Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Trends Immunol ; 45(6): 442-453, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38782625

ABSTRACT

Activated CD8+ T cells directly kill target cells. Therefore, the regulation of their function is central to avoiding immunopathology. Mechanisms that curb effector functions in CD4+ and CD8+ T cells are mostly shared, yet important differences occur. Here, we focus on the control of CD8+ T cell activity and discuss the importance of a poorly understood aspect of tolerance that directly impairs engagement of target cells: the downregulation of CD8. We contextualize this process and propose that it represents a key element during CD8+ T cell modulation.


Subject(s)
CD8-Positive T-Lymphocytes , Immune Tolerance , Animals , Humans , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/metabolism , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Down-Regulation/immunology , Lymphocyte Activation/immunology
2.
Arthritis Rheumatol ; 75(6): 961-972, 2023 06.
Article in English | MEDLINE | ID: mdl-36575804

ABSTRACT

OBJECTIVE: Variants in STAT4 are associated with systemic lupus erythematosus (SLE) and other autoimmune diseases. We undertook this study to investigate how disease-associated variants affect STAT4 expression, in particular in CD4+ T cells where STAT4 plays an essential role. METHODS: We compared Th1 differentiation between naive CD4+ T cells from healthy donors homozygous for the risk (R/R) or nonrisk (NR/NR) alleles. We analyzed epigenetic marks in STAT4 and evaluated the relevance of its third intron, assessed the consequences of Stat4 overexpression in vivo in mice, and analyzed the effects of the STAT4 genotype in patients with lupus nephritis. RESULTS: Naive CD4+ T cells from NR/NR healthy donors down-regulated STAT4 in response to interleukin-12 (IL-12). In contrast, cells from R/R healthy donors maintained high levels. R/R cells exhibited a higher abundance of transcriptionally active STAT4 and increased interferon-γ production. Accordingly, R/R healthy donors exhibited a stronger induction of local active enhancer marks. Genetic editing confirmed the presence of a negative regulatory region in the STAT4 third intron, where most of the SLE-associated STAT4 single-nucleotide polymorphisms (SNPs) are located. In vivo forced expression demonstrated that increases in Stat4 levels in T cells enhanced glomerulonephritis in mice. Accordingly, the R/R genotype was associated with suboptimal response to treatment and with worse clinical outcomes in patients with proliferative lupus nephritis. CONCLUSION: The SLE-associated STAT4 haplotype correlates with an abnormal IL-12-mediated STAT4 transcriptional regulation. Carriers of the risk variant exhibit exaggerated CD4+ proinflammatory capacities that, in the context of SLE, contribute to more severe disease. R/R patients may benefit from blockade of the IL-12/STAT4 pathway.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Animals , Mice , CD4-Positive T-Lymphocytes/metabolism , Down-Regulation , Haplotypes , Interferon-gamma/genetics , Interleukin-12 , Lupus Erythematosus, Systemic/genetics , Lupus Nephritis/genetics , Polymorphism, Single Nucleotide , STAT4 Transcription Factor/genetics , Humans
3.
Proc Natl Acad Sci U S A ; 119(49): e2203454119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36442116

ABSTRACT

The development of innate lymphoid cell (ILC) transcription factor reporter mice has shown a previously unexpected complexity in ILC hematopoiesis. Using novel polychromic mice to achieve higher phenotypic resolution, we have characterized bone marrow progenitors that are committed to the group 1 ILC lineage. These common ILC1/NK cell progenitors (ILC1/NKP), which we call "aceNKPs", are defined as lineage-Id2+IL-7Rα+CD25-α4ß7-NKG2A/C/E+Bcl11b-. In vitro, aceNKPs differentiate into group 1 ILCs, including NK-like cells that express Eomes without the requirement for IL-15, and produce IFN-γ and perforin upon IL-15 stimulation. Following reconstitution of Rag2-/-Il2rg-/- hosts, aceNKPs give rise to a spectrum of mature ILC1/NK cells (regardless of their tissue location) that cannot be clearly segregated into the traditional ILC1 and NK subsets, suggesting that group 1 ILCs constitute a dynamic continuum of ILCs that can develop from a common progenitor. In addition, aceNKP-derived ILC1/NK cells effectively ameliorate tumor burden in a model of lung metastasis, where they acquired a cytotoxic NK cell phenotype. Our results identify the primary ILC1/NK progenitor that lacks ILC2 or ILC3 potential and is strictly committed to ILC1/NK cell production irrespective of tissue homing.


Subject(s)
Immunity, Innate , Interleukin-15 , Animals , Mice , Interleukin-15/genetics , Killer Cells, Natural , Perforin , Transcription Factors , Repressor Proteins , Tumor Suppressor Proteins
4.
Entropy (Basel) ; 24(11)2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36359673

ABSTRACT

Cryptocurrency markets have attracted many interest for global investors because of their novelty, wide on-line availability, increasing capitalization, and potential profits. In the econophysics tradition, we show that many of the most available cryptocurrencies have return statistics that do not follow Gaussian distributions, instead following heavy-tailed distributions. Entropy measures are applied, showing that portfolio diversification is a reasonable practice for decreasing return uncertainty.

5.
Front Immunol ; 13: 981479, 2022.
Article in English | MEDLINE | ID: mdl-36263033

ABSTRACT

Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide, and is largely refractory to current immunotherapeutic interventions. The lack of efficacy of existing cancer immunotherapies in CRC reflects the complex nature of the unique intestinal immune environment, which serves to maintain barrier integrity against pathogens and harmful environmental stimuli while sustaining host-microbe symbiosis during homeostasis. With their expression by barrier epithelial cells, the cytokines interleukin-25 (IL-25) and IL-33 play key roles in intestinal immune responses, and have been associated with inappropriate allergic reactions, autoimmune diseases and cancer pathology. Studies in the past decade have begun to uncover the important roles of IL-25 and IL-33 in shaping the CRC tumour immune microenvironment, where they may promote or inhibit tumorigenesis depending on the specific CRC subtype. Notably, both IL-25 and IL-33 have been shown to act on group 2 innate lymphoid cells (ILC2s), but can also stimulate an array of other innate and adaptive immune cell types. Though sometimes their functions can overlap they can also produce distinct phenotypes dependent on the differential distribution of their receptor expression. Furthermore, both IL-25 and IL-33 modulate pathways previously known to contribute to CRC tumorigenesis, including angiogenesis, tumour stemness, invasion and metastasis. Here, we review our current understanding of IL-25 and IL-33 in CRC tumorigenesis, with specific focus on dissecting their individual function in the context of distinct subtypes of CRC, and the potential prospects for targeting these pathways in CRC immunotherapy.


Subject(s)
Colorectal Neoplasms , Interleukin-33 , Humans , Interleukin-17/metabolism , Immunity, Innate , Colorectal Neoplasms/metabolism , Lymphocytes/metabolism , Carcinogenesis , Cell Transformation, Neoplastic/genetics , Cytokines , Tumor Microenvironment
6.
Sci Immunol ; 7(72): eabn0175, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35658010

ABSTRACT

Interleukin-25 (IL-25) and group 2 innate lymphoid cells (ILC2s) defend the host against intestinal helminth infection and are associated with inappropriate allergic reactions. IL-33-activated ILC2s were previously found to augment protective tissue-specific pancreatic cancer immunity. Here, we showed that intestinal IL-25-activated ILC2s created an innate cancer-permissive microenvironment. Colorectal cancer (CRC) patients with higher tumor IL25 expression had reduced survival and increased IL-25R-expressing tumor-resident ILC2s and myeloid-derived suppressor cells (MDSCs) associated with impaired antitumor responses. Ablation of IL-25 signaling reduced tumors, virtually doubling life expectancy in an Apc mutation-driven model of spontaneous intestinal tumorigenesis. Mechanistically, IL-25 promoted intratumoral ILC2s, which sustained tumor-infiltrating MDSCs to suppress antitumor immunity. Therapeutic antibody-mediated blockade of IL-25 signaling decreased intratumoral ILC2s, MDSCs, and adenoma/adenocarcinoma while increasing antitumor adaptive T cell and interferon-γ (IFN-γ)-mediated immunity. Thus, the roles of innate epithelium-derived cytokines IL-25 and IL-33 as well as ILC2s in cancer cannot be generalized. The protumoral nature of the IL-25-ILC2 axis in CRC highlights this pathway as a potential therapeutic target against CRC.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Interleukin-33 , Myeloid-Derived Suppressor Cells , Carcinogenesis , Humans , Immunity, Innate , Interleukin-17 , Interleukin-33/genetics , Lymphocytes , Mutation , Tumor Microenvironment
7.
Nat Rev Rheumatol ; 18(4): 232-244, 2022 04.
Article in English | MEDLINE | ID: mdl-35075294

ABSTRACT

Adaptive immune responses rely on the proliferation of T lymphocytes able to recognize and eliminate pathogens. The magnitude and duration of the expansion of activated T cell clones are finely regulated to minimize immunopathology and avoid autoimmunity. In patients with rheumatic autoimmune diseases, such as systemic lupus erythematosus and rheumatoid arthritis, activated lymphocytes survive and exert effector functions for prolonged periods, defying the mechanisms that normally curb their capacities during acute and chronic infections. Here, we review the molecular mechanisms that limit the duration of immune responses in health and discuss the factors that alter such regulation in the setting of systemic lupus erythematosus and rheumatoid arthritis. We highlight defects that could contribute to the development and progression of autoimmune disease and describe how chronic inflammation can alter the regulation of activated lymphocyte survival, promoting its perpetuation. These concepts might contribute to the understanding of the mechanisms that underlie the chronicity of inflammation in the context of autoimmunity.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Lupus Erythematosus, Systemic , Rheumatic Diseases , Autoimmunity , Cell Survival , Humans , Inflammation , T-Lymphocytes
8.
Front Immunol ; 12: 635862, 2021.
Article in English | MEDLINE | ID: mdl-33841416

ABSTRACT

Activation of self-reactive CD8+ T cells induces a peripheral tolerance mechanism that involves loss of CD8 expression. Because genetic deficiency of Fas and Fasl causes the accumulation of double-negative (DN; CD3+ TCR-αß+ CD4- CD8-) T cells that have been proposed to derive from CD8+ cells, we decided to explore the role of Fas and FasL in self-antigen-induced CD8 downregulation. To this end, we quantified Fas and FasL induction by different stimuli and analyzed the effects of Fas/FasL deficiency during a protective immune response and after exposure to self-antigens. Our data describes how Fas and FasL upregulation differs depending on the setting of CD8 T cell activation and demonstrates that Fas/FasL signaling maintains CD8 expression during repetitive antigen stimulation and following self-antigen encounter. Together, our results reveal an unexpected role of Fas/FasL signaling and offer a new insight into the role of these molecules in the regulation of immune tolerance.


Subject(s)
Autoantigens/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Fas Ligand Protein/metabolism , Immune Tolerance , Lymphocyte Activation , fas Receptor/metabolism , Adoptive Transfer , Animals , Autoantigens/immunology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Down-Regulation , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Kinetics , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Signal Transduction , fas Receptor/genetics , fas Receptor/immunology
9.
Annu Rev Immunol ; 39: 167-198, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33534604

ABSTRACT

Type 2 immunity helps protect the host from infection, but it also plays key roles in tissue homeostasis, metabolism, and repair. Unfortunately, inappropriate type 2 immune reactions may lead to allergy and asthma. Group 2 innate lymphoid cells (ILC2s) in the lungs respond rapidly to local environmental cues, such as the release of epithelium-derived type 2 initiator cytokines/alarmins, producing type 2 effector cytokines such as IL-4, IL-5, and IL-13 in response to tissue damage and infection. ILC2s are associated with the severity of allergic asthma, and experimental models of lung inflammation have shown how they act as playmakers, receiving signals variously from stromal and immune cells as well as the nervous system and then distributing cytokine cues to elicit type 2 immune effector functions and potentiate CD4+ T helper cell activation, both of which characterize the pathology of allergic asthma. Recent breakthroughs identifying stromal- and neuronal-derived microenvironmental cues that regulate ILC2s, along with studies recognizing the potential plasticity of ILC2s, have improved our understanding of the immunoregulation of asthma and opened new avenues for drug discovery.


Subject(s)
Asthma , Hypersensitivity , Animals , Asthma/etiology , Humans , Immunity, Innate , Interleukin-13 , Lymphocytes
10.
Mucosal Immunol ; 14(1): 26-37, 2021 01.
Article in English | MEDLINE | ID: mdl-32457448

ABSTRACT

Type-2 immunity is characterised by interleukin (IL)-4, IL-5 and IL-13, eosinophilia, mucus production, IgE, and alternatively activated macrophages (AAM). However, despite the lack of neutrophil chemoattractants such as CXCL1, neutrophils, a feature of type-1 immunity, are observed in type-2 responses. Consequently, alternative mechanisms must exist to ensure that neutrophils can contribute to type-2 immune reactions without escalation of deleterious inflammation. We now demonstrate that type-2 immune-associated neutrophil infiltration is regulated by the mouse RNase A homologue, eosinophil-associated ribonuclease 11 (Ear11), which is secreted by AAM downstream of IL-25-stimulated ILC2. Transgenic overexpression of Ear11 resulted in tissue neutrophilia, whereas Ear11-deficient mice have fewer resting tissue neutrophils, whilst other type-2 immune responses are not impaired. Notably, administration of recombinant mouse Ear11 increases neutrophil motility and recruitment. Thus, Ear11 helps maintain tissue neutrophils at homoeostasis and during type-2 reactions when chemokine-producing classically activated macrophages are infrequently elicited.


Subject(s)
Immunity, Innate , Lymphocytes/physiology , Macrophage Activation/immunology , Macrophages/physiology , Neutrophil Infiltration/immunology , Neutrophils/physiology , Ribonucleases/biosynthesis , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Eosinophils/immunology , Eosinophils/metabolism , Immunomodulation , Immunophenotyping , Interleukin-13/biosynthesis , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Transgenic , Ribonucleases/genetics
11.
J Clin Invest ; 130(11): 5989-6004, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32750040

ABSTRACT

How T cells integrate environmental cues into signals that limit the magnitude and length of immune responses is poorly understood. Here, we provide data that demonstrate that B55ß, a regulatory subunit of protein phosphatase 2A, represents a molecular link between cytokine concentration and apoptosis in activated CD8+ T cells. Through the modulation of AKT, B55ß induced the expression of the proapoptotic molecule Hrk in response to cytokine withdrawal. Accordingly, B55ß and Hrk were both required for in vivo and in vitro contraction of activated CD8+ lymphocytes. We show that this process plays a role during clonal contraction, establishment of immune memory, and preservation of peripheral tolerance. This regulatory pathway may represent an unexplored opportunity to end unwanted immune responses or to promote immune memory.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Protein Phosphatase 2/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Mice , Mice, Transgenic , Neuropeptides/genetics , Neuropeptides/immunology , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology
12.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32591431

ABSTRACT

BACKGROUND: Tumor-infiltrating lymphocytes (TILs), mainly CD8+ cytotoxic T lymphocytes (CTL), are linked to immune-mediated control of human cancers and response to immunotherapy. Tumors have nonetheless developed specific mechanisms that selectively restrict T cell entry into the tumor microenvironment. The extracellular superoxide dismutase (SOD3) is an anti-oxidant enzyme usually downregulated in tumors. We hypothesize that upregulation of SOD3 in the tumor microenvironment might be a mechanism to boost T cell infiltration by normalizing the tumor-associated endothelium. RESULTS: Here we show that SOD3 overexpression in endothelial cells increased in vitro transmigration of naïve and activated CD4+ and CD8+ T cells, but not of myeloid cells. Perivascular expression of SOD3 also specifically increased CD4+ and CD8+ effector T cell infiltration into tumors and improved the effectiveness of adoptively transferred tumor-specific CD8+ T cells. SOD3-induced enhanced transmigration in vitro and tumor infiltration in vivo were not associated to upregulation of T cell chemokines such as CXCL9 or CXCL10, nor to changes in the levels of endothelial adhesion receptors such as intercellular adhesion molecule-1 (ICAM-1) or vascular cell adhesion molecule-1 (VCAM-1). Instead, SOD3 enhanced T cell infiltration via HIF-2α-dependent induction of specific WNT ligands in endothelial cells; this led to WNT signaling pathway activation in the endothelium, FOXM1 stabilization, and transcriptional induction of laminin-α4 (LAMA4), an endothelial basement membrane component permissive for T cell infiltration. In patients with stage II colorectal cancer, SOD3 was associated with increased CD8+ TIL density and disease-free survival. SOD3 expression was also linked to a T cell-inflamed gene signature using the COAD cohort from The Cancer Genome Atlas program. CONCLUSION: Our findings suggest that SOD3-induced upregulation of LAMA4 in endothelial cells boosts selective tumor infiltration by T lymphocytes, thus transforming immunologically "cold" into "hot" tumors. High SOD3 levels are associated with human colon cancer infiltration by CD8+ T cells, with potential consequences for the clinical outcome of these patients. Our results also uncover a cell type-specific, distinct activity of the WNT pathway for the regulation of T cell infiltration into tumors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/immunology , Superoxide Dismutase/metabolism , Superoxide Dismutase/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Female , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Superoxide Dismutase/genetics , Tumor Cells, Cultured , Tumor Microenvironment
13.
JCI Insight ; 5(5)2020 03 12.
Article in English | MEDLINE | ID: mdl-32161189

ABSTRACT

Protein phosphatase 2A (PP2A), a serine/threonine phosphatase, has been shown to control T cell function. We found that in vitro-activated B cells and B cells from various lupus-prone mice and patients with systemic lupus erythematosus display increased PP2A activity. To understand the contribution of PP2A to B cell function, we generated a Cd19CrePpp2r1afl/fl (flox/flox) mouse which lacks functional PP2A only in B cells. Flox/flox mice displayed reduced spontaneous germinal center formation and decreased responses to T cell-dependent and T-independent antigens, while their B cells responded poorly in vitro to stimulation with an anti-CD40 antibody or CpG in the presence of IL-4. Transcriptome and metabolome studies revealed altered nicotinamide adenine dinucleotide (NAD) and purine/pyrimidine metabolism and increased expression of purine nucleoside phosphorylase in PP2A-deficient B cells. Our results demonstrate that PP2A is required for optimal B cell function and may contribute to increased B cell activity in systemic autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Protein Phosphatase 2/metabolism , Animals , Autoantibodies/biosynthesis , Autoimmunity , B-Lymphocytes/enzymology , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Germinal Center/immunology , Humans , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation , Mice , Mice, Transgenic , T-Lymphocytes/immunology
14.
J Leukoc Biol ; 108(3): 851-857, 2020 09.
Article in English | MEDLINE | ID: mdl-32052478

ABSTRACT

The cellular origin of CD4- CD8- (double negative, DNT) TCR-α/ß+ T cells remains unknown. Available evidence indicates that they may derive from CD8+ T cells, but most published data have been obtained using cells that bear an invariant transgenic T cell receptor that recognizes an Ag that is not present in normal mice. Here, we have used complementary fate mapping and adoptive transfer experiments to identify the cellular lineage of origin of DNT cells in wild-type mice with a polyclonal T cell repertoire. We show that TCR-α/ß+ DNT cells can be traced back to CD8+ and CD4+ CD8+ double positive cells in the thymus. We also demonstrate that polyclonal DNT cells generated in secondary lymphoid organs proliferate upon adoptive transfer and can regain CD8 expression in lymphopenic environment. These results demonstrate the cellular origin of DNT cells and provide a conceptual framework to understand their presence in pathological circumstances.


Subject(s)
CD4 Antigens/analysis , CD8 Antigens/analysis , CD8-Positive T-Lymphocytes/cytology , Lymphopoiesis , Receptors, Antigen, T-Cell, alpha-beta/analysis , T-Lymphocyte Subsets/cytology , Adoptive Transfer , Animals , CD8 Antigens/biosynthesis , CD8 Antigens/genetics , CD8-Positive T-Lymphocytes/classification , Cell Lineage , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Specific Pathogen-Free Organisms , Up-Regulation
15.
Clin Immunol ; 212: 108240, 2020 03.
Article in English | MEDLINE | ID: mdl-31299381

ABSTRACT

CD8 T cells can kill malignant cells in an antigen-specific manner. However, anti-tumoral responses are usually limited by suppressive factors that curb the effector responses of tumor-infiltrating CD8 T cells. Therapeutic strategies to overcome intra-tumoral T cell suppression, for example immune checkpoint inhibition, have been clinically effective in patients with cancer. Here, we provide data that demonstrates that GK-1, a peptide derived from the parasite Taenia crassiceps, promotes an anti-melanoma CD8 T cell response with heightened effector characteristics that leads to an increased amount of tumor-infiltrating CD44+ IFN-γ-producing CD8 T cells. The response induced by GK-1 was associated with a reduction in the expression of PD-1 and PD-L1 on tumor-infiltrating CD8 and dendritic cells, respectively, effects that led to a dramatic decrease in tumor burden. Our results suggest that the immunomodulatory properties of GK-1 may promote a CD8 T cell response that may be therapeutically useful in the setting of cancer.


Subject(s)
B7-H1 Antigen/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dendritic Cells/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma, Experimental/immunology , Peptides, Cyclic/pharmacology , Programmed Cell Death 1 Receptor/drug effects , Skin Neoplasms/immunology , Tumor Microenvironment/drug effects , Adoptive Transfer , Animals , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Down-Regulation , Hyaluronan Receptors/immunology , Interferon-gamma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/transplantation , Taenia , Tumor Microenvironment/immunology
16.
JCI Insight ; 52019 07 23.
Article in English | MEDLINE | ID: mdl-31335320

ABSTRACT

Chronic inflammation causes target organ damage in patients with systemic autoimmune diseases. The factors that allow this protracted response are poorly understood. We analyzed the transcriptional regulation of PPP2R2B (B55ß), a molecule necessary for the termination of the immune response, in patients with autoimmune diseases. Altered expression of B55ß conditioned resistance to cytokine withdrawal-induced death (CWID) in patients with autoimmune diseases. The impaired upregulation of B55ß was caused by inflammation-driven hypermethylation of specific cytosines located within a regulatory element of PPP2R2B preventing CTCF binding. This phenotype could be induced in healthy T cells by exposure to TNF-α. Our results reveal a gene whose expression is affected by an acquired defect, through an epigenetic mechanism, in the setting of systemic autoimmunity. Because failure to remove activated T cells through CWID could contribute to autoimmune pathology, this mechanism illustrates a vicious cycle through which autoimmune inflammation contributes to its own perpetuation.


Subject(s)
Apoptosis/drug effects , Autoimmune Diseases/metabolism , DNA Methylation , Nerve Tissue Proteins/metabolism , Protein Phosphatase 2/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CCCTC-Binding Factor/metabolism , Cytokines/metabolism , Cytosine/metabolism , DNA Methylation/drug effects , Gene Expression Regulation , Humans , Inflammation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/pharmacology , T-Lymphocytes , Up-Regulation
17.
J Exp Med ; 216(9): 1999-2009, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31248899

ABSTRACT

Group-2 innate lymphoid cells (ILC2), type-2 cytokines, and eosinophils have all been implicated in sustaining adipose tissue homeostasis. However, the interplay between the stroma and adipose-resident immune cells is less well understood. We identify that white adipose tissue-resident multipotent stromal cells (WAT-MSCs) can act as a reservoir for IL-33, especially after cell stress, but also provide additional signals for sustaining ILC2. Indeed, we demonstrate that WAT-MSCs also support ICAM-1-mediated proliferation and activation of LFA-1-expressing ILC2s. Consequently, ILC2-derived IL-4 and IL-13 feed back to induce eotaxin secretion from WAT-MSCs, supporting eosinophil recruitment. Thus, MSCs provide a niche for multifaceted dialogue with ILC2 to sustain a type-2 immune environment in WAT.


Subject(s)
Adipose Tissue, White/cytology , Immunity, Innate , Lymphocytes/cytology , Lymphocytes/immunology , Animals , Cell Proliferation , Eosinophils/metabolism , Interleukin-33 , Interleukin-5/biosynthesis , Mice, Inbred BALB C , Mice, Inbred C57BL , Stromal Cells/cytology
18.
Immunity ; 51(1): 104-118.e7, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31128961

ABSTRACT

Innate lymphoid cells (ILCs) play strategic roles in tissue homeostasis and immunity. ILCs arise from lymphoid progenitors undergoing lineage restriction and the development of specialized ILC subsets. We generated "5x polychromILC" transcription factor reporter mice to delineate ILC precursor states by revealing the multifaceted expression of key ILC-associated transcription factors (Id2, Bcl11b, Gata3, RORγt, and RORα) during ILC development in the bone marrow. This approach allowed previously unattained enrichment of rare progenitor subsets and revealed hitherto unappreciated ILC precursor heterogeneity. In vivo and in vitro assays identified precursors with potential to generate all ILC subsets and natural killer (NK) cells, and also permitted discrimination of elusive ILC3 bone marrow antecedents. Single-cell gene expression analysis identified a discrete ILC2-committed population and delineated transition states between early progenitors and a highly heterogeneous ILC1, ILC3, and NK precursor cell cluster. This diversity might facilitate greater lineage potential upon progenitor recruitment to peripheral tissues.


Subject(s)
Bone Marrow/immunology , Lymphocyte Subsets/physiology , Lymphocytes/physiology , Lymphoid Progenitor Cells/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , Gene Expression Regulation, Developmental , Genes, Reporter , Immunity, Innate , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Single-Cell Analysis , Transcription Factors/genetics
19.
Eur J Immunol ; 46(6): 1383-91, 2016 06.
Article in English | MEDLINE | ID: mdl-27060346

ABSTRACT

TCR-αß(+) double negative (DN) T cells (CD3(+) TCR-αß(+) CD4(-) CD8(-) NK1.1(-) CD49b(-) ) represent a minor heterogeneous population in healthy humans and mice. These cells have been ascribed pro-inflammatory and regulatory capacities and are known to expand during the course of several autoimmune diseases. Importantly, previous studies have shown that self-reactive CD8(+) T cells become DN after activation by self-antigens, suggesting that self-reactive T cells may exist within the DN T-cell population. Here, we demonstrate that programmed cell death 1 (PD-1) expression in unmanipulated mice identifies a subset of DN T cells with expression of activation-associated markers and a phenotype that strongly suggests they are derived from self-reactive CD8(+) cells. We also found that, within DN T cells, the PD-1(+) subset generates the majority of pro-inflammatory cytokines. Finally, using a TCR-activation reporter mouse (Nur77-GFP), we confirmed that in the steady-state PD-1(+) DN T cells engage endogenous antigens in healthy mice. In conclusion, we provide evidence that indicates that the PD-1(+) fraction of DN T cells represents self-reactive cells.


Subject(s)
Autoimmunity , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Autoantigens/metabolism , Biomarkers , Cells, Cultured , Cytokines/metabolism , Gene Expression , Immunophenotyping , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/genetics
20.
Nat Immunol ; 17(5): 556-64, 2016 May.
Article in English | MEDLINE | ID: mdl-26974206

ABSTRACT

Homeostasis of the immune system depends on the proper function of regulatory T cells (T(reg) cells). Compromised suppressive activity of T(reg) cells leads to autoimmune disease and graft rejection and promotes anti-tumor immunity. Here we report a previously unrecognized requirement for the serine-threonine phosphatase PP2A in the function of T(reg) cells. T(reg) cells exhibited high PP2A activity, and T(reg) cell-specific ablation of the PP2A complex resulted in a severe, multi-organ, lymphoproliferative autoimmune disorder. Mass spectrometry revealed that PP2A associated with components of the mTOR metabolic-checkpoint kinase pathway and suppressed the activity of the mTORC1 complex. In the absence of PP2A, T(reg) cells altered their metabolic and cytokine profile and were unable to suppress effector immune responses. Therefore, PP2A is required for the function of T(reg) cells and the prevention of autoimmunity.


Subject(s)
Autoimmune Diseases/immunology , Lymphoproliferative Disorders/immunology , Protein Phosphatase 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Autoimmunity/genetics , Autoimmunity/immunology , Cells, Cultured , Ceramides/immunology , Ceramides/metabolism , Female , Flow Cytometry , Humans , Immunoblotting , Jurkat Cells , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Phosphorylation/immunology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...