Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Immunity ; 43(5): 945-58, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26588780

ABSTRACT

Epithelial cells orchestrate pulmonary homeostasis and pathogen defense and play a crucial role in the initiation of allergic immune responses. Maintaining the balance between homeostasis and inappropriate immune activation and associated pathology is particularly complex at mucosal sites that are exposed to billions of potentially antigenic particles daily. We demonstrated that epithelial cell-derived cytokine TGF-ß had a central role in the generation of the pulmonary immune response. Mice that specifically lacked epithelial cell-derived TGF-ß1 displayed a reduction in type 2 innate lymphoid cells (ILCs), resulting in suppression of interleukin-13 and hallmark features of the allergic response including airway hyperreactivity. ILCs in the airway lumen were primed to respond to TGF-ß by expressing the receptor TGF-ßRII and ILC chemoactivity was enhanced by TGF-ß. These data demonstrate that resident epithelial cells instruct immune cells, highlighting the central role of the local environmental niche in defining the nature and magnitude of immune reactions.


Subject(s)
Epithelial Cells/immunology , Immunity, Innate/immunology , Lung/immunology , Lymphocytes/immunology , Transforming Growth Factor beta1/immunology , Animals , Cells, Cultured , Interleukin-13/immunology , Mice , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/immunology , Respiratory Hypersensitivity/immunology
2.
Am J Respir Cell Mol Biol ; 60(3): 259-268, 2019 03.
Article in English | MEDLINE | ID: mdl-30372120

ABSTRACT

The human lung is constantly exposed to the environment and potential pathogens. As the interface between host and environment, the respiratory epithelium has evolved sophisticated sensing mechanisms as part of its defense against pathogens. In this review, we examine how the respiratory epithelium senses and responds to influenza A virus, the biggest cause of respiratory viral deaths worldwide.


Subject(s)
Influenza A virus/immunology , Influenza, Human/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Humans , Influenza, Human/virology , Lung/immunology , Lung/virology
3.
Eur Respir J ; 54(2)2019 08.
Article in English | MEDLINE | ID: mdl-31164437

ABSTRACT

Children with severe therapy-resistant asthma (STRA) have poor control despite maximal treatment, while those with difficult asthma (DA) have poor control from failure to implement basic management, including adherence to therapy. Although recognised as clinically distinct, the airway molecular phenotype, including the role of innate lymphoid cells (ILCs) and their response to steroids in DA and STRA is unknown.Immunophenotyping of sputum and blood ILCs and T-cells from STRA, DA and non-asthmatic controls was undertaken. Leukocytes were analysed longitudinally pre- and post-intramuscular triamcinolone in children with STRA. Cultured ILCs were evaluated to assess steroid responsiveness in vitroAirway eosinophils, type 2 T-helper (Th2) cells and ILC2s were significantly higher in STRA patients compared to DA and disease controls, while IL-17+ lymphoid cells were similar. ILC2s and Th2 cells were significantly reduced in vivo following intramuscular triamcinolone and in vitro with steroids. Furthermore, asthma attacks and symptoms reduced after systemic steroids despite persistence of steroid-resistant IL-17+ cells and eosinophils.Paediatric STRA and DA have distinct airway molecular phenotypes with STRA characterised by elevated type-2 cells. Systemic corticosteroids, but not maintenance inhaled steroids resulted in improved symptom control and exacerbations concomitant with a reduction in functional ILC2s despite persistently elevated IL-17+ lymphoid cells.


Subject(s)
Asthma/physiopathology , Lymphocytes/immunology , Steroids/therapeutic use , Th2 Cells/immunology , Adolescent , Asthma/therapy , Child , Eosinophils/immunology , Female , Humans , Immunity, Innate , Immunophenotyping , Interleukin-13/metabolism , Interleukin-17/metabolism , Leukocytes/immunology , Leukocytes, Mononuclear/immunology , Lung , Male , Pediatrics , Phenotype , Th17 Cells/immunology , Triamcinolone/therapeutic use
4.
J Allergy Clin Immunol ; 139(5): 1496-1507.e3, 2017 May.
Article in English | MEDLINE | ID: mdl-27623174

ABSTRACT

BACKGROUND: Genome-wide association studies have identified the ORM (yeast)-like protein isoform 3 (ORMDL3) gene locus on human chromosome 17q to be a highly significant risk factor for childhood-onset asthma. OBJECTIVE: We sought to investigate in vivo the functional role of ORMDL3 in disease inception. METHODS: An Ormdl3-deficient mouse was generated and the role of ORMDL3 in the generation of allergic airways disease to the fungal aeroallergen Alternaria alternata was determined. An adeno-associated viral vector was also used to reconstitute ORMDL3 expression in airway epithelial cells of Ormdl3 knockout mice. RESULTS: Ormdl3 knockout mice were found to be protected from developing allergic airways disease and showed a marked decrease in pathophysiology, including lung function and airway eosinophilia induced by Alternaria. Alternaria is a potent inducer of cellular stress and the unfolded protein response, and ORMDL3 was found to play a critical role in driving the activating transcription factor 6-mediated arm of this response through Xbp1 and downstream activation of the endoplasmic reticulum-associated degradation pathway. In addition, ORMDL3 mediated uric acid release, another marker of cellular stress. In the knockout mice, reconstitution of Ormdl3 transcript levels specifically in the bronchial epithelium resulted in reinstatement of susceptibility to fungal allergen-induced allergic airways disease. CONCLUSIONS: This study demonstrates that ORMDL3, an asthma susceptibility gene identified by genome-wide association studies, contributes to key pathways that promote changes in airway physiology during allergic immune responses.


Subject(s)
Asthma/immunology , Membrane Proteins/immunology , Allergens/immunology , Alternaria/immunology , Animals , Asthma/physiopathology , Disease Models, Animal , Lung/immunology , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Respiratory Mucosa/immunology , Uric Acid/metabolism
6.
J Immunol ; 189(2): 551-7, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22685310

ABSTRACT

Neuropathology in multiple sclerosis is closely linked to presence of macrophages in the CNS. Both M1 (inflammatory) and M2 (alternatively activated, noninflammatory) macrophages are found in the inflamed CNS and thought to differentiate from infiltrating monocytes. It is unclear whether the balance of M1 and M2 macrophages can be altered and whether this affects disease outcome. We show in this article that Ly6C(hi) inflammatory monocytes are the early and dominant infiltrating cells in the CNS during experimental autoimmune encephalomyelitis, a model for the acute phase of multiple sclerosis. Activation of invariant NKT (iNKT) cells reduced the frequency of Ly6C(hi) monocytes and increased the proportion of M2 macrophages in the CNS with associated improvement in neurologic impairment. In contrast, iNKT-deficient mice showed higher numbers of Ly6C(hi) monocytes, reduced M2, and much more severe disease. Adoptive transfer of M2-enriched cells to iNKT-deficient mice markedly improved neurologic impairment. In vitro and in vivo experiments showed that iNKT cells promote differentiation of monocytes to M2 macrophages in an IL-4 and CD1d-dependent process. These findings indicate that infiltrating Ly6C(hi) inflammatory monocytes are early players in acute neuroinflammation and that their frequency and differentiation can be influenced by activation of iNKT cells with resultant improvement in disease outcome.


Subject(s)
Antigens, Ly/biosynthesis , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Inflammation Mediators/physiology , Lymphocyte Activation/immunology , Macrophages/immunology , Monocytes/immunology , Natural Killer T-Cells/immunology , Acute Disease , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Movement/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/pathology
7.
J Allergy Clin Immunol ; 132(3): 676-685.e13, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23759184

ABSTRACT

BACKGROUND: TH2 cytokines are not responsible for the ongoing symptoms and pathology in children with severe therapy-resistant asthma (STRA). IL-33 induces airway hyperresponsiveness, but its role in airway remodeling and steroid resistance is unknown. OBJECTIVE: We sought to investigate the relationship between IL-33 and airway remodeling in pediatric patients with STRA. METHODS: IL-33 levels were quantified in neonatal mice given inhaled house dust mite (HDM), and the effect of blocking IL-13 on remodeling and IL-33 levels was assessed. HDM-induced allergic airways disease (AAD) in neonatal ST2(-/-) mice lacking the IL-33 receptor was assessed, together with collagen production after IL-33 administration. The effect of steroid therapy on IL-33 levels in patients with neonatal AAD was explored. IL-33 expression was quantified in endobronchial biopsy (EB) specimens from children with STRA and related to remodeling, and collagen production by airway fibroblasts from pediatric patients stimulated with IL-33 and budesonide was quantified. RESULTS: Blocking IL-13 after AAD was established in neonatal mice and did not reduce remodeling or IL-33 levels; airway hyperresponsiveness was only partially reduced. IL-33 promoted collagen synthesis both from asthmatic fibroblasts from pediatric patients and after intranasal administration in mice. Increased cellular expression of IL-33, but not IL-13, was associated with increased reticular basement membrane thickness in EB specimens from children with STRA, whereas remodeling was absent in HDM-exposed ST2(-/-) mice. IL-33 levels were maintained, whereas IL-13 levels were abrogated by steroid treatment in neonatal HDM-exposed mice and in EB specimens from children with STRA. CONCLUSION: IL-33 is a relatively steroid-resistant mediator that promotes airway remodeling in patients with STRA and is an important therapeutic target.


Subject(s)
Airway Remodeling , Asthma/pathology , Bronchial Hyperreactivity/immunology , Interleukins/immunology , Adolescent , Airway Remodeling/physiology , Animals , Animals, Newborn , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Asthma/immunology , Bronchial Hyperreactivity/pathology , Budesonide/therapeutic use , Child , Collagen/immunology , Drug Resistance , Female , Glucocorticoids/therapeutic use , Humans , Interleukin-13/immunology , Interleukin-33 , Interleukins/administration & dosage , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Pyroglyphidae/immunology
8.
Am J Respir Crit Care Med ; 186(12): 1292-7, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23087026

ABSTRACT

RATIONALE: T-cell responses have been implicated in control and exacerbation of lung injury during influenza A virus (IAV) infection. OBJECTIVES: To examine the breadth and magnitude of influenza-specific CD4(+) and CD8(+) T-cell responses during acute phase of infection. METHODS: Influenza-specific T-cell response to the entire pandemic H1N1/09 IAV proteome and T cell-related cytokine levels were measured in blood from previously healthy individuals with mild (n = 32) and severe (n = 16) IAV infection during the 2009 influenza pandemic. Virus-specific T-cell response in lung and blood was also performed in two acutely infected, severely ill patients using fluorescent-conjugated pdmH1N1/09 Matrix-MHC-I tetrameric complexes. MEASUREMENTS AND MAIN RESULTS: Strong and broad CD4(+) but not CD8(+) T-cell responses were observed in the blood, and were higher in those with severe disease. Antigen-specific CD8(+) T cells in the lungs were on average 45-fold higher compared with blood in severely ill patients. Paradoxically, in patients with severe disease, IL-17, IL-2, IL-4, and IFN-γ levels were significantly decreased. CONCLUSIONS: High levels of circulating virus-specific CD4(+) T cells to two viral internal proteins (nucleoprotein and matrix) in the first phase of infection are associated with subsequent development of severe IAV infection. This finding could be an early and specific marker for ensuing clinical deterioration. Contrasting levels of antigen-specific CD8(+) T cells in lungs and blood have implications on design and analysis of clinical trials for T-cell vaccines because measurements of T cells in the periphery may not reflect events in the lungs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Adult , Biomarkers/blood , Bronchoalveolar Lavage , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/virology , China/epidemiology , Female , Humans , Influenza, Human/blood , Influenza, Human/epidemiology , Influenza, Human/virology , Male , Middle Aged , Pandemics , Severity of Illness Index
9.
JCI Insight ; 8(2)2023 Jan 24.
Article in English | MEDLINE | ID: mdl-36472908

ABSTRACT

Severe lung damage resulting from COVID-19 involves complex interactions between diverse populations of immune and stromal cells. In this study, we used a spatial transcriptomics approach to delineate the cells, pathways, and genes present across the spectrum of histopathological damage in COVID-19-affected lung tissue. We applied correlation network-based approaches to deconvolve gene expression data from 46 areas of interest covering more than 62,000 cells within well-preserved lung samples from 3 patients. Despite substantial interpatient heterogeneity, we discovered evidence for a common immune-cell signaling circuit in areas of severe tissue that involves crosstalk between cytotoxic lymphocytes and pro-inflammatory macrophages. Expression of IFNG by cytotoxic lymphocytes was associated with induction of chemokines, including CXCL9, CXCL10, and CXCL11, which are known to promote the recruitment of CXCR3+ immune cells. The TNF superfamily members BAFF (TNFSF13B) and TRAIL (TNFSF10) were consistently upregulated in the areas with severe tissue damage. We used published spatial and single-cell SARS-CoV-2 data sets to validate our findings in the lung tissue from additional cohorts of patients with COVID-19. The resulting model of severe COVID-19 immune-mediated tissue pathology may inform future therapeutic strategies.


Subject(s)
COVID-19 , Pneumonia , Humans , Transcriptome , SARS-CoV-2 , Lung
10.
Nat Commun ; 14(1): 7216, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37940670

ABSTRACT

Single cell spatial interrogation of the immune-structural interactions in COVID -19 lungs is challenging, mainly because of the marked cellular infiltrate and architecturally distorted microstructure. To address this, we develop a suite of mathematical tools to search for statistically significant co-locations amongst immune and structural cells identified using 37-plex imaging mass cytometry. This unbiased method reveals a cellular map interleaved with an inflammatory network of immature neutrophils, cytotoxic CD8 T cells, megakaryocytes and monocytes co-located with regenerating alveolar progenitors and endothelium. Of note, a highly active cluster of immature neutrophils and CD8 T cells, is found spatially linked with alveolar progenitor cells, and temporally with the diffuse alveolar damage stage. These findings offer further insights into how immune cells interact in the lungs of severe COVID-19 disease. We provide our pipeline [Spatial Omics Oxford Pipeline (SpOOx)] and visual-analytical tool, Multi-Dimensional Viewer (MDV) software, as a resource for spatial analysis.


Subject(s)
COVID-19 , Neutrophils , Humans , CD8-Positive T-Lymphocytes , Lung , T-Lymphocytes, Cytotoxic
12.
J Leukoc Biol ; 110(1): 107-114, 2021 07.
Article in English | MEDLINE | ID: mdl-33155728

ABSTRACT

Mϕs are the main innate immune cells in the lung at homeostasis, with important roles in host defence and immune modulation. Alveolar Mϕs (AMs) and interstitial Mϕs (IMs) are the two lung Mϕ subsets, so called according to the sites they reside in. These subsets are also defined by their origins and immunological microenvironment, which endow these cells with distinct features and plasticity. This review summarizes the latest definitions and functions of lung Mϕs during homeostasis and provides exemplar of their divergent roles in lung fibrosis.


Subject(s)
Immunity, Innate , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Animals , Cellular Microenvironment , Disease Susceptibility , Homeostasis , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/pathology , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
13.
Front Immunol ; 12: 623430, 2021.
Article in English | MEDLINE | ID: mdl-33746960

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is the most severe form of chronic lung fibrosis. Circulating monocytes have been implicated in immune pathology in IPF but their phenotype is unknown. In this work, we determined the immune phenotype of monocytes in IPF using multi-colour flow cytometry, RNA sequencing and corresponding serum factors, and mapped the main findings to amount of lung fibrosis and single cell transcriptomic landscape of myeloid cells in IPF lungs. We show that monocytes from IPF patients displayed increased expression of CD64 (FcγR1) which correlated with amount of lung fibrosis, and an amplified type I IFN response ex vivo. These were accompanied by markedly raised CSF-1 levels, IL-6, and CCL-2 in serum of IPF patients. Interrogation of single cell transcriptomic data from human IPF lungs revealed increased proportion of CD64hi monocytes and "transitional macrophages" with higher expression of CCL-2 and type I IFN genes. Our study shows that monocytes in IPF patients are phenotypically distinct from age-matched controls, with a primed type I IFN pathway that may contribute to driving chronic inflammation and fibrosis. These findings strengthen the potential role of monocytes in the pathogenesis of IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis/immunology , Interferon Type I/metabolism , Lung/immunology , Monocytes/immunology , Case-Control Studies , Cells, Cultured , Chemokine CCL2/blood , Flow Cytometry , Gene Expression Profiling , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Immunophenotyping , Interferon Type I/genetics , Interleukin-6/blood , Lung/metabolism , Lung/pathology , Macrophage Colony-Stimulating Factor/blood , Macrophages/immunology , Macrophages/metabolism , Monocytes/metabolism , Phenotype , Receptors, IgG/genetics , Receptors, IgG/metabolism , Single-Cell Analysis
14.
Sci Immunol ; 4(36)2019 06 07.
Article in English | MEDLINE | ID: mdl-31175176

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are enriched in mucosal tissues (e.g., lung) and respond to epithelial cell-derived cytokines initiating type 2 inflammation. During inflammation, ILC2 numbers are increased in the lung. However, the mechanisms controlling ILC2 trafficking and motility within inflamed lungs remain unclear and are crucial for understanding ILC2 function in pulmonary immunity. Using several approaches, including lung intravital microscopy, we demonstrate that pulmonary ILC2s are highly dynamic, exhibit amoeboid-like movement, and aggregate in the lung peribronchial and perivascular spaces. They express distinct chemokine receptors, including CCR8, and actively home to CCL8 deposits located around the airway epithelium. Within lung tissue, ILC2s were particularly motile in extracellular matrix-enriched regions. We show that collagen-I drives ILC2 to markedly change their morphology by remodeling their actin cytoskeleton to promote environmental exploration critical for regulating eosinophilic inflammation. Our study provides previously unappreciated insights into ILC2 migratory patterns during inflammation and highlights the importance of environmental guidance cues in the lung in controlling ILC2 dynamics.


Subject(s)
Lung/immunology , Lymphocytes/immunology , Animals , Cell Movement/drug effects , Collagen/immunology , Eosinophils/immunology , Extracellular Matrix/immunology , Female , Fibronectins/immunology , Humans , Immunity, Innate , Inflammation/immunology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Mice, Inbred BALB C , Mice, Transgenic , Recombinant Proteins/pharmacology
15.
Biomed J ; 41(4): 218-233, 2018 08.
Article in English | MEDLINE | ID: mdl-30348265

ABSTRACT

The respiratory epithelium is the major interface between the environment and the host. Sophisticated barrier, sensing, anti-microbial and immune regulatory mechanisms have evolved to help maintain homeostasis and to defend the lung against foreign substances and pathogens. During influenza virus infection, these specialised structural cells and populations of resident immune cells come together to mount the first response to the virus, one which would play a significant role in the immediate and long term outcome of the infection. In this review, we focus on the immune defence machinery of the respiratory epithelium and briefly explore how it repairs and regenerates after infection.


Subject(s)
Influenza, Human/immunology , Respiratory Mucosa/immunology , Cell Polarity , Chemokines/physiology , Cytokines/physiology , DEAD Box Protein 58/physiology , Humans , Interferons/physiology , NLR Proteins/physiology , Tight Junctions/physiology , Toll-Like Receptors/physiology
16.
Sci Immunol ; 3(27)2018 09 07.
Article in English | MEDLINE | ID: mdl-30194239

ABSTRACT

Airway hyperresponsiveness (AHR) is a critical feature of wheezing and asthma in children, but the initiating immune mechanisms remain unconfirmed. We demonstrate that both recombinant interleukin-33 (rIL-33) and allergen [house dust mite (HDM) or Alternaria alternata] exposure from day 3 of life resulted in significantly increased pulmonary IL-13+CD4+ T cells, which were indispensable for the development of AHR. In contrast, adult mice had a predominance of pulmonary LinnegCD45+CD90+IL-13+ type 2 innate lymphoid cells (ILC2s) after administration of rIL-33. HDM exposure of neonatal IL-33 knockout (KO) mice still resulted in AHR. However, neonatal CD4creIL-13 KO mice (lacking IL-13+CD4+ T cells) exposed to allergen from day 3 of life were protected from AHR despite persistent pulmonary eosinophilia, elevated IL-33 levels, and IL-13+ ILCs. Moreover, neonatal mice were protected from AHR when inhaled Acinetobacter lwoffii (an environmental bacterial isolate found in cattle farms, which is known to protect from childhood asthma) was administered concurrent with HDM. A. lwoffii blocked the expansion of pulmonary IL-13+CD4+ T cells, whereas IL-13+ ILCs and IL-33 remained elevated. Administration of A. lwoffii mirrored the findings from the CD4creIL-13 KO mice, providing a translational approach for disease protection in early life. These data demonstrate that IL-13+CD4+ T cells, rather than IL-13+ ILCs or IL-33, are critical for inception of allergic AHR in early life.


Subject(s)
Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-13/immunology , Respiratory Hypersensitivity/immunology , Acinetobacter/immunology , Alternaria/immunology , Animals , Animals, Newborn , Female , Interleukin-33/genetics , Male , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Pyroglyphidae/immunology
17.
Nat Commun ; 7: 11653, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27337592

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are abundant in humans and recognize bacterial ligands. Here, we demonstrate that MAIT cells are also activated during human viral infections in vivo. MAIT cells activation was observed during infection with dengue virus, hepatitis C virus and influenza virus. This activation-driving cytokine release and Granzyme B upregulation-is TCR-independent but dependent on IL-18 in synergy with IL-12, IL-15 and/or interferon-α/ß. IL-18 levels and MAIT cell activation correlate with disease severity in acute dengue infection. Furthermore, HCV treatment with interferon-α leads to specific MAIT cell activation in vivo in parallel with an enhanced therapeutic response. Moreover, TCR-independent activation of MAIT cells leads to a reduction of HCV replication in vitro mediated by IFN-γ. Together these data demonstrate MAIT cells are activated following viral infections, and suggest a potential role in both host defence and immunopathology.


Subject(s)
Lymphocyte Activation/physiology , Mucosal-Associated Invariant T Cells/physiology , Virus Diseases/immunology , Adult , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Female , Humans , Leukocytes, Mononuclear/physiology , Male
18.
J Leukoc Biol ; 91(3): 357-68, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22003207

ABSTRACT

Little is known of how a strong immune response in the lungs is regulated to minimize tissue injury during severe influenza A virus (IAV) infection. Here, using a model of lethal, high-pathogenicity IAV infection, we first show that Ly6C(hi)Ly6G(-) inflammatory monocytes, and not neutrophils, are the main infiltrate in lungs of WT mice. Mice devoid of iNKT cells (Jα18(-/-) mice) have increased levels of inflammatory monocytes, which correlated with increased lung injury and mortality (but not viral load). Activation of iNKT cells correlated with reduction of MCP-1 levels and improved outcome. iNKT cells were able to selectively lyse infected, MCP-1-producing monocytes in vitro, in a CD1d-dependent process. Our study provides a detailed profile and kinetics of innate immune cells in the lungs during severe IAV infection, highlighting inflammatory monocytes as the major infiltrate and identifying a role for iNKT cells in control of these cells and lung immune-pathology.


Subject(s)
Inflammation/immunology , Influenza A virus/immunology , Lung/immunology , Monocytes/immunology , Natural Killer T-Cells/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/immunology , Antigens, CD1d/metabolism , Antigens, Ly/metabolism , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Humans , Immunophenotyping , Inflammation/metabolism , Inflammation/pathology , Lung/metabolism , Lung/pathology , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/pathology , Monocytes/virology , Orthomyxoviridae Infections/mortality , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology
19.
PLoS One ; 5(5): e10675, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20502691

ABSTRACT

BACKGROUND: The cause of severe disease in some patients infected with pandemic influenza A virus is unclear. METHODOLOGY/PRINCIPAL FINDINGS: We present the cellular immunology profile in the blood, and detailed clinical (and post-mortem) findings of three patients with rapidly progressive infection, including a pregnant patient who died. The striking finding is of reduction in natural killer (NK) cells but preservation of activated effector CD8 T lymphocytes; with viraemia in the patient who had no NK cells. Comparison with control groups suggests that the reduction of NK cells is unique to these severely ill patients. CONCLUSION/SIGNIFICANCE: Our report shows markedly reduced NK cells in the three patients that we sampled and raises the hypothesis that NK may have a more significant role than T lymphocytes in controlling viral burden when the host is confronted with a new influenza A virus subtype.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , Killer Cells, Natural/immunology , Adult , CD8-Positive T-Lymphocytes/virology , Fatal Outcome , Female , Freezing , Health , Humans , Influenza, Human/blood , Influenza, Human/diagnostic imaging , Killer Cells, Natural/virology , Lung/immunology , Lung/pathology , Lung/virology , Male , Middle Aged , Postmortem Changes , Pregnancy , RNA, Viral/blood , Radiography, Thoracic , Young Adult
20.
Eur J Immunol ; 38(7): 1913-22, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18521958

ABSTRACT

Invariant NKT (iNKT) cells have an indubitable role in antiviral immunity, although the mechanisms by which these cells exert their functions are not fully elucidated. With the emerging importance of high-pathogenicity influenza A virus infections in humans, we questioned whether iNKT cells contribute to immune defence against influenza A virus and whether activation of these cells influences outcome. We show that activation of iNKT cells with alpha-galactosylceramide (alpha-GC) during influenza virus infection transiently enhanced early innate immune response without affecting T cell immunity, and reduced early viral titres in lungs of C57BL/6 mice. This is accompanied by a better disease course with improved weight loss profile. Temporal changes in iNKT cells in the liver, blood and lungs suggest activation and migration of iNKT cells from the liver to the lungs in mice that were administered alpha-GC. Improvement in viral titres appears dependent on activation of iNKT cells via the intraperitoneal route since intranasal administration of alpha-GC did not have the same effect. We conclude that activation of iNKT cells enhances early innate immune response in the lungs and contribute to antiviral immunity and improved disease course in influenza A virus infection.


Subject(s)
Cytokines/metabolism , Immunity, Innate , Inflammation Mediators/metabolism , Influenza A virus/immunology , Influenza, Human/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/virology , Cytokines/immunology , Disease Models, Animal , Galactosylceramides/immunology , Humans , Inflammation Mediators/immunology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/immunology , Influenza A virus/growth & development , Influenza, Human/virology , Liver/immunology , Lung/cytology , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL