Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Immunity ; 43(1): 187-99, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26200014

ABSTRACT

The role of intestinal eosinophils in immune homeostasis is enigmatic and the molecular signals that drive them from protective to tissue damaging are unknown. Most commonly associated with Th2 cell-mediated diseases, we describe a role for eosinophils as crucial effectors of the interleukin-23 (IL-23)-granulocyte macrophage colony-stimulating factor (GM-CSF) axis in colitis. Chronic intestinal inflammation was characterized by increased bone marrow eosinopoiesis and accumulation of activated intestinal eosinophils. IL-5 blockade or eosinophil depletion ameliorated colitis, implicating eosinophils in disease pathogenesis. GM-CSF was a potent activator of eosinophil effector functions and intestinal accumulation, and GM-CSF blockade inhibited chronic colitis. By contrast neutrophil accumulation was GM-CSF independent and dispensable for colitis. In addition to TNF secretion, release of eosinophil peroxidase promoted colitis identifying direct tissue-toxic mechanisms. Thus, eosinophils are key perpetrators of chronic inflammation and tissue damage in IL-23-mediated immune diseases and it suggests the GM-CSF-eosinophil axis as an attractive therapeutic target.


Subject(s)
Colitis/immunology , Eosinophils/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-23 Subunit p19/immunology , Animals , Cell Movement/immunology , Cytokine Receptor Common beta Subunit/genetics , Eosinophil Peroxidase/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Inflammation/immunology , Interleukin-5/antagonists & inhibitors , Intestines/cytology , Intestines/immunology , Intestines/pathology , Leukocyte Reduction Procedures , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Tumor Necrosis Factors/metabolism
3.
Immunity ; 37(6): 1116-29, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23200826

ABSTRACT

In interleukin-23 (IL-23)-dependent colitis, there is excessive accumulation of short-lived neutrophils and inflammatory monocytes in the intestine. It is unknown whether this reflects changes in mature cell populations or whether the IL-23-driven colitogenic T cell program regulates upstream hematopoietic stem and progenitor cells (HSPC). Here we have shown dysregulation of hematopoiesis in colitis mediated by inflammatory cytokines. First, there was an interferon-gamma-dependent accumulation of proliferating hematopoietic stem cells in the bone marrow and spleen. Second, there was a strong skew toward granulocyte-monocyte progenitor (GMP) production at the expense of erythroid and lymphoid progenitors. Extramedullary hematopoiesis was also evident, and granulocyte macrophage-colony stimulating factor (GM-CSF) blockade reduced the accumulation of splenic and colonic GMPs, resulting in amelioration of colitis. Importantly, transfer of GMPs exacerbated colitis. These data identify HSPCs as a major target of the IL-23-driven inflammatory axis suggesting therapeutic strategies for the treatment of inflammatory bowel disease.


Subject(s)
Colitis/immunology , Colitis/metabolism , Hematopoietic Stem Cells/metabolism , Interleukin-23/physiology , Animals , Bone Marrow Cells/metabolism , Cell Lineage , Cell Proliferation , Colitis/chemically induced , Disease Models, Animal , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoiesis, Extramedullary , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-23/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/physiology , Mice , Mice, Knockout , Spleen/physiology
4.
Nature ; 513(7519): 564-568, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25043027

ABSTRACT

FOXP3(+) regulatory T cells (Treg cells) are abundant in the intestine, where they prevent dysregulated inflammatory responses to self and environmental stimuli. It is now appreciated that Treg cells acquire tissue-specific adaptations that facilitate their survival and function; however, key host factors controlling the Treg response in the intestine are poorly understood. The interleukin (IL)-1 family member IL-33 is constitutively expressed in epithelial cells at barrier sites, where it functions as an endogenous danger signal, or alarmin, in response to tissue damage. Recent studies in humans have described high levels of IL-33 in inflamed lesions of inflammatory bowel disease patients, suggesting a role for this cytokine in disease pathogenesis. In the intestine, both protective and pathological roles for IL-33 have been described in murine models of acute colitis, but its contribution to chronic inflammation remains ill defined. Here we show in mice that the IL-33 receptor ST2 is preferentially expressed on colonic Treg cells, where it promotes Treg function and adaptation to the inflammatory environment. IL-33 signalling in T cells stimulates Treg responses in several ways. First, it enhances transforming growth factor (TGF)-ß1-mediated differentiation of Treg cells and, second, it provides a necessary signal for Treg-cell accumulation and maintenance in inflamed tissues. Strikingly, IL-23, a key pro-inflammatory cytokine in the pathogenesis of inflammatory bowel disease, restrained Treg responses through inhibition of IL-33 responsiveness. These results demonstrate a hitherto unrecognized link between an endogenous mediator of tissue damage and a major anti-inflammatory pathway, and suggest that the balance between IL-33 and IL-23 may be a key controller of intestinal immune responses.


Subject(s)
Interleukins/immunology , Intestines/cytology , Intestines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Colitis/immunology , Colitis/pathology , Colon/cytology , Colon/immunology , Colon/pathology , Disease Models, Animal , Female , Immunity, Mucosal , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-23/immunology , Interleukin-33 , Interleukins/antagonists & inhibitors , Interleukins/metabolism , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Transforming Growth Factor beta/metabolism
5.
Immunity ; 30(2): 289-99, 2009 Feb 20.
Article in English | MEDLINE | ID: mdl-19217323

ABSTRACT

Invariant natural killer T (iNKT) cells promote immune responses to various pathogens, but exactly how iNKT cells control antiviral responses is unclear. Here, we showed that iNKT cells induced tissue-specific antiviral effects in mice infected by lymphocytic choriomeningitis virus (LCMV). Indeed, iNKT cells inhibited viral replication in the pancreas and liver but not in the spleen. In the pancreas, iNKT cells expressed the OX40 molecule and promoted type I interferon (IFN) production by plasmacytoid dendritic cells (pDCs) through OX40-OX40 ligand interaction. Subsequently, this iNKT cell-pDC cooperation attenuated the antiviral adaptive immune response in the pancreas but not in the spleen. The dampening of pancreatic anti-LCMV CD8(+) T cell response prevented tissue damage in transgenic mice expressing LCMV protein in islet beta cells. Thus, this study identifies pDCs as an essential partner of iNKT cells for mounting an efficient, nondeleterious antiviral response in peripheral tissue.


Subject(s)
Dendritic Cells/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Natural Killer T-Cells/immunology , Receptors, OX40/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus/etiology , Diabetes Mellitus/immunology , Diabetes Mellitus/virology , Liver/immunology , Liver/virology , Lymphocytic Choriomeningitis/complications , Mice , OX40 Ligand/immunology , Organ Specificity/immunology , Pancreas/immunology , Pancreas/virology , Signal Transduction/immunology , Spleen/immunology , Spleen/virology , Virus Replication
6.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32520308

ABSTRACT

An important comorbidity of chronic inflammation is anemia, which may be related to dysregulated activity of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow (BM). Among HSPCs, we found that the receptor for IL-33, ST2, is expressed preferentially and highly on erythroid progenitors. Induction of inflammatory spondyloarthritis in mice increased IL-33 in BM plasma, and IL-33 was required for inflammation-dependent suppression of erythropoiesis in BM. Conversely, administration of IL-33 in healthy mice suppressed erythropoiesis, decreased hemoglobin expression, and caused anemia. Using purified erythroid progenitors in vitro, we show that IL-33 directly inhibited terminal maturation. This effect was dependent on NF-κB activation and associated with altered signaling events downstream of the erythropoietin receptor. Accordingly, IL-33 also suppressed erythropoietin-accelerated erythropoiesis in vivo. These results reveal a role for IL-33 in pathogenesis of anemia during inflammatory disease and define a new target for its treatment.


Subject(s)
Anemia/pathology , Cell Differentiation , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/pathology , Inflammation/pathology , Interleukin-33/metabolism , Anemia/complications , Animals , Annexin A5/metabolism , Bone Marrow/pathology , Chronic Disease , Erythropoiesis , Erythropoietin/pharmacology , Hematopoiesis , Inflammation/complications , Injections , Interleukin-1 Receptor-Like 1 Protein/metabolism , Ki-67 Antigen/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Myelopoiesis , NF-kappa B/metabolism , Phosphorylation , Receptors, Erythropoietin/metabolism , Signal Transduction , Spondylarthritis/pathology , beta-Glucans
7.
Nat Commun ; 11(1): 155, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31919358

ABSTRACT

Dysregulated hematopoiesis occurs in several chronic inflammatory diseases, but it remains unclear how hematopoietic stem cells (HSCs) in the bone marrow (BM) sense peripheral inflammation and contribute to tissue damage in arthritis. Here, we show the HSC gene expression program is biased toward myelopoiesis and differentiation skewed toward granulocyte-monocyte progenitors (GMP) during joint and intestinal inflammation in experimental spondyloarthritis (SpA). GM-CSF-receptor is increased on HSCs and multipotent progenitors, favoring a striking increase in myelopoiesis at the earliest hematopoietic stages. GMP accumulate in the BM in SpA and, unexpectedly, at extramedullary sites: in the inflamed joints and spleen. Furthermore, we show that GM-CSF promotes extramedullary myelopoiesis, tissue-toxic neutrophil accumulation in target organs, and GM-CSF prophylactic or therapeutic blockade substantially decreases SpA severity. Surprisingly, besides CD4+ T cells and innate lymphoid cells, mast cells are a source of GM-CSF in this model, and its pathogenic production is promoted by the alarmin IL-33.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoiesis, Extramedullary/physiology , Hematopoietic Stem Cells/metabolism , Myelopoiesis/physiology , Spondylarthritis/pathology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cells, Cultured , Female , Interleukin-33/immunology , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Spondylarthritis/immunology
8.
Int Rev Immunol ; 26(1-2): 49-72, 2007.
Article in English | MEDLINE | ID: mdl-17454264

ABSTRACT

Type 1 diabetes is an autoimmune disease due to the destruction of insulin-producing pancreatic beta cells. Natural Killer T (NKT) cells are a T-cell subset that links the innate and adaptive immune systems. NKT cells play a key regulatory role in type 1 diabetes. The absence of NKT cells correlates with exacerbation of type 1 diabetes, whereas an increased frequency and/or activation of NKT cells prevents beta-cell autoimmunity. Various mechanisms are involved in the protective effect of NKT cells. The goal is now to translate knowledge gained from mouse models into human therapeutics.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/immunology , Animals , Disease Models, Animal , Humans , Islets of Langerhans/immunology , Mice , Mice, Inbred NOD
9.
Elife ; 5: e10066, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26780670

ABSTRACT

Innate lymphoid cells (ILCs) contribute to host defence and tissue repair but can induce immunopathology. Recent work has revealed tissue-specific roles for ILCs; however, the question of how a small population has large effects on immune homeostasis remains unclear. We identify two mechanisms that ILC3s utilise to exert their effects within intestinal tissue. ILC-driven colitis depends on production of granulocyte macrophage-colony stimulating factor (GM-CSF), which recruits and maintains intestinal inflammatory monocytes. ILCs present in the intestine also enter and exit cryptopatches in a highly dynamic process. During colitis, ILC3s mobilize from cryptopatches, a process that can be inhibited by blocking GM-CSF, and mobilization precedes inflammatory foci elsewhere in the tissue. Together these data identify the IL-23R/GM-CSF axis within ILC3 as a key control point in the accumulation of innate effector cells in the intestine and in the spatio-temporal dynamics of ILCs in the intestinal inflammatory response.


Subject(s)
Colitis/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate , Intestines/immunology , Lymphocytes/immunology , Humans , Interleukin-23 Subunit p19/metabolism
10.
J Exp Med ; 207(4): 699-709, 2010 Apr 12.
Article in English | MEDLINE | ID: mdl-20368580

ABSTRACT

The immune response in the gastrointestinal tract is a tightly controlled balance between effector and regulatory cell responses. Here, we have investigated the role of OX40 in influencing the balance between conventional T cells and Foxp3+ regulatory T (T reg) cells. Under steady-state conditions, OX40 was required by T reg cells for their accumulation in the colon, but not peripheral lymphoid organs. Strikingly, under inflammatory conditions OX40 played an essential role in T reg cell-mediated suppression of colitis. OX40(-/-) T reg cells showed reduced accumulation in the colon and peripheral lymphoid organs, resulting in their inability to keep pace with the effector response. In the absence of OX40 signaling, T reg cells underwent enhanced activation-induced cell death, indicating that OX40 delivers an important survival signal to T reg cells after activation. As OX40 also promoted the colitogenic Th1 response, its expression on T reg cells may be required for effective competition with OX40-dependent effector responses. These results newly identify a key role for OX40 in the homeostasis of intestinal Foxp3+ T reg cells and in suppression of colitis. These fi ndings should be taken into account when considering OX40 blockade for treatment of IBD.


Subject(s)
Colitis/immunology , Receptors, OX40/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Count , Cell Movement/immunology , Cell Proliferation , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression/genetics , Gene Expression/immunology , Homeodomain Proteins/genetics , Integrins/metabolism , Interferon-gamma/metabolism , Interleukin-17/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Leukocyte Common Antigens/genetics , Lymphocyte Activation/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , OX40 Ligand , Receptors, OX40/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Tumor Necrosis Factors/genetics
11.
J Immunol ; 178(3): 1332-40, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17237379

ABSTRACT

Invariant NKT (iNKT) cells can prevent diabetes by inhibiting the differentiation of anti-islet T cells. We recently showed that neither iNKT cell protection against diabetes nor iNKT cell inhibition of T cell differentiation in vitro requires cytokines such as IL-4, IL-10, IL-13, and TGF-beta. In contrast, cell-cell contacts were required for iNKT cell inhibition of T cell differentiation in vitro. The present study was designed to determine whether the CD1d molecule is involved in the inhibitory function of iNKT cells. Experiments were performed in vitro and in vivo, using cells lacking CD1d expression. The in vivo experiments used CD1d-deficient mice that were either reconstituted with iNKT cells or expressed a CD1d transgene exclusively in the thymus. Both mouse models had functional iNKT cells in the periphery, even though CD1d was not expressed in peripheral tissues. Surprisingly, both in vitro inhibition of T cell differentiation by iNKT cells and mouse protection against diabetes by iNKT cells were CD1d-independent. These results reveal that iNKT cells can exert critical immunoregulatory effects in the absence of CD1d recognition and that different molecular interactions are involved in iNKT cell functions.


Subject(s)
Antigens, CD1/genetics , Diabetes Mellitus, Type 1/prevention & control , Killer Cells, Natural/immunology , Animals , Antigens, CD1d , Cell Differentiation , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Mice , Mice, Knockout , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland , Transgenes
12.
J Immunol ; 174(4): 1954-61, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699123

ABSTRACT

NKT cells are potent regulatory T cells that prevent the development of several autoimmune diseases. Analysis of NKT cell regulatory function in the NOD mouse has revealed that NKT cells inhibit the development of type 1 diabetes by impairing the differentiation of anti-islet T cells into Th1 effector cells. In the present study, we have performed in vitro and in vivo experiments to determine the respective role of cytokines and cell contacts in the blockade of T cell differentiation by NKT cells. These experiments reveal that cytokines such as IL-4, IL-10, IL-13, and TGF-beta, that have been involved in other functions of NKT cells, play only a minor role if any in the blockade of T cell differentiation by NKT cells. Diabetes is still prevented by NKT cells in the absence of functional IL-4, IL-10, IL-13, and TGF-beta. In contrast, we show for the first time that cell contacts are crucial for the immunoregulatory function of NKT cells.


Subject(s)
Cell Communication/immunology , Cell Differentiation/immunology , Immunosuppression Therapy , Killer Cells, Natural/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Animals , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Interleukin-10/physiology , Interleukin-13/physiology , Interleukin-4/physiology , Killer Cells, Natural/cytology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Transforming Growth Factor beta/physiology
13.
J Immunol ; 175(4): 2091-101, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16081775

ABSTRACT

Invariant NKT (iNKT) cells have been implicated in the regulation of autoimmune diseases. In several models of type 1 diabetes, increasing the number of iNKT cells prevents the development of disease. Because CD8 T cells play a crucial role in the pathogenesis of diabetes, we have investigated the influence of iNKT cells on diabetogenic CD8 T cells. In the present study, type 1 diabetes was induced by the transfer of CD8 T cells specific for the influenza virus hemagglutinin into recipient mice expressing the hemagglutinin Ag specifically in their beta pancreatic cells. In contrast to previous reports, high frequency of iNKT cells promoted severe insulitis and exacerbated diabetes. Analysis of diabetogenic CD8 T cells showed that iNKT cells enhance their activation, their expansion, and their differentiation into effector cells producing IFN-gamma. This first analysis of the influence of iNKT cells on diabetogenic CD8 T cells reveals that iNKT cells not only fail to regulate but in fact exacerbate the development of diabetes. Thus, iNKT cells can induce opposing effects dependent on the model of type 1 diabetes that is being studied. This prodiabetogenic capacity of iNKT cells should be taken into consideration when developing therapeutic approaches based on iNKT cell manipulation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Diabetes Mellitus, Type 1/immunology , Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Epitopes, T-Lymphocyte/immunology , Hindlimb , Injections, Intravenous , Interferon-gamma/biosynthesis , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Killer Cells, Natural/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Activation/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL