Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956379

ABSTRACT

The functional diversity of natural killer (NK) cell repertoires stems from differentiation, homeostatic, receptor-ligand interactions and adaptive-like responses to viral infections. In the present study, we generated a single-cell transcriptional reference map of healthy human blood- and tissue-derived NK cells, with temporal resolution and fate-specific expression of gene-regulatory networks defining NK cell differentiation. Transfer learning facilitated incorporation of tumor-infiltrating NK cell transcriptomes (39 datasets, 7 solid tumors, 427 patients) into the reference map to analyze tumor microenvironment (TME)-induced perturbations. Of the six functionally distinct NK cell states identified, a dysfunctional stressed CD56bright state susceptible to TME-induced immunosuppression and a cytotoxic TME-resistant effector CD56dim state were commonly enriched across tumor types, the ratio of which was predictive of patient outcome in malignant melanoma and osteosarcoma. This resource may inform the design of new NK cell therapies and can be extended through transfer learning to interrogate new datasets from experimental perturbations or disease conditions.

2.
Immunity ; 54(2): 259-275.e7, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33382972

ABSTRACT

The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.


Subject(s)
Hematopoietic Stem Cells/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Monocytes/immunology , Antigens, CD34/metabolism , Biodiversity , Cell Differentiation , Cell Movement , Cells, Cultured , Fetal Blood/cytology , Humans , Lipopolysaccharide Receptors/metabolism , Lung/blood supply , Receptors, IgG/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Stem Cell Niche
3.
Immunity ; 46(2): 287-300, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28214226

ABSTRACT

Tissue-resident memory T (Trm) cells form a heterogeneous population that provides localized protection against pathogens. Here, we identify CD49a as a marker that differentiates CD8+ Trm cells on a compartmental and functional basis. In human skin epithelia, CD8+CD49a+ Trm cells produced interferon-γ, whereas CD8+CD49a- Trm cells produced interleukin-17 (IL-17). In addition, CD8+CD49a+ Trm cells from healthy skin rapidly induced the expression of the effector molecules perforin and granzyme B when stimulated with IL-15, thereby promoting a strong cytotoxic response. In skin from patients with vitiligo, where melanocytes are eradicated locally, CD8+CD49a+ Trm cells that constitutively expressed perforin and granzyme B accumulated both in the epidermis and dermis. Conversely, CD8+CD49a- Trm cells from psoriasis lesions predominantly generated IL-17 responses that promote local inflammation in this skin disease. Overall, CD49a expression delineates CD8+ Trm cell specialization in human epithelial barriers and correlates with the effector cell balance found in distinct inflammatory skin diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic/immunology , Integrin alpha1/immunology , Skin/immunology , T-Lymphocyte Subsets/immunology , Cell Separation , Flow Cytometry , Humans , Immunologic Memory/immunology , Integrin alpha1/biosynthesis , Lymphocyte Activation/immunology , Microscopy, Confocal , Psoriasis/immunology , Vitiligo/immunology
4.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Article in English | MEDLINE | ID: mdl-33836578

ABSTRACT

Human adaptive-like "memory" CD56dimCD16+ natural killer (NK) cells in peripheral blood from cytomegalovirus-seropositive individuals have been extensively investigated in recent years and are currently explored as a treatment strategy for hematological cancers. However, treatment of solid tumors remains limited due to insufficient NK cell tumor infiltration, and it is unknown whether large expansions of adaptive-like NK cells that are equipped for tissue residency and tumor homing exist in peripheral tissues. Here, we show that human lung and blood contains adaptive-like CD56brightCD16- NK cells with hallmarks of tissue residency, including expression of CD49a. Expansions of adaptive-like lung tissue-resident NK (trNK) cells were found to be present independently of adaptive-like CD56dimCD16+ NK cells and to be hyperresponsive toward target cells. Together, our data demonstrate that phenotypically, functionally, and developmentally distinct subsets of adaptive-like NK cells exist in human lung and blood. Given their tissue-related character and hyperresponsiveness, human lung adaptive-like trNK cells might represent a suitable alternative for therapies targeting solid tumors.


Subject(s)
Killer Cells, Natural/immunology , Lung/immunology , Adaptation, Physiological/immunology , Flow Cytometry , Humans , Immunophenotyping , Integrin alpha1/immunology , Lung Neoplasms/immunology , Lung Neoplasms/therapy
5.
J Hepatol ; 71(2): 301-312, 2019 08.
Article in English | MEDLINE | ID: mdl-31100314

ABSTRACT

BACKGROUND & AIMS: Hepatitis delta virus (HDV) infection is the most severe form of viral hepatitis. Although HDV-associated liver disease is considered immune-mediated, adaptive immune responses against HDV are weak. Thus, the role of several other cell-mediated mechanisms such as those driven by mucosa-associated invariant T (MAIT) cells, a group of innate-like T cells highly enriched in the human liver, has not been extensively studied in clinical HDV infection. METHODS: MAIT cells from a sizeable cohort of patients with chronic HDV were analyzed ex vivo and in vitro after stimulation. Results were compared with MAIT cells from hepatitis B virus (HBV) monoinfected patients and healthy controls. RESULTS: Circulating MAIT cells were dramatically decreased in the peripheral blood of HDV-infected patients. Signs of decline were also observed in the liver. In contrast, only a modest decrease of circulating MAIT cells was noted in HBV monoinfection. Unsupervised high-dimensional analysis of residual circulating MAIT cells in chronic HDV infection revealed the appearance of a compound phenotype of CD38hiPD-1hiCD28loCD127loPLZFloEomesloHelioslo cells indicative of activation. Corroborating these results, MAIT cells exhibited a functionally impaired responsiveness. In parallel to MAIT cell loss, HDV-infected patients exhibited signs of monocyte activation and increased levels of proinflammatory cytokines IL-12 and IL-18. In vitro, IL-12 and IL-18 induced an activated MAIT cell phenotype similar to the one observed ex vivo in HDV-infected patients. These cytokines also promoted MAIT cell death, suggesting that they may contribute to MAIT cell activation and subsequent loss during HDV infection. CONCLUSIONS: These results suggest that chronic HDV infection engages the MAIT cell compartment causing activation, functional impairment, and subsequent progressive loss of MAIT cells as the HDV-associated liver disease progresses. LAY SUMMARY: Hepatitis delta virus (HDV) infection is the most severe form of viral hepatitis. We found that in patients with HDV, a subset of innate-like T cells called mucosa-associated invariant T cells (or MAIT cells), which are normally abundant in peripheral blood and the liver, are activated, functionally impaired and severely depleted.


Subject(s)
Hepatitis D, Chronic/immunology , Hepatitis Delta Virus/physiology , Lymphocyte Activation/immunology , Mucosal-Associated Invariant T Cells/immunology , Adult , Aged , CD8-Positive T-Lymphocytes/immunology , Cohort Studies , Female , Hep G2 Cells , Hepatitis D, Chronic/virology , Histocompatibility Antigens Class I/metabolism , Humans , Interleukin-12/blood , Interleukin-18/blood , Liver/pathology , Male , Middle Aged , Minor Histocompatibility Antigens/metabolism , Phenotype , Receptors, Antigen, T-Cell/metabolism , Young Adult
6.
J Immunol ; 197(8): 3069-3075, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27591320

ABSTRACT

Amniotic fluid (AF) surrounds the growing fetus, and cells derived from AF are commonly used for diagnosis of genetic diseases. Intra-amniotic infections are strongly linked to preterm birth, which is the leading cause of perinatal mortality worldwide. Surprisingly little is known, however, about mature hematopoietic cells in AF, which could potentially be involved in immune responses during pregnancy. In this study, we show that the dominating population of viable CD45+ cells in AF is represented by a subset of fetal CD103+ group 3 innate lymphoid cells (ILCs) producing high levels of IL-17 and TNF. Fetal CD103+ ILC3s could also be detected at high frequency in second-trimester mucosal tissues (e.g., the intestine and lung). Taken together, our data indicate that CD103+ ILC3s accumulate with gestation in the fetal intestine and subsequently egress to the AF. The dominance of ILC3s producing IL-17 and TNF in AF suggests that they could be involved in controlling intra-amniotic infections and inflammation and as such could be important players in regulating subsequent premature birth.


Subject(s)
Amniotic Fluid/immunology , Intestinal Mucosa/immunology , Lymphocyte Subsets/immunology , Lymphocytes/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Respiratory Mucosa/immunology , Antigens, CD/metabolism , Cells, Cultured , Female , Fetus , Humans , Immunity, Innate , Infant, Newborn , Integrin alpha Chains/metabolism , Interleukin-17/metabolism , Leukocyte Common Antigens/metabolism , Pregnancy , Pregnancy Trimester, Second , Tumor Necrosis Factor-alpha/metabolism
7.
J Allergy Clin Immunol ; 139(4): 1321-1330.e4, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27670241

ABSTRACT

BACKGROUND: In contrast to the extensive knowledge about human natural killer (NK) cells in peripheral blood, relatively little is known about NK cells in the human lung. Knowledge about the composition, differentiation, and function of human lung NK cells is critical to better understand their role in diseases affecting the lung, including asthma, chronic obstructive pulmonary disease, infections, and cancer. OBJECTIVE: We sought to analyze and compare the phenotypic and functional characteristics of NK cells in the human lung and peripheral blood at the single-cell level. METHODS: NK cells in human lung tissue and matched peripheral blood from 132 subjects were analyzed by using 16-color flow cytometry and confocal microscopy. RESULTS: CD56dimCD16+ NK cells made up the vast majority of NK cells in human lungs, had a more differentiated phenotype, and more frequently expressed educating killer cell immunoglobulin-like receptors compared with NK cells in peripheral blood. Despite this, human lung NK cells were hyporesponsive toward target cell stimulation, even after priming with IFN-α. Furthermore, we detected a small subset of NK cells expressing CD69, a marker of tissue residency. These CD69+ NK cells in the lung consisted predominantly of immature CD56brightCD16- NK cells and less differentiated CD56dimCD16+ NK cells. CONCLUSION: Here, we characterize the major NK cell populations in the human lung. Our data suggest a model in which the majority of NK cells in the human lung dynamically move between blood and the lung rather than residing in the lung as bona fide tissue-resident CD69+ NK cells.


Subject(s)
Killer Cells, Natural/cytology , Lung/cytology , Lung/immunology , Lymphocyte Subsets/cytology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD56 Antigen/immunology , Cell Differentiation/immunology , Flow Cytometry , Humans , Killer Cells, Natural/immunology , Lectins, C-Type/immunology , Lymphocyte Subsets/immunology , Microscopy, Confocal
8.
J Immunol ; 195(7): 3262-72, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26283480

ABSTRACT

NK cells play an important role in the defense against viral infections. However, little is known about the regulation of NK cell responses during the first days of acute viral infections in humans. In this study, we used the live attenuated yellow fever virus (YFV) vaccine 17D as a human in vivo model to study the temporal dynamics and regulation of NK cell responses in an acute viral infection. YFV induced a robust NK cell response in vivo, with an early activation and peak in NK cell function at day 6, followed by a delayed peak in Ki67 expression, which was indicative of proliferation, at day 10. The in vivo NK cell response correlated positively with plasma type I/III IFN levels at day 6, as well as with the viral load. YFV induced an increased functional responsiveness to IL-12 and IL-18, as well as to K562 cells, indicating that the NK cells were primed in vivo. The NK cell responses were associated primarily with the stage of differentiation, because the magnitude of induced Ki67 and CD69 expression was distinctly higher in CD57(-) NK cells. In contrast, NK cells expressing self- and nonself-HLA class I-binding inhibitory killer cell Ig-like receptors contributed, to a similar degree, to the response. Taken together, our results indicate that NK cells are primed by type I/III IFN in vivo early after YFV infection and that their response is governed primarily by the differentiation stage, independently of killer cell Ig-like receptor/HLA class I-mediated inhibition or education.


Subject(s)
Interferon Type I/immunology , Killer Cells, Natural/immunology , Receptors, KIR/immunology , Yellow Fever Vaccine/immunology , Yellow fever virus/immunology , Adult , Antibodies, Neutralizing/immunology , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , B-Lymphocytes/immunology , CD57 Antigens/metabolism , Cell Differentiation/immunology , Cell Proliferation , Histocompatibility Antigens Class I/immunology , Humans , Interferon Type I/blood , Interleukin-12 Subunit p35/immunology , Interleukin-18/immunology , K562 Cells , Ki-67 Antigen/biosynthesis , Killer Cells, Natural/cytology , Lectins, C-Type/biosynthesis , Lymphocyte Activation/immunology , Middle Aged , T-Lymphocytes/immunology , Vaccines, Attenuated/immunology , Viral Load/immunology , Viral Vaccines/immunology
9.
J Immunol ; 194(6): 2467-71, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25672754

ABSTRACT

Although NK cells are considered innate, recent studies in mice revealed the existence of a unique lineage of hepatic CD49a(+)DX5(-) NK cells with adaptive-like features. Development of this NK cell lineage is, in contrast to conventional NK cells, dependent on T-bet but not Eomes. In this study, we describe the identification of a T-bet(+)Eomes(-)CD49a(+) NK cell subset readily detectable in the human liver, but not in afferent or efferent hepatic venous or peripheral blood. Human intrahepatic CD49a(+) NK cells express killer cell Ig-like receptor and NKG2C, indicative of having undergone clonal-like expansion, are CD56(bright), and express low levels of CD16, CD57, and perforin. After stimulation, CD49a(+) NK cells express high levels of inflammatory cytokines but degranulate poorly. CD49a(+) NK cells retain their phenotype after expansion in long-term in vitro cultures. These results demonstrate the presence of a likely human counterpart of mouse intrahepatic NK cells with adaptive-like features.


Subject(s)
Cell Proliferation , Integrin alpha1/immunology , Killer Cells, Natural/immunology , Liver/immunology , Adult , CD56 Antigen/immunology , CD56 Antigen/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Integrin alpha1/metabolism , Killer Cells, Natural/metabolism , Liver/metabolism , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Receptors, KIR/immunology , Receptors, KIR/metabolism
10.
Cell Commun Signal ; 12: 63, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25266361

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) are increasingly considered to be used as biological immunosuppressants in hematopoietic stem cell transplantation (HSCT). In the early reconstitution phase following HSCT, natural killer (NK) cells represent the major lymphocyte population in peripheral blood and display graft-vs-leukemia (GvL) effects. The functional interactions between NK cells and MSCs have the potential to influence the leukemia relapse rate after HSCT. Until date, MSC-NK cell interaction studies are largely focussed on bone marrow derived (BM)-MSCs. Umbilical cord derived (UC)-MSCs might be an alternative source of therapeutic MSCs. Thus, we studied the interaction of UC-MSCs with unstimulated allogeneic NK cells. RESULTS: UC-MSCs could potently suppress NK cell cytotoxicity in overnight cultures via soluble factors. The main soluble immunosuppressant was identified as prostaglandin (PG)-E2. Maximal PGE2 release involved IL-1ß priming of MSCs after close contact between the NK cells and UC-MSCs. Interestingly, blocking gamma-secretase activation alleviated the immunosuppression by controlling PGE2 production. IL-1 receptor activation and subsequent downstream signalling events were found to require gamma-secretase activity. CONCLUSION: Although the role of PGE2 in NK cell-MSC has been reported, the requirement of cell-cell contact for PGE2 induced immunosuppression remained unexplained. Our findings shed light on this puzzling observation and identify new players in the NK cell-MSC crosstalk.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Dinoprostone/metabolism , Killer Cells, Natural/metabolism , Mesenchymal Stem Cells/metabolism , Cell Communication , Cell Degranulation , Cells, Cultured , Cytotoxicity Tests, Immunologic , Humans , Infant, Newborn , Interleukin-1/metabolism , K562 Cells , Killer Cells, Natural/physiology , Mesenchymal Stem Cells/physiology , Phenotype , Receptors, Interleukin-1/metabolism , Umbilical Cord/cytology
11.
J Allergy Clin Immunol ; 140(1): 318, 2017 07.
Article in English | MEDLINE | ID: mdl-28528925
12.
Oncoimmunology ; 12(1): 2233402, 2023.
Article in English | MEDLINE | ID: mdl-37448786

ABSTRACT

Lung cancer is a leading cause of cancer-related death worldwide. Despite recent advances in tissue immunology, little is known about the spatial distribution of tissue-resident lymphocyte subsets in lung tumors. Using high-parameter flow cytometry, we identified an accumulation of tissue-resident lymphocytes including tissue-resident NK (trNK) cells and CD8+ tissue-resident memory T (TRM) cells toward the center of human non-small cell lung carcinomas (NSCLC). Chemokine receptor expression patterns indicated different modes of tumor-infiltration and/or residency between trNK cells and CD8+ TRM cells. In contrast to CD8+ TRM cells, trNK cells and ILCs generally expressed low levels of immune checkpoint receptors independent of location in the tumor. Additionally, granzyme expression in trNK cells and CD8+ TRM cells was highest in the tumor center, and intratumoral CD49a+CD16- NK cells were functional and responded stronger to target cell stimulation than their CD49a- counterparts, indicating functional relevance of trNK cells in lung tumors. In summary, the present spatial mapping of lymphocyte subsets in human NSCLC provides novel insights into the composition and functionality of tissue-resident immune cells, suggesting a role for trNK cells and CD8+ TRM cells in lung tumors and their potential relevance for future therapeutic approaches.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , CD8-Positive T-Lymphocytes , Immunity, Innate , Integrin alpha1/metabolism , Killer Cells, Natural/metabolism
13.
Front Immunol ; 13: 834862, 2022.
Article in English | MEDLINE | ID: mdl-35371005

ABSTRACT

Respiratory viral infections with SARS-CoV-2 and influenza viruses commonly induce a strong infiltration of immune cells into the human lung, with potential detrimental effects on the integrity of the lung tissue. Despite comprising the largest fractions of circulating lymphocytes in the lung, rather little is known about how peripheral blood natural killer (NK) cell and T cell subsets are equipped for lung-homing in COVID-19 and influenza. Here, we provide a detailed comparative analysis of NK cells and T cells in patients infected with SARS-CoV-2 or influenza virus, focusing on the protein and gene expression of chemokine receptors known to be involved in recruitment to the lung. For this, we used 28-colour flow cytometry as well as re-analysis of a publicly available single-cell RNA-seq dataset from bronchoalveolar lavage (BAL) fluid. Frequencies of NK cells and T cells expressing CXCR3, CXCR6, and CCR5 were altered in peripheral blood of COVID-19 and influenza patients, in line with increased transcript expression of CXCR3, CXCR6, and CCR5 and their respective ligands in BAL fluid. NK cells and T cells expressing lung-homing receptors displayed stronger phenotypic signs of activation compared to cells lacking lung-homing receptors, and activation was overall stronger in influenza compared to COVID-19. Together, our results indicate a role for CXCR3+, CXCR6+, and/or CCR5+ NK cells and T cells that potentially migrate to the lungs in moderate COVID-19 and influenza patients, identifying common targets for future therapeutic interventions in respiratory viral infections.


Subject(s)
COVID-19 , Influenza, Human , Gene Expression , Humans , Influenza, Human/metabolism , Killer Cells, Natural , Lung , SARS-CoV-2 , T-Lymphocyte Subsets
14.
Cell Rep Med ; 3(2): 100508, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35243416

ABSTRACT

Few approaches have been made toward exploring autologous NK cells in settings of cancer immunotherapy. Here, we demonstrate the feasibility of infusing multiple doses of ex vivo activated and expanded autologous NK cells in patients with multiple myeloma (MM) post-autologous stem cell transplantation. Infused NK cells were detected in circulation up to 4 weeks after the last infusion. Elevations in plasma granzyme B levels were observed following each consecutive NK cell infusion. Moreover, increased granzyme B levels were detected in bone marrow 4 weeks after the last infusion. All measurable patients had objective, detectable responses after NK cell infusions in terms of reduction in M-component and/or minimal residual disease. The present study demonstrates that autologous NK cell-based immunotherapy is feasible in a setting of MM consolidation therapy. It opens up the possibility for usage of autologous NK cells in clinical settings where patients are not readily eligible for allogeneic NK cell-based immunotherapies.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma , Consolidation Chemotherapy , Granzymes , Humans , Killer Cells, Natural , Multiple Myeloma/therapy , Stem Cell Transplantation , Transplantation, Autologous
15.
Eur J Immunol ; 40(5): 1428-39, 2010 May.
Article in English | MEDLINE | ID: mdl-20186880

ABSTRACT

Human NK cells can be subdivided into CD56(dim) and CD56(bright) NK cells, which exhibit different phenotypical and functional characteristics. As murine NK cells lack CD56 or a distinct correlate, direct comparative studies of NK cells in mice and humans are limited. Although CD27 is currently proposed as a feasible subset marker in mice, we assume that the usage of this marker alone is insufficient. We rather investigated the expression of the chemokine receptor CXCR3 for its suitability for distinguishing murine NK-cell subsets with simultaneous consideration of CD27. Compared with CXCR3(-) NK cells, exerting stronger cytotoxic capability, CXCR3+ NK cells displayed an activated phenotype with a lower expression of Ly49 receptors, corresponding to human CD56(bright) NK cells. Also in common with human CD56(bright) NK cells, murine CXCR3+ NK cells exhibit prolific expansion as well as robust IFN-gamma, TNF-alpha and MIP-1alpha production. We additionally demonstrated changes in both CXCR3 and CD27 expression upon NK-cell activation. In summary, CXCR3 serves as an additional applicable marker for improved discrimination of functionally distinct murine NK-cell subsets that comply with those in humans.


Subject(s)
CD56 Antigen/analysis , Killer Cells, Natural/classification , Lymphocyte Subsets/chemistry , Receptors, CXCR3/analysis , Tumor Necrosis Factor Receptor Superfamily, Member 7/analysis , Animals , Biomarkers , Chemokine CCL3/biosynthesis , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Cytotoxicity, Immunologic , Female , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukins/pharmacology , Ionomycin/pharmacology , Killer Cells, Natural/chemistry , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily A/analysis , RNA, Messenger/biosynthesis , Species Specificity , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
Brain Behav Immun ; 25(6): 1233-41, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21447379

ABSTRACT

Previous studies have demonstrated the utility of S100B as a surrogate marker of brain-related pathologies, e.g. neuropsychiatric disorders, and melanoma progression, which have an inflammatory component. This study addresses the relevance of S100B(+) lymphocytes in mediating such responses. S100B expression was determined in human peripheral blood leukocytes isolated from healthy volunteers using flow cytometry. S100B(+) lymphocytes were characterised for phenotype, cytokine production and S100B secretion. In addition, we investigated whether S100B activates monocytes and neutrophils. S100B(+) cells comprised 2-4% of all lymphocytes and the majority displayed a CD3(+) CD8(+) phenotype; fewer cells were CD3(-) CD56(+) NK lymphocytes. Comparison of S100B(+) and S100B(-) CD3(+) CD8(+) cells revealed no differences in production of interferon gamma (IFNγ) and interleukin-2 (IL-2). Stimulation of S100B(+) CD3(+) CD8(+) lymphocytes with anti-CD3 or phytohaemagglutinin resulted in release of S100B. High concentrations of recombinant human S100B triggered upregulation of CD11b and membrane shedding of CD62L in granulocytes and monocytes. These findings set the stage for a new field of research addressing a S100B-mediated crosstalk between the innate and adaptive immune systems if close proximity of effector and responder cells accomplishes sufficient local S100B levels. In various physiological and pathological conditions S100B might function as an interface to immunological processes, distinct from known cytokine- and chemokine-mediated pathways.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Killer Cells, Natural/metabolism , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Adaptive Immunity , Adult , Biomarkers , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , CD3 Complex/analysis , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Humans , Immunity, Innate , Intracellular Fluid/metabolism , Ionomycin/pharmacology , Killer Cells, Natural/drug effects , L-Selectin/metabolism , Lymphocyte Activation/drug effects , Monocytes/drug effects , Muromonab-CD3/pharmacology , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Nerve Growth Factors/pharmacology , Neutrophils/drug effects , Phytohemagglutinins/pharmacology , Recombinant Proteins/pharmacology , S100 Calcium Binding Protein beta Subunit , S100 Proteins/biosynthesis , S100 Proteins/genetics , S100 Proteins/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Up-Regulation/drug effects
17.
Front Immunol ; 12: 752104, 2021.
Article in English | MEDLINE | ID: mdl-34867984

ABSTRACT

Innate lymphoid cells (ILCs) contribute to immune defense, yet it is poorly understood how ILCs develop and are strategically positioned in the lung. This applies especially to human ILCs due to the difficulty of studying them in vivo. Here we investigated the ontogeny and migration of human ILCs in vivo with a humanized mouse model ("MISTRG") expressing human cytokines. In addition to known tissue-resident ILC subsets, we discovered CD5-expressing ILCs that predominantly resided within the lung vasculature and in the circulation. CD5+ ILCs contained IFNγ-producing mature ILC1s as well as immature ILCs that produced ILC effector cytokines under polarizing conditions in vitro. CD5+ ILCs had a distinct ontogeny compared to conventional CD5- ILCs because they first appeared in the thymus, spleen and liver rather than in the bone marrow after transplantation of MISTRG mice with human CD34+ hematopoietic stem and progenitor cells. Due to their strategic location, human CD5+ ILCs could serve as blood-borne sentinels, ready to be recruited into the lung to respond to environmental challenges. This work emphasizes the uniqueness of human CD5+ ILCs in terms of their anatomical localization and developmental origin compared to well-studied CD5- ILCs.


Subject(s)
CD5 Antigens/immunology , Lung/immunology , Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Animals , Animals, Genetically Modified , Cell Differentiation , Cell Movement , Cytokines/immunology , Female , Hematopoietic Stem Cell Transplantation , Humans , Immunity, Innate , Male , Mice , Middle Aged , Spleen/immunology
18.
Sci Immunol ; 5(50)2020 08 21.
Article in English | MEDLINE | ID: mdl-32826343

ABSTRACT

Understanding innate immune responses in COVID-19 is important to decipher mechanisms of host responses and interpret disease pathogenesis. Natural killer (NK) cells are innate effector lymphocytes that respond to acute viral infections but might also contribute to immunopathology. Using 28-color flow cytometry, we here reveal strong NK cell activation across distinct subsets in peripheral blood of COVID-19 patients. This pattern was mirrored in scRNA-seq signatures of NK cells in bronchoalveolar lavage from COVID-19 patients. Unsupervised high-dimensional analysis of peripheral blood NK cells furthermore identified distinct NK cell immunotypes that were linked to disease severity. Hallmarks of these immunotypes were high expression of perforin, NKG2C, and Ksp37, reflecting increased presence of adaptive NK cells in circulation of patients with severe disease. Finally, arming of CD56bright NK cells was observed across COVID-19 disease states, driven by a defined protein-protein interaction network of inflammatory soluble factors. This study provides a detailed map of the NK cell activation landscape in COVID-19 disease.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/immunology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Killer Cells, Natural/immunology , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Severity of Illness Index , Adaptive Immunity , CD56 Antigen/metabolism , COVID-19 , Coronavirus Infections/blood , Coronavirus Infections/pathology , Female , Flow Cytometry/methods , Humans , Lymphocyte Activation , Male , Middle Aged , Pandemics , Phenotype , Pneumonia, Viral/blood , Pneumonia, Viral/pathology , Polymerase Chain Reaction , Prospective Studies , Protein Interaction Maps/immunology , Receptors, KIR/metabolism , SARS-CoV-2 , Serologic Tests , Sweden/epidemiology
19.
Nat Commun ; 10(1): 3841, 2019 08 26.
Article in English | MEDLINE | ID: mdl-31451696

ABSTRACT

Human lung tissue-resident NK cells (trNK cells) are likely to play an important role in host responses towards viral infections, inflammatory conditions and cancer. However, detailed insights into these cells are still largely lacking. Here we show, using RNA sequencing and flow cytometry-based analyses, that subsets of human lung CD69+CD16- NK cells display hallmarks of tissue-residency, including high expression of CD49a, CD103, and ZNF683, and reduced expression of SELL, S1PR5, and KLF2/3. CD49a+CD16- NK cells are functionally competent, and produce IFN-γ, TNF, MIP-1ß, and GM-CSF. After stimulation with IL-15, they upregulate perforin, granzyme B, and Ki67 to a similar degree as CD49a-CD16- NK cells. Comparing datasets from trNK cells in human lung and bone marrow with tissue-resident memory CD8+ T cells identifies core genes co-regulated either by tissue-residency, cell-type or location. Together, our data indicate that human lung trNK cells have distinct features, likely regulating their function in barrier immunity.


Subject(s)
Immunity, Mucosal , Killer Cells, Natural/metabolism , Lung Diseases/immunology , Lung/cytology , Transcriptome/immunology , Aged , Aged, 80 and over , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Datasets as Topic , Female , Humans , Killer Cells, Natural/immunology , Lung/immunology , Lung/surgery , Lung Diseases/pathology , Lung Diseases/surgery , Male , Middle Aged , Pneumonectomy , RNA-Seq , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism
20.
Front Immunol ; 10: 1116, 2019.
Article in English | MEDLINE | ID: mdl-31156653

ABSTRACT

NK cells in the human lung respond to influenza A virus- (IAV-) infected target cells. However, the detailed functional capacity of human lung and peripheral blood NK cells remains to be determined in IAV and other respiratory viral infections. Here, we investigated the effects of IAV infection on human lung and peripheral blood NK cells in vitro and ex vivo following clinical infection. IAV infection of lung- and peripheral blood-derived mononuclear cells in vitro induced NK cell hyperresponsiveness to K562 target cells, including increased degranulation and cytokine production particularly in the CD56brightCD16- subset of NK cells. Furthermore, lung CD16- NK cells showed increased IAV-mediated but target cell-independent activation compared to CD16+ lung NK cells or total NK cells in peripheral blood. IAV infection rendered peripheral blood NK cells responsive toward the normally NK cell-resistant lung epithelial cell line A549, indicating that NK cell activation during IAV infection could contribute to killing of surrounding non-infected epithelial cells. In vivo, peripheral blood CD56dimCD16+ and CD56brightCD16- NK cells were primed during acute IAV infection, and a small subset of CD16-CD49a+CXCR3+ NK cells could be identified, with CD49a and CXCR3 potentially promoting homing to and tissue-retention in the lung during acute infection. Together, we show that IAV respiratory viral infections prime otherwise hyporesponsive lung NK cells, indicating that both CD16+ and CD16- NK cells including CD16-CD49a+ tissue-resident NK cells could contribute to host immunity but possibly also tissue damage in clinical IAV infection.


Subject(s)
Influenza A virus/physiology , Influenza, Human/immunology , Lung/physiology , Antigen Presentation , Blood Circulation , Bronchial Hyperreactivity/metabolism , Cytotoxicity, Immunologic , Humans , K562 Cells , Killer Cells, Natural/immunology , Lymphocyte Activation
SELECTION OF CITATIONS
SEARCH DETAIL