Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
ACS Nano ; 17(17): 17451-17467, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37643371

ABSTRACT

Nanoparticles (NPs) elicit sterile inflammation, but the underlying signaling pathways are poorly understood. Here, we report that human monocytes are particularly vulnerable to amorphous silica NPs, as evidenced by single-cell-based analysis of peripheral blood mononuclear cells using cytometry by time-of-flight (CyToF), while silane modification of the NPs mitigated their toxicity. Using human THP-1 cells as a model, we observed cellular internalization of silica NPs by nanoscale secondary ion mass spectrometry (nanoSIMS) and this was confirmed by transmission electron microscopy. Lipid droplet accumulation was also noted in the exposed cells. Furthermore, time-of-flight secondary ion mass spectrometry (ToF-SIMS) revealed specific changes in plasma membrane lipids, including phosphatidylcholine (PC) in silica NP-exposed cells, and subsequent studies suggested that lysophosphatidylcholine (LPC) acts as a cell autonomous signal for inflammasome activation in the absence of priming with a microbial ligand. Moreover, we found that silica NPs elicited NLRP3 inflammasome activation in monocytes, whereas cell death transpired through a non-apoptotic, lipid peroxidation-dependent mechanism. Together, these data further our understanding of the mechanism of sterile inflammation.


Subject(s)
Inflammasomes , Nanoparticles , Humans , Leukocytes, Mononuclear , Spectrometry, Mass, Secondary Ion , Inflammation , Silicon Dioxide/pharmacology
2.
Respir Res ; 24(1): 62, 2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36829233

ABSTRACT

BACKGROUND: COVID-19 remains a major public health challenge, requiring the development of tools to improve diagnosis and inform therapeutic decisions. As dysregulated inflammation and coagulation responses have been implicated in the pathophysiology of COVID-19 and sepsis, we studied their plasma proteome profiles to delineate similarities from specific features. METHODS: We measured 276 plasma proteins involved in Inflammation, organ damage, immune response and coagulation in healthy controls, COVID-19 patients during acute and convalescence phase, and sepsis patients; the latter included (i) community-acquired pneumonia (CAP) caused by Influenza, (ii) bacterial CAP, (iii) non-pneumonia sepsis, and (iv) septic shock patients. RESULTS: We identified a core response to infection consisting of 42 proteins altered in both COVID-19 and sepsis, although higher levels of cytokine storm-associated proteins were evident in sepsis. Furthermore, microbiologic etiology and clinical endotypes were linked to unique signatures. Finally, through machine learning, we identified biomarkers, such as TRIM21, PTN and CASP8, that accurately differentiated COVID-19 from CAP-sepsis with higher accuracy than standard clinical markers. CONCLUSIONS: This study extends the understanding of host responses underlying sepsis and COVID-19, indicating varying disease mechanisms with unique signatures. These diagnostic and severity signatures are candidates for the development of personalized management of COVID-19 and sepsis.


Subject(s)
COVID-19 , Community-Acquired Infections , Pneumonia , Sepsis , Humans , COVID-19/complications , Proteomics , Inflammation/complications , Biomarkers
3.
iScience ; 26(2): 106071, 2023 Feb 17.
Article in English | MEDLINE | ID: mdl-36818285

ABSTRACT

Here we extend the understanding of how chemical inhibition of SHIP paralogs controls obesity. We compare different classes of SHIP inhibitors and find that selective inhibitors of SHIP1 or SHIP2 are unable to prevent weight gain and body fat accumulation during increased caloric intake. Surprisingly, only pan-SHIP1/2 inhibitors (pan-SHIPi) prevent diet-induced obesity. We confirm that pan-SHIPi is essential by showing that dual treatment with SHIP1 and SHIP2 selective inhibitors reduced adiposity during excess caloric intake. Consistent with this, genetic inactivation of both SHIP paralogs in eosinophils or myeloid cells also reduces obesity and adiposity. In fact, pan-SHIPi requires an eosinophil compartment to prevent diet-induced adiposity, demonstrating that pan-SHIPi acts via an immune mechanism. We also find that pan-SHIPi increases ILC2 cell function in aged, obese mice to reduce their obesity. Finally, we show that pan-SHIPi also reduces hyperglycemia, but not via eosinophils, indicating a separate mechanism for glucose control.

5.
iScience ; 25(10): 105137, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36185379

ABSTRACT

Although PD-1 was shown to be a hallmark of T cells exhaustion, controversial studies have been reported on the role of PD-1 on NK cells. Here, we found by flow cytometry and single cell RNA sequencing analysis that PD-1 can be expressed on MHC class I-deficient tumor-infiltrating NK cells in vivo. We also demonstrate distinct alterations in the phenotype of PD-1-deficient NK cells and a more mature phenotype which might reduce their capacity to migrate and kill in vivo. Tumor-infiltrating NK cells that express PD-1 were highly associated with the expression of CXCR6. Furthermore, our results demonstrate that PD-L1 molecules in membranes of PD-1-deficient NK cells migrate faster than in NK cells from wild-type mice, suggesting that PD-1 and PD-L1 form cis interactions with each other on NK cells. These data demonstrate that there may be a role for the PD-1/PD-L1 axis in tumor-infiltrating NK cells in vivo.

6.
Nat Commun ; 13(1): 5093, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064840

ABSTRACT

The hypoxia-inducible factors (HIFs) regulate the main transcriptional pathway of response to hypoxia in T cells and are negatively regulated by von Hippel-Lindau factor (VHL). But the role of HIFs in the regulation of CD4 T cell responses during infection with M. tuberculosis isn't well understood. Here we show that mice lacking VHL in T cells (Vhl cKO) are highly susceptible to infection with M. tuberculosis, which is associated with a low accumulation of mycobacteria-specific T cells in the lungs that display reduced proliferation, altered differentiation and enhanced expression of inhibitory receptors. In contrast, HIF-1 deficiency in T cells is redundant for M. tuberculosis control. Vhl cKO mice also show reduced responses to vaccination. Further, VHL promotes proper MYC-activation, cell-growth responses, DNA synthesis, proliferation and survival of CD4 T cells after TCR activation. The VHL-deficient T cell responses are rescued by the loss of HIF-1α, indicating that the increased susceptibility to M. tuberculosis infection and the impaired responses of Vhl-deficient T cells are HIF-1-dependent.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Tuberculosis , Von Hippel-Lindau Tumor Suppressor Protein , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Hypoxia , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Mice , T-Lymphocytes/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/prevention & control , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/immunology
7.
Scand J Immunol ; : e13195, 2022 Jun 02.
Article in English | MEDLINE | ID: mdl-35652743

ABSTRACT

The Karolinska KI/K COVID-19 Immune Atlas project was conceptualized in March 2020 as a part of the academic research response to the developing SARS-CoV-2 pandemic. The aim was to rapidly provide a curated dataset covering the acute immune response towards SARS-CoV-2 infection in humans, as it occurred during the first wave. The Immune Atlas was built as an open resource for broad research and educational purposes. It contains a presentation of the response evoked by different immune and inflammatory cells in defined naïve patient-groups as they presented with moderate and severe COVID-19 disease. The present Resource Article describes how the Karolinska KI/K COVID-19 Immune Atlas allow scientists, students, and other interested parties to freely explore the nature of the immune response towards human SARS-CoV-2 infection in an online setting.

8.
Front Immunol ; 13: 854312, 2022.
Article in English | MEDLINE | ID: mdl-35757763

ABSTRACT

Natural killer (NK) cells play roles in viral clearance and early surveillance against malignant transformation, yet our knowledge of the underlying mechanisms controlling their development and functions remain incomplete. To reveal cell fate-determining pathways in NK cell progenitors (NKP), we utilized an unbiased approach and generated comprehensive gene expression profiles of NK cell progenitors. We found that the NK cell program was gradually established in the CLP to preNKP and preNKP to rNKP transitions. In line with FOXO1 and FOXO3 being co-expressed through the NK developmental trajectory, the loss of both perturbed the establishment of the NK cell program and caused stalling in both NK cell development and maturation. In addition, we found that the combined loss of FOXO1 and FOXO3 caused specific changes to the composition of the non-cytotoxic innate lymphoid cell (ILC) subsets in bone marrow, spleen, and thymus. By combining transcriptome and chromatin profiling, we revealed that FOXO TFs ensure proper NK cell development at various lineage-commitment stages through orchestrating distinct molecular mechanisms. Combined FOXO1 and FOXO3 deficiency in common and innate lymphoid cell progenitors resulted in reduced expression of genes associated with NK cell development including ETS-1 and their downstream target genes. Lastly, we found that FOXO1 and FOXO3 controlled the survival of committed NK cells via gene regulation of IL-15Rß (CD122) on rNKPs and bone marrow NK cells. Overall, we revealed that FOXO1 and FOXO3 function in a coordinated manner to regulate essential developmental genes at multiple stages during murine NK cell and ILC lineage commitment.


Subject(s)
Forkhead Box Protein O1 , Forkhead Box Protein O3 , Killer Cells, Natural , Lymphoid Progenitor Cells , Animals , Cell Differentiation/immunology , Forkhead Box Protein O1/immunology , Forkhead Box Protein O3/immunology , Immunity, Innate , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL
9.
Eur J Immunol ; 52(3): 503-510, 2022 03.
Article in English | MEDLINE | ID: mdl-34837225

ABSTRACT

Corona disease 2019 (COVID-19) affects multiple organ systems. Recent studies have indicated perturbations in the circulating metabolome linked to COVID-19 severity. However, several questions pertain with respect to the metabolome in COVID-19. We performed an in-depth assessment of 1129 unique metabolites in 27 hospitalized COVID-19 patients and integrated results with large-scale proteomic and immunology data to capture multiorgan system perturbations. More than half of the detected metabolic alterations in COVID-19 were driven by patient-specific confounding factors ranging from comorbidities to xenobiotic substances. Systematically adjusting for this, a COVID-19-specific metabolic imprint was defined which, over time, underwent a switch in response to severe acute respiratory syndrome coronavirus-2 seroconversion. Integration of the COVID-19 metabolome with clinical, cellular, molecular, and immunological severity scales further revealed a network of metabolic trajectories aligned with multiple pathways for immune activation, and organ damage including neurological inflammation and damage. Altogether, this resource refines our understanding of the multiorgan system perturbations in severe COVID-19 patients.


Subject(s)
COVID-19/immunology , COVID-19/metabolism , Metabolome/immunology , SARS-CoV-2 , Adolescent , Adult , Aged , COVID-19/complications , Case-Control Studies , Central Nervous System Diseases/etiology , Central Nervous System Diseases/immunology , Central Nervous System Diseases/metabolism , Cohort Studies , Female , Humans , Male , Metabolomics , Middle Aged , Organ Specificity , Pandemics , Phenotype , Proteomics , Severity of Illness Index , Young Adult
10.
Front Immunol ; 12: 727300, 2021.
Article in English | MEDLINE | ID: mdl-34887849

ABSTRACT

Upon infection with Mycobacterium tuberculosis (Mtb) the host immune response might clear the bacteria, control its growth leading to latent tuberculosis (LTB), or fail to control its growth resulting in active TB (ATB). There is however no clear understanding of the features underlying a more or less effective response. Mtb glycolipids are abundant in the bacterial cell envelope and modulate the immune response to Mtb, but the patterns of response to glycolipids are still underexplored. To identify the CD45+ leukocyte activation landscape induced by Mtb glycolipids in peripheral blood of ATB and LTB, we performed a detailed assessment of the immune response of PBMCs to the Mtb glycolipids lipoarabinomannan (LAM) and its biosynthetic precursor phosphatidyl-inositol mannoside (PIM), and purified-protein derivate (PPD). At 24 h of stimulation, cell profiling and secretome analysis was done using mass cytometry and high-multiplex immunoassay. PIM induced a diverse cytokine response, mainly affecting antigen-presenting cells to produce both pro-inflammatory and anti-inflammatory cytokines, but not IFN-γ, contrasting with PPD that was a strong inducer of IFN-γ. The effect of PIM on the antigen-presenting cells was partly TLR2-dependent. Expansion of monocyte subsets in response to PIM or LAM was reduced primarily in LTB as compared to healthy controls, suggesting a hyporesponsive/tolerance pattern derived from Mtb infection.


Subject(s)
Latent Tuberculosis/immunology , Tuberculosis/immunology , Adult , Aged , Aged, 80 and over , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , B-Lymphocytes/classification , B-Lymphocytes/immunology , Case-Control Studies , Cohort Studies , Cytokines/biosynthesis , Female , Glycolipids/administration & dosage , Glycolipids/immunology , Humans , In Vitro Techniques , Killer Cells, Natural/immunology , Male , Middle Aged , Mycobacterium tuberculosis/immunology , Myeloid Cells/immunology , Phosphatidylinositols/administration & dosage , Phosphatidylinositols/immunology , Prospective Studies , T-Lymphocytes/classification , T-Lymphocytes/immunology , Toll-Like Receptor 2/immunology , Tuberculin/administration & dosage , Tuberculin/immunology , Young Adult
11.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Article in English | MEDLINE | ID: mdl-34548411

ABSTRACT

Since the outset of the COVID-19 pandemic, increasing evidence suggests that the innate immune responses play an important role in the disease development. A dysregulated inflammatory state has been proposed as a key driver of clinical complications in COVID-19, with a potential detrimental role of granulocytes. However, a comprehensive phenotypic description of circulating granulocytes in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients is lacking. In this study, we used high-dimensional flow cytometry for granulocyte immunophenotyping in peripheral blood collected from COVID-19 patients during acute and convalescent phases. Severe COVID-19 was associated with increased levels of both mature and immature neutrophils, and decreased counts of eosinophils and basophils. Distinct immunotypes were evident in COVID-19 patients, with altered expression of several receptors involved in activation, adhesion, and migration of granulocytes (e.g., CD62L, CD11a/b, CD69, CD63, CXCR4). Paired sampling revealed recovery and phenotypic restoration of the granulocytic signature in the convalescent phase. The identified granulocyte immunotypes correlated with distinct sets of soluble inflammatory markers, supporting pathophysiologic relevance. Furthermore, clinical features, including multiorgan dysfunction and respiratory function, could be predicted using combined laboratory measurements and immunophenotyping. This study provides a comprehensive granulocyte characterization in COVID-19 and reveals specific immunotypes with potential predictive value for key clinical features associated with COVID-19.


Subject(s)
COVID-19/immunology , Granulocytes/immunology , COVID-19/blood , COVID-19/diagnosis , COVID-19/physiopathology , Granulocytes/cytology , Humans , Immunity, Innate , Immunophenotyping , Leukocyte Count , Lung/physiopathology , Models, Biological , Organ Dysfunction Scores , SARS-CoV-2 , Severity of Illness Index
12.
J Cell Sci ; 134(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34085696

ABSTRACT

NKG2D (also known as KLRK1) is a crucial natural killer (NK) cell-activating receptor, and the murine cytomegalovirus (MCMV) employs multiple immunoevasins to avoid NKG2D-mediated activation. One of the MCMV immunoevasins, gp40 (m152), downregulates the cell surface NKG2D ligand RAE-1γ (also known as Raet1c) thus limiting NK cell activation. This study establishes the molecular mechanism by which gp40 retains RAE-1γ in the secretory pathway. Using flow cytometry and pulse-chase analysis, we demonstrate that gp40 retains RAE-1γ in the early secretory pathway, and that this effect depends on the binding of gp40 to a host protein, TMED10, a member of the p24 protein family. We also show that the TMED10-based retention mechanism can be saturated, and that gp40 has a backup mechanism as it masks RAE-1γ on the cell surface, blocking the interaction with the NKG2D receptor and thus NK cell activation.


Subject(s)
Muromegalovirus , Animals , Ligands , Membrane Proteins , Mice , NK Cell Lectin-Like Receptor Subfamily K/genetics , Viral Proteins
13.
J Leukoc Biol ; 110(4): 617-628, 2021 10.
Article in English | MEDLINE | ID: mdl-34028876

ABSTRACT

Protective cytotoxic and proinflammatory cytokine responses by NK cells impact the outcome of infections by Toxoplasma gondii, a common parasite in humans and other vertebrates. However, T. gondii can also sequester within NK cells and downmodulate their effector functions. Recently, the implication of GABA signaling in infection and inflammation-related responses of mononuclear phagocytes and T cells has become evident. Yet, the role of GABAergic signaling in NK cells has remained unknown. Here, we report that human and murine NK cells synthesize and secrete GABA in response to infection challenge. Parasitized NK cells secreted GABA, whereas activation stimuli, such as IL-12/IL-18 or parasite lysates, failed to induce GABA secretion. GABA secretion by NK cells was associated to a transcriptional up-regulation of GABA synthesis enzymes (glutamate decarboxylases [GAD65/67]) and was abrogated by GAD inhibition. Further, NK cells expressed GABA-A receptor subunits and GABA signaling regulators, with transcriptional modulations taking place upon challenge with T. gondii. Exogenous GABA and GABA-containing supernatants from parasitized dendritic cells (DCs) impacted NK cell function by reducing the degranulation and cytotoxicity of NK cells. Conversely, GABA-containing supernatants from NK cells enhanced the migratory responses of parasitized DCs. This enhanced DC migration was abolished by GABA-A receptor antagonism or GAD inhibition and was reconstituted by exogenous GABA. Jointly, the data show that NK cells are GABAergic cells and that GABA hampers NK cell cytotoxicity in vitro. We hypothesize that GABA secreted by parasitized immune cells modulates the immune responses to T. gondii infection.


Subject(s)
Killer Cells, Natural/metabolism , Killer Cells, Natural/parasitology , Signal Transduction , Toxoplasma/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Cell Death , Cell Degranulation/physiology , Cell Line , Cell Movement , Dendritic Cells/parasitology , Humans , Killer Cells, Natural/physiology , Mice, Inbred C57BL , Transcription, Genetic
14.
Proc Natl Acad Sci U S A ; 117(50): 32005-32016, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33229588

ABSTRACT

Tumor-associated macrophages (TAMs) can have protumor properties, including suppressing immune responses, promoting vascularization and, consequently, augmenting tumor progression. To stop TAM-mediated immunosuppression, we use a novel treatment by injecting antibodies specific for scavenger receptor MARCO, which is expressed on a specific subpopulation of TAMs in the tumor. We now report the location of this TAM as well as the pleiotropic mechanism of action of anti-MARCO antibody treatment on tumor progression and further show that this is potentially relevant to humans. Using specific targeting, we observed decreased tumor vascularization, a switch in the metabolic program of MARCO-expressing macrophages, and activation of natural killer (NK) cell killing through TNF-related apoptosis-inducing ligand (TRAIL). This latter activity reverses the effect of melanoma cell-conditioned macrophages in blocking NK activation and synergizes with T cell-directed immunotherapy, such as antibodies to PD-1 or PD-L1, to enhance tumor killing. Our study thus reveals an approach to targeting the immunosuppressive tumor microenvironment with monoclonal antibodies to enhance NK cell activation and NK cell-mediated killing. This can complement existing T cell-directed immunotherapy, providing a promising approach to combinatorial immunotherapy for cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Killer Cells, Natural/immunology , Melanoma/drug therapy , Receptors, Immunologic/antagonists & inhibitors , Tumor-Associated Macrophages/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/metabolism , Male , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Knockout , Primary Cell Culture , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
15.
Eur J Immunol ; 50(4): 494-504, 2020 04.
Article in English | MEDLINE | ID: mdl-31834938

ABSTRACT

NK cells are innate immune cells characterized by their ability to spontaneously lyse tumor and virally infected cells. We have recently demonstrated that IL-15-sufficient DC regulate NK cell effector functions in mice. Here, we established that among ITAM-proximal signaling molecules, the expression levels of the scaffold molecule Linker for Activation of T cells (LAT) and its transcription factor ELF-1 were reduced 4 days after in vivo depletion of DC. Addition of IL-15, a cytokine presented by DC to NK cells, regulates LAT expression in NK cells with a significant effect on the DNAM1+ subset compared to DNAM1- cells. We also found that LAT expression is regulated via interaction of the DNAM1 receptor with its ligand CD155 in both immature and mature NK cells, independently of NK cell education. Finally, we found that LAT expression within DNAM1+ NK cells might be responsible for enhanced calcium mobilization following the triggering of activating receptors on NK cells. Altogether, we found that LAT expression is tightly regulated in DNAM1+ NK cells, via interaction(s) with DC, which express CD155 and IL-15, resulting in rapid activation of the DNAM1+ subset during activating receptor triggering.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Interleukin-15/metabolism , Killer Cells, Natural/immunology , Large Neutral Amino Acid-Transporter 1/metabolism , Receptors, Virus/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Calcium Signaling , Cells, Cultured , Cytotoxicity, Immunologic , DNA-Binding Proteins/genetics , Interleukin-15/genetics , Large Neutral Amino Acid-Transporter 1/genetics , Lymphocyte Activation , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Receptors, Virus/genetics , Transcription Factors/genetics , Transcriptional Activation
16.
Immun Inflamm Dis ; 8(1): 8-23, 2020 03.
Article in English | MEDLINE | ID: mdl-31742928

ABSTRACT

INTRODUCTION: Innate lymphoid cells (ILCs) can provide early cytokine help against a variety of pathogens in the lungs and gastrointestinal tract. Type 2 ILC (ILC2) are comparable to T helper 2 cells found in the adaptive immune system, which secrete cytokines such as interleukin 5 (IL-5) and IL-13 and have been found to play roles in host defense against helminth infections and in allergic responses. Recent studies have identified that programmed cell death protein 1 (PD-1) and peroxisome proliferator activated receptor-γ (PPAR-γ) are highly expressed by ILC2. We examined whether PD-1 plays a role in ILC2 function and whether there was any connection between PD-1 and PPAR-γ METHODS: To ensure that only innate immune cells were present, ILC2 cells were examined from RAG1-/- and PD-1-/- xRAG1-/- mice under steady-state or following inoculation with IL-33. We also tested ILC2 generated from bone marrow of RAG1-/- and PD-1-/- xRAG1-/- mice for their production of cytokines. These in vitro-derived ILC2 were also exposed to agonist and antagonist of PPAR-γ. RESULTS: We found that ILC2 from PD-1-/- xRAG1-/- mice had reduced frequencies of IL-5 and IL-13 producing cells both in vitro upon IL-33 stimulation and in vivo following intraperitoneal administration of IL-33 when compared with ILC2 from RAG1-/- mice. However, by adding IL-2, IL-25, and thymic stromal lymphopoietin to the in vitro cultures, the frequency of IL-5 and IL-13 expressing ILC2 from PD-1-/- xRAG1-/- mice became similar to the frequency observed for ILC2 from RAG1-/- mice. In addition, PPAR-γ agonists and antagonists were found to increase and decrease PD-1 expression on ILC2 respectively. CONCLUSIONS: These findings illustrate that chronic loss of PD-1 plays a role in ILC2 function and PD-1 expression can be modulated by PPAR-γ.


Subject(s)
Cytokines/metabolism , Immunity, Innate , Lymphocytes/cytology , Lymphoid Progenitor Cells/cytology , PPAR gamma/metabolism , Programmed Cell Death 1 Receptor/metabolism , Animals , Homeodomain Proteins/genetics , Hypersensitivity/immunology , Lymphocytes/metabolism , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Th2 Cells/immunology
17.
Sci Rep ; 9(1): 16362, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31704965

ABSTRACT

The contribution of natural killer (NK) cells to the clearance of hepatic viral infections is well recognized. The recently discovered heterogeneity of NK cell populations renders them interesting targets for immune interventions. Invariant natural killer T (iNKT) cells represent a key interaction partner for hepatic NK cells. The present study addressed whether characteristics of NK cells in the liver can be shaped by targeting iNKT cells. For this, the CD1d-binding pegylated glycolipid αGalCerMPEG was assessed for its ability to modulate the features of NK cells permanently or transiently residing in the liver. In vivo administration resulted in enhanced functionality of educated and highly differentiated CD27+ Mac-1+ NK cells accompanied by an increased proliferation. Improved liver homing was supported by serum-derived and cellular factors. Reduced viral loads in a mCMV infection model confirmed the beneficial effect of NK cells located in the liver upon stimulation with αGalCerMPEG. Thus, targeting iNKT cell-mediated NK cell activation in the liver represents a promising approach for the establishment of liver-directed immune interventions.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Killer Cells, Natural/immunology , Liver/immunology , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/metabolism , Cell Movement , Cell Proliferation , Cytomegalovirus/drug effects , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Galactosylceramides/pharmacology , Glycolipids/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Polyethylene Glycols/chemistry
18.
Front Immunol ; 10: 1849, 2019.
Article in English | MEDLINE | ID: mdl-31440243

ABSTRACT

Non-NK group 1 innate lymphoid cells (ILC1s), mainly investigated in the mucosal areas of the intestine, are well-known to contribute to anti-parasitic and anti-bacterial immune responses. Recently, our group revealed that lung ILC1s become activated during murine influenza infection, thereby contributing to viral clearance. In this context, worldwide seasonal influenza infections often result in severe disease outbreaks leading to high morbidity and mortality. Therefore, new immune interventions are urgently needed. In contrast to NK cells, the potential of non-NK ILC1s to become functionally tailored by immune modulators to contribute to the combat against mucosal-transmitted viral pathogens has not yet been addressed. The present study aimed at assessing the potential of ILC1s to become modulated by iNKT cells activated through the CD1d agonist αGalCerMPEG. Our results demonstrate an improved functional responsiveness of murine lung and splenic ILC1s following iNKT cell stimulation by the mucosal route, as demonstrated by enhanced surface expression of TNF-related apoptosis-inducing ligand (TRAIL), CD49a and CD28, and increased secretion of IFNγ. Interestingly, iNKT cell stimulation also induced the expression of the immune checkpoint molecules GITR and CTLA-4, which represent crucial points of action for immune regulation. An in vivo influenza infection model revealed that intranasal activation of ILC1s by αGalCerMPEG contributed to increased viral clearance as shown by reduced viral loads in the lungs. The findings that ILC1s can become modulated by mucosally activated iNKT cells in a beneficial manner emphasize their up to now underestimated potential and renders them to be considered as targets for novel immune interventions.


Subject(s)
Immunity, Mucosal , Influenza A virus/immunology , Natural Killer T-Cells/immunology , Orthomyxoviridae Infections/immunology , Animals , Antigens, Differentiation/immunology , Female , Mice , Mice, Knockout , Natural Killer T-Cells/pathology , Orthomyxoviridae Infections/pathology
19.
Cancer Immunol Res ; 6(11): 1417-1425, 2018 11.
Article in English | MEDLINE | ID: mdl-30143536

ABSTRACT

Metastatic breast cancer is a fatal disease that responds poorly to treatment. Cancer vaccines targeting antigens expressed by metastatic breast cancer cells and cancer stem cells could function as anticancer therapies. Cripto-1 is an oncofetal protein overexpressed in invasive breast cancer and cancer-initiating cells. In this study, we explored the potential of a Cripto-1-encoding DNA vaccine to target breast cancer in preclinical mouse models. BALB/c mice and BALB-neuT mice were treated with a DNA vaccine encoding mouse Cripto-1 (mCr-1). BALB/c mice were challenged with murine breast cancer 4T1 cells or TUBO spheres; BALB-neuT mice spontaneously developed breast cancer. Tumor growth was followed in all mouse models and lung metastases were evaluated. In vitro assays were performed to identify the immune response elicited by vaccination. Vaccination against mCr-1 reduced primary tumor growth in the 4T1 metastatic breast cancer model and reduced lung metastatic burden. In BALB-neuT mice, because the primary tumors are Cripto-1 negative, vaccination against mCr-1 did not affect primary tumors but did reduce lung metastatic burden. Spheroid-cultured TUBO cells, derived from a BALB/neuT primary tumor, develop a cancer stem cell-like phenotype and express mCr-1. We observed reduced tumor growth in vaccinated mice after challenge with TUBO spheres. Our data indicate that vaccination against Cripto-1 results in a protective immune response against mCr-1 expressing and metastasizing cells. Targeting Cripto-1 by vaccination holds promise as an immunotherapy for treatment of metastatic breast cancer. Cancer Immunol Res; 6(11); 1417-25. ©2018 AACR.


Subject(s)
Cancer Vaccines/pharmacology , Epidermal Growth Factor/genetics , Mammary Neoplasms, Experimental/therapy , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Vaccines, DNA/pharmacology , Animals , Cell Line, Tumor , Epidermal Growth Factor/immunology , Female , Immunity, Humoral , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Membrane Glycoproteins/immunology , Mice, Inbred BALB C , Neoplasm Proteins/immunology , Neoplastic Stem Cells/immunology
20.
Front Immunol ; 9: 505, 2018.
Article in English | MEDLINE | ID: mdl-29623077

ABSTRACT

Innate lymphoid cells (ILCs) represent diversified subsets of effector cells as well as immune regulators of mucosal immunity and are classified into group 1 ILCs, group 2 ILCs, and group 3 ILCs. Group 1 ILCs encompass natural killer (NK) cells and non-NK ILCs (ILC1s) and mediate their functionality via the rapid production of IFN-γ and TNF-α. The current knowledge of ILC1s mainly associates them to inflammatory processes. Much less is known about their regulation during infection and their capacity to interact with cells of the adaptive immune system. The present study dissected the role of ILC1s during early influenza A virus infection, thereby revealing their impact on the antiviral response. Exploiting in vitro and in vivo H1N1 infection systems, a cross-talk of ILC1s with cells of the innate and the adaptive immunity was demonstrated, which contributes to anti-influenza immunity. A novel association of ILC1 functionality and the expression of the glucocorticoid-induced TNFR-related protein (GITR) was observed, which hints toward a so far undescribed role of GITR in regulating ILC1 responsiveness. Overexpression of GITR inhibits IFN-γ production by ILC1s, whereas partial reduction of GITR expression can reverse this effect, thereby regulating ILC1 functionality. These new insights into ILC1 biology define potential intervention targets to modulate the functional properties of ILC1s, thus contributing toward the development of new immune interventions against influenza.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/immunology , Influenza A Virus, H1N1 Subtype , Lymphocytes/immunology , Orthomyxoviridae Infections/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Coculture Techniques , DNA-Binding Proteins/genetics , Dendritic Cells/immunology , Female , Immunity, Innate , Interleukin-12/immunology , Interleukin-18/immunology , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...