Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956325

ABSTRACT

Antigen discovery technologies have largely focused on major histocompatibility complex (MHC) class I-restricted human T cell receptors (TCRs), leaving methods for MHC class II-restricted and mouse TCR reactivities relatively undeveloped. Here we present TCR mapping of antigenic peptides (TCR-MAP), an antigen discovery method that uses a synthetic TCR-stimulated circuit in immortalized T cells to activate sortase-mediated tagging of engineered antigen-presenting cells (APCs) expressing processed peptides on MHCs. Live, tagged APCs can be directly purified for deconvolution by sequencing, enabling TCRs with unknown specificity to be queried against barcoded peptide libraries in a pooled screening context. TCR-MAP accurately captures self-reactivities or viral reactivities with high throughput and sensitivity for both MHC class I-restricted and class II-restricted TCRs. We elucidate problematic cross-reactivities of clinical TCRs targeting the cancer/testis melanoma-associated antigen A3 and discover targets of myocarditis-inciting autoreactive T cells in mice. TCR-MAP has the potential to accelerate T cell antigen discovery efforts in the context of cancer, infectious disease and autoimmunity.

2.
Cell ; 186(25): 5569-5586.e21, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38016469

ABSTRACT

CD4+ T cells play fundamental roles in orchestrating immune responses and tissue homeostasis. However, our inability to associate peptide human leukocyte antigen class-II (HLA-II) complexes with their cognate T cell receptors (TCRs) in an unbiased manner has hampered our understanding of CD4+ T cell function and role in pathologies. Here, we introduce TScan-II, a highly sensitive genome-scale CD4+ antigen discovery platform. This platform seamlessly integrates the endogenous HLA-II antigen-processing machinery in synthetic antigen-presenting cells and TCR signaling in T cells, enabling the simultaneous screening of multiple HLAs and TCRs. Leveraging genome-scale human, virome, and epitope mutagenesis libraries, TScan-II facilitates de novo antigen discovery and deep exploration of TCR specificity. We demonstrate TScan-II's potential for basic and translational research by identifying a non-canonical antigen for a cancer-reactive CD4+ T cell clone. Additionally, we identified two antigens for clonally expanded CD4+ T cells in Sjögren's disease, which bind distinct HLAs and are expressed in HLA-II-positive ductal cells within affected salivary glands.


Subject(s)
CD4-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Humans , Antigen-Presenting Cells , CD4 Antigens/metabolism , HLA Antigens/metabolism , Receptors, Antigen, T-Cell/metabolism , Cell Line , Genome, Human
3.
Nat Immunol ; 22(2): 179-192, 2021 02.
Article in English | MEDLINE | ID: mdl-33462452

ABSTRACT

Metabolic programming controls immune cell lineages and functions, but little is known about γδ T cell metabolism. Here, we found that γδ T cell subsets making either interferon-γ (IFN-γ) or interleukin (IL)-17 have intrinsically distinct metabolic requirements. Whereas IFN-γ+ γδ T cells were almost exclusively dependent on glycolysis, IL-17+ γδ T cells strongly engaged oxidative metabolism, with increased mitochondrial mass and activity. These distinct metabolic signatures were surprisingly imprinted early during thymic development and were stably maintained in the periphery and within tumors. Moreover, pro-tumoral IL-17+ γδ T cells selectively showed high lipid uptake and intracellular lipid storage and were expanded in obesity and in tumors of obese mice. Conversely, glucose supplementation enhanced the antitumor functions of IFN-γ+ γδ T cells and reduced tumor growth upon adoptive transfer. These findings have important implications for the differentiation of effector γδ T cells and their manipulation in cancer immunotherapy.


Subject(s)
Breast Neoplasms/metabolism , Colonic Neoplasms/metabolism , Energy Metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma, Experimental/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism , Tumor Microenvironment , Animals , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Cell Lineage , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Female , Glucose/metabolism , Glycolysis , Humans , Immunotherapy, Adoptive , Interferon-gamma/metabolism , Interleukin-17/metabolism , Lipid Metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Obesity/immunology , Obesity/metabolism , Organ Culture Techniques , Phenotype , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Thymus Gland/immunology , Tumor Burden
4.
Cell Metab ; 32(2): 243-258.e6, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32516575

ABSTRACT

Adipose tissue invariant natural killer T (iNKT) cells are phenotypically different from other iNKT cells because they produce IL-10 and control metabolic homeostasis. Why that is the case is unclear. Here, using single-cell RNA sequencing, we found several adipose iNKT clusters, which we grouped into two functional populations based on NK1.1 expression. NK1.1NEG cells almost exclusively produced IL-10 and other regulatory cytokines, while NK1.1POS iNKT cells predominantly produced IFNγ. Mechanistically, biochemical fractionation revealed that free fatty acids drive IL-10 production primarily in NK1.1NEG iNKT cells via the IRE1α-XBP1s arm of the unfolded protein response. Correspondingly, adoptive transfer of adipose tissue NK1.1NEG iNKT cells selectively restored metabolic function in obese mice. Further, we found an unexpected role for NK1.1POS iNKT cells in lean adipose tissue, as IFNγ licenses natural killer cell-mediated macrophage killing to limit pathological macrophage expansion. Together, these two iNKT cell populations utilize non-redundant pathways to preserve metabolic integrity.


Subject(s)
Adipose Tissue/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Natural Killer T-Cells/metabolism , Animals , Endoplasmic Reticulum Stress , Homeostasis , Interferon-gamma/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
5.
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.

6.
J Immunol ; 201(7): 1827-1834, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30224362

ABSTRACT

During the past 25 y, the immune system has appeared as a key regulator of adipose tissue biology and metabolic homeostasis. In lean animals, adipose-resident leukocytes maintain an anti-inflammatory microenvironment that preserves the proper functioning of the tissue. In this review, we describe two populations of innate T cells enriched in adipose tissue, invariant NKT and γδ T cells, and how they serve overlapping and nonredundant roles in controlling adipose tissue functions. These cells interact with and expand anti-inflammatory regulatory T cells and M2 macrophages, thereby driving a metabolically beneficial tissue milieu. Surprisingly, we have found that adipose invariant NKT and γδ T cells also promote weight loss and heat production in a process called "nonshivering thermogenesis." The data surrounding these two cell types highlight their powerful ability to regulate not only other leukocytes, but also tissue-wide processes that affect an entire organism.


Subject(s)
Adipose Tissue/physiology , Immunity, Innate , Macrophages/immunology , Natural Killer T-Cells/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cellular Microenvironment , Cytokines/metabolism , Homeostasis , Humans , Obesity/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Th2 Cells/immunology
7.
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
8.
Int J Radiat Oncol Biol Phys ; 99(2): 468-475, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28871998

ABSTRACT

PURPOSE: Few tools exist to predict clinical outcomes after radiopharmaceutical therapy. Our goal was to identify associations between blood-based biomarkers of radiation effect and clinical outcomes after 131I-metaiodobenzylguanidine (131I-MIBG) therapy in patients with neuroblastoma. METHODS AND MATERIALS: We conducted a prospective, single-center cohort study in children with advanced neuroblastoma treated with 131I-MIBG as monotherapy or in combination with systemic putative radiation sensitizers. We collected serial peripheral blood samples after 131I-MIBG infusions and quantified a panel of protein and messenger RNA markers. We plotted relative change from baseline to assess degree of modulation over time and then evaluated association of marker modulation with toxicity and response endpoints. RESULTS: The cohort included 40 patients (30 male/10 female; median age 7 years). We observed significant modulation of the majority of markers between baseline and hour 72 after 131I-MIBG. Greater fold increase of plasma FLT3 ligand was associated with subsequent grade 4 neutropenia (P=.039). Modulation of peripheral blood BCLXL and DDB2 was associated with grade 3+ nonhematologic toxicity (P=.043 and .048, respectively). No markers were associated with tumor response. Greater plasma FLT3 ligand, BCLXL, and BCL2 modulation was observed in patients receiving 131I-MIBG in combination with radiation sensitizers. Among 9 patients who received 2 courses, the degree of modulation in serum amylase was significantly lower after the second course (P=.012). CONCLUSIONS: Peripheral blood biomarkers relevant to radiation exposure are significantly modulated during the acute period after 131I-MIBG. The degree of modulation of a subset of these markers is associated with toxicity and receipt of concomitant radiation sensitizers.


Subject(s)
3-Iodobenzylguanidine/adverse effects , Biomarkers/blood , Neuroblastoma/blood , Radiopharmaceuticals/adverse effects , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Male , Membrane Proteins/blood , Neutropenia/etiology , Pilot Projects , Prospective Studies , Radiation-Sensitizing Agents/therapeutic use
9.
J Clin Invest ; 127(9): 3300-3312, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28758901

ABSTRACT

M2 macrophages, innate lymphoid type 2 cells (ILC2s), eosinophils, Tregs, and invariant NK T cells (iNKT cells) all help to control adipose tissue inflammation, while M1 macrophages, TNF, and other inflammatory cytokines drive inflammation and insulin resistance in obesity. Stromal cells regulate leukocyte responses in lymph nodes, but the role of stromal cells in adipose tissue inflammation is unknown. PDGFRα+ stromal cells are major producers of IL-33 in adipose tissue. Here, we show that mesenchymal cadherin-11 modulates stromal fibroblast function. Cadherin-11-deficient mice displayed increased stromal production of IL-33, with concomitant enhancements in ILC2s and M2 macrophages that helped control adipose tissue inflammation. Higher expression levels of IL-33 in cadherin-11-deficient mice mediated ILC2 activation, resulting in higher IL-13 expression levels and M2 macrophage expansion in adipose tissue. Consistent with reduced adipose tissue inflammation, cadherin-11-deficient mice were protected from obesity-induced glucose intolerance and adipose tissue fibrosis. Importantly, anti-cadherin-11 mAb blockade similarly improved inflammation and glycemic control in obese WT mice. These results suggest that stromal fibroblasts expressing cadherin-11 regulate adipose tissue inflammation and thus highlight cadherin-11 as a potential therapeutic target for the management of obesity.


Subject(s)
Adipose Tissue/physiopathology , Cadherins/metabolism , Diabetes Mellitus, Experimental/physiopathology , Inflammation/physiopathology , Adipocytes/cytology , Adipose Tissue/metabolism , Animals , Cell Differentiation , Crosses, Genetic , Diabetes Mellitus, Experimental/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Glucose Intolerance/metabolism , Inflammation/metabolism , Insulin Resistance , Interleukin-13/metabolism , Interleukin-33/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Obesity/metabolism , Phenotype
10.
Semin Immunol ; 28(5): 431-440, 2016 10.
Article in English | MEDLINE | ID: mdl-27789087

ABSTRACT

At the simplest interpretation of the word, Immunometabolism describes the intersection of the fields of immunology and metabolism. With rapidly growing interest in this field, the term has expanded, and now encompasses a variety of concepts and definitions shaped by an individual's scientific area of expertise, cell-type and tissue of interest, and biological approach. One scientist may be interested in investigating the intrinsic metabolic checkpoints that drive a M1 versus M2 macrophage response, while another may be interested in how macrophages affect systemic metabolism during obesity. Although both interests have very different foci, they both reflect the current interests in immunometabolism and studies over the last decade have uncovered new metabolic nodes that dictate the course of effector fate within cells, as well as an unexpected role for the immune system in controlling systemic metabolism. Thus, immunometabolism is at the frontier for many novel therapeutic targets to control both cell intrinsic and whole body metabolism in many diseases including cancer, diabetes, obesity, and sepsis among others. In this review, we hope to break down the word immunometabolism into two main themes: whole-body metabolism and cellular bioenergetics. In each instance we will focus on the adipose tissue and its resident immune cells to illustrate recent advances in both sectors of immunometabolism.


Subject(s)
Adipose Tissue/immunology , Adipose Tissue/metabolism , Energy Metabolism , Immunity , Immunomodulation , Animals , Disease Susceptibility , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Metabolic Networks and Pathways , Signal Transduction
11.
Immunogenetics ; 68(8): 649-63, 2016 08.
Article in English | MEDLINE | ID: mdl-27457886

ABSTRACT

Invariant natural killer T (iNKT) cells are a specialized T cell subset that plays an important role in host defense, orchestrating both innate and adaptive immune effector responses against a variety of microbes. Specific microbial lipids and mammalian self lipids displayed by the antigen-presenting molecule CD1d can activate iNKT cells through their semi-invariant αß T cell receptors (TCRs). iNKT cells also constitutively express receptors for inflammatory cytokines typically secreted by antigen-presenting cells (APCs) after recognition of pathogen-associated molecular patterns (PAMPs), and they can be activated through these cytokine receptors either in combination with TCR signals, or in some cases even in the absence of TCR signaling. During infection, experimental evidence suggests that both TCR-driven and cytokine-driven mechanisms contribute to iNKT cell activation. While the relative contributions of these two signaling mechanisms can vary widely depending on the infectious context, both lipid antigens and PAMPs mediate reciprocal activation of iNKT cells and APCs, leading to downstream activation of multiple other immune cell types to promote pathogen clearance. In this review, we discuss the mechanisms involved in iNKT cell activation during infection, focusing on the central contributions of both lipid antigens and PAMP-induced inflammatory cytokines, and highlight in vivo examples of activation during bacterial, viral, and fungal infections.


Subject(s)
Antigen Presentation/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Humans
12.
J Am Chem Soc ; 133(8): 2390-3, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21294571

ABSTRACT

One essential requirement for more sensitive gadolinium-based MRI contrast agents is to slow the molecular tumbling of the gadolinium(III) ion, which increases the gadolinium's relaxivity (i.e., its ability to speed up the NMR relaxation of nearby water molecules). One route to this is through conjugation to high-molecular-weight polymers such as dendrimers. In this work, amine-functionalized TREN-bis(1,2-HOPO)-TAM-ethylamine and TREN-bis(1-Me-3,2-HOPO)-TAM-ethylamine ligands have been synthesized and attached to biocompatible 40 kDa esteramide (EA)- and poly-l-lysine (PLL)-based dendrimers capable of binding up to eight gadolinium complexes. These conjugates have T(1) relaxivities of up to 38.14 ± 0.02 mM(-1) s(-1) per gadolinium at 37 °C, corresponding to relaxivities of up to 228 mM(-1) s(-1) per dendrimer molecule. This relaxivity expressed on a "per Gd" basis is several times that of the small-molecule complexes and an order of magnitude higher than that of current commercial agents. Because of their high performance and low toxicity, these macromolecules may constitute an attractive complement to currently available gadolinium(III)-based contrast agents.


Subject(s)
Amides/chemistry , Contrast Media/chemistry , Dendrimers/chemistry , Esters/chemistry , Gadolinium/chemistry , Pyridones/chemistry , Contrast Media/chemical synthesis , Dendrimers/chemical synthesis , Ligands , Magnetic Resonance Imaging , Molecular Structure , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...