Homologous , Vascular Cell Adhesion Molecule-1/metabolism , Interleukin-22
9.
Nat Commun ; 9(1): 863, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491406

ABSTRACT

CD103+ dendritic cells (DC) are crucial for regulation of intestinal tolerance in humans. However, upon infection of the lamina propria this tolerogenic response is converted to an inflammatory response. Here we show that immunoglobulin A (IgA) immune complexes (IgA-IC), which are present after bacterial infection of the lamina propria, are important for the induction of inflammation by the human CD103+SIRPα+ DC subset. IgA-IC, by recognition through FcαRI, selectively amplify the production of proinflammatory cytokines TNF, IL-1ß and IL-23 by human CD103+ DCs. These cells then enhance inflammation by promoting Th17 responses and activating human intestinal innate lymphoid cells 3. Moreover, FcαRI-induced cytokine production is orchestrated via upregulation of cytokine translation and caspase-1 activation, which is dependent on glycolytic reprogramming mediated by kinases Syk, PI3K and TBK1-IKKε. Our data suggest that the formation of IgA-IC in the human intestine provides an environmental cue for the conversion of a tolerogenic to an inflammatory response.


Subject(s)
Antigens, CD/immunology , Dendritic Cells/immunology , Integrin alpha Chains/immunology , Intestines/immunology , Receptors, Fc/immunology , Cellular Reprogramming , Glycolysis , Humans , Immunoglobulin A/immunology , Interleukin-1beta/immunology , Interleukin-23/immunology , Intestines/cytology , Th17 Cells/immunology
10.
Immunity ; 46(5): 756-757, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28514676
11.
Cell Res ; 26(11): 1178-1179, 2016 11.
Article in English | MEDLINE | ID: mdl-27633062

ABSTRACT

A recent paper published in Nature demonstrates a multifaceted relation between enteric glial cells (EGC), intestinal epithelia, and ILC3, via the EGC release of neurotrophic factors, a structurally related group of ligands within the TGF-ß superfamily of signaling molecules and IL-22 produced by ILC3.


Subject(s)
Immunity, Innate , Lymphocytes , Neuroglia , Neurons , Neurotransmitter Agents
12.
Nat Immunol ; 17(7): 758-64, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27328005

ABSTRACT

Innate lymphoid cells (ILCs) are effectors and regulators of innate immunity and tissue modeling and repair. Researchers have identified subsets of ILCs with differing functional activities, capacities to produce cytokines and transcription factors required for development and function. Natural killer (NK) cells represent the prototypical member of the ILC family. Together with ILC1s, NK cells constitute group 1 ILCs, which are characterized by their capacity to produce interferon-γ and their functional dependence on the transcription factor T-bet. NK cells and ILC1s are developmentally distinct but share so many features that they are difficult to distinguish, particularly under conditions of infection and inflammation. Here we review current knowledge of NK cells and the various ILC1 subsets.


Subject(s)
Infections/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Lymphocytes/immunology , T-Box Domain Proteins/metabolism , Animals , Cell Differentiation , Host-Pathogen Interactions , Humans , Immunity, Innate , Interferon-gamma/metabolism , Lymphocyte Activation , T-Box Domain Proteins/genetics
13.
Nat Immunol ; 17(6): 636-45, 2016 06.
Article in English | MEDLINE | ID: mdl-27111145

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) secrete type 2 cytokines, which protect against parasites but can also contribute to a variety of inflammatory airway diseases. We report here that interleukin 1ß (IL-1ß) directly activated human ILC2s and that IL-12 induced the conversion of these activated ILC2s into interferon-γ (IFN-γ)-producing ILC1s, which was reversed by IL-4. The plasticity of ILCs was manifested in diseased tissues of patients with severe chronic obstructive pulmonary disease (COPD) or chronic rhinosinusitis with nasal polyps (CRSwNP), which displayed IL-12 or IL-4 signatures and the accumulation of ILC1s or ILC2s, respectively. Eosinophils were a major cellular source of IL-4, which revealed cross-talk between IL-5-producing ILC2s and IL-4-producing eosinophils. We propose that IL-12 and IL-4 govern ILC2 functional identity and that their imbalance results in the perpetuation of type 1 or type 2 inflammation.


Subject(s)
Cell Plasticity , Eosinophils/immunology , Immunity, Innate , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-4/metabolism , Lymphocytes/immunology , Nasal Polyps/immunology , Pneumonia/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Rhinitis/immunology , Sinusitis/immunology , Animals , Cell Differentiation , Cells, Cultured , Female , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, SCID , Th1 Cells/immunology , Th1-Th2 Balance , Th2 Cells/immunology
14.
Immunity ; 43(1): 146-60, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26187413

ABSTRACT

Human group 1 ILCs consist of at least three phenotypically distinct subsets, including NK cells, CD127(+) ILC1, and intraepithelial CD103(+) ILC1. In inflamed intestinal tissues from Crohn's disease patients, numbers of CD127(+) ILC1 increased at the cost of ILC3. Here we found that differentiation of ILC3 to CD127(+) ILC1 is reversible in vitro and in vivo. CD127(+) ILC1 differentiated to ILC3 in the presence of interleukin-2 (IL-2), IL-23, and IL-1ß dependent on the transcription factor RORγt, and this process was enhanced in the presence of retinoic acid. Furthermore, we observed in resection specimen from Crohn's disease patients a higher proportion of CD14(+) dendritic cells (DC), which in vitro promoted polarization from ILC3 to CD127(+) ILC1. In contrast, CD14(-) DCs promoted differentiation from CD127(+) ILC1 toward ILC3. These observations suggest that environmental cues determine the composition, function, and phenotype of CD127(+) ILC1 and ILC3 in the gut.


Subject(s)
Interleukin-12 Subunit p35/immunology , Interleukin-23 Subunit p19/immunology , Interleukin-7 Receptor alpha Subunit/immunology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Crohn Disease/immunology , Dendritic Cells/immunology , Humans , Interleukin-1beta/immunology , Interleukin-2/immunology , Intestinal Mucosa/cytology , Killer Cells, Natural/immunology , Lipopolysaccharide Receptors/immunology , Lymphocyte Transfusion , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptor gamma/metabolism , Tretinoin/pharmacology , Retinoic Acid Receptor gamma
15.
Curr Opin Immunol ; 31: 115-20, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25459003

ABSTRACT

Group 2 innate lymphoid cells (ILC2) which are strategically interspersed throughout barrier surfaces are important regulators of type 2 immune reactions, particularly against helminthic parasites. ILC2 are also involved in tissue homeostasis and repair. Studies in a variety of animal models have demonstrated that when dysregulated or chronically activated, ILC2 can contribute to the pathology of allergic inflammatory diseases such as allergic asthma and atopic dermatitis. Here we discuss new findings of the cross talk of ILC2 with other hematopoietic cells, in particular T cells, and review recent information on the role of ILC2 in type 2 inflammatory diseases.


Subject(s)
Asthma/immunology , Dermatitis, Atopic/immunology , Hematopoietic Stem Cells/immunology , Immunity, Innate , Lymphocytes/immunology , Animals , Asthma/pathology , Dermatitis, Atopic/pathology , Hematopoietic Stem Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Lymphocytes/pathology
16.
Blood ; 124(5): 812-21, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24855210

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) is widely used to treat hematopoietic cell disorders but is often complicated by graft-versus-host disease (GVHD), which causes severe epithelial damage. Here we have investigated longitudinally the effects of induction chemotherapy, conditioning radiochemotherapy, and allogeneic HSCT on composition, phenotype, and recovery of circulating innate lymphoid cells (ILCs) in 51 acute leukemia patients. We found that reconstitution of ILC1, ILC2, and NCR(-)ILC3 was slow compared with that of neutrophils and monocytes. NCR(+) ILC3 cells, which are not present in the circulation of healthy persons, appeared both after induction chemotherapy and after allogeneic HSCT. Circulating patient ILCs before transplantation, as well as donor ILCs after transplantation, expressed activation (CD69), proliferation (Ki-67), and tissue homing markers for gut (α4ß7, CCR6) and skin (CCR10 and CLA). The proportion of ILCs expressing these markers was associated with a decreased susceptibility to therapy-induced mucositis and acute GVHD. Taken together, these data suggest that ILC recovery and treatment-related tissue damage are interrelated and affect the development of GVHD.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Immunity, Innate , Leukemia/therapy , Lymphocytes/immunology , Mucositis/immunology , Acute Disease , Adult , Aged , Allografts , Antigens, Differentiation/immunology , Antigens, Differentiation/metabolism , Female , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Humans , Ki-67 Antigen/immunology , Ki-67 Antigen/metabolism , Leukemia/immunology , Leukemia/metabolism , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Mucositis/metabolism , Mucositis/pathology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology
17.
J Invest Dermatol ; 134(9): 2351-2360, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24658504

ABSTRACT

Innate lymphoid cells (ILCs) are increasingly appreciated as important regulators of tissue homeostasis and inflammation. However, their role in human skin remains obscure. We found that healthy peripheral blood CD117(+) ILC3, lacking the natural cytotoxicity receptor (NCR) NKp44 (NCR(-) ILC3), CD117(-)NCR(-)CRTH2(-)CD161(+) ILC1, and CRTH2(+) ILC2, express the skin-homing receptor cutaneous lymphocyte antigen (CLA). NCR(+) ILC3 were scarce in peripheral blood. Consistently, we identified in normal skin ILC2 and NCR(-) ILC3, a small proportion of CD161(+) ILC1, and hardly any NCR(+) ILC3, whereas NCR(+) ILC3 were present in cultured dermal explants. The skin ILC2 and NCR(+) ILC3 subsets produced IL-13 and IL-22, respectively, upon cytokine stimulation. Remarkably, dermal NCR(-) ILC3 converted to NCR(+) ILC3 upon culture in IL-1ß plus IL-23, cytokines known to be involved in psoriatic inflammation. In line with this observation, significantly increased proportions of NCR(+) ILC3 were present in lesional skin and peripheral blood of psoriasis patients as compared with skin and blood of healthy individuals, respectively, whereas the proportions of ILC2 and CD161(+) ILC1 remained unchanged. NCR(+) ILC3 from skin and blood of psoriasis patients produced IL-22, which is regarded as a key driver of epidermal thickening, suggesting that NCR(+) ILC3 may participate in psoriasis pathology.


Subject(s)
Dermis/immunology , Epidermis/immunology , Lymphocytes/immunology , Natural Cytotoxicity Triggering Receptor 2/immunology , Psoriasis/immunology , Adult , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Line, Transformed , Cell Lineage/immunology , Dermis/cytology , Epidermal Cells , Humans , Immunophenotyping , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukins/immunology , Interleukins/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Natural Cytotoxicity Triggering Receptor 2/metabolism , Proto-Oncogene Proteins c-kit/immunology , Proto-Oncogene Proteins c-kit/metabolism , Psoriasis/blood , Psoriasis/pathology , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/immunology , Receptors, Prostaglandin/metabolism , Interleukin-22
18.
Proc Natl Acad Sci U S A ; 110(32): 13091-6, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23878224

ABSTRACT

The receptor tyrosine kinases Axl and Mer, belonging to the Tyro3, Axl and Mer (TAM) receptor family, are expressed in a number of tumor cells and have well-characterized oncogenic roles. The therapeutic targeting of these kinases is considered an anticancer strategy, and various inhibitors are currently under development. At the same time, Axl and Mer are expressed in dendritic cells and macrophages and have an essential function in limiting inflammation. Inflammation is an enabling characteristic of multiple cancer hallmarks. These contrasting oncogenic and anti-inflammatory functions of Axl and Mer posit a potential paradox in terms of anticancer therapy. Here we demonstrate that azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced inflammation-associated cancer is exacerbated in mice lacking Axl and Mer. Ablation of Axl and Mer signaling is associated with increased production of proinflammatory cytokines and failure to clear apoptotic neutrophils in the intestinal lamina propria, thereby favoring a tumor-promoting environment. Interestingly, loss of these genes in the hematopoietic compartment is not associated with increased colitis. Axl and Mer are expressed in radioresistant intestinal macrophages, and the loss of these genes is associated with an increased inflammatory signature in this compartment. Our results raise the possibility of potential adverse effects of systemic anticancer therapies with Axl and Mer inhibitors, and underscore the importance of understanding their tissue and cell type-specific functions in cancer.


Subject(s)
Colitis/immunology , Colonic Neoplasms/immunology , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Azoxymethane , Colitis/chemically induced , Colitis/genetics , Colon/immunology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Cytokines/genetics , Cytokines/immunology , Dextran Sulfate , Female , Flow Cytometry , Gene Expression/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/pathology , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
19.
Nat Immunol ; 14(3): 221-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334791

ABSTRACT

Innate lymphoid cells (ILCs) are effectors of innate immunity and regulators of tissue modeling. Recently identified ILC populations have a cytokine expression pattern that resembles that of the helper T cell subsets T(H)2, T(H)17 and T(H)22. Here we describe a distinct ILC subset similar to T(H)1 cells, which we call 'ILC1'. ILC1 cells expressed the transcription factor T-bet and responded to interleukin 12 (IL-12) by producing interferon-γ (IFN-γ). ILC1 cells were distinct from natural killer (NK) cells as they lacked perforin, granzyme B and the NK cell markers CD56, CD16 and CD94, and could develop from RORγt(+) ILC3 under the influence of IL-12. The frequency of the ILC1 subset was much higher in inflamed intestine of people with Crohn's disease, which indicated a role for these IFN-γ-producing ILC1 cells in the pathogenesis of gut mucosal inflammation.


Subject(s)
Crohn Disease/immunology , Interleukin-12/metabolism , Intestinal Mucosa/immunology , Lymphocyte Subsets/immunology , Lymphocytes/immunology , T-Box Domain Proteins/biosynthesis , Animals , CD56 Antigen/analysis , Cell Differentiation , Cells, Cultured , Colitis/immunology , Cytokines/immunology , Cytokines/metabolism , Granzymes/analysis , Humans , Inflammation , Interferon-gamma/biosynthesis , Intestinal Mucosa/metabolism , Intestines/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily D/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Perforin/analysis , Receptors, IgG/analysis , Th1 Cells/immunology , Th1 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...