Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
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.
Gan To Kagaku Ryoho ; 47(4): 712-714, 2020 Apr.
Article Ja | MEDLINE | ID: mdl-32389993

A 60-year-old woman was referred to our hospital due to pancreatic head cancer with right ureter invasion. We considered that it was difficult to achieve R0 resection for the patient by operation because of a wide range of retroperitoneal invasions involving the right ureter. She was treated with chemotherapy(gemcitabine plus nab-paclitaxel: GnP). GnP therapy was administered 3-weeks on/1-week off for 1 course. After 3 courses, we performed pancreaticoduodenectomy, right nephrectomy and partial transverse colectomy. We achieved R0 resection and considered the GnP therapy to be effective.


Pancreatic Neoplasms , Ureter , Antineoplastic Combined Chemotherapy Protocols , Deoxycytidine , Female , Humans , Middle Aged , Neoadjuvant Therapy , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/surgery
3.
Cell ; 180(4): 749-763.e13, 2020 02 20.
Article En | MEDLINE | ID: mdl-32059780

Immune responses in diverse tissue sites are critical for protective immunity and homeostasis. Here, we investigate how tissue localization regulates the development and function of human natural killer (NK) cells, innate lymphocytes important for anti-viral and tumor immunity. Integrating high-dimensional analysis of NK cells from blood, lymphoid organs, and mucosal tissue sites from 60 individuals, we identify tissue-specific patterns of NK cell subset distribution, maturation, and function maintained across age and between individuals. Mature and terminally differentiated NK cells with enhanced effector function predominate in blood, bone marrow, spleen, and lungs and exhibit shared transcriptional programs across sites. By contrast, precursor and immature NK cells with reduced effector capacity populate lymph nodes and intestines and exhibit tissue-resident signatures and site-specific adaptations. Together, our results reveal anatomic control of NK cell development and maintenance as tissue-resident populations, whereas mature, terminally differentiated subsets mediate immunosurveillance through diverse peripheral sites. VIDEO ABSTRACT.


Aging/immunology , Killer Cells, Natural/cytology , Lymphopoiesis , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Child , Female , Humans , Immunity, Innate , Intestinal Mucosa/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/physiology , Lung/cytology , Lymph Nodes/cytology , Male , Middle Aged , Spleen/cytology
4.
Nat Commun ; 10(1): 4706, 2019 10 17.
Article En | MEDLINE | ID: mdl-31624246

Human T cells coordinate adaptive immunity in diverse anatomic compartments through production of cytokines and effector molecules, but it is unclear how tissue site influences T cell persistence and function. Here, we use single cell RNA-sequencing (scRNA-seq) to define the heterogeneity of human T cells isolated from lungs, lymph nodes, bone marrow and blood, and their functional responses following stimulation. Through analysis of >50,000 resting and activated T cells, we reveal tissue T cell signatures in mucosal and lymphoid sites, and lineage-specific activation states across all sites including distinct effector states for CD8+ T cells and an interferon-response state for CD4+ T cells. Comparing scRNA-seq profiles of tumor-associated T cells to our dataset reveals predominant activated CD8+ compared to CD4+ T cell states within multiple tumor types. Our results therefore establish a high dimensional reference map of human T cell activation in health for analyzing T cells in disease.


Lung/metabolism , Lymph Nodes/metabolism , Neoplasms/genetics , Single-Cell Analysis/methods , T-Lymphocytes/metabolism , Transcriptome/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Lymph Nodes/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Neoplasms/pathology , T-Lymphocytes/immunology
5.
Sci Immunol ; 4(33)2019 03 08.
Article En | MEDLINE | ID: mdl-30850393

Tissue-resident memory T cells (TRM) maintain immunity in diverse sites as determined in mouse models, whereas their establishment and role in human tissues have been difficult to assess. Here, we investigated human lung TRM generation, maintenance, and function in airway samples obtained longitudinally from human leukocyte antigen (HLA)-disparate lung transplant recipients, where donor and recipient T cells could be localized and tracked over time. Donor T cells persist specifically in the lungs (and not blood) of transplant recipients and express high levels of TRM signature markers including CD69, CD103, and CD49a, whereas lung-infiltrating recipient T cells gradually acquire TRM phenotypes over months in vivo. Single-cell transcriptome profiling of airway T cells reveals that donor T cells comprise two TRM-like subsets with varying levels of expression of TRM-associated genes, whereas recipient T cells comprised non-TRM and similar TRM-like subpopulations, suggesting de novo TRM generation. Transplant recipients exhibiting higher frequencies of persisting donor TRM experienced fewer adverse clinical events such as primary graft dysfunction and acute cellular rejection compared with recipients with low donor TRM persistence, suggesting that monitoring TRM dynamics could be clinically informative. Together, our results provide spatial and temporal insights into how human TRM develop, function, persist, and affect tissue integrity within the complexities of lung transplantation.


Immunologic Memory , Lung Transplantation , Lung/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Female , Graft Rejection , Humans , Male , Middle Aged , Transcriptome
6.
Gan To Kagaku Ryoho ; 46(13): 2518-2520, 2019 Dec.
Article Ja | MEDLINE | ID: mdl-32156984

INTRODUCTION: Primary small-cell carcinomas occur commonly in the lungs but rarely in the other organs. We studied the treatment outcomes in 6 cases of primary small-cell carcinoma of the digestive tract at our hospital. PATIENTS: Six patients were diagnosed with small-cell carcinoma of the digestive tract histopathologically and treated at our hospital from September 2000 to December 2018. RESULTS: The average age of the patients was 61.5 years(range: 40-80 years). Patients were 3 men and 3 women. The occurrence sites were the esophagus, stomach, and colon in 1, 2, and 3 patients, respectively. The patient with esophageal cancer underwent chemoradiotherapy without surgery. Other patients, except for 1 patient with colon cancer, underwent adjuvant chemotherapy after the surgery. Two of the 6 patients survived for over 5 years. DISCUSSION: For small-cell carcinomas of the digestive tract with poor prognosis, long-term survival can be expected using multidisciplinary treatments depending on the case.


Carcinoma, Small Cell , Adult , Aged , Aged, 80 and over , Colonic Neoplasms , Esophageal Neoplasms , Female , Humans , Male , Middle Aged , Retrospective Studies , Stomach Neoplasms
7.
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
8.
Cell Stem Cell ; 24(2): 227-239.e8, 2019 02 07.
Article En | MEDLINE | ID: mdl-30503142

Human intestinal transplantation often results in long-term mixed chimerism of donor and recipient blood in transplant patients. We followed the phenotypes of chimeric peripheral blood cells in 21 patients receiving intestinal allografts over 5 years. Donor lymphocyte phenotypes suggested a contribution of hematopoietic stem and progenitor cells (HSPCs) from the graft. Surprisingly, we detected donor-derived HSPCs in intestinal mucosa, Peyer's patches, mesenteric lymph nodes, and liver. Human gut HSPCs are phenotypically similar to bone marrow HSPCs and have multilineage differentiation potential in vitro and in vivo. Analysis of circulating post-transplant donor T cells suggests that they undergo selection in recipient lymphoid organs to acquire immune tolerance. Our longitudinal study of human HSPCs carried in intestinal allografts demonstrates their turnover kinetics and gradual replacement of donor-derived HSPCs from a circulating pool. Thus, we have demonstrated the existence of functioning HSPCs in human intestines with implications for promoting tolerance in transplant recipients.


Cell Movement , Hematopoietic Stem Cells/cytology , Intestines/cytology , Intestines/transplantation , Animals , Cell Differentiation , Cell Line , Cell Lineage , Chimerism , Graft vs Host Disease/immunology , Humans , Immune Tolerance , Intestinal Mucosa/cytology , Liver/cytology , Lymph Nodes/cytology , Mice , Peyer's Patches/cytology , Phenotype , T-Lymphocytes/cytology , Tissue Donors , Transplantation, Homologous
9.
JCI Insight ; 3(22)2018 11 15.
Article En | MEDLINE | ID: mdl-30429372

Tissue-resident memory T cells (TRMs) accelerate pathogen clearance through rapid and enhanced functional responses in situ. TRMs are prevalent in diverse anatomic sites throughout the human lifespan, yet their phenotypic and functional diversity has not been fully described. Here, we identify subpopulations of human TRMs based on the ability to efflux fluorescent dyes [efflux(+) TRMs] located within mucosal and lymphoid sites with distinct transcriptional profiles, turnover, and functional capacities. Compared with efflux(-) TRMs, efflux(+) TRMs showed transcriptional and phenotypic features of quiescence including reduced turnover, decreased expression of exhaustion markers, and increased proliferative capacity and signaling in response to homeostatic cytokines. Moreover, upon activation, efflux(+) TRMs secreted lower levels of inflammatory cytokines such as IFN-γ and IL-2 and underwent reduced degranulation. Interestingly, analysis of TRM subsets following activation revealed that both efflux(+) and efflux(-) TRMs undergo extensive transcriptional changes following TCR ligation but retain core TRM transcriptional properties including retention markers, suggesting that TRMs carry out effector function in situ. Overall, our results suggest a model for tissue-resident immunity wherein heterogeneous subsets have differential capacities for longevity and effector function.


CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Fluorescent Dyes , Humans , Lymphoid Tissue/cytology , Mitochondria/metabolism , Models, Immunological , Phenotype , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tissue Distribution , Transcriptome
10.
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
11.
Nature ; 549(7671): 282-286, 2017 09 14.
Article En | MEDLINE | ID: mdl-28869965

The type 2 cytokines interleukin (IL)-4, IL-5, IL-9 and IL-13 have important roles in stimulating innate and adaptive immune responses that are required for resistance to helminth infection, promotion of allergic inflammation, metabolic homeostasis and tissue repair. Group 2 innate lymphoid cells (ILC2s) produce type 2 cytokines, and although advances have been made in understanding the cytokine milieu that promotes ILC2 responses, how ILC2 responses are regulated by other stimuli remains poorly understood. Here we demonstrate that ILC2s in the mouse gastrointestinal tract co-localize with cholinergic neurons that express the neuropeptide neuromedin U (NMU). In contrast to other haematopoietic cells, ILC2s selectively express the NMU receptor 1 (NMUR1). In vitro stimulation of ILC2s with NMU induced rapid cell activation, proliferation, and secretion of the type 2 cytokines IL-5, IL-9 and IL-13 that was dependent on cell-intrinsic expression of NMUR1 and Gαq protein. In vivo administration of NMU triggered potent type 2 cytokine responses characterized by ILC2 activation, proliferation and eosinophil recruitment that was associated with accelerated expulsion of the gastrointestinal nematode Nippostrongylus brasiliensis or induction of lung inflammation. Conversely, worm burden was higher in Nmur1-/- mice than in control mice. Furthermore, use of gene-deficient mice and adoptive cell transfer experiments revealed that ILC2s were necessary and sufficient to mount NMU-elicited type 2 cytokine responses. Together, these data indicate that the NMU-NMUR1 neuronal signalling circuit provides a selective mechanism through which the enteric nervous system and innate immune system integrate to promote rapid type 2 cytokine responses that can induce anti-microbial, inflammatory and tissue-protective type 2 responses at mucosal sites.


Cytokines/immunology , Immunity, Innate , Inflammation/immunology , Lymphocytes/immunology , Neuropeptides/metabolism , Adoptive Transfer , Animals , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Cytokines/metabolism , Eosinophils/cytology , Eosinophils/drug effects , Eosinophils/immunology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gastrointestinal Tract/cytology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/innervation , Immunity, Innate/drug effects , Inflammation/chemically induced , Inflammation/pathology , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-5/immunology , Interleukin-5/metabolism , Interleukin-9/immunology , Interleukin-9/metabolism , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Mice , Neuropeptides/pharmacology , Nippostrongylus/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Pneumonia/pathology , Receptors, Neurotransmitter/deficiency , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Signal Transduction/drug effects
12.
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
13.
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
...