Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 610
Filter
Add more filters

Publication year range
1.
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
2.
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.

4.
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
5.
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
6.
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
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.
Cell ; 161(1): 5-8, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25815978

ABSTRACT

Chefs and scientists exploring biophysical processes have given rise to molecular gastronomy. In this Commentary, we describe how a scientific understanding of recipes and techniques facilitates the development of new textures and expands the flavor palette. The new dishes that result engage our senses in unexpected ways. PAPERCLIP.


Subject(s)
Dietary Proteins/chemistry , Food Analysis , Taste , Biophysics , Cooking , Fermentation , Food , Humans
9.
Nature ; 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987586

ABSTRACT

Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.

10.
Nature ; 611(7934): 68-73, 2022 11.
Article in English | MEDLINE | ID: mdl-36289343

ABSTRACT

Objects that deform a liquid interface are subject to capillary forces, which can be harnessed to assemble the objects1-4. Once assembled, such structures are generally static. Here we dynamically modulate these forces to move objects in programmable two-dimensional patterns. We 3D-print devices containing channels that trap floating objects using repulsive capillary forces5,6, then move these devices vertically in a water bath. Because the channel cross-sections vary with height, the trapped objects can be steered in two dimensions. The device and interface therefore constitute a simple machine that converts vertical to lateral motion. We design machines that translate, rotate and separate multiple floating objects and that do work on submerged objects through cyclic vertical motion. We combine these elementary machines to make centimetre-scale compound machines that braid micrometre-scale filaments into prescribed topologies, including non-repeating braids. Capillary machines are distinct from mechanical, optical or fluidic micromanipulators in that a meniscus links the object to the machine. Therefore, the channel shapes need only be controlled on the scale of the capillary length (a few millimetres), even when the objects are microscopic. Consequently, such machines can be built quickly and inexpensively. This approach could be used to manipulate micrometre-scale particles or to braid microwires for high-frequency electronics.

11.
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
12.
Proc Natl Acad Sci U S A ; 121(27): e2311891121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38913891

ABSTRACT

Direct design of complex functional materials would revolutionize technologies ranging from printable organs to novel clean energy devices. However, even incremental steps toward designing functional materials have proven challenging. If the material is constructed from highly complex components, the design space of materials properties rapidly becomes too computationally expensive to search. On the other hand, very simple components such as uniform spherical particles are not powerful enough to capture rich functional behavior. Here, we introduce a differentiable materials design model with components that are simple enough to design yet powerful enough to capture complex materials properties: rigid bodies composed of spherical particles with directional interactions (patchy particles). We showcase the method with self-assembly designs ranging from open lattices to self-limiting clusters, all of which are notoriously challenging design goals to achieve using purely isotropic particles. By directly optimizing over the location and interaction of the patches on patchy particles using gradient descent, we dramatically reduce the computation time for finding the optimal building blocks.

13.
Proc Natl Acad Sci U S A ; 121(23): e2320007121, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38820003

ABSTRACT

A dynamical systems approach to turbulence envisions the flow as a trajectory through a high-dimensional state space [Hopf, Commun. Appl. Maths 1, 303 (1948)]. The chaotic dynamics are shaped by the unstable simple invariant solutions populating the inertial manifold. The hope has been to turn this picture into a predictive framework where the statistics of the flow follow from a weighted sum of the statistics of each simple invariant solution. Two outstanding obstacles have prevented this goal from being achieved: 1) paucity of known solutions and 2) the lack of a rational theory for predicting the required weights. Here, we describe a method to substantially solve these problems, and thereby provide compelling evidence that the probability density functions (PDFs) of a fully developed turbulent flow can be reconstructed with a set of unstable periodic orbits. Our method for finding solutions uses automatic differentiation, with high-quality guesses constructed by minimizing a trajectory-dependent loss function. We use this approach to find hundreds of solutions in turbulent, two-dimensional Kolmogorov flow. Robust statistical predictions are then computed by learning weights after converting a turbulent trajectory into a Markov chain for which the states are individual solutions, and the nearest solution to a given snapshot is determined using a deep convolutional autoencoder. In this study, the PDFs of a spatiotemporally chaotic system have been successfully reproduced with a set of simple invariant states, and we provide a fascinating connection between self-sustaining dynamical processes and the more well-known statistical properties of turbulence.

14.
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
15.
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
16.
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
17.
Proc Natl Acad Sci U S A ; 120(31): e2303928120, 2023 08.
Article in English | MEDLINE | ID: mdl-37494398

ABSTRACT

Although sensor technologies have allowed us to outperform the human senses of sight, hearing, and touch, the development of artificial noses is significantly behind their biological counterparts. This largely stems from the sophistication of natural olfaction, which relies on both fluid dynamics within the nasal anatomy and the response patterns of hundreds to thousands of unique molecular-scale receptors. We designed a sensing approach to identify volatiles inspired by the fluid dynamics of the nose, allowing us to extract information from a single sensor (here, the reflectance spectra from a mesoporous one-dimensional photonic crystal) rather than relying on a large sensor array. By accentuating differences in the nonequilibrium mass-transport dynamics of vapors and training a machine learning algorithm on the sensor output, we clearly identified polar and nonpolar volatile compounds, determined the mixing ratios of binary mixtures, and accurately predicted the boiling point, flash point, vapor pressure, and viscosity of a number of volatile liquids, including several that had not been used for training the model. We further implemented a bioinspired active sniffing approach, in which the analyte delivery was performed in well-controlled 'inhale-exhale' sequences, enabling an additional modality of differentiation and reducing the duration of data collection and analysis to seconds. Our results outline a strategy to build accurate and rapid artificial noses for volatile compounds that can provide useful information such as the composition and physical properties of chemicals, and can be applied in a variety of fields, including disease diagnosis, hazardous waste management, and healthy building monitoring.


Subject(s)
Nose , Smell , Humans , Electronic Nose , Machine Learning , Gases
18.
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
19.
Nat Immunol ; 14(6): 619-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23644507

ABSTRACT

The differentiation of αßT cells from thymic precursors is a complex process essential for adaptive immunity. Here we exploited the breadth of expression data sets from the Immunological Genome Project to analyze how the differentiation of thymic precursors gives rise to mature T cell transcriptomes. We found that early T cell commitment was driven by unexpectedly gradual changes. In contrast, transit through the CD4(+)CD8(+) stage involved a global shutdown of housekeeping genes that is rare among cells of the immune system and correlated tightly with expression of the transcription factor c-Myc. Selection driven by major histocompatibility complex (MHC) molecules promoted a large-scale transcriptional reactivation. We identified distinct signatures that marked cells destined for positive selection versus apoptotic deletion. Differences in the expression of unexpectedly few genes accompanied commitment to the CD4(+) or CD8(+) lineage, a similarity that carried through to peripheral T cells and their activation, demonstrated by mass cytometry phosphoproteomics. The transcripts newly identified as encoding candidate mediators of key transitions help define the 'known unknowns' of thymocyte differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , Cells, Cultured , Cluster Analysis , Flow Cytometry , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Phosphorylation/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Thymocytes/cytology , Thymocytes/immunology , Thymocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
20.
Nat Immunol ; 14(6): 633-43, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23624555

ABSTRACT

The differentiation of hematopoietic stem cells into cells of the immune system has been studied extensively in mammals, but the transcriptional circuitry that controls it is still only partially understood. Here, the Immunological Genome Project gene-expression profiles across mouse immune lineages allowed us to systematically analyze these circuits. To analyze this data set we developed Ontogenet, an algorithm for reconstructing lineage-specific regulation from gene-expression profiles across lineages. Using Ontogenet, we found differentiation stage-specific regulators of mouse hematopoiesis and identified many known hematopoietic regulators and 175 previously unknown candidate regulators, as well as their target genes and the cell types in which they act. Among the previously unknown regulators, we emphasize the role of ETV5 in the differentiation of γδ T cells. As the transcriptional programs of human and mouse cells are highly conserved, it is likely that many lessons learned from the mouse model apply to humans.


Subject(s)
Algorithms , Gene Expression Regulation/immunology , Immune System/metabolism , Transcription, Genetic/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Expression Profiling , Gene Regulatory Networks/immunology , Humans , Immune System/cytology , Mice , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Repressor Proteins/genetics , Repressor Proteins/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Trans-Activators/genetics , Trans-Activators/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcriptome/genetics , Transcriptome/immunology
SELECTION OF CITATIONS
SEARCH DETAIL