Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Nat Med ; 28(12): 2622-2632, 2022 12.
Article En | MEDLINE | ID: mdl-36411343

Older people are particularly susceptible to infectious and neoplastic diseases of the lung and it is unclear how lifelong exposure to environmental pollutants affects respiratory immune function. In an analysis of human lymph nodes (LNs) from 84 organ donors aged 11-93 years, we found a specific age-related decline in lung-associated, but not gut-associated, LN immune function linked to the accumulation of inhaled atmospheric particulate matter. Increasing densities of particulates were found in lung-associated LNs with age, but not in the corresponding gut-associated LNs. Particulates were specifically contained within CD68+CD169- macrophages, which exhibited decreased activation, phagocytic capacity, and altered cytokine production compared with non-particulate-containing macrophages. The structures of B cell follicles and lymphatic drainage were also disrupted in lung-associated LNs with particulates. Our results reveal that the cumulative effects of environmental exposure and age may compromise immune surveillance of the lung via direct effects on immune cell function and lymphoid architecture.


Lung , Lymph Nodes , Humans , Aged , Lymph Nodes/pathology , Disease Susceptibility/pathology , Dust , Immunity
2.
Mucosal Immunol ; 12(2): 378-389, 2019 03.
Article En | MEDLINE | ID: mdl-30523311

Defining adaptive immunity with the complex structures of the human gastrointestinal (GI) tract over life is essential for understanding immune responses to ingested antigens, commensal and pathogenic microorganisms, and dysfunctions in disease. We present here an analysis of lymphocyte localization and T cell subset composition across the human GI tract including mucosal sites (jejunum, ileum, colon), gut-associated lymphoid tissues (isolated lymphoid follicles (ILFs), Peyer's patches (PPs), appendix), and mesenteric lymph nodes (MLNs) from a total of 68 donors spanning eight decades of life. In pediatric donors, ILFs and PP containing naïve T cells and regulatory T cells (Tregs) are prevalent in the jejunum and ileum, respectively; these decline in frequency with age, contrasting stable frequencies of ILFs and T cell subsets in the colon. In the mucosa, tissue resident memory T cells develop during childhood, and persist in high frequencies into advanced ages, while T cell composition changes with age in GALT and MLN. These spatial and temporal features of human intestinal T cell immunity define signatures that can be used to train predictive machine learning algorithms. Our findings demonstrate an anatomic basis for age-associated alterations in immune responses, and establish a quantitative baseline for intestinal immunity to define disease pathologies.


Aging/physiology , Duodenum/immunology , Ileum/immunology , Intestinal Mucosa/immunology , Jejunum/immunology , Lymph Nodes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Cells, Cultured , Child , Humans , Immunity, Cellular , Organ Specificity , Peyer's Patches/immunology
3.
J Immunol ; 201(7): 2132-2140, 2018 10 01.
Article En | MEDLINE | ID: mdl-30111633

Translating studies on T cell function and modulation from mouse models to humans requires extrapolating in vivo results on mouse T cell responses in lymphoid organs (spleen and lymph nodes [LN]) to human peripheral blood T cells. However, our understanding of T cell responses in human lymphoid sites and their relation to peripheral blood remains sparse. In this study, we used a unique human tissue resource to study human T cells in different anatomical compartments within individual donors and identify a subset of memory CD8+ T cells in LN, which maintain a distinct differentiation and functional profile compared with memory CD8+ T cells in blood, spleen, bone marrow, and lungs. Whole-transcriptome and high-dimensional cytometry by time-of-flight profiling reveals that LN memory CD8+ T cells express signatures of quiescence and self-renewal compared with corresponding populations in blood, spleen, bone marrow, and lung. LN memory T cells exhibit a distinct transcriptional signature, including expression of stem cell-associated transcription factors TCF-1 and LEF-1, T follicular helper cell markers CXCR5 and CXCR4, and reduced expression of effector molecules. LN memory T cells display high homology to a subset of mouse CD8+ T cells identified in chronic infection models that respond to checkpoint blockade immunotherapy. Functionally, human LN memory T cells exhibit increased proliferation to TCR-mediated stimulation and maintain higher TCR clonal diversity compared with memory T cells from blood and other sites. These findings establish human LN as reservoirs for memory T cells with high capacities for expansion and diverse recognition and important targets for immunotherapies.


CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Lymph Nodes/immunology , T Cell Transcription Factor 1/metabolism , Animals , Antibodies, Monoclonal , Biodiversity , Cell Self Renewal , Clone Cells , Costimulatory and Inhibitory T-Cell Receptors/immunology , Humans , Immunologic Memory , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transcriptome
4.
Front Immunol ; 9: 1472, 2018.
Article En | MEDLINE | ID: mdl-30008715

B cell clones expand and contract during adaptive immune responses and can persist or grow uncontrollably in lymphoproliferative disorders. One way to monitor and track B cell clones is to perform large-scale sampling of bulk cell populations, amplifying, and sequencing antibody gene rearrangements by next-generation sequencing (NGS). Here, we describe a series of computational approaches for estimating B cell clone size in NGS immune repertoire profiling data of antibody heavy chain gene rearrangements. We define three different measures of B cell clone size-copy numbers, instances, and unique sequences-and show how these measures can be used to rank clones, analyze their diversity, and study their distribution within and between individuals. We provide a detailed, step-by-step procedure for performing these analyses using two different data sets of spleen samples from human organ donors. In the first data set, 19 independently generated biological replicates from a single individual are analyzed for B cell clone size, diversity and sampling sufficiency for clonal overlap analysis. In the second data set, B cell clones are compared in eight different organ donors. We comment upon frequently encountered pitfalls and offer practical advice with alternative approaches. Overall, we provide a series of pragmatic analytical approaches and show how different clone size measures can be used to study the clonal landscape in bulk B cell immune repertoire profiling data.

5.
Cell Rep ; 20(12): 2921-2934, 2017 Sep 19.
Article En | MEDLINE | ID: mdl-28930685

Tissue-resident memory T cells (TRMs) in mice mediate optimal protective immunity to infection and vaccination, while in humans, the existence and properties of TRMs remain unclear. Here, we use a unique human tissue resource to determine whether human tissue memory T cells constitute a distinct subset in diverse mucosal and lymphoid tissues. We identify a core transcriptional profile within the CD69+ subset of memory CD4+ and CD8+ T cells in lung and spleen that is distinct from that of CD69- TEM cells in tissues and circulation and defines human TRMs based on homology to the transcriptional profile of mouse CD8+ TRMs. Human TRMs in diverse sites exhibit increased expression of adhesion and inhibitory molecules, produce both pro-inflammatory and regulatory cytokines, and have reduced turnover compared with circulating TEM, suggesting unique adaptations for in situ immunity. Together, our results provide a unifying signature for human TRM and a blueprint for designing tissue-targeted immunotherapies.


Gene Expression Profiling , Immunologic Memory , Lymphoid Tissue/immunology , Mucous Membrane/immunology , T-Lymphocytes/immunology , Transcription, Genetic , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Lineage/genetics , Clone Cells , Humans , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Mice , Phenotype , Transcriptome/genetics
6.
Nat Biotechnol ; 35(9): 879-884, 2017 Sep.
Article En | MEDLINE | ID: mdl-28829438

B-cell responses result in clonal expansion, and can occur in a variety of tissues. To define how B-cell clones are distributed in the body, we sequenced 933,427 B-cell clonal lineages and mapped them to eight different anatomic compartments in six human organ donors. We show that large B-cell clones partition into two broad networks-one spans the blood, bone marrow, spleen and lung, while the other is restricted to tissues within the gastrointestinal (GI) tract (jejunum, ileum and colon). Notably, GI tract clones display extensive sharing of sequence variants among different portions of the tract and have higher frequencies of somatic hypermutation, suggesting extensive and serial rounds of clonal expansion and selection. Our findings provide an anatomic atlas of B-cell clonal lineages, their properties and tissue connections. This resource serves as a foundation for studies of tissue-based immunity, including vaccine responses, infections, autoimmunity and cancer.


B-Lymphocytes/cytology , Cell Lineage/genetics , Organ Specificity/genetics , Adult , Clone Cells , Female , Humans , Male , Middle Aged , Young Adult
7.
Immunity ; 46(3): 504-515, 2017 03 21.
Article En | MEDLINE | ID: mdl-28329707

Maturation and migration to lymph nodes (LNs) constitutes a central paradigm in conventional dendritic cell (cDC) biology but remains poorly defined in humans. Using our organ donor tissue resource, we analyzed cDC subset distribution, maturation, and migration in mucosal tissues (lungs, intestines), associated lymph nodes (LNs), and other lymphoid sites from 78 individuals ranging from less than 1 year to 93 years of age. The distribution of cDC1 (CD141hiCD13hi) and cDC2 (Sirp-α+CD1c+) subsets was a function of tissue site and was conserved between donors. We identified cDC2 as the major mature (HLA-DRhi) subset in LNs with the highest frequency in lung-draining LNs. Mature cDC2 in mucosal-draining LNs expressed tissue-specific markers derived from the paired mucosal site, reflecting their tissue-migratory origin. These distribution and maturation patterns were largely maintained throughout life, with site-specific variations. Our findings provide evidence for localized DC tissue surveillance and reveal a lifelong division of labor between DC subsets, with cDC2 functioning as guardians of the mucosa.


Dendritic Cells/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Infant , Male , Middle Aged , Young Adult
8.
J Exp Med ; 214(3): 651-667, 2017 03 06.
Article En | MEDLINE | ID: mdl-28130404

T cell responses to viruses are initiated and maintained in tissue sites; however, knowledge of human antiviral T cells is largely derived from blood. Cytomegalovirus (CMV) persists in most humans, requires T cell immunity to control, yet tissue immune responses remain undefined. Here, we investigated human CMV-specific T cells, virus persistence and CMV-associated T cell homeostasis in blood, lymphoid, mucosal and secretory tissues of 44 CMV seropositive and 28 seronegative donors. CMV-specific T cells were maintained in distinct distribution patterns, highest in blood, bone marrow (BM), or lymph nodes (LN), with the frequency and function in blood distinct from tissues. CMV genomes were detected predominantly in lung and also in spleen, BM, blood and LN. High frequencies of activated CMV-specific T cells were found in blood and BM samples with low virus detection, whereas in lung, CMV-specific T cells were present along with detectable virus. In LNs, CMV-specific T cells exhibited quiescent phenotypes independent of virus. Overall, T cell differentiation was enhanced in sites of viral persistence with age. Together, our results suggest tissue T cell reservoirs for CMV control shaped by both viral and tissue-intrinsic factors, with global effects on homeostasis of tissue T cells over the lifespan.


Cytomegalovirus Infections/immunology , T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Humans , Interferon-gamma/biosynthesis , Lymph Nodes/immunology , Viremia/immunology
9.
Nat Med ; 22(1): 72-7, 2016 Jan.
Article En | MEDLINE | ID: mdl-26657141

It is unclear how the immune response in early life becomes appropriately stimulated to provide protection while also avoiding excessive activation as a result of diverse new antigens. T cells are integral to adaptive immunity; mouse studies indicate that tissue localization of T cell subsets is important for both protective immunity and immunoregulation. In humans, however, the early development and function of T cells in tissues remain unexplored. We present here an analysis of lymphoid and mucosal tissue T cells derived from pediatric organ donors in the first two years of life, as compared to adult organ donors, revealing early compartmentalization of T cell differentiation and regulation. Whereas adult tissues contain a predominance of memory T cells, in pediatric blood and tissues the main subset consists of naive recent thymic emigrants, with effector memory T cells (T(EM)) found only in the lungs and small intestine. Additionally, regulatory T (T(reg)) cells comprise a high proportion (30-40%) of CD4(+) T cells in pediatric tissues but are present at much lower frequencies (1-10%) in adult tissues. Pediatric tissue T(reg) cells suppress endogenous T cell activation, and early T cell functionality is confined to the mucosal sites that have the lowest T(reg):T(EM) cell ratios, which suggests control in situ of immune responses in early life.


Cell Differentiation/immunology , Cytokines/immunology , Lymphoid Tissue/immunology , Mucous Membrane/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Female , Flow Cytometry , Humans , Infant , Intestinal Mucosa/immunology , Intestines/immunology , Lung/immunology , Lymph Nodes/immunology , Male , Middle Aged , Respiratory Mucosa/immunology , Spleen/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Young Adult
10.
Mol Ther ; 22(1): 112-22, 2014 Jan.
Article En | MEDLINE | ID: mdl-24025748

Tumors are theoretically capable of eliciting an antitumor immune response, but are often poorly immunogenic. Oncolytic viruses (OVs) have recently emerged as a promising strategy for the immunogenic delivery of tumor-associated antigens (TAAs) to cancer patients. However, safe and effective OV/TAA therapies have not yet been established. We have previously demonstrated that vectors based on Sindbis virus (SV) can inhibit tumor growth and activate the innate immune system in mice. Here, we demonstrate that SV vectors carrying a TAA generate a dramatically enhanced therapeutic effect in mice bearing subcutaneous, intraperitoneal, and lung cancers. Notably, SV/TAA efficacy was not dependent on tumor cell targeting, but was characterized by the transient expression of TAAs in lymph nodes draining the injection site. Early T-cell activation at this site was followed by a robust influx of NKG2D expressing antigen-specific cytotoxic CD8+ T cells into the tumor site, subsequently leading to the generation of long-lasting memory T cells which conferred protection against rechallenge with TAA-positive as well as TAA-negative tumor cells. By combining in vivo imaging, flow cytometry, cytotoxicity/cytokine assays, and tetramer analysis, we investigated the relationship between these events and propose a model for CD8+ T-cell activation during SV/TAA therapy.


Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/genetics , Lymph Nodes/immunology , Neoplasms/genetics , Neoplasms/immunology , Sindbis Virus/genetics , Animals , Cricetinae , Cytotoxicity, Immunologic , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Immunologic Memory , Lymph Nodes/metabolism , Lymphocyte Activation , Mice , Neoplasms/pathology , Neoplasms/therapy , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Sindbis Virus/immunology , Tumor Burden/genetics , Tumor Burden/immunology
11.
PLoS One ; 6(6): e20598, 2011.
Article En | MEDLINE | ID: mdl-21674047

Oncolytic viruses (OVs) represent a relatively novel anti-cancer modality. Like other new cancer treatments, effective OV therapy will likely require combination with conventional treatments. In order to design combinatorial treatments that work well together, a greater scrutiny of the mechanisms behind the individual treatments is needed. Sindbis virus (SV) based vectors have previously been shown to target and kill tumors in xenograft, syngeneic, and spontaneous mouse models. However, the effect of SV treatment on the immune system has not yet been studied. Here we used a variety of methods, including FACS analysis, cytotoxicity assays, cell depletion, imaging of tumor growth, cytokine blockade, and survival experiments, to study how SV therapy affects Natural Killer (NK) cell function in SCID mice bearing human ovarian carcinoma tumors. Surprisingly, we found that SV anti-cancer efficacy is largely NK cell-dependent. Furthermore, the enhanced therapeutic effect previously observed from Sin/IL12 vectors, which carry the gene for interleukin 12, is also NK cell dependent, but works through a separate IFNγ-dependent mechanism, which also induces the activation of peritoneal macrophages. These results demonstrate the multimodular nature of SV therapy, and open up new possibilities for potential synergistic or additive combinatorial therapies with other treatments.


Cytotoxicity, Immunologic/immunology , Genetic Vectors/genetics , Killer Cells, Natural/immunology , Oncolytic Virotherapy , Sindbis Virus/genetics , Sindbis Virus/immunology , Animals , Cell Count , Cell Line, Tumor , Cell Transformation, Neoplastic/immunology , Cricetinae , Cytotoxicity, Immunologic/genetics , Female , Genes, MHC Class II/genetics , Genetic Vectors/immunology , Humans , Interferon-gamma/metabolism , Interleukin-12/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Lac Operon/genetics , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Peritoneum/immunology , Peritoneum/metabolism , Sindbis Virus/physiology , Up-Regulation/immunology
...