Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 20(3): 373, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30728493

ABSTRACT

In the version of this article initially published, three authors (Hui-Fern Kuoy, Adam P. Uldrich and Dale. I. Godfrey) and their affiliations, acknowledgments and contributions were not included. The correct information is as follows:Ayano C. Kohlgruber1,2, Shani T. Gal-Oz3, Nelson M. LaMarche1,2, Moto Shimazaki1, Danielle Duquette4, Hui-Fern Koay5,6, Hung N. Nguyen1, Amir I. Mina4, Tyler Paras1, Ali Tavakkoli7, Ulrich von Andrian2,8, Adam P. Uldrich5,6, Dale I. Godfrey5,6, Alexander S. Banks4, Tal Shay3, Michael B. Brenner1,10* and Lydia Lynch1,4,9,10*1Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA. 2Division of Medical Sciences, Harvard Medical School, Boston, MA, USA. 3Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel. 4Division of Endocrinology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA. 5Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Australia. 6ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Australia. 7Department of General and Gastrointestinal Surgery, Brigham and Women's Hospital, Boston, MA, USA. 8Department of Microbiology and Immunology, Harvard Medical School, Boston, MA, USA. 9School of Biochemistry and Immunology, Trinity College, Dublin, Ireland. 10These authors jointly supervised this work: Michael B. Brenner, Lydia Lynch. *e-mail: mbrenner@research.bwh.harvard.edu; llynch@bwh.harvard.eduAcknowledgementsWe thank A.T. Chicoine, flow cytometry core manager at the Human Immunology Center at BWH, for flow cytometry sorting. We thank D. Sant'Angelo (Rutgers Cancer Institute) for providing Zbtb16-/- mice and R. O'Brien (National Jewish Health) for providing Vg4/6-/- mice. Supported by NIH grant R01 AI11304603 (to M.B.B.), ERC Starting Grant 679173 (to L.L.), the National Health and Medical Research Council of Australia (1013667), an Australian Research Council Future Fellowship (FT140100278 for A.P.U.) and a National Health and Medical Research Council of Australia Senior Principal Research Fellowship (1117766 for D.I.G.).Author contributionsA.C.K., L.L., and M.B.B. conceived and designed the experiments, and wrote the manuscript. A.C.K., N.M.L., L.L., H.N.N., M.S., T.P., and D.D. performed the experiments. S.T.G.-O. and T.S. performed the RNA-seq analysis. A.S.B. and A.I.M. provided advice and performed the CLAMS experiments. A.T. provided human bariatric patient samples. Parabiosis experiments were performed in the laboratory of U.v.A. H.-F.K., A.P.U. and D.I.G provided critical insight into the TCR chain usage of PLZF+ γδ T cells. M.B.B., N.M.L., and L.L. critically reviewed the manuscript.The errors have been corrected in the HTML and PDF version of the article.Correction to: Nature Immunology doi:10.1038/s41590-018-0094-2 (2018), published online 18 April 2018.

3.
Nat Immunol ; 20(7): 928-942, 2019 07.
Article in English | MEDLINE | ID: mdl-31061532

ABSTRACT

To define the cell populations that drive joint inflammation in rheumatoid arthritis (RA), we applied single-cell RNA sequencing (scRNA-seq), mass cytometry, bulk RNA sequencing (RNA-seq) and flow cytometry to T cells, B cells, monocytes, and fibroblasts from 51 samples of synovial tissue from patients with RA or osteoarthritis (OA). Utilizing an integrated strategy based on canonical correlation analysis of 5,265 scRNA-seq profiles, we identified 18 unique cell populations. Combining mass cytometry and transcriptomics revealed cell states expanded in RA synovia: THY1(CD90)+HLA-DRAhi sublining fibroblasts, IL1B+ pro-inflammatory monocytes, ITGAX+TBX21+ autoimmune-associated B cells and PDCD1+ peripheral helper T (TPH) cells and follicular helper T (TFH) cells. We defined distinct subsets of CD8+ T cells characterized by GZMK+, GZMB+, and GNLY+ phenotypes. We mapped inflammatory mediators to their source cell populations; for example, we attributed IL6 expression to THY1+HLA-DRAhi fibroblasts and IL1B production to pro-inflammatory monocytes. These populations are potentially key mediators of RA pathogenesis.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Gene Expression Profiling , Synovial Membrane/metabolism , Transcriptome , Arthritis, Rheumatoid/pathology , Autoimmunity/genetics , Biomarkers , Computational Biology/methods , Cross-Sectional Studies , Cytokines/metabolism , Fibroblasts/metabolism , Flow Cytometry , Gene Expression , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Leukocytes/immunology , Leukocytes/metabolism , Monocytes/immunology , Monocytes/metabolism , Signal Transduction , Single-Cell Analysis/methods , Synovial Membrane/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Workflow
4.
Nat Immunol ; 20(7): 902-914, 2019 07.
Article in English | MEDLINE | ID: mdl-31209404

ABSTRACT

Lupus nephritis is a potentially fatal autoimmune disease for which the current treatment is ineffective and often toxic. To develop mechanistic hypotheses of disease, we analyzed kidney samples from patients with lupus nephritis and from healthy control subjects using single-cell RNA sequencing. Our analysis revealed 21 subsets of leukocytes active in disease, including multiple populations of myeloid cells, T cells, natural killer cells and B cells that demonstrated both pro-inflammatory responses and inflammation-resolving responses. We found evidence of local activation of B cells correlated with an age-associated B-cell signature and evidence of progressive stages of monocyte differentiation within the kidney. A clear interferon response was observed in most cells. Two chemokine receptors, CXCR4 and CX3CR1, were broadly expressed, implying a potentially central role in cell trafficking. Gene expression of immune cells in urine and kidney was highly correlated, which would suggest that urine might serve as a surrogate for kidney biopsies.


Subject(s)
Kidney/immunology , Lupus Nephritis/immunology , Biomarkers , Biopsy , Cluster Analysis , Computational Biology/methods , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunophenotyping , Interferons/metabolism , Kidney/metabolism , Kidney/pathology , Leukocytes/immunology , Leukocytes/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Molecular Sequence Annotation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Single-Cell Analysis , Transcriptome
5.
Nat Immunol ; 19(12): 1330-1340, 2018 12.
Article in English | MEDLINE | ID: mdl-30420624

ABSTRACT

Up to 49% of certain types of cancer are attributed to obesity, and potential mechanisms include overproduction of hormones, adipokines, and insulin. Cytotoxic immune cells, including natural killer (NK) cells and CD8+ T cells, are important in tumor surveillance, but little is known about the impact of obesity on immunosurveillance. Here, we show that obesity induces robust peroxisome proliferator-activated receptor (PPAR)-driven lipid accumulation in NK cells, causing complete 'paralysis' of their cellular metabolism and trafficking. Fatty acid administration, and PPARα and PPARδ (PPARα/δ) agonists, mimicked obesity and inhibited mechanistic target of rapamycin (mTOR)-mediated glycolysis. This prevented trafficking of the cytotoxic machinery to the NK cell-tumor synapse. Inhibiting PPARα/δ or blocking the transport of lipids into mitochondria reversed NK cell metabolic paralysis and restored cytotoxicity. In vivo, NK cells had blunted antitumor responses and failed to reduce tumor growth in obesity. Our results demonstrate that the lipotoxic obese environment impairs immunosurveillance and suggest that metabolic reprogramming of NK cells may improve cancer outcomes in obesity.


Subject(s)
Immunologic Surveillance/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Melanoma, Experimental/immunology , Obesity/immunology , Adult , Animals , Female , Humans , Killer Cells, Natural/pathology , Male , Melanoma, Experimental/complications , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/complications , Young Adult
6.
Nat Immunol ; 19(5): 464-474, 2018 05.
Article in English | MEDLINE | ID: mdl-29670241

ABSTRACT

γδ T cells are situated at barrier sites and guard the body from infection and damage. However, little is known about their roles outside of host defense in nonbarrier tissues. Here, we characterize a highly enriched tissue-resident population of γδ T cells in adipose tissue that regulate age-dependent regulatory T cell (Treg) expansion and control core body temperature in response to environmental fluctuations. Mechanistically, innate PLZF+ γδ T cells produced tumor necrosis factor and interleukin (IL) 17 A and determined PDGFRα+ and Pdpn+ stromal-cell production of IL-33 in adipose tissue. Mice lacking γδ T cells or IL-17A exhibited decreases in both ST2+ Treg cells and IL-33 abundance in visceral adipose tissue. Remarkably, these mice also lacked the ability to regulate core body temperature at thermoneutrality and after cold challenge. Together, these findings uncover important physiological roles for resident γδ T cells in adipose tissue immune homeostasis and body-temperature control.


Subject(s)
Adipose Tissue/cytology , Homeostasis/physiology , Interleukin-17/metabolism , T-Lymphocytes, Regulatory/physiology , Thermogenesis/physiology , Adipose Tissue/physiology , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta , T-Lymphocyte Subsets/physiology
7.
Nat Immunol ; 16(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25436972

ABSTRACT

Invariant natural killer T cells (iNKT cells) are lipid-sensing innate T cells that are restricted by the antigen-presenting molecule CD1d and express the transcription factor PLZF. iNKT cells accumulate in adipose tissue, where they are anti-inflammatory, but the factors that contribute to their anti-inflammatory nature, as well as their targets in adipose tissue, are unknown. Here we found that iNKT cells in adipose tissue had a unique transcriptional program and produced interleukin 2 (IL-2) and IL-10. Unlike other iNKT cells, they lacked PLZF but expressed the transcription factor E4BP4, which controlled their IL-10 production. The adipose iNKT cells were a tissue-resident population that induced an anti-inflammatory phenotype in macrophages and, through the production of IL-2, controlled the number, proliferation and suppressor function of regulatory T cells (Treg cells) in adipose tissue. Thus, iNKT cells in adipose tissue are unique regulators of immunological homeostasis in this tissue.


Subject(s)
Adipose Tissue/immunology , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/immunology , Natural Killer T-Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Adipose Tissue/cytology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cell Growth Processes/immunology , Female , Flow Cytometry , Gene Expression Regulation , Homeostasis/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Macrophages/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
8.
Immunity ; 46(2): 220-232, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228280

ABSTRACT

Fibroblasts are major contributors to and regulators of inflammation and dominant producers of interleukin-6 (IL-6) in inflammatory diseases like rheumatoid arthritis. Yet, compared to leukocytes, the regulation of inflammatory pathways in fibroblasts is largely unknown. Here, we report that analyses of genes coordinately upregulated with IL-6 pointed to STAT4 and leukemia inhibitory factor (LIF) as potentially linked. Gene silencing revealed that STAT4 was required for IL-6 transcription. STAT4 was recruited to the IL-6 promoter after fibroblast activation, and LIF receptor (LIFR) and STAT4 formed a molecular complex that, together with JAK1 and TYK2 kinases, controlled STAT4 activation. Importantly, a positive feedback loop involving autocrine LIF, LIFR, and STAT4 drove sustained IL-6 transcription. Besides IL-6, this autorine loop also drove the production of other key inflammatory factors including IL-8, granulocyte-colony stimulating factor (G-CSF), IL-33, IL-11, IL-1α, and IL-1ß. These findings define the transcriptional regulation of fibroblast-mediated inflammation as distinct from leukocytes.


Subject(s)
Autocrine Communication/immunology , Fibroblasts/immunology , Gene Expression Regulation/immunology , Leukemia Inhibitory Factor/immunology , Receptors, OSM-LIF/immunology , Arthritis, Rheumatoid/immunology , Cells, Cultured , Cytokines/biosynthesis , Gene Expression Profiling , Humans , Inflammation/immunology , Interleukin-6/immunology , STAT4 Transcription Factor/immunology , Synovial Membrane/immunology , Transcriptome
9.
Immunity ; 46(2): 273-286, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228283

ABSTRACT

Adipose tissue has a dynamic immune system that adapts to changes in diet and maintains homeostatic tissue remodeling. Adipose type 1 innate lymphoid cells (AT1-ILCs) promote pro-inflammatory macrophages in obesity, but little is known about their functions at steady state. Here we found that human and murine adipose tissue harbor heterogeneous populations of AT1-ILCs. Experiments using parabiotic mice fed a high-fat diet (HFD) showed differential trafficking of AT1-ILCs, particularly in response to short- and long-term HFD and diet restriction. At steady state, AT1-ILCs displayed cytotoxic activity toward adipose tissue macrophages (ATMs). Depletion of AT1-ILCs and perforin deficiency resulted in alterations in the ratio of inflammatory to anti-inflammatory ATMs, and adoptive transfer of AT1-ILCs exacerbated metabolic disorder. Diet-induced obesity impaired AT1-ILC killing ability. Our findings reveal a role for AT1-ILCs in regulating ATM homeostasis through cytotoxicity and suggest that this function is relevant in both homeostasis and metabolic disease.


Subject(s)
Adipose Tissue/immunology , Cytotoxicity, Immunologic/immunology , Homeostasis/immunology , Lymphocytes/immunology , Macrophages/immunology , Obesity/immunology , Adipose Tissue/cytology , Animals , Female , Humans , Immunity, Innate , Male , Mice , Mice, Inbred C57BL , Obesity/pathology
10.
Nature ; 582(7811): 259-264, 2020 06.
Article in English | MEDLINE | ID: mdl-32499639

ABSTRACT

The synovium is a mesenchymal tissue composed mainly of fibroblasts, with a lining and sublining that surround the joints. In rheumatoid arthritis the synovial tissue undergoes marked hyperplasia, becomes inflamed and invasive, and destroys the joint1,2. It has recently been shown that a subset of fibroblasts in the sublining undergoes a major expansion in rheumatoid arthritis that is linked to disease activity3-5; however, the molecular mechanism by which these fibroblasts differentiate and expand is unknown. Here we identify a critical role for NOTCH3 signalling in the differentiation of perivascular and sublining fibroblasts that express CD90 (encoded by THY1). Using single-cell RNA sequencing and synovial tissue organoids, we found that NOTCH3 signalling drives both transcriptional and spatial gradients-emanating from vascular endothelial cells outwards-in fibroblasts. In active rheumatoid arthritis, NOTCH3 and Notch target genes are markedly upregulated in synovial fibroblasts. In mice, the genetic deletion of Notch3 or the blockade of NOTCH3 signalling attenuates inflammation and prevents joint damage in inflammatory arthritis. Our results indicate that synovial fibroblasts exhibit a positional identity that is regulated by endothelium-derived Notch signalling, and that this stromal crosstalk pathway underlies inflammation and pathology in inflammatory arthritis.


Subject(s)
Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Receptor, Notch3/metabolism , Signal Transduction , Synovial Membrane/pathology , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Endothelial Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Receptor, Notch3/antagonists & inhibitors , Receptor, Notch3/deficiency , Receptor, Notch3/genetics , Thy-1 Antigens/metabolism
11.
Nat Immunol ; 14(1): 90-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202270

ABSTRACT

Invariant natural killer T cells (iNKT cells) are innate-like T lymphocytes that act as critical regulators of the immune response. To better characterize this population, we profiled gene expression in iNKT cells during ontogeny and in peripheral subsets as part of the Immunological Genome Project. High-resolution comparative transcriptional analyses defined developmental and subset-specific programs of gene expression by iNKT cells. In addition, we found that iNKT cells shared an extensive transcriptional program with NK cells, similar in magnitude to that shared with major histocompatibility complex (MHC)-restricted T cells. Notably, the program shared by NK cells and iNKT cells also operated constitutively in γδ T cells and in adaptive T cells after activation. Together our findings highlight a core effector program regulated distinctly in innate and adaptive lymphocytes.


Subject(s)
Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Transcriptome , Adaptive Immunity/genetics , Animals , Cell Differentiation , Cell Lineage , Genome, Human/immunology , Humans , Immunity, Innate/genetics , Immunologic Memory/genetics , Mice , Microarray Analysis , Thymus Gland/growth & development
12.
Nature ; 570(7760): 246-251, 2019 06.
Article in English | MEDLINE | ID: mdl-31142839

ABSTRACT

The identification of lymphocyte subsets with non-overlapping effector functions has been pivotal to the development of targeted therapies in immune-mediated inflammatory diseases (IMIDs)1,2. However, it remains unclear whether fibroblast subclasses with non-overlapping functions also exist and are responsible for the wide variety of tissue-driven processes observed in IMIDs, such as inflammation and damage3-5. Here we identify and describe the biology of distinct subsets of fibroblasts responsible for mediating either inflammation or tissue damage in arthritis. We show that deletion of fibroblast activation protein-α (FAPα)+ fibroblasts suppressed both inflammation and bone erosions in mouse models of resolving and persistent arthritis. Single-cell transcriptional analysis identified two distinct fibroblast subsets within the FAPα+ population: FAPα+THY1+ immune effector fibroblasts located in the synovial sub-lining, and FAPα+THY1- destructive fibroblasts restricted to the synovial lining layer. When adoptively transferred into the joint, FAPα+THY1- fibroblasts selectively mediate bone and cartilage damage with little effect on inflammation, whereas transfer of FAPα+ THY1+ fibroblasts resulted in a more severe and persistent inflammatory arthritis, with minimal effect on bone and cartilage. Our findings describing anatomically discrete, functionally distinct fibroblast subsets with non-overlapping functions have important implications for cell-based therapies aimed at modulating inflammation and tissue damage.


Subject(s)
Arthritis, Rheumatoid/pathology , Fibroblasts/pathology , Animals , Bone and Bones/pathology , Endopeptidases , Female , Fibroblasts/classification , Fibroblasts/metabolism , Gelatinases/metabolism , Humans , Inflammation/pathology , Joints/pathology , Male , Membrane Proteins/metabolism , Mice , RNA-Seq , Serine Endopeptidases/metabolism , Single-Cell Analysis , Synovial Membrane/pathology , Thy-1 Antigens/metabolism
13.
Nat Immunol ; 13(5): 511-8, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22473038

ABSTRACT

Innate γδ T cells function in the early phase of immune responses. Although innate γδ T cells have often been studied as one homogenous population, they can be functionally classified into effector subsets on the basis of the production of signature cytokines, analogous to adaptive helper T cell subsets. However, unlike the function of adaptive T cells, γδ effector T cell function correlates with genomically encoded T cell antigen receptor (TCR) chains, which suggests that clonal TCR selection is not the main determinant of the differentiation of γδ effector cells. A high-resolution transcriptome analysis of all emergent γδ thymocyte subsets segregated on the basis of use of the TCR γ-chain or δ-chain indicated the existence of three separate subtypes of γδ effector cells in the thymus. The immature γδ subsets were distinguished by unique transcription-factor modules that program effector function.


Subject(s)
Cell Differentiation/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Transcriptome/immunology , Age Factors , Animals , CD24 Antigen/immunology , CD24 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/immunology , Fetus/cytology , Fetus/immunology , Flow Cytometry , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Models, Immunological , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Principal Component Analysis , Receptors, Antigen, T-Cell, gamma-delta/classification , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptome/genetics
14.
Nat Immunol ; 13(5): 499-510, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22466668

ABSTRACT

Lymph node stromal cells (LNSCs) closely regulate immunity and self-tolerance, yet key aspects of their biology remain poorly elucidated. Here, comparative transcriptomic analyses of mouse LNSC subsets demonstrated the expression of important immune mediators, growth factors and previously unknown structural components. Pairwise analyses of ligands and cognate receptors across hematopoietic and stromal subsets suggested a complex web of crosstalk. Fibroblastic reticular cells (FRCs) showed enrichment for higher expression of genes relevant to cytokine signaling, relative to their expression in skin and thymic fibroblasts. LNSCs from inflamed lymph nodes upregulated expression of genes encoding chemokines and molecules involved in the acute-phase response and the antigen-processing and antigen-presentation machinery. Poorly studied podoplanin (gp38)-negative CD31(-) LNSCs showed similarities to FRCs but lacked expression of interleukin 7 (IL-7) and were identified as myofibroblastic pericytes that expressed integrin α(7). Together our data comprehensively describe the transcriptional characteristics of LNSC subsets.


Subject(s)
Gene Expression/immunology , Inflammation/immunology , Lymph Nodes/immunology , Stromal Cells/immunology , Stromal Cells/metabolism , Transcriptome , Acute-Phase Reaction/immunology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Cytokines/immunology , Cytokines/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Homeostasis/immunology , Inflammation/genetics , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Lymph Nodes/cytology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Pericytes/immunology , Pericytes/metabolism , Self Tolerance/immunology , Tissue Array Analysis/methods
15.
EMBO J ; 38(14): e101260, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31304630

ABSTRACT

Tissue-resident iNKT cells maintain tissue homeostasis and peripheral surveillance against pathogens; however, studying these cells is challenging due to their low abundance and poor recovery from tissues. We here show that iNKT transnuclear mice, generated by somatic cell nuclear transfer, have increased tissue resident iNKT cells. We examined expression of PLZF, T-bet, and RORγt, as well as cytokine/chemokine profiles, and found that both monoclonal and polyclonal iNKT cells differentiated into functional subsets that faithfully replicated those seen in wild-type mice. We detected iNKT cells from tissues in which they are rare, including adipose, lung, skin-draining lymph nodes, and a previously undescribed population in Peyer's patches (PP). PP-NKT cells produce the majority of the IL-4 in Peyer's patches and provide indirect help for B-cell class switching to IgG1 in both transnuclear and wild-type mice. Oral vaccination with α-galactosylceramide shows enhanced fecal IgG1 titers in iNKT cell-sufficient mice. Transcriptional profiling reveals a unique signature of PP-NKT cells, characterized by tissue residency. We thus define PP-NKT as potentially important for surveillance for mucosal pathogens.


Subject(s)
Gene Expression Profiling/methods , Immunoglobulin Class Switching , Immunoglobulin G/genetics , Natural Killer T-Cells/metabolism , Peyer's Patches/immunology , Animals , Cell Differentiation , Cells, Cultured , Female , Galactosylceramides/administration & dosage , Galactosylceramides/immunology , Interleukin-4/genetics , Mice , Natural Killer T-Cells/cytology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Transfer Techniques , Promyelocytic Leukemia Zinc Finger Protein/genetics , T-Box Domain Proteins/genetics , Vaccination
16.
Nat Immunol ; 12(12): 1202-11, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037601

ABSTRACT

Invariant natural killer T cells (iNKT cells) have a prominent role during infection and other inflammatory processes, and these cells can be activated through their T cell antigen receptors by microbial lipid antigens. However, increasing evidence shows that they are also activated in situations in which foreign lipid antigens would not be present, which suggests a role for lipid self antigen. We found that an abundant endogenous lipid, ß-D-glucopyranosylceramide (ß-GlcCer), was a potent iNKT cell self antigen in mouse and human and that its activity depended on the composition of the N-acyl chain. Furthermore, ß-GlcCer accumulated during infection and in response to Toll-like receptor agonists, contributing to iNKT cell activation. Thus, we propose that recognition of ß-GlcCer by the invariant T cell antigen receptor translates innate danger signals into iNKT cell activation.


Subject(s)
Autoantigens/immunology , Bacterial Infections/immunology , Glycosphingolipids/immunology , Natural Killer T-Cells/immunology , Animals , Autoimmunity/immunology , Cell Line , Glycosphingolipids/metabolism , Humans , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
17.
Nat Immunol ; 13(1): 44-50, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22120118

ABSTRACT

Mouse invariant natural killer T cells (iNKT cells) provide cognate and noncognate help for lipid and protein-specific B cells, respectively. However, the long-term outcome for B cells after cognate help is provided by iNKT cells is unknown at present. Here we found that cognate iNKT cell help resulted in a B cell differentiation program characterized by extrafollicular plasmablasts, germinal-center formation, affinity maturation and a robust primary immunoglobulin G (IgG) antibody response that was uniquely dependent on iNKT cell-derived interleukin 21 (IL-21). However, cognate help from iNKT cells did not generate an enhanced humoral memory response. Thus, cognate iNKT cell help for lipid-specific B cells induces a unique signature that is a hybrid of classic T cell-dependent and T cell-independent type 2 B cell responses.


Subject(s)
Antigens/immunology , B-Lymphocytes/immunology , Interleukins/physiology , Lipids/immunology , Natural Killer T-Cells/immunology , Animals , Germinal Center/immunology , Immunity, Humoral , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Spleen/immunology
18.
Nature ; 542(7639): 110-114, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28150777

ABSTRACT

CD4+ T cells are central mediators of autoimmune pathology; however, defining their key effector functions in specific autoimmune diseases remains challenging. Pathogenic CD4+ T cells within affected tissues may be identified by expression of markers of recent activation. Here we use mass cytometry to analyse activated T cells in joint tissue from patients with rheumatoid arthritis, a chronic immune-mediated arthritis that affects up to 1% of the population. This approach revealed a markedly expanded population of PD-1hiCXCR5-CD4+ T cells in synovium of patients with rheumatoid arthritis. However, these cells are not exhausted, despite high PD-1 expression. Rather, using multidimensional cytometry, transcriptomics, and functional assays, we define a population of PD-1hiCXCR5- 'peripheral helper' T (TPH) cells that express factors enabling B-cell help, including IL-21, CXCL13, ICOS, and MAF. Like PD-1hiCXCR5+ T follicular helper cells, TPH cells induce plasma cell differentiation in vitro through IL-21 secretion and SLAMF5 interaction (refs 3, 4). However, global transcriptomics highlight differences between TPH cells and T follicular helper cells, including altered expression of BCL6 and BLIMP1 and unique expression of chemokine receptors that direct migration to inflamed sites, such as CCR2, CX3CR1, and CCR5, in TPH cells. TPH cells appear to be uniquely poised to promote B-cell responses and antibody production within pathologically inflamed non-lymphoid tissues.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Arthritis, Rheumatoid/blood , B-Lymphocytes/pathology , Cell Differentiation , Cell Movement , Chemokine CXCL13/metabolism , Gene Expression Profiling , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukins/metabolism , Macrophage-Activating Factors , Positive Regulatory Domain I-Binding Factor 1 , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/deficiency , Receptors, CXCR5/metabolism , Receptors, Chemokine/metabolism , Repressor Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , Synovial Fluid/immunology , T-Lymphocytes, Helper-Inducer/metabolism
19.
Proc Natl Acad Sci U S A ; 117(16): 8900-8911, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32253314

ABSTRACT

Signaling pathways that sense amino acid abundance are integral to tissue homeostasis and cellular defense. Our laboratory has previously shown that halofuginone (HF) inhibits the prolyl-tRNA synthetase catalytic activity of glutamyl-prolyl-tRNA synthetase (EPRS), thereby activating the amino acid response (AAR). We now show that HF treatment selectively inhibits inflammatory responses in diverse cell types and that these therapeutic benefits occur in cells that lack GCN2, the signature effector of the AAR. Depletion of arginine, histidine, or lysine from cultured fibroblast-like synoviocytes recapitulates key aspects of HF treatment, without utilizing GCN2 or mammalian target of rapamycin complex 1 pathway signaling. Like HF, the threonyl-tRNA synthetase inhibitor borrelidin suppresses the induction of tissue remodeling and inflammatory mediators in cytokine-stimulated fibroblast-like synoviocytes without GCN2, but both aminoacyl-tRNA synthetase (aaRS) inhibitors are sensitive to the removal of GCN1. GCN1, an upstream component of the AAR pathway, binds to ribosomes and is required for GCN2 activation. These observations indicate that aaRS inhibitors, like HF, can modulate inflammatory response without the AAR/GCN2 signaling cassette, and that GCN1 has a role that is distinct from its activation of GCN2. We propose that GCN1 participates in a previously unrecognized amino acid sensor pathway that branches from the canonical AAR.


Subject(s)
Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Piperidines/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/surgery , Cell Line , Fibroblasts , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Lung/cytology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Piperidines/therapeutic use , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Quinazolinones/therapeutic use , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Seq , Signal Transduction/immunology , Synovial Membrane/cytology , Synovial Membrane/pathology , Synoviocytes , Trans-Activators/genetics , Trans-Activators/metabolism
20.
Proc Natl Acad Sci U S A ; 117(10): 5532-5541, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32079724

ABSTRACT

The role of stromal fibroblasts in chronic inflammation is unfolding. In rheumatoid arthritis, leukocyte-derived cytokines TNF and IL-17A work together, activating fibroblasts to become a dominant source of the hallmark cytokine IL-6. However, IL-17A alone has minimal effect on fibroblasts. To identify key mediators of the synergistic response to TNF and IL-17A in human synovial fibroblasts, we performed time series, dose-response, and gene-silencing transcriptomics experiments. Here we show that in combination with TNF, IL-17A selectively induces a specific set of genes mediated by factors including cut-like homeobox 1 (CUX1) and IκBζ (NFKBIZ). In the promoters of CXCL1, CXCL2, and CXCL3, we found a putative CUX1-NF-κB binding motif not found elsewhere in the genome. CUX1 and NF-κB p65 mediate transcription of these genes independent of LIFR, STAT3, STAT4, and ELF3. Transcription of NFKBIZ, encoding the atypical IκB factor IκBζ, is IL-17A dose-dependent, and IκBζ only mediates the transcriptional response to TNF and IL-17A, but not to TNF alone. In fibroblasts, IL-17A response depends on CUX1 and IκBζ to engage the NF-κB complex to produce chemoattractants for neutrophil and monocyte recruitment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Arthritis, Rheumatoid/metabolism , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Inflammation/metabolism , Interleukin-17/physiology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcriptome/physiology , Tumor Necrosis Factor-alpha/physiology , Adaptor Proteins, Signal Transducing/genetics , Arthritis, Rheumatoid/genetics , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL2/genetics , Chemokines, CXC/genetics , Chemotactic Factors/genetics , Fibroblasts/drug effects , Homeodomain Proteins/genetics , Humans , Inflammation/genetics , Interleukin-17/pharmacology , Interleukin-6/genetics , Matrix Metalloproteinase 3/metabolism , Monocytes/drug effects , Monocytes/physiology , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Stromal Cells/drug effects , Stromal Cells/metabolism , Synovial Fluid , Transcription Factor RelA/metabolism , Transcription Factors/genetics , Transcriptome/radiation effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL