Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(11): e0248034, 2021.
Article in English | MEDLINE | ID: mdl-34752458

ABSTRACT

Retinoic acid receptor-related orphan nuclear receptor (ROR) γt is a member of the RORC nuclear hormone receptor family of transcription factors. RORγt functions as a critical regulator of thymopoiesis and immune responses. RORγt is expressed in multiple immune cell populations including Th17 cells, where its primary function is regulation of immune responses to bacteria and fungi through IL-17A production. However, excessive IL-17A production has been linked to numerous autoimmune diseases. Moreover, Th17 cells have been shown to elicit both pro- and anti-tumor effects. Thus, modulation of the RORγt/IL-17A axis may represent an attractive therapeutic target for the treatment of autoimmune disorders and some cancers. Herein we report the design, synthesis and characterization of three selective allosteric RORγt inhibitors in preclinical models of inflammation and tumor growth. We demonstrate that these compounds can inhibit Th17 differentiation and maintenance in vitro and Th17-dependent inflammation and associated gene expression in vivo, in a dose-dependent manner. Finally, RORγt inhibitors were assessed for efficacy against tumor formation. While, RORγt inhibitors were shown to inhibit tumor formation in pancreatic ductal adenocarcinoma (PDAC) organoids in vitro and modulate RORγt target genes in vivo, this activity was not sufficient to delay tumor volume in a KP/C human tumor mouse model of pancreatic cancer.


Subject(s)
Gene Expression/drug effects , Inflammation/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Th17 Cells/drug effects , Animals , Carcinogenesis/drug effects , Carcinogenesis/genetics , Inflammation/metabolism , Interleukin-17/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Th17 Cells/metabolism
2.
Am J Respir Crit Care Med ; 201(1): 63-72, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31394048

ABSTRACT

Rationale: Primary graft dysfunction (PGD) is the leading cause of early morbidity and mortality after lung transplantation, but the immunologic mechanisms are poorly understood. Innate lymphoid cells (ILC) are a heterogeneous family of immune cells regulating pathologic inflammation and beneficial tissue repair. However, whether changes in donor-derived lung ILC populations are associated with PGD development has never been examined.Objectives: To determine whether PGD in chronic obstructive pulmonary disease or interstitial lung disease transplant recipients is associated with alterations in ILC subset composition within the allograft.Methods: We performed a single-center cohort study of lung transplantation patients with surgical biopsies of donor tissue taken before, and immediately after, allograft reperfusion. Donor immune cells from 18 patients were characterized phenotypically by flow cytometry for single-cell resolution of distinct ILC subsets. Changes in the percentage of ILC subsets with reperfusion or PGD (grade 3 within 72 h) were assessed.Measurements and Main Results: Allograft reperfusion resulted in significantly decreased frequencies of natural killer cells and a trend toward reduced ILC populations, regardless of diagnosis (interstitial lung disease or chronic obstructive pulmonary disease). Seven patients developed PGD (38.9%), and PGD development was associated with selective reduction of the ILC2 subset after reperfusion. Conversely, patients without PGD exhibited significantly higher ILC1 frequencies before reperfusion, accompanied by elevated ILC2 frequencies after allograft reperfusion.Conclusions: The composition of donor ILC subsets is altered after allograft reperfusion and is associated with PGD development, suggesting that ILCs may be involved in regulating lung injury in lung transplant recipients.


Subject(s)
Immunity, Innate , Lung Transplantation/adverse effects , Lymphocytes/immunology , Primary Graft Dysfunction/etiology , Primary Graft Dysfunction/immunology , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Risk Factors
3.
J Neuroimmunol ; 294: 41-5, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27138097

ABSTRACT

Innate lymphoid cells (ILCs) play an important role in immunity, inflammation, and tissue remodeling and their dysregulation is implicated in autoimmune and inflammatory disorders. We analyzed the impact of daclizumab, a humanized monoclonal anti-CD25 antibody, on circulating natural killer (NK) cells and ILCs in a cohort of multiple sclerosis patients. An increase in CD56(bright) NK cells and CD56(hi)CD16(intermediate) transitional NK cells was observed. No significant change in total ILCs or major ILC subpopulations was observed. These results refine our understanding of the impact of daclizumab on innate lymphoid cell populations.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use , Killer Cells, Natural/drug effects , Lymphocytes/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/metabolism , Cohort Studies , Daclizumab , Female , Flow Cytometry , Humans , Immunoglobulin G/pharmacology , Immunosuppressive Agents/pharmacology , Lymphocytes/metabolism , Male , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
4.
Nat Immunol ; 17(6): 656-65, 2016 06.
Article in English | MEDLINE | ID: mdl-27043409

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair after activation by cell-extrinsic factors such as host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme arginase-1 (Arg1) during acute or chronic lung inflammation is a conserved trait of mouse and human ILC2s. Deletion of mouse ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics that controls proliferative capacity and proinflammatory functions promoting type 2 inflammation.


Subject(s)
Arginase/metabolism , Lymphocytes/physiology , Pneumonia/immunology , Animals , Arginase/genetics , Cell Proliferation/genetics , Cells, Cultured , Cytokines/metabolism , Glycolysis/genetics , Humans , Immunity, Innate , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Polyamines/metabolism , Th2 Cells/immunology
5.
Nature ; 519(7542): 242-6, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25533952

ABSTRACT

Obesity is an increasingly prevalent disease regulated by genetic and environmental factors. Emerging studies indicate that immune cells, including monocytes, granulocytes and lymphocytes, regulate metabolic homeostasis and are dysregulated in obesity. Group 2 innate lymphoid cells (ILC2s) can regulate adaptive immunity and eosinophil and alternatively activated macrophage responses, and were recently identified in murine white adipose tissue (WAT) where they may act to limit the development of obesity. However, ILC2s have not been identified in human adipose tissue, and the mechanisms by which ILC2s regulate metabolic homeostasis remain unknown. Here we identify ILC2s in human WAT and demonstrate that decreased ILC2 responses in WAT are a conserved characteristic of obesity in humans and mice. Interleukin (IL)-33 was found to be critical for the maintenance of ILC2s in WAT and in limiting adiposity in mice by increasing caloric expenditure. This was associated with recruitment of uncoupling protein 1 (UCP1)(+) beige adipocytes in WAT, a process known as beiging or browning that regulates caloric expenditure. IL-33-induced beiging was dependent on ILC2s, and IL-33 treatment or transfer of IL-33-elicited ILC2s was sufficient to drive beiging independently of the adaptive immune system, eosinophils or IL-4 receptor signalling. We found that ILC2s produce methionine-enkephalin peptides that can act directly on adipocytes to upregulate Ucp1 expression in vitro and that promote beiging in vivo. Collectively, these studies indicate that, in addition to responding to infection or tissue damage, ILC2s can regulate adipose function and metabolic homeostasis in part via production of enkephalin peptides that elicit beiging.


Subject(s)
Adipose Tissue, White/cytology , Adipose Tissue, White/immunology , Immunity, Innate/immunology , Lymphocytes/physiology , Obesity/immunology , Adipocytes/cytology , Adipocytes/drug effects , Animals , Energy Metabolism/immunology , Enkephalin, Methionine/biosynthesis , Enkephalin, Methionine/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Female , Homeostasis/drug effects , Humans , Interleukins/immunology , Interleukins/pharmacology , Ion Channels/metabolism , Lymphocytes/cytology , Lymphocytes/immunology , Male , Mice , Mitochondrial Proteins/metabolism , Obesity/pathology , Receptors, Interleukin-4/immunology , Receptors, Interleukin-4/metabolism , Uncoupling Protein 1
6.
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
7.
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
8.
Nature ; 504(7478): 153-7, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24185009

ABSTRACT

The development and severity of inflammatory bowel diseases and other chronic inflammatory conditions can be influenced by host genetic and environmental factors, including signals derived from commensal bacteria. However, the mechanisms that integrate these diverse cues remain undefined. Here we demonstrate that mice with an intestinal epithelial cell (IEC)-specific deletion of the epigenome-modifying enzyme histone deacetylase 3 (HDAC3(ΔIEC) mice) exhibited extensive dysregulation of IEC-intrinsic gene expression, including decreased basal expression of genes associated with antimicrobial defence. Critically, conventionally housed HDAC3(ΔIEC) mice demonstrated loss of Paneth cells, impaired IEC function and alterations in the composition of intestinal commensal bacteria. In addition, HDAC3(ΔIEC) mice showed significantly increased susceptibility to intestinal damage and inflammation, indicating that epithelial expression of HDAC3 has a central role in maintaining intestinal homeostasis. Re-derivation of HDAC3(ΔIEC) mice into germ-free conditions revealed that dysregulated IEC gene expression, Paneth cell homeostasis and intestinal barrier function were largely restored in the absence of commensal bacteria. Although the specific mechanisms through which IEC-intrinsic HDAC3 expression regulates these complex phenotypes remain to be determined, these data indicate that HDAC3 is a critical factor that integrates commensal-bacteria-derived signals to calibrate epithelial cell responses required to establish normal host-commensal relationships and maintain intestinal homeostasis.


Subject(s)
Gene Expression Regulation , Histone Deacetylases/metabolism , Homeostasis , Intestinal Mucosa/enzymology , Intestines/microbiology , Symbiosis , Adult , Animals , Bacteria/genetics , Colitis, Ulcerative/enzymology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Crohn Disease/enzymology , Crohn Disease/genetics , Crohn Disease/microbiology , Female , Gene Deletion , Gene Expression Profiling , Histone Deacetylases/genetics , Humans , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , Paneth Cells/metabolism , RNA, Ribosomal, 16S/genetics , Signal Transduction
9.
J Exp Med ; 210(9): 1823-37, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23960191

ABSTRACT

The predominantly epithelial cell-derived cytokines IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) can promote CD4(+) Th2 cell-dependent immunity, inflammation, and tissue repair at barrier surfaces through the induction of multiple innate immune cell populations. IL-25 and IL-33 were previously shown to elicit four innate cell populations, named natural helper cells, nuocytes, innate type 2 helper cells, and multipotent progenitor type 2 (MPP(type2)) cells, now collectively termed group 2 innate lymphoid cells (ILC2). In contrast to other types of ILC2, MPP(type2) cells exhibit multipotent potential and do not express T1/ST2 or IL-7Rα, suggesting that MPP(type2) cells may be a distinct population. Here, we show that IL-33 elicits robust ILC2 responses, whereas IL-25 predominantly promotes MPP(type2) cell responses at multiple tissue sites with limited effects on ILC2 responses. MPP(type2) cells were distinguished from ILC2 by their differential developmental requirements for specific transcription factors, distinct genome-wide transcriptional profile, and functional potential. Furthermore, IL-25-induced MPP(type2) cells promoted Th2 cytokine-associated inflammation after depletion of ILC2. These findings indicate that IL-25 simultaneously elicits phenotypically and functionally distinct innate lymphoid- and nonlymphoid-associated cell populations and implicate IL-25-elicited MPP(type2) cells and extramedullary hematopoiesis in the promotion of Th2 cytokine responses at mucosal surfaces.


Subject(s)
Immunity, Innate/drug effects , Interleukin-17/pharmacology , Lymphocytes/cytology , Lymphocytes/immunology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Animals , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/pathology , Inhibitor of Differentiation Protein 2/metabolism , Interleukin-33 , Interleukins/metabolism , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/drug effects , Phenotype , Protein Binding/drug effects , Protein Binding/immunology , Receptors, Interleukin/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Th2 Cells/immunology , Transcription, Genetic/drug effects , Transcriptome
10.
Immunity ; 38(4): 694-704, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23601684

ABSTRACT

Group 2 innate lymphoid cells (ILC2) are innate lymphocytes that confer protective type 2 immunity during helminth infection and are also involved in allergic airway inflammation. Here we report that ILC2 development required T cell factor 1 (TCF-1, the product of the Tcf7 gene), a transcription factor also implicated in T cell lineage specification. Tcf7(-/-) mice lack ILC2, and were unable to mount ILC2-mediated innate type 2 immune responses. Forced expression of TCF-1 in bone marrow progenitors partially bypassed the requirement for Notch signaling in the generation of ILC2 in vivo. TCF-1 acted through both GATA-3-dependent and GATA-3-independent pathways to promote the generation of ILC2. These results are reminiscent of the critical roles of TCF-1 in early T cell development. Hence, transcription factors that underlie early steps of T cell development are also implicated in the development of innate lymphoid cells.


Subject(s)
Asthma/immunology , Bone Marrow Cells/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphocytes/immunology , Nippostrongylus/immunology , Strongylida Infections/immunology , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Hepatocyte Nuclear Factor 1-alpha/genetics , Immunity, Innate , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Transgenes/genetics
11.
Sci Transl Med ; 5(170): 170ra16, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23363980

ABSTRACT

Innate lymphoid cells (ILCs) are a recently identified family of heterogeneous immune cells that can be divided into three groups based on their differential developmental requirements and expression of effector cytokines. Among these, group 2 ILCs produce the type 2 cytokines interleukin-5 (IL-5) and IL-13 and promote type 2 inflammation in the lung and intestine. However, whether group 2 ILCs reside in the skin and contribute to skin inflammation has not been characterized. We identify a population of skin-resident group 2 ILCs present in healthy human skin that are enriched in lesional human skin from atopic dermatitis (AD) patients. Group 2 ILCs were also found in normal murine skin and were critical for the development of inflammation in a murine model of AD-like disease. Remarkably, in contrast to group 2 ILC responses in the intestine and lung, which are critically regulated by IL-33 and IL-25, group 2 ILC responses in the skin and skin-draining lymph nodes were independent of these canonical cytokines but were critically dependent on thymic stromal lymphopoietin (TSLP). Collectively, these results demonstrate an essential role for IL-33- and IL-25-independent group 2 ILCs in promoting skin inflammation.


Subject(s)
Cytokines/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Interleukins/metabolism , Lymphocytes/immunology , Skin/pathology , Adaptive Immunity , Animals , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Disease Models, Animal , Humans , Inflammation/pathology , Interleukin-33 , Mice , Mice, Inbred C57BL , Protein Binding , Skin/immunology , Thymic Stromal Lymphopoietin
12.
J Immunol ; 187(11): 5505-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22025549

ABSTRACT

Natural helper (NH) cells are recently discovered innate immune cells that confer protective type 2 immunity during helminth infection and mediate influenza-induced airway hypersensitivity. Little is known about the ontogeny of NH cells. We report in this study that NH cells derive from bone marrow lymphoid progenitors. Using RAG-1Cre/ROSA26(YFP) mice, we show that most NH cells are marked with a history of RAG-1 expression, implying lymphoid developmental origin. The development of NH cells depends on the cytokine receptor Flt3, which is required for the efficient generation of bone marrow lymphoid progenitors. Finally, we demonstrate that lymphoid progenitors, but not myeloid-erythroid progenitors, give rise to NH cells in vivo. This work therefore expands the lymphocyte family, currently comprising T, B, and NK cells, to include NH cells as another type of innate lymphocyte that derives from bone marrow lymphoid progenitors.


Subject(s)
Cell Lineage/immunology , Hematopoietic Stem Cells/cytology , Immunity, Innate , Lymphocyte Subsets/cytology , Lymphocytes/cytology , Animals , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/immunology , Lung/cytology , Lung/immunology , Lymphocyte Subsets/immunology , Lymphocytes/immunology , Mice , Mice, Mutant Strains , Polymerase Chain Reaction
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.
Trends Immunol ; 31(11): 407-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20951092

ABSTRACT

The recent identification of previously unrecognized innate cell populations, termed natural helper cells (NHCs), multi-potent progenitor type 2 (MPP(type2)) cells, nuocytes, and innate type 2 helper (Ih2) cells has provided new insights into our understanding of the cellular mechanisms that lead to the development of CD4(+) Th2 cell-dependent immunity and/or inflammation at mucosal sites. In this review, we focus on the functional significance, similarities, and differences between NHCs, MPP(type2) cells, nuocytes and Ih2 cells. All four cell populations are activated by interleukin (IL)-25 and/or IL-33 and are capable of promoting Th2 cytokine responses. Collectively, the identification of these cell populations might illuminate ancient evolutionary conserved pathways that are involved in the development of Th2 cytokine responses, and could be of benefit in the development of therapeutic approaches that target helminth infections and allergic diseases.


Subject(s)
Cell Differentiation , Cytokines/immunology , Immunity, Innate , Th2 Cells/immunology , Animals , Humans , Th2 Cells/cytology
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.
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
17.
J Exp Med ; 205(10): 2191-8, 2008 Sep 29.
Article in English | MEDLINE | ID: mdl-18762568

ABSTRACT

Alterations in the composition of intestinal commensal bacteria are associated with enhanced susceptibility to multiple inflammatory diseases, including those conditions associated with interleukin (IL)-17-producing CD4(+) T helper (Th17) cells. However, the relationship between commensal bacteria and the expression of proinflammatory cytokines remains unclear. Using germ-free mice, we show that the frequency of Th17 cells in the large intestine is significantly elevated in the absence of commensal bacteria. Commensal-dependent expression of the IL-17 family member IL-25 (IL-17E) by intestinal epithelial cells limits the expansion of Th17 cells in the intestine by inhibiting expression of macrophage-derived IL-23. We propose that acquisition of, or alterations in, commensal bacteria influences intestinal immune homeostasis via direct regulation of the IL-25-IL-23-IL-17 axis.


Subject(s)
Interleukin-17/metabolism , Interleukin-23/metabolism , Interleukins/metabolism , Intestines/immunology , Intestines/microbiology , Animals , Epithelial Cells/immunology , Epithelial Cells/microbiology , Germ-Free Life , Homeostasis , Interleukin-17/genetics , Interleukin-23/genetics , Interleukins/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestines/anatomy & histology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Symbiosis , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology
18.
Immunol Rev ; 226: 172-90, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19161424

ABSTRACT

There is compelling evidence that epithelial cells (ECs) at mucosal surfaces, beyond their role in creating a physical barrier, are integral components of innate and adaptive immunity. The capacity of these cells to license the functions of specific immune cell populations in the airway and gastrointestinal tract offers the prospect of novel therapeutic strategies to target multiple inflammatory diseases in which barrier immunity is dysregulated. In this review, we discuss the critical functions of EC-derived thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and IL-33 in the development and regulation of T-helper 2 (Th2) cytokine-dependent immune responses. We first highlight recent data that have provided new insights into the factors that control expression of this triad of cytokines and their receptors. In addition, we review their proinflammatory and immunoregulatory functions in models of mucosal infection and inflammation. Lastly, we discuss new findings indicating that despite their diverse structural features and differential expression of their receptors, TSLP, IL-25, and IL-33 cross-regulate one another and share overlapping properties that influence Th2 cytokine-dependent responses at mucosal sites.


Subject(s)
Cytokines/immunology , Epithelial Cells/immunology , Helminthiasis/immunology , Interleukin-17/immunology , Interleukins/immunology , Animals , Humans , Immunity, Active , Immunity, Innate , Interleukin-33 , Mucous Membrane/immunology , Mucous Membrane/parasitology , Signal Transduction/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Thymic Stromal Lymphopoietin
19.
Proc Natl Acad Sci U S A ; 103(32): 12011-6, 2006 Aug 08.
Article in English | MEDLINE | ID: mdl-16877545

ABSTRACT

The phylogenetic enigma of snail hemoglobin, its isolated occurrence in a single gastropod family, the Planorbidae, and the lack of sequence data, stimulated the present study. We present here the complete cDNA and predicted amino acid sequence of two hemoglobin polypeptides from the planorbid Biomphalaria glabrata (intermediate host snail for the human parasite Schistosoma mansoni). Both isoforms contain 13 different, cysteine-free globin domains, plus a small N-terminal nonglobin "plug" domain with three cysteines for subunit dimerization (total M(r) approximately 238 kDa). We also identified the native hemoglobin molecule and present here a preliminary 3D reconstruction from electron microscopical images (3 nm resolution); it suggests a 3 x 2-mer quaternary structure (M(r) approximately 1.43 MDa). Moreover, we identified a previously undescribed rosette-like hemolymph protein that has been mistaken for hemoglobin. We also detected expression of an incomplete hemocyanin as trace component. The combined data show that B. glabrata hemoglobin evolved from pulmonate myoglobin, possibly to replace a less-efficient hemocyanin, and reveals a surprisingly simple evolutionary mechanism to create a high molecular mass respiratory protein from 78 similar globin domains.


Subject(s)
Hemoglobins/chemistry , Amino Acid Sequence , Animals , Biomphalaria , Evolution, Molecular , Hemoglobins/genetics , Hemolymph/chemistry , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Molecular Sequence Data , Phylogeny , Protein Isoforms , Protein Structure, Tertiary , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...