Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 176(5): 967-981.e19, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30739797

ABSTRACT

Tissue-resident lymphocytes play a key role in immune surveillance, but it remains unclear how these inherently stable cell populations respond to chronic inflammation. In the setting of celiac disease (CeD), where exposure to dietary antigen can be controlled, gluten-induced inflammation triggered a profound depletion of naturally occurring Vγ4+/Vδ1+ intraepithelial lymphocytes (IELs) with innate cytolytic properties and specificity for the butyrophilin-like (BTNL) molecules BTNL3/BTNL8. Creation of a new niche with reduced expression of BTNL8 and loss of Vγ4+/Vδ1+ IELs was accompanied by the expansion of gluten-sensitive, interferon-γ-producing Vδ1+ IELs bearing T cell receptors (TCRs) with a shared non-germline-encoded motif that failed to recognize BTNL3/BTNL8. Exclusion of dietary gluten restored BTNL8 expression but was insufficient to reconstitute the physiological Vγ4+/Vδ1+ subset among TCRγδ+ IELs. Collectively, these data show that chronic inflammation permanently reconfigures the tissue-resident TCRγδ+ IEL compartment in CeD. VIDEO ABSTRACT.


Subject(s)
Celiac Disease/immunology , Inflammation/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Antigens , Butyrophilins/metabolism , Celiac Disease/physiopathology , Chronic Disease , Diet, Gluten-Free , Glutens/metabolism , HEK293 Cells , Humans , Inflammation/metabolism , Intestinal Mucosa/immunology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism
2.
Nature ; 578(7796): 600-604, 2020 02.
Article in English | MEDLINE | ID: mdl-32051586

ABSTRACT

Coeliac disease is a complex, polygenic inflammatory enteropathy caused by exposure to dietary gluten that occurs in a subset of genetically susceptible individuals who express either the HLA-DQ8 or HLA-DQ2 haplotypes1,2. The need to develop non-dietary treatments is now widely recognized3, but no pathophysiologically relevant gluten- and HLA-dependent preclinical model exists. Furthermore, although studies in humans have led to major advances in our understanding of the pathogenesis of coeliac disease4, the respective roles of disease-predisposing HLA molecules, and of adaptive and innate immunity in the development of tissue damage, have not been directly demonstrated. Here we describe a mouse model that reproduces the overexpression of interleukin-15 (IL-15) in the gut epithelium and lamina propria that is characteristic of active coeliac disease, expresses the predisposing HLA-DQ8 molecule, and develops villous atrophy after ingestion of gluten. Overexpression of IL-15 in both the epithelium and the lamina propria is required for the development of villous atrophy, which demonstrates the location-dependent central role of IL-15 in the pathogenesis of coeliac disease. In addition, CD4+ T cells and HLA-DQ8 have a crucial role in the licensing of cytotoxic T cells to mediate intestinal epithelial cell lysis. We also demonstrate a role for the cytokine interferon-γ (IFNγ) and the enzyme transglutaminase 2 (TG2) in tissue destruction. By reflecting the complex interaction between gluten, genetics and IL-15-driven tissue inflammation, this mouse model provides the opportunity to both increase our understanding of coeliac disease, and develop new therapeutic strategies.


Subject(s)
Celiac Disease/immunology , Celiac Disease/pathology , Glutens/immunology , HLA-DQ Antigens/immunology , Interleukin-15/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Female , HLA-DQ Antigens/genetics , Humans , Interferon-gamma/immunology , Interleukin-15/genetics , Male , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism
3.
Cell Commun Signal ; 22(1): 23, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38195510

ABSTRACT

Cerebral cavernous malformation (CCM) is a hemorrhagic neurovascular disease with no currently available therapeutics. Prior evidence suggests that different cell types may play a role in CCM pathogenesis. The contribution of each cell type to the dysfunctional cellular crosstalk remains unclear. Herein, RNA-seq was performed on fluorescence-activated cell sorted endothelial cells (ECs), pericytes, and neuroglia from CCM lesions and non-lesional brain tissue controls. Differentially Expressed Gene (DEG), pathway and Ligand-Receptor (LR) analyses were performed to characterize the dysfunctional genes of respective cell types within CCMs. Common DEGs among all three cell types were related to inflammation and endothelial-to-mesenchymal transition (EndMT). DEG and pathway analyses supported a role of lesional ECs in dysregulated angiogenesis and increased permeability. VEGFA was particularly upregulated in pericytes. Further pathway and LR analyses identified vascular endothelial growth factor A/ vascular endothelial growth factor receptor 2 signaling in lesional ECs and pericytes that would result in increased angiogenesis. Moreover, lesional pericytes and neuroglia predominantly showed DEGs and pathways mediating the immune response. Further analyses of cell specific gene alterations in CCM endorsed potential contribution to EndMT, coagulation, and a hypoxic microenvironment. Taken together, these findings motivate mechanistic hypotheses regarding non-endothelial contributions to lesion pathobiology and may lead to novel therapeutic targets. Video Abstract.


Subject(s)
Hemangioma, Cavernous, Central Nervous System , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/genetics , Hemangioma, Cavernous, Central Nervous System/genetics , Endothelial Cells , Gene Expression Profiling , Transcriptome , Tumor Microenvironment
4.
Gastroenterology ; 158(3): 625-637.e13, 2020 02.
Article in English | MEDLINE | ID: mdl-31622625

ABSTRACT

BACKGROUND & AIMS: Gamma chain (γc) cytokines (interleukin [IL]2, IL4, IL7, IL9, IL15, and IL21) signal via a common γc receptor. IL2 regulates the immune response, whereas IL21 and IL15 contribute to development of autoimmune disorders, including celiac disease. We investigated whether BNZ-2, a peptide designed to inhibit IL15 and IL21, blocks these cytokines selectively and its effects on intraepithelial cytotoxic T cells. METHODS: We obtained duodenal biopsies from 9 patients with potential celiac disease (positive results from tests for anti-TG2 but no villous atrophy), 30 patients with untreated celiac disease (with villous atrophy), and 5 patients with treated celiac disease (on a gluten-free diet), as well as 43 individuals without celiac disease (controls). We stimulated primary intestinal intraepithelial CD8+ T-cell lines, or CD8+ T cells directly isolated from intestinal biopsies, with γc cytokines in presence or absence of BNZ-2. Cells were analyzed by immunoblots, flow cytometry, or RNA-sequencing analysis for phosphorylation of signaling molecules, gene expression profiles, proliferation, and levels of granzyme B. RESULTS: Duodenal tissues from patients with untreated celiac disease had increased levels of messenger RNAs encoding IL15 receptor subunit alpha (IL15RA) and IL21 compared with tissues from patients with potential celiac disease and controls. Activation of intraepithelial cytotoxic T cells with IL15 or IL21 induced separate signaling pathways; incubation of the cells with IL15 and IL21 cooperatively increased their transcriptional activity, proliferation, and cytolytic properties. BNZ-2 specifically inhibited the effects of IL15 and IL21, but not of other γc cytokines. CONCLUSIONS: We found increased expression of IL15RA and IL21 in duodenal tissues from patients with untreated celiac disease compared with controls. IL15 and IL21 cooperatively activated intestinal intraepithelial cytotoxic T cells. In particular, they increased their transcriptional activity, proliferation, and cytolytic activity. The peptide BNZ-2 blocked these effects, but not those of other γc cytokines, including IL2. BNZ-2 might be used to prevent cytotoxic T-cell-mediated tissue damage in complex immune disorders exhibiting upregulation of IL15 and IL21.


Subject(s)
Benzodiazepines/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/physiology , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Interleukin-15/pharmacology , Interleukins/pharmacology , Case-Control Studies , Celiac Disease/immunology , Cell Line , Cell Proliferation/drug effects , Cellular Reprogramming/drug effects , Duodenum/pathology , Humans , Interleukin-15/genetics , Interleukins/genetics , Primary Cell Culture , RNA, Messenger , Receptors, Interleukin-15/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects
5.
J Autoimmun ; 108: 102422, 2020 03.
Article in English | MEDLINE | ID: mdl-32033836

ABSTRACT

The respective effects of tissue alarmins interleukin (IL)-15 and interferon beta (IFNß), and IL-21 produced by T cells on the reprogramming of cytotoxic T lymphocytes (CTLs) that cause tissue destruction in celiac disease is poorly understood. Transcriptomic and epigenetic profiling of primary intestinal CTLs showed massive and distinct temporal transcriptional changes in response to tissue alarmins, while the impact of IL-21 was limited. Only anti-viral pathways were induced in response to all the three stimuli, albeit with differences in dynamics and strength. Moreover, changes in gene expression were primarily independent of changes in H3K27ac, suggesting that other regulatory mechanisms drive the robust transcriptional response. Finally, we found that IL-15/IFNß/IL-21 transcriptional signatures could be linked to transcriptional alterations in risk loci for complex immune diseases. Together these results provide new insights into molecular mechanisms that fuel the activation of CTLs under conditions that emulate the inflammatory environment in patients with autoimmune diseases.


Subject(s)
Alarmins/metabolism , Cytokines/metabolism , Gene Expression Regulation , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Autoimmunity , Celiac Disease/etiology , Celiac Disease/metabolism , Celiac Disease/pathology , Gene Expression Profiling , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interleukin-15/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Promoter Regions, Genetic
6.
Gastroenterology ; 149(3): 681-91.e10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26001928

ABSTRACT

BACKGROUND & AIMS: The mechanisms of tissue destruction during progression of celiac disease are poorly defined. It is not clear how tissue stress and adaptive immunity contribute to the activation of intraepithelial cytotoxic T cells and the development of villous atrophy. We analyzed epithelial cells and intraepithelial cytotoxic T cells in family members of patients with celiac disease, who were without any signs of adaptive antigluten immunity, and in potential celiac disease patients, who have antibodies against tissue transglutaminase 2 in the absence of villous atrophy. METHODS: We collected blood and intestinal biopsy specimens from 268 patients at tertiary medical centers in the United States and Italy from 2004 to 2012. All subjects had normal small intestinal histology. Study groups included healthy individuals with no family history of celiac disease or antibodies against tissue transglutaminase 2 (controls), healthy family members of patients with celiac disease, and potential celiac disease patients. Intraepithelial cytotoxic T cells were isolated and levels of inhibitory and activating natural killer (NK) cells were measured by flow cytometry. Levels of heat shock protein (HSP) and interleukin 15 were measured by immunohistochemistry, and ultrastructural alterations in intestinal epithelial cells (IECs) were assessed by electron microscopy. RESULTS: IECs from subjects with a family history of celiac disease, but not from subjects who already had immunity to gluten, expressed higher levels of HS27, HSP70, and interleukin-15 than controls; their IECs also had ultrastructural alterations. Intraepithelial cytotoxic T cells from relatives of patients with celiac disease expressed higher levels of activating NK receptors than cells from controls, although at lower levels than patients with active celiac disease, and without loss of inhibitory receptors for NK cells. Intraepithelial cytotoxic T cells from potential celiac disease patients failed to up-regulate activating NK receptors. CONCLUSIONS: A significant subset of healthy family members of patients with celiac disease with normal intestinal architecture had epithelial alterations, detectable by immunohistochemistry and electron microscopy. The adaptive immune response to gluten appears to act in synergy with epithelial stress to allow intraepithelial cytotoxic T cells to kill epithelial cells and induce villous atrophy in patients with active celiac disease.


Subject(s)
Adaptive Immunity , Celiac Disease/immunology , Cell Communication , Epithelial Cells/immunology , Intestinal Mucosa/immunology , Intestine, Small/immunology , Stress, Physiological , T-Lymphocytes, Cytotoxic/immunology , Autoantibodies/blood , Case-Control Studies , Celiac Disease/blood , Celiac Disease/pathology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , GTP-Binding Proteins/immunology , HSP27 Heat-Shock Proteins/immunology , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/immunology , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Interleukin-15/immunology , Interleukin-15/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/ultrastructure , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Italy , Molecular Chaperones , Phenotype , Protein Glutamine gamma Glutamyltransferase 2 , Risk Factors , Signal Transduction , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/ultrastructure , Transglutaminases/immunology , United States
7.
J Immunol ; 193(2): 610-6, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24935923

ABSTRACT

The NKG2 family of NK receptors includes activating and inhibitory members. With the exception of the homodimer-forming NKG2D, NKG2 receptors recognize the nonclassical MHC class I molecule HLA-E, and they can be subdivided into two groups: those that associate with and signal through DAP12 to activate cells, and those that contain an ITIM motif to promote inhibition. The function of NKG2 family member NKG2E is unclear in humans, and its surface expression has never been conclusively established, largely because there is no Ab that binds specifically to NKG2E. Seeking to determine a role for this molecule, we chose to investigate its expression and ability to form complexes with intracellular signaling molecules. We found that NKG2E was capable of associating with CD94 and DAP12 but that the complex was retained intracellularly at the endoplasmic reticulum instead of being expressed on cell surfaces, and that this localization was dependent on a sequence of hydrophobic amino acids in the extracellular domain of NKG2E. Because this particular sequence has emerged and been conserved selectively among higher order primates evolutionarily, this observation raises the intriguing possibility that NKG2E may function as an intracellular protein.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoplasm/metabolism , Membrane Proteins/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/genetics , Amino Acids/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/genetics , Microscopy, Confocal , Molecular Sequence Data , Mutation , NK Cell Lectin-Like Receptor Subfamily C/classification , NK Cell Lectin-Like Receptor Subfamily C/genetics , NK Cell Lectin-Like Receptor Subfamily D/genetics , Phylogeny , Protein Binding , Sequence Homology, Amino Acid
8.
Proc Natl Acad Sci U S A ; 110(20): 8158-63, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23637340

ABSTRACT

A major challenge of cancer immunotherapy is the persistence and outgrowth of subpopulations that lose expression of the target antigen. IL-15 is a potent cytokine that can promote organ-specific autoimmunity when up-regulated on tissue cells. Here we report that T cells eradicated 2-wk-old solid tumors that expressed IL-15, eliminating antigen-negative cells. In contrast, control tumors that lacked IL-15 expression consistently relapsed. Interestingly, even tumors lacking expression of cognate antigen were rejected when expressing IL-15, indicating that rejection after adoptive T-cell transfer was independent of cognate antigen expression. Nevertheless, the T-cell receptor of the transferred T cells influenced the outcome, consistent with the notion that T-cell receptor activation and effector status determine whether IL-15 can confer lymphokine killer activity-like properties to T cells. The effect was limited to the microenvironment of tumors expressing IL-15; there were no noticeable effects on contralateral tumors lacking IL-15. Taken together, these results indicate that expression of IL-15 in the tumor microenvironment may prevent the escape of antigen loss variants and subsequent tumor recurrence by enabling T cells to eliminate cancer cells lacking cognate antigen expression in a locally restricted manner.


Subject(s)
Gene Expression Regulation, Neoplastic , Interleukin-15/metabolism , Neoplasms/metabolism , Receptors, Antigen, T-Cell/metabolism , Tumor Microenvironment , Animals , Antigens, Neoplasm/metabolism , Autoimmunity , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Green Fluorescent Proteins/metabolism , Humans , Interleukin-15/genetics , Killer Cells, Natural/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Perforin/metabolism , Spleen/cytology , Stromal Cells/cytology
9.
Nature ; 456(7221): 534-8, 2008 Nov 27.
Article in English | MEDLINE | ID: mdl-19037317

ABSTRACT

Major histocompatibility complex (MHC) class II alleles HLA-DQ8 and the mouse homologue I-A(g7) lacking a canonical aspartic acid residue at position beta57 are associated with coeliac disease and type I diabetes. However, the role of this single polymorphism in disease initiation and progression remains poorly understood. The lack of Asp 57 creates a positively charged P9 pocket, which confers a preference for negatively charged peptides. Gluten lacks such peptides, but tissue transglutaminase (TG2) introduces negatively charged residues at defined positions into gluten T-cell epitopes by deamidating specific glutamine residues on the basis of their spacing to proline residues. The commonly accepted model, proposing that HLA-DQ8 simply favours binding of negatively charged peptides, does not take into account the fact that TG2 requires inflammation for activation and that T-cell responses against native gluten peptides are found, particularly in children. Here we show that beta57 polymorphism promotes the recruitment of T-cell receptors bearing a negative signature charge in the complementary determining region 3beta (CDR3beta) during the response against native gluten peptides presented by HLA-DQ8 in coeliac disease. These T cells showed a crossreactive and heteroclitic (stronger) response to deamidated gluten peptides. Furthermore, gluten peptide deamidation extended the T-cell-receptor repertoire by relieving the requirement for a charged residue in CDR3beta. Thus, the lack of a negative charge at position beta57 in MHC class II was met by negatively charged residues in the T-cell receptor or in the peptide, the combination of which might explain the role of HLA-DQ8 in amplifying the T-cell response against dietary gluten.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Celiac Disease/genetics , Celiac Disease/immunology , Glutens/immunology , HLA-DQ Antigens/genetics , Polymorphism, Genetic/genetics , Amides/chemistry , Animals , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Cross Reactions , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Gliadin/chemistry , Gliadin/immunology , Glutens/chemistry , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/immunology , Humans , Hybridomas/immunology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Static Electricity
10.
Nat Med ; 29(6): 1520-1529, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37322120

ABSTRACT

Primary sclerosing cholangitis (PSC) is an immune-mediated disease of the bile ducts that co-occurs with inflammatory bowel disease (IBD) in almost 90% of cases. Colorectal cancer is a major complication of patients with PSC and IBD, and these patients are at a much greater risk compared to patients with IBD without concomitant PSC. Combining flow cytometry, bulk and single-cell transcriptomics, and T and B cell receptor repertoire analysis of right colon tissue from 65 patients with PSC, 108 patients with IBD and 48 healthy individuals we identified a unique adaptive inflammatory transcriptional signature associated with greater risk and shorter time to dysplasia in patients with PSC. This inflammatory signature is characterized by antigen-driven interleukin-17A (IL-17A)+ forkhead box P3 (FOXP3)+ CD4 T cells that express a pathogenic IL-17 signature, as well as an expansion of IgG-secreting plasma cells. These results suggest that the mechanisms that drive the emergence of dysplasia in PSC and IBD are distinct and provide molecular insights that could guide prevention of colorectal cancer in individuals with PSC.


Subject(s)
Cholangitis, Sclerosing , Colorectal Neoplasms , Inflammatory Bowel Diseases , Humans , Cholangitis, Sclerosing/complications , Cholangitis, Sclerosing/pathology , Inflammation/complications , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/pathology , Colorectal Neoplasms/pathology
11.
J Exp Med ; 203(5): 1343-55, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16682498

ABSTRACT

Celiac disease is an intestinal inflammatory disorder induced by dietary gluten in genetically susceptible individuals. The mechanisms underlying the massive expansion of interferon gamma-producing intraepithelial cytotoxic T lymphocytes (CTLs) and the destruction of the epithelial cells lining the small intestine of celiac patients have remained elusive. We report massive oligoclonal expansions of intraepithelial CTLs that exhibit a profound genetic reprogramming of natural killer (NK) functions. These CTLs aberrantly expressed cytolytic NK lineage receptors, such as NKG2C, NKp44, and NKp46, which associate with adaptor molecules bearing immunoreceptor tyrosine-based activation motifs and induce ZAP-70 phosphorylation, cytokine secretion, and proliferation independently of T cell receptor signaling. This NK transformation of CTLs may underlie both the self-perpetuating, gluten-independent tissue damage and the uncontrolled CTL expansion leading to malignant lymphomas in severe forms of celiac disease. Because similar changes were detected in a subset of CTLs from cytomegalovirus-seropositive patients, we suggest that a stepwise transformation of CTLs into NK-like cells may underlie immunopathology in various chronic infectious and inflammatory diseases.


Subject(s)
Celiac Disease/immunology , Cell Differentiation/immunology , Cell Proliferation , Killer Cells, Natural/immunology , Signal Transduction/immunology , T-Lymphocytes, Cytotoxic/immunology , Base Sequence , Celiac Disease/complications , Celiac Disease/genetics , Celiac Disease/pathology , Chronic Disease , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Interferon-gamma/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/pathology , Killer Cells, Natural/pathology , Lymphoma/etiology , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Molecular Sequence Data , Phosphorylation , Protein Processing, Post-Translational/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Cytotoxic/pathology , ZAP-70 Protein-Tyrosine Kinase/immunology
12.
J Immunol ; 185(10): 5732-42, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20926796

ABSTRACT

Deficiencies of the T cell and NK cell CD3ζ signaling adapter protein in patients with cancer and autoimmune diseases are well documented, but mechanistic explanations are fragmentary. The stimulatory NKG2D receptor on T and NK cells mediates tumor immunity but can also promote local and systemic immune suppression in conditions of persistent NKG2D ligand induction that include cancer and certain autoimmune diseases. In this paper, we provide evidence that establishes a causative link between CD3ζ impairment and chronic NKG2D stimulation due to pathological ligand expression. We describe a mechanism whereby NKG2D signaling in human T and NK cells initiates Fas ligand/Fas-mediated caspase-3/-7 activation and resultant CD3ζ degradation. As a consequence, the functional capacities of the TCR, the low-affinity Fc receptor for IgG, and the NKp30 and NKp46 natural cytotoxicity receptors, which all signal through CD3ζ, are impaired. These findings are extended to ex vivo phenotypes of T and NK cells among tumor-infiltrating lymphocytes and in peripheral blood from patients with juvenile-onset lupus. Collectively, these results indicate that pathological NKG2D ligand expression leads to simultaneous impairment of multiple CD3ζ-dependent receptor functions, thus offering an explanation that may be applicable to CD3ζ deficiencies associated with diverse disease conditions.


Subject(s)
Autoimmune Diseases/metabolism , CD3 Complex/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasms/metabolism , Receptors, Immunologic/metabolism , Autoimmune Diseases/immunology , Caspases/metabolism , Cell Separation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoblotting , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/immunology , Receptors, Immunologic/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Sci Adv ; 5(8): eaaw7756, 2019 08.
Article in English | MEDLINE | ID: mdl-31457091

ABSTRACT

Celiac disease (CeD), caused by immune reactions to cereal gluten, is treated with gluten -elimination diets. Within hours of gluten exposure, either perorally or extraorally by intradermal injection, treated patients experience gastrointestinal symptoms. To test whether gluten exposure leads to systemic cytokine production time -related to symptoms, series of multiplex cytokine measurements were obtained in CeD patients after gluten challenge. Peptide injection elevated at least 15 plasma cytokines, with IL-2, IL-8, and IL-10 being most prominent (fold-change increase at 4 hours of 272, 11, and 1.2, respectively). IL-2 and IL-8 were the only cytokines elevated at 2 hours, preceding onset of symptoms. After gluten ingestion, IL-2 was the earliest and most prominent cytokine (15-fold change at 4 hours). Supported by studies of patient-derived gluten-specific T cell clones and primary lymphocytes, our observations indicate that gluten-specific CD4+ T cells are rapidly reactivated by antigen -exposure likely causing CeD-associated gastrointestinal symptoms.


Subject(s)
Celiac Disease/pathology , Cytokines/blood , Glutens/administration & dosage , Adult , Aged , CD4-Positive T-Lymphocytes/classification , CD4-Positive T-Lymphocytes/metabolism , Celiac Disease/immunology , Celiac Disease/metabolism , Double-Blind Method , Female , Genotype , Glutens/adverse effects , HLA Antigens/genetics , Humans , Interleukin-10/blood , Interleukin-2/blood , Interleukin-8/blood , Male , Middle Aged , Placebo Effect , Vomiting/etiology , Young Adult
14.
J Exp Med ; 212(10): 1487-95, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26304964

ABSTRACT

Eicosanoids are inflammatory mediators that play a key but incompletely understood role in linking the innate and adaptive immune systems. Here, we show that cytotoxic effector T cells (CTLs) are capable of both producing and responding to cysteinyl leukotrienes (CystLTs), allowing for the killing of target cells in a T cell receptor-independent manner. This process is dependent on the natural killer receptor NKG2D and exposure to IL-15, a cytokine induced in distressed tissues. IL-15 and NKG2D signaling drives the up-regulation of key enzymes implicated in the synthesis of CystLTs, as well as the expression of CystLT receptors, suggesting a positive feedback loop. Finally, although the CystLT pathway has been previously linked to various allergic disorders, we provide unexpected evidence for its involvement in the pathogenesis of celiac disease (CD), a T helper 1 cell-mediated enteropathy induced by gluten. These findings provide new insights into the cytolytic signaling pathway of NKG2D and the pathogenesis of organ-specific immune disorders. Furthermore, they suggest that the blockade of CystLT receptors may represent a potent therapeutic target for CD or potentially other autoimmune disorders in which NKG2D has been implicated.


Subject(s)
Celiac Disease/immunology , Cysteine/metabolism , Interleukin-15/immunology , Leukotrienes/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Arachidonate 5-Lipoxygenase/genetics , Arachidonate 5-Lipoxygenase/metabolism , Case-Control Studies , Celiac Disease/physiopathology , Cells, Cultured , Cysteine/immunology , Female , Humans , Interleukin-15/metabolism , Leukotrienes/immunology , Male , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Leukotriene/genetics , Receptors, Leukotriene/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Up-Regulation
15.
PLoS One ; 8(9): e76292, 2013.
Article in English | MEDLINE | ID: mdl-24086722

ABSTRACT

NK cells are large granular lymphocytes that form a critical component of the innate immune system, whose functions include the killing of cells expressing stress-induced molecules. It is increasingly accepted that despite being considered prototypical effector cells, NK cells require signals to reach their full cytotoxic potential. We previously showed that IL-15 is capable of arming CD8 effector T cells to kill independently of their TCR via NKG2D in a cPLA2-dependent process. As NK cells also express NKG2D, we wanted to investigate whether this pathway functioned in an analogous manner and if resting NK cells could be primed to the effector phase by IL-15. Furthermore, to establish relevance to human disease we studied a possible role for this pathway in the pathogenesis of psoriatic arthritis, since there are aspects of this disease that suggest a potential effector role for the innate immune system. We found that PsA patients had upregulated IL-15 and MIC in their affected synovial tissues, and that this unique inflammatory environment enabled NK cell activation and killing via NKG2D and cPLA2. Moreover, we were able to reproduce the phenotype of joint NK cells from blood NK cells by incubating them with IL-15. Altogether, these findings suggest a destructive role for NK cells when activated by environmental stress signals during the pathogenesis of PsA and demonstrate that IL-15 is capable of priming resting NK cells in tissues to the effector phase.


Subject(s)
Arthritis, Psoriatic/immunology , Gene Expression Regulation/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phospholipases A2, Cytosolic/metabolism , Signal Transduction/immunology , Arthritis, Psoriatic/metabolism , Cytokines/genetics , Cytokines/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , RNA, Small Interfering/genetics , Transfection
16.
J Exp Med ; 206(3): 707-19, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19237603

ABSTRACT

IL-15 and NKG2D promote autoimmunity and celiac disease by arming cytotoxic T lymphocytes (CTLs) to cause tissue destruction. However, the downstream signaling events underlying these functional properties remain unclear. Here, we identify cytosolic phospholipase A(2) (cPLA(2)) as a central molecule in NKG2D-mediated cytolysis in CTLs. Furthermore, we report that NKG2D induces, upon recognition of MIC(+) target cells, the release of arachidonic acid (AA) by CTLs to promote tissue inflammation in association with target killing. Interestingly, IL-15, which licenses NKG2D-mediated lymphokine killer activity in CTLs, cooperates with NKG2D to induce cPLA(2) activation and AA release. Finally, cPLA(2) activation in intraepithelial CTLs of celiac patients provides an in vivo pathophysiological dimension to cPLA(2) activation in CTLs. These results reveal an unrecognized link between NKG2D and tissue inflammation, which may underlie the emerging role of NKG2D in various immunopathological conditions and define new therapeutic targets.


Subject(s)
Celiac Disease/enzymology , Celiac Disease/immunology , Interleukin-15/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phospholipases A2, Cytosolic/metabolism , T-Lymphocytes, Cytotoxic/enzymology , Arachidonic Acid/metabolism , Celiac Disease/pathology , Cell Degranulation , Cytotoxicity, Immunologic , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Models, Immunological , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/physiology , Up-Regulation
17.
Immunity ; 21(3): 357-66, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15357947

ABSTRACT

A major function of NKG2D linking innate and adaptive immunity is to upregulate antigen-specific CTL-mediated cytotoxicity in tissues expressing stress-induced NKG2D ligands, such as MIC, by coactivating TCR signaling. Here, we show that, under conditions of dysregulated IL15 expression in vivo in patients with celiac disease and in vitro in healthy individuals, multiple steps of the NKG2D/DAP10 signaling pathway leading to ERK and JNK activation are coordinately primed to activate direct cytolytic function independent of TCR specificity in effector CD8 T cells. These findings may not only explain previous reports of transformation of CTL into NK-like "lymphokine-activated killers" (LAK cells) under high doses of IL2 (a substitute for IL15) but may also have significant implications for understanding and treating immunopathological diseases.


Subject(s)
Celiac Disease/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Cytotoxic/immunology , Adolescent , Adult , Aged , Flow Cytometry , Humans , Immunohistochemistry , Interleukin-15/metabolism , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Middle Aged , NK Cell Lectin-Like Receptor Subfamily K , Receptors, Antigen, T-Cell/immunology , Receptors, Immunologic/metabolism , Receptors, Natural Killer Cell , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL