Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nat Immunol ; 24(2): 309-319, 2023 02.
Article in English | MEDLINE | ID: mdl-36658238

ABSTRACT

T lymphocytes migrate to barrier sites after exposure to pathogens, providing localized immunity and long-term protection. Here, we obtained blood and tissues from human organ donors to examine T cells across major barrier sites (skin, lung, jejunum), associated lymph nodes, lymphoid organs (spleen, bone marrow), and in circulation. By integrating single-cell protein and transcriptome profiling, we demonstrate that human barrier sites contain tissue-resident memory T (TRM) cells that exhibit site-adapted profiles for residency, homing and function distinct from circulating memory T cells. Incorporating T cell receptor and transcriptome analysis, we show that circulating memory T cells are highly expanded, display extensive overlap between sites and exhibit effector and cytolytic functional profiles, while TRM clones exhibit site-specific expansions and distinct functional capacities. Together, our findings indicate that circulating T cells are more disseminated and differentiated, while TRM cells exhibit tissue-specific adaptation and clonal segregation, suggesting that strategies to promote barrier immunity require tissue targeting.


Subject(s)
Immunologic Memory , Memory T Cells , Humans , Lymph Nodes , Clone Cells , Cell Differentiation , CD8-Positive T-Lymphocytes
2.
Cell ; 180(4): 749-763.e13, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32059780

ABSTRACT

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.


Subject(s)
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
3.
Nat Immunol ; 22(1): 25-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33154590

ABSTRACT

Clinical manifestations of COVID-19 caused by the new coronavirus SARS-CoV-2 are associated with age1,2. Adults develop respiratory symptoms, which can progress to acute respiratory distress syndrome (ARDS) in the most severe form, while children are largely spared from respiratory illness but can develop a life-threatening multisystem inflammatory syndrome (MIS-C)3-5. Here, we show distinct antibody responses in children and adults after SARS-CoV-2 infection. Adult COVID-19 cohorts had anti-spike (S) IgG, IgM and IgA antibodies, as well as anti-nucleocapsid (N) IgG antibody, while children with and without MIS-C had reduced breadth of anti-SARS-CoV-2-specific antibodies, predominantly generating IgG antibodies specific for the S protein but not the N protein. Moreover, children with and without MIS-C had reduced neutralizing activity as compared to both adult COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children independent of whether they develop MIS-C, with implications for developing age-targeted strategies for testing and protecting the population.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation/immunology , COVID-19/immunology , Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adolescent , Adult , Aged , COVID-19/virology , Child , Child, Preschool , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Middle Aged , SARS-CoV-2/physiology , Young Adult
4.
Immunity ; 56(8): 1894-1909.e5, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37421943

ABSTRACT

Infancy and childhood are critical life stages for generating immune memory to protect against pathogens; however, the timing, location, and pathways for memory development in humans remain elusive. Here, we investigated T cells in mucosal sites, lymphoid tissues, and blood from 96 pediatric donors aged 0-10 years using phenotypic, functional, and transcriptomic profiling. Our results revealed that memory T cells preferentially localized in the intestines and lungs during infancy and accumulated more rapidly in mucosal sites compared with blood and lymphoid organs, consistent with site-specific antigen exposure. Early life mucosal memory T cells exhibit distinct functional capacities and stem-like transcriptional profiles. In later childhood, they progressively adopt proinflammatory functions and tissue-resident signatures, coincident with increased T cell receptor (TCR) clonal expansion in mucosal and lymphoid sites. Together, our findings identify staged development of memory T cells targeted to tissues during the formative years, informing how we might promote and monitor immunity in children.


Subject(s)
Lymphoid Tissue , Memory T Cells , Child , Humans , Infant , CD8-Positive T-Lymphocytes , Immunologic Memory , Lymphoid Tissue/metabolism , Mucous Membrane , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Infant, Newborn , Child, Preschool
5.
Immunity ; 54(4): 797-814.e6, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33765436

ABSTRACT

Immune response dynamics in coronavirus disease 2019 (COVID-19) and their severe manifestations have largely been studied in circulation. Here, we examined the relationship between immune processes in the respiratory tract and circulation through longitudinal phenotypic, transcriptomic, and cytokine profiling of paired airway and blood samples from patients with severe COVID-19 relative to heathy controls. In COVID-19 airways, T cells exhibited activated, tissue-resident, and protective profiles; higher T cell frequencies correlated with survival and younger age. Myeloid cells in COVID-19 airways featured hyperinflammatory signatures, and higher frequencies of these cells correlated with mortality and older age. In COVID-19 blood, aberrant CD163+ monocytes predominated over conventional monocytes, and were found in corresponding airway samples and in damaged alveoli. High levels of myeloid chemoattractants in airways suggest recruitment of these cells through a CCL2-CCR2 chemokine axis. Our findings provide insights into immune processes driving COVID-19 lung pathology with therapeutic implications for targeting inflammation in the respiratory tract.


Subject(s)
COVID-19/immunology , Lung/immunology , Myeloid Cells/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , COVID-19/blood , COVID-19/mortality , COVID-19/pathology , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation , Longitudinal Studies , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Middle Aged , Monocytes/immunology , Monocytes/pathology , Myeloid Cells/pathology , SARS-CoV-2 , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcriptome , Young Adult
6.
Immunol Rev ; 316(1): 23-37, 2023 07.
Article in English | MEDLINE | ID: mdl-37211646

ABSTRACT

Tissue-resident memory T cells (TRM) represent a dedicated layer of localized immune memory in virtually every organ throughout the human body. By virtue of their long-term residence in disparate tissues, TRM are shaped by a myriad of site-specific influences and exhibit remarkable heterogeneity in form and function. Here, we review the major axes by which TRM vary, including their surface phenotypes, transcriptional programming, and the tissue-specific adaptations that accrue over their tenancy. We discuss how localization within distinct anatomic niches both within and across major organ systems shapes TRM identity and examine mechanisms and prevailing models for TRM generation. Understanding the drivers of differentiation, function and maintenance of the various subpopulations that together define the TRM lineage may hold the key to unlocking the full potential of TRM to promote localized and protective tissue immunity throughout the body.


Subject(s)
Immunologic Memory , Memory T Cells , Humans , Phenotype , Cell Differentiation , CD8-Positive T-Lymphocytes
7.
PLoS Biol ; 15(6): e2001930, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28632753

ABSTRACT

Superantigens (SAgs) are potent exotoxins secreted by Staphylococcus aureus and Streptococcus pyogenes. They target a large fraction of T cell pools to set in motion a "cytokine storm" with severe and sometimes life-threatening consequences typically encountered in toxic shock syndrome (TSS). Given the rapidity with which TSS develops, designing timely and truly targeted therapies for this syndrome requires identification of key mediators of the cytokine storm's initial wave. Equally important, early host responses to SAgs can be accompanied or followed by a state of immunosuppression, which in turn jeopardizes the host's ability to combat and clear infections. Unlike in mouse models, the mechanisms underlying SAg-associated immunosuppression in humans are ill-defined. In this work, we have identified a population of innate-like T cells, called mucosa-associated invariant T (MAIT) cells, as the most powerful source of pro-inflammatory cytokines after exposure to SAgs. We have utilized primary human peripheral blood and hepatic mononuclear cells, mouse MAIT hybridoma lines, HLA-DR4-transgenic mice, MAIThighHLA-DR4+ bone marrow chimeras, and humanized NOD-scid IL-2Rγnull mice to demonstrate for the first time that: i) mouse and human MAIT cells are hyperresponsive to SAgs, typified by staphylococcal enterotoxin B (SEB); ii) the human MAIT cell response to SEB is rapid and far greater in magnitude than that launched by unfractionated conventional T, invariant natural killer T (iNKT) or γδ T cells, and is characterized by production of interferon (IFN)-γ, tumor necrosis factor (TNF)-α and interleukin (IL)-2, but not IL-17A; iii) high-affinity MHC class II interaction with SAgs, but not MHC-related protein 1 (MR1) participation, is required for MAIT cell activation; iv) MAIT cell responses to SEB can occur in a T cell receptor (TCR) Vß-specific manner but are largely contributed by IL-12 and IL-18; v) as MAIT cells are primed by SAgs, they also begin to develop a molecular signature consistent with exhaustion and failure to participate in antimicrobial defense. Accordingly, they upregulate lymphocyte-activation gene 3 (LAG-3), T cell immunoglobulin and mucin-3 (TIM-3), and/or programmed cell death-1 (PD-1), and acquire an anergic phenotype that interferes with their cognate function against Klebsiella pneumoniae and Escherichia coli; vi) MAIT cell hyperactivation and anergy co-utilize a signaling pathway that is governed by p38 and MEK1/2. Collectively, our findings demonstrate a pathogenic, rather than protective, role for MAIT cells during infection. Furthermore, we propose a novel mechanism of SAg-associated immunosuppression in humans. MAIT cells may therefore provide an attractive therapeutic target for the management of both early and late phases of severe SAg-mediated illnesses.


Subject(s)
Antigens, Bacterial/toxicity , Clonal Anergy , Models, Immunological , Mucosal-Associated Invariant T Cells/immunology , Staphylococcus aureus/immunology , Streptococcus pyogenes/immunology , Superantigens/toxicity , Animals , Antigens, Bacterial/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Clonal Anergy/drug effects , Crosses, Genetic , Enterotoxins/metabolism , Enterotoxins/toxicity , Female , Humans , Hybridomas , Immunity, Innate , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Mucosal-Associated Invariant T Cells/cytology , Mucosal-Associated Invariant T Cells/drug effects , Mucosal-Associated Invariant T Cells/metabolism , Specific Pathogen-Free Organisms , Staphylococcus aureus/metabolism , Streptococcus pyogenes/metabolism , Superantigens/metabolism , Transplantation Chimera/blood , Transplantation Chimera/immunology , Transplantation Chimera/metabolism
8.
J Immunol ; 198(7): 2805-2818, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28219889

ABSTRACT

Toxic shock syndrome (TSS) is caused by staphylococcal and streptococcal superantigens (SAgs) that provoke a swift hyperinflammatory response typified by a cytokine storm. The precipitous decline in the host's clinical status and the lack of targeted therapies for TSS emphasize the need to identify key players of the storm's initial wave. Using a humanized mouse model of TSS and human cells, we herein demonstrate that SAgs elicit in vitro and in vivo IL-17A responses within hours. SAg-triggered human IL-17A production was characterized by remarkably high mRNA stability for this cytokine. A distinct subpopulation of CD4+ effector memory T (TEM) cells that secrete IL-17A, but not IFN-γ, was responsible for early IL-17A production. We found mouse "TEM-17" cells to be enriched within the intestinal epithelium and among lamina propria lymphocytes. Furthermore, interfering with IL-17A receptor signaling in human PBMCs attenuated the expression of numerous inflammatory mediators implicated in the TSS-associated cytokine storm. IL-17A receptor blockade also abrogated the secondary effect of SAg-stimulated PBMCs on human dermal fibroblasts as judged by C/EBP δ expression. Finally, the early IL-17A response to SAgs was pathogenic because in vivo neutralization of IL-17A in humanized mice ameliorated hepatic and intestinal damage and reduced mortality. Together, our findings identify CD4+ TEM cells as a key effector of TSS and reveal a novel role for IL-17A in TSS immunopathogenesis. Our work thus elucidates a pathogenic, as opposed to protective, role for IL-17A during Gram-positive bacterial infections. Accordingly, the IL-17-IL-17R axis may provide an attractive target for the management of SAg-mediated illnesses.


Subject(s)
Interleukin-17/immunology , Shock, Septic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Flow Cytometry , Gram-Positive Bacterial Infections/immunology , Humans , Immunologic Memory/immunology , Interleukin-17/biosynthesis , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Shock, Septic/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism
9.
J Infect Dis ; 215(5): 824-829, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28035011

ABSTRACT

During toxic shock syndrome (TSS), bacterial superantigens trigger a polyclonal T -cell response leading to a potentially catastrophic "cytokine storm". Whether innate-like invariant natural killer T (iNKT) cells, with remarkable immunomodulatory properties, participate in TSS is unclear. Using genetic and cell depletion approaches, we generated iNKT cell-deficient, superantigen-sensitive HLA-DR4-transgenic (DR4tg) mice, which were compared with their iNKT-sufficient counterparts for responsiveness to staphylococcal enterotoxin B (SEB). Both approaches indicate that iNKT cells are pathogenic in TSS. Importantly, treating DR4tg mice with a TH2-polarizing glycolipid agonist of iNKT cells reduced SEB-inflicted morbidity/mortality. Therefore, iNKT cells may constitute an attractive therapeutic target in superantigen-mediated illnesses.


Subject(s)
HLA-DR4 Antigen/genetics , Natural Killer T-Cells/immunology , Shock, Septic/immunology , Shock, Septic/prevention & control , Animals , Disease Models, Animal , Enterotoxins/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superantigens/blood , Superantigens/immunology
10.
J Infect Dis ; 213(12): 1990-5, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26908735

ABSTRACT

Toxic shock syndrome (TSS) and other superantigen-mediated illnesses are associated with 'systemic' immunosuppression that jeopardizes the host's ability to fight pathogens. Here, we define a novel mechanism of 'local' immunosuppression that may benefit the host. Systemic exposure to staphylococcal enterotoxin B (SEB) rapidly and selectively recruited CD11b(+)Gr-1(high)Ly-6C(+) granulocytic myeloid-derived suppressor cells (MDSCs) to the liver of HLA-DR4 transgenic mice. Hepatic MDSCs inhibited SEB-triggered T cell proliferation in a reactive oxygen species-dependent manner, and ex vivo-generated human MDSCs also similarly attenuated the proliferative response of autologous T cells to SEB. We propose a role for MDSCs in mitigating excessive tissue injury during TSS.


Subject(s)
Enterotoxins/immunology , HLA-DR4 Antigen/genetics , Myeloid-Derived Suppressor Cells/immunology , Shock, Septic/immunology , Superantigens/immunology , Animals , CD11b Antigen/immunology , Cell Proliferation , Disease Models, Animal , Female , Granulocytes/immunology , HLA-DR4 Antigen/immunology , Humans , Immunosuppression Therapy , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reactive Oxygen Species/immunology
11.
Infect Immun ; 82(9): 3588-98, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24914221

ABSTRACT

Staphylococcus aureus is a versatile bacterial pathogen that produces T cell-activating toxins known as superantigens (SAgs). Although excessive immune activation by SAgs can induce a dysregulated cytokine storm as a component of what is known as toxic shock syndrome (TSS), the contribution of SAgs to the staphylococcal infection process is not well defined. Here, we evaluated the role of the bacterial superantigen staphylococcal enterotoxin A (SEA) in a bacteremia model using humanized transgenic mice expressing SAg-responsive HLA-DR4 molecules. Infection with S. aureus Newman induced SEA-dependent Vß skewing of T cells and enhanced bacterial survival in the liver compared with infection by sea knockout strain. SEA-induced gamma interferon, interleukin-12, and chemokine responses resulted in increased infiltration of CD11b(+) Ly6G(+) neutrophils into the liver, promoting the formation of abscesses that contained large numbers of viable staphylococci. Hepatic abscesses occurred significantly more frequently in S. aureus Newman-infected livers than in livers infected with the Newman sea knockout strain, promoting the survival of S. aureus in vivo. This represents a novel mechanism during infection whereby S. aureus utilizes SAgs to form a specialized niche and manipulate the immune system.


Subject(s)
Abscess/immunology , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Superantigens/immunology , Abscess/microbiology , Animals , Antigens, Ly/immunology , CD11b Antigen/immunology , Enterotoxins/immunology , HLA-DR4 Antigen/immunology , Interferon-gamma/immunology , Interleukin-12/immunology , Liver/immunology , Liver/microbiology , Lymphocyte Activation/immunology , Male , Mice , Mice, Transgenic , Neutrophils/microbiology , Staphylococcal Infections/microbiology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology
12.
bioRxiv ; 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-37461466

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is invaluable for profiling cellular heterogeneity and dissecting transcriptional states, but transcriptomic profiles do not always delineate subsets defined by surface proteins, as in cells of the immune system. Cellular Indexing of Transcriptomes and Epitopes (CITE-seq) enables simultaneous profiling of single-cell transcriptomes and surface proteomes; however, accurate cell type annotation requires a classifier that integrates multimodal data. Here, we describe MultiModal Classifier Hierarchy (MMoCHi), a marker-based approach for classification, reconciling gene and protein expression without reliance on reference atlases. We benchmark MMoCHi using sorted T lymphocyte subsets and annotate a cross-tissue human immune cell dataset. MMoCHi outperforms leading transcriptome-based classifiers and multimodal unsupervised clustering in its ability to identify immune cell subsets that are not readily resolved and to reveal novel subset markers. MMoCHi is designed for adaptability and can integrate annotation of cell types and developmental states across diverse lineages, samples, or modalities.

13.
bioRxiv ; 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38260588

ABSTRACT

The immune system comprises multiple cell lineages and heterogeneous subsets found in blood and tissues throughout the body. While human immune responses differ between sites and over age, the underlying sources of variation remain unclear as most studies are limited to peripheral blood. Here, we took a systems approach to comprehensively profile RNA and surface protein expression of over 1.25 million immune cells isolated from blood, lymphoid organs, and mucosal tissues of 24 organ donors aged 20-75 years. We applied a multimodal classifier to annotate the major immune cell lineages (T cells, B cells, innate lymphoid cells, and myeloid cells) and their corresponding subsets across the body, leveraging probabilistic modeling to define bases for immune variations across donors, tissue, and age. We identified dominant tissue-specific effects on immune cell composition and function across lineages for lymphoid sites, intestines, and blood-rich tissues. Age-associated effects were intrinsic to both lineage and site as manifested by macrophages in mucosal sites, B cells in lymphoid organs, and T and NK cells in blood-rich sites. Our results reveal tissue-specific signatures of immune homeostasis throughout the body and across different ages. This information provides a basis for defining the transcriptional underpinnings of immune variation and potential associations with disease-associated immune pathologies across the human lifespan.

14.
Nat Med ; 28(12): 2622-2632, 2022 12.
Article in English | MEDLINE | ID: mdl-36411343

ABSTRACT

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.


Subject(s)
Lung , Lymph Nodes , Humans , Aged , Lymph Nodes/pathology , Disease Susceptibility/pathology , Dust , Immunity
15.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35446789

ABSTRACT

Respiratory failure in COVID-19 is characterized by widespread disruption of the lung's alveolar gas exchange interface. To elucidate determinants of alveolar lung damage, we performed epithelial and immune cell profiling in lungs from 24 COVID-19 autopsies and 43 uninfected organ donors ages 18-92 years. We found marked loss of type 2 alveolar epithelial (T2AE) cells and increased perialveolar lymphocyte cytotoxicity in all fatal COVID-19 cases, even at early stages before typical patterns of acute lung injury are histologically apparent. In lungs from uninfected organ donors, there was also progressive loss of T2AE cells with increasing age, which may increase susceptibility to COVID-19-mediated lung damage in older individuals. In the fatal COVID-19 cases, macrophage infiltration differed according to the histopathological pattern of lung injury. In cases with acute lung injury, we found accumulation of CD4+ macrophages that expressed distinctly high levels of T cell activation and costimulation genes and strongly correlated with increased extent of alveolar epithelial cell depletion and CD8+ T cell cytotoxicity. Together, our results show that T2AE cell deficiency may underlie age-related COVID-19 risk and initiate alveolar dysfunction shortly after infection, and we define immune cell mediators that may contribute to alveolar injury in distinct pathological stages of fatal COVID-19.


Subject(s)
Acute Lung Injury , COVID-19 , Acute Lung Injury/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Alveolar Epithelial Cells/pathology , Autopsy , Humans , Lung/pathology , Middle Aged , Young Adult
16.
Cell Rep ; 37(9): 110071, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34852222

ABSTRACT

The persistence of anti-viral immunity is essential for protection and exhibits profound heterogeneity across individuals. Here, we elucidate the factors that shape maintenance and function of anti-viral T cell immunity in the body by comprehensive profiling of virus-specific T cells across blood, lymphoid organs, and mucosal tissues of organ donors. We use flow cytometry, T cell receptor sequencing, single-cell transcriptomics, and cytokine analysis to profile virus-specific CD8+ T cells recognizing the ubiquitous pathogens influenza and cytomegalovirus. Our results reveal that virus specificity determines overall magnitude, tissue distribution, differentiation, and clonal repertoire of virus-specific T cells. Age and sex influence T cell differentiation and dissemination in tissues, while T cell tissue residence and functionality are highly correlated with the site. Together, our results demonstrate how the covariates of virus, tissue, age, and sex impact the anti-viral immune response, which is important for targeting, monitoring, and predicting immune responses to existing and emerging viruses.


Subject(s)
Antiviral Agents/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immunologic Memory/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Adult , Age Factors , Child , Child, Preschool , Cytokines/metabolism , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Female , Humans , Infant , Influenza, Human/drug therapy , Influenza, Human/virology , Lymphocyte Activation , Male , Receptors, Antigen, T-Cell/immunology , Sex Factors , Single-Cell Analysis , Transcriptome
17.
Sci Immunol ; 6(65): eabl9105, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34618554

ABSTRACT

Adaptive immune responses to SARS-CoV-2 infection have been extensively characterized in blood; however, most functions of protective immunity must be accomplished in tissues. Here, we report from examination of SARS-CoV-2 seropositive organ donors (ages 10 to 74) that CD4+ T, CD8+ T, and B cell memory generated in response to infection is present in the bone marrow, spleen, lung, and multiple lymph nodes (LNs) for up to 6 months after infection. Lungs and lung-associated LNs were the most prevalent sites for SARS-CoV-2­specific memory T and B cells with significant correlations between circulating and tissue-resident memory T and B cells in all sites. We further identified SARS-CoV-2­specific germinal centers in the lung-associated LNs up to 6 months after infection. SARS-CoV-2­specific follicular helper T cells were also abundant in lung-associated LNs and lungs. Together, the results indicate local tissue coordination of cellular and humoral immune memory against SARS-CoV-2 for site-specific protection against future infectious challenges.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Immunity, Cellular , Immunologic Memory , Lymphocytes/immunology , SARS-CoV-2/immunology , Female , Humans , Male , Organ Specificity/immunology
18.
medRxiv ; 2020 Oct 18.
Article in English | MEDLINE | ID: mdl-33106817

ABSTRACT

Immune responses to respiratory viruses like SARS-CoV-2 originate and function in the lung, yet assessments of human immunity are often limited to blood. Here, we conducted longitudinal, high-dimensional profiling of paired airway and blood samples from patients with severe COVID-19, revealing immune processes in the respiratory tract linked to disease pathogenesis. Survival from severe disease was associated with increased CD4 + T cells and decreased monocyte/macrophage frequencies in the airway, but not in blood. Airway T cells and macrophages exhibited tissue-resident phenotypes and activation signatures, including high level expression and secretion of monocyte chemoattractants CCL2 and CCL3 by airway macrophages. By contrast, monocytes in blood expressed the CCL2-receptor CCR2 and aberrant CD163 + and immature phenotypes. Extensive accumulation of CD163 + monocyte/macrophages within alveolar spaces in COVID-19 lung autopsies suggested recruitment from circulation. Our findings provide evidence that COVID-19 pathogenesis is driven by respiratory immunity, and rationale for site-specific treatment and prevention strategies.

19.
medRxiv ; 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32699861

ABSTRACT

Clinical manifestations of COVID-19 caused by the novel coronavirus SARS-CoV-2 are associated with age. While children are largely spared from severe respiratory disease, they can present with a SARS-CoV-2-associated multisystem inflammatory syndrome (MIS-C) similar to Kawasaki's disease. Here, we show distinct antibody (Ab) responses in children with MIS-C compared to adults with severe COVID-19 causing acute respiratory distress syndrome (ARDS), and those who recovered from mild disease. There was a reduced breadth and specificity of anti-SARS-CoV-2-specific antibodies in MIS-C patients compared to the COVID patient groups; MIS-C predominantly generated IgG Abs specific for the Spike (S) protein but not for the nucleocapsid (N) protein, while both COVID-19 cohorts had anti-S IgG, IgM and IgA Abs, as well as anti-N IgG Abs. Moreover, MIS-C patients had reduced neutralizing activity compared to COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children and adults who develop severe disease, with implications for optimizing treatments based on symptom and age.

20.
Sci Immunol ; 4(34)2019 04 05.
Article in English | MEDLINE | ID: mdl-30952804

ABSTRACT

The discovery of T cells resident in diverse tissues has altered our understanding of adaptive immunity to encompass site-specific responses mediated by tissue-adapted memory T cells throughout the body. Here, we discuss the key phenotypic, transcriptional, and functional features of these tissue-resident memory T cells (TRM) as established in mouse models of infection and translated to humans by novel tissue sampling approaches. Integration of findings from mouse and human studies may hold the key to unlocking the potential of TRM for promoting tissue immunity and preventing infection.


Subject(s)
Immunologic Memory , Infections/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL