Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 15(1): e1007560, 2019 01.
Article in English | MEDLINE | ID: mdl-30682165

ABSTRACT

Bacterial lung infections, particularly with methicillin-resistant Staphylococcus aureus (MRSA), increase mortality following influenza infection, but the mechanisms remain unclear. Here we show that expression of TLR9, a microbial DNA sensor, is increased in murine lung macrophages, dendritic cells, CD8+ T cells and epithelial cells post-influenza infection. TLR9-/- mice did not show differences in handling influenza nor MRSA infection alone. However, TLR9-/- mice have improved survival and bacterial clearance in the lung post-influenza and MRSA dual infection, with no difference in viral load during dual infection. We demonstrate that TLR9 is upregulated on macrophages even when they are not themselves infected, suggesting that TLR9 upregulation is related to soluble mediators. We rule out a role for elevations in interferon-γ (IFNγ) in mediating the beneficial MRSA clearance in TLR9-/- mice. While macrophages from WT and TLR9-/- mice show similar phagocytosis and bacterial killing to MRSA alone, following influenza infection, there is a marked upregulation of scavenger receptor A and MRSA phagocytosis as well as inducible nitric oxide synthase (Inos) and improved bacterial killing that is specific to TLR9-deficient cells. Bone marrow transplant chimera experiments and in vitro experiments using TLR9 antagonists suggest TLR9 expression on non-hematopoietic cells, rather than the macrophages themselves, is important for regulating myeloid cell function. Interestingly, improved bacterial clearance post-dual infection was restricted to MRSA, as there was no difference in the clearance of Streptococcus pneumoniae. Taken together these data show a surprising inhibitory role for TLR9 signaling in mediating clearance of MRSA that manifests following influenza infection.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/immunology , Methicillin-Resistant Staphylococcus aureus/metabolism , Toll-Like Receptor 9/metabolism , Animals , Humans , Influenza, Human/immunology , Lung/immunology , Macrophages , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Knockout , Orthomyxoviridae Infections/immunology , Phagocytosis , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Toll-Like Receptor 9/genetics
2.
Mucosal Immunol ; 12(2): 518-530, 2019 03.
Article in English | MEDLINE | ID: mdl-30498200

ABSTRACT

Post influenza bacterial pneumonia is associated with significant mortality and morbidity. Dendritic cells (DCs) play a crucial role in host defense against bacterial pneumonia, but their contribution to post influenza-susceptibility to secondary bacterial pneumonia is incompletely understood. WT and CCR2-/- mice were infected with 100 plaque forming units (pfu) H1N1 intranasally alone or were challenged on day 5 with 7 × 107 colony forming units (cfu) methicillin-resistant Staphylococcus aureus intratracheally. WT mice express abundant CCL2 mRNA and protein post-H1N1 alone or dual infection. CCR2-/- mice had significantly higher survival as compared to WT mice, associated with significantly improved bacterial clearance at 24 and 48 h (10-fold and 14-fold, respectively) post bacterial challenge. There was robust upregulation of IL-23 and IL-17 as well as downregulation of IL-27 expression in CCR2-/- mice following sequential infection as compared to WT mice, which was also associated with significantly greater accumulation of CD103+ DC. Finally, WT mice treated with a CCR2 inhibitor showed improved bacterial clearance in association with similar cytokine profiles as CCR2-/- mice. Thus, CCR2 significantly contributes to increased susceptibility to bacterial infection after influenza pneumonia likely via altered dendritic cell responses and thus, CCR2 antagonism represents a potential therapeutic strategy.


Subject(s)
Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Interleukin-17/metabolism , Orthomyxoviridae Infections/immunology , Pneumonia, Bacterial/immunology , Receptors, CCR2/metabolism , Th17 Cells/physiology , Animals , Antigens, CD/metabolism , Cells, Cultured , Disease Susceptibility , Humans , Influenza, Human/therapy , Integrin alpha Chains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neutrophils/immunology , Orthomyxoviridae Infections/therapy , Pneumonia, Bacterial/therapy , Receptors, CCR2/genetics
3.
Am J Physiol Lung Cell Mol Physiol ; 310(5): L465-75, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26589478

ABSTRACT

Postinfluenza bacterial pneumonia is associated with significant mortality and morbidity. MicroRNAs (miRNAs) are small, noncoding RNAs that regulate gene expression posttranscriptionally. miR-155 has recently emerged as a crucial regulator of innate immunity and inflammatory responses and is induced in macrophages during infection. We hypothesized upregulation of miR-155 inhibits IL-17 and increases susceptibility to secondary bacterial pneumonia. Mice were challenged with 100 plaque-forming units H1N1 intranasally and were infected with 10(7) colony-forming units of MRSA intratracheally at day 5 postviral challenge. Lungs were harvested 24 h later, and expression of miR-155, IL-17, and IL-23 was measured by real-time RT-PCR. Induction of miR-155 was 3.6-fold higher in dual-infected lungs compared with single infection. miR-155(-/-) mice were protected with significantly lower (4-fold) bacterial burden and no differences in viral load, associated with robust induction of IL-23 and IL-17 (2.2- and 4.8-fold, respectively) postsequential challenge with virus and bacteria, compared with WT mice. Treatment with miR-155 antagomir improved lung bacterial clearance by 4.2-fold compared with control antagomir postsequential infection with virus and bacteria. Moreover, lung macrophages collected from patients with postviral bacterial pneumonia also had upregulation of miR-155 expression compared with healthy controls, consistent with observations in our murine model. This is the first demonstration that cellular miRNAs regulate postinfluenza immune response to subsequent bacterial challenge by suppressing the IL-17 pathway in the lung. Our findings suggest that antagonizing certain microRNA might serve as a potential therapeutic strategy against secondary bacterial infection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , MicroRNAs/genetics , Pneumonia, Bacterial/microbiology , Animals , Humans , Immunity, Innate/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Lung/immunology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/microbiology , Orthomyxoviridae Infections/virology , Pneumonia, Bacterial/genetics
4.
PLoS One ; 10(1): e0114574, 2015.
Article in English | MEDLINE | ID: mdl-25635685

ABSTRACT

INTRODUCTION: Post influenza pneumonia is a leading cause of mortality and morbidity, with mortality rates approaching 60% when bacterial infections are secondary to multi-drug resistant (MDR) pathogens. Staphylococcus aureus, in particular community acquired MRSA (cMRSA), has emerged as a leading cause of post influenza pneumonia. HYPOTHESIS: Linezolid (LZD) prevents acute lung injury in murine model of post influenza bacterial pneumonia. METHODS: Mice were infected with HINI strain of influenza and then challenged with cMRSA at day 7, treated with antibiotics (LZD or Vanco) or vehicle 6 hours post bacterial challenge and lungs and bronchoalveolar lavage fluid (BAL) harvested at 24 hours for bacterial clearance, inflammatory cell influx, cytokine/chemokine analysis and assessment of lung injury. RESULTS: Mice treated with LZD or Vanco had lower bacterial burden in the lung and no systemic dissemination, as compared to the control (no antibiotic) group at 24 hours post bacterial challenge. As compared to animals receiving Vanco, LZD group had significantly lower numbers of neutrophils in the BAL (9×10(3) vs. 2.3×10(4), p < 0.01), which was associated with reduced levels of chemotactic chemokines and inflammatory cytokines KC, MIP-2, IFN-γ, TNF-α and IL-1ß in the BAL. Interestingly, LZD treatment also protected mice from lung injury, as assessed by albumin concentration in the BAL post treatment with H1N1 and cMRSA when compared to vanco treatment. Moreover, treatment with LZD was associated with significantly lower levels of PVL toxin in lungs. CONCLUSION: Linezolid has unique immunomodulatory effects on host inflammatory response and lung injury in a murine model of post-viral cMRSA pneumonia.


Subject(s)
Acetamides/administration & dosage , Influenza, Human/drug therapy , Methicillin-Resistant Staphylococcus aureus/drug effects , Oxazolidinones/administration & dosage , Pneumonia, Bacterial/drug therapy , Animals , Humans , Immunomodulation , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/complications , Linezolid , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/microbiology , Staphylococcal Infections/drug therapy
5.
J Immunol ; 194(4): 1894-904, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595781

ABSTRACT

Idiopathic pulmonary fibrosis is a devastating lung disease characterized by inflammation and the development of excessive extracellular matrix deposition. Currently, there are only limited therapeutic intervenes to offer patients diagnosed with pulmonary fibrosis. Although previous studies focused on structural cells in promoting fibrosis, our study assessed the contribution of macrophages. Recently, TLR signaling has been identified as a regulator of pulmonary fibrosis. IL-1R-associated kinase-M (IRAK-M), a MyD88-dependent inhibitor of TLR signaling, suppresses deleterious inflammation, but may paradoxically promote fibrogenesis. Mice deficient in IRAK-M (IRAK-M(-/-)) were protected against bleomycin-induced fibrosis and displayed diminished collagen deposition in association with reduced production of IL-13 compared with wild-type (WT) control mice. Bone marrow chimera experiments indicated that IRAK-M expression by bone marrow-derived cells, rather than structural cells, promoted fibrosis. After bleomycin, WT macrophages displayed an alternatively activated phenotype, whereas IRAK-M(-/-) macrophages displayed higher expression of classically activated macrophage markers. Using an in vitro coculture system, macrophages isolated from in vivo bleomycin-challenged WT, but not IRAK-M(-/-), mice promoted increased collagen and α-smooth muscle actin expression from lung fibroblasts in an IL-13-dependent fashion. Finally, IRAK-M expression is upregulated in peripheral blood cells from idiopathic pulmonary fibrosis patients and correlated with markers of alternative macrophage activation. These data indicate expression of IRAK-M skews lung macrophages toward an alternatively activated profibrotic phenotype, which promotes collagen production, leading to the progression of experimental pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-1 Receptor-Associated Kinases/metabolism , Macrophage Activation/physiology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Blotting, Western , Cell Separation , Coculture Techniques , Collagen , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Transcriptome
6.
J Immunol ; 190(1): 349-56, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23180821

ABSTRACT

Hypersensitivity pneumonitis (HP) is an inflammatory lung disease that develops after repeated exposure to inhaled particulate Ag. Stachybotrys chartarum is a dimorphic fungus that has been implicated in a number of respiratory illnesses, including HP. In this study, we have developed a murine model of S. chartarum-induced HP that reproduces pathology observed in human HP, and we have hypothesized that TLR9-mediated IL-23 and IL-17 responses are required for the generation of granulomatous inflammation induced by inhaled S. chartarum. Mice that undergo i.p. sensitization and intratracheal challenge with 10(6) S. chartarum spores developed granulomatous inflammation with multinucleate giant cells, accompanied by increased accumulation of T cells. S. chartarum sensitization and challenge resulted in robust pulmonary expression of IL-17 and IL-23. S. chartarum-mediated granulomatous inflammation required intact IL-23 or IL-17 responses and required TLR9, because TLR9(-/-) mice displayed reduced IL-17 and IL-23 expression in whole lung associated with decreased accumulation of IL-17 expressing CD4(+) and γδ T cells. Compared with S. chartarum-sensitized dendritic cells (DC) isolated from WT mice, DCs isolated from TLR9(-/-) mice had a reduced ability to produce IL-23 in responses to S. chartarum. Moreover, shRNA knockdown of IL-23 in DCs abolished IL-17 production from splenocytes in response to Ag challenge. Finally, the intratracheal reconstitution of IL-23 in TLR9(-/-) mice recapitulated the immunopathology observed in WT mice. In conclusion, our studies suggest that TLR9 is critical for the development of Th17-mediated granulomatous inflammation in the lung in response to S. chartarum.


Subject(s)
Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/pathology , Interleukin-17/physiology , Interleukin-23/physiology , Stachybotrys/immunology , Toll-Like Receptor 9/physiology , Alveolitis, Extrinsic Allergic/genetics , Animals , Antigens, Fungal/administration & dosage , Antigens, Fungal/immunology , Disease Models, Animal , Humans , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukin-23/administration & dosage , Interleukin-23/genetics , Intubation, Intratracheal , Mice , Mice, Inbred BALB C , Mice, Knockout , Recombinant Proteins/administration & dosage , Stachybotrys/pathogenicity , Toll-Like Receptor 9/deficiency
7.
J Immunol ; 189(1): 356-64, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22661086

ABSTRACT

Mechanical ventilation using high oxygen tensions is often necessary to treat patients with respiratory failure. Recently, TLRs were identified as regulators of noninfectious oxidative lung injury. IRAK-M is an inhibitor of MyD88-dependent TLR signaling. Exposure of mice deficient in IRAK-M (IRAK-M(-/-)) to 95% oxygen resulted in reduced mortality compared with wild-type mice and occurred in association with decreased alveolar permeability and cell death. Using a bone marrow chimera model, we determined that IRAK-M's effects were mediated by structural cells rather than bone marrow-derived cells. We confirmed the expression of IRAK-M in alveolar epithelial cells (AECs) and showed that hyperoxia can induce the expression of this protein. In addition, IRAK-M(-/-) AECs exposed to hyperoxia experienced a decrease in cell death. IRAK-M may potentiate hyperoxic injury by suppression of key antioxidant pathways, because lungs and AECs isolated from IRAK-M(-/-) mice have increased expression/activity of heme oxygenase-1, a phase II antioxidant, and NF (erythroid-derived)-related factor-2, a transcription factor that initiates antioxidant generation. Treatment of IRAK-M(-/-) mice in vivo and IRAK-M(-/-) AECs in vitro with the heme oxygenase-1 inhibitor, tin protoporphyrin, substantially decreased survival and significantly reduced the number of live cells after hyperoxia exposure. Collectively, our data suggest that IRAK-M inhibits the induction of antioxidants essential for protecting the lungs against cell death, resulting in enhanced susceptibility to hyperoxic lung injury.


Subject(s)
Acute Lung Injury/immunology , Hyperoxia/immunology , Oxidants/physiology , Pulmonary Alveoli/immunology , Respiratory Mucosa/immunology , Signal Transduction/immunology , Toll-Like Receptors/physiology , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Cell Death/genetics , Cell Death/immunology , Cell Line , Hyperoxia/pathology , Hyperoxia/prevention & control , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/physiology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction/genetics , Toll-Like Receptors/antagonists & inhibitors
8.
J Immunol ; 189(1): 304-11, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22634613

ABSTRACT

Cathelicidins are a family of endogenous antimicrobial peptides that exert diverse immune functions, including both direct bacterial killing and immunomodulatory effects. In this study, we examined the contribution of the murine cathelicidin, cathelicidin-related antimicrobial peptide (CRAMP), to innate mucosal immunity in a mouse model of Gram-negative pneumonia. CRAMP expression is induced in the lung in response to infection with Klebsiella pneumoniae. Mice deficient in the gene encoding CRAMP (Cnlp(-/-)) demonstrate impaired lung bacterial clearance, increased bacterial dissemination, and reduced survival in response to intratracheal K. pneumoniae administration. Neutrophil influx into the alveolar space during K. pneumoniae infection was delayed early but increased by 48 h in CRAMP-deficient mice, which was associated with enhanced expression of inflammatory cytokines and increased lung injury. Bone marrow chimera experiments indicated that CRAMP derived from bone marrow cells rather than structural cells was responsible for antimicrobial effects in the lung. Additionally, CRAMP exerted bactericidal activity against K. pneumoniae in vitro. Similar defects in lung bacterial clearance and delayed early neutrophil influx were observed in CRAMP-deficient mice infected with Pseudomonas aeruginosa, although this did not result in increased bacterial dissemination, increased lung injury, or changes in lethality. Taken together, our findings demonstrate that CRAMP is an important contributor to effective host mucosal immunity in the lung in response to Gram-negative bacterial pneumonia.


Subject(s)
Cathelicidins/physiology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/prevention & control , Respiratory Mucosa/immunology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides , Cathelicidins/deficiency , Disease Models, Animal , Klebsiella pneumoniae/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Pneumonia, Bacterial/microbiology , Pseudomonas aeruginosa/immunology , Respiratory Mucosa/microbiology
9.
J Immunol ; 188(8): 3940-8, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22422883

ABSTRACT

We investigated mechanisms by which TLR9 signaling promoted the development of the protective response to Cryptococcus neoformans in mice with cryptococcal pneumonia. The afferent (week 1) and efferent (week 3) phase immune parameters were analyzed in the infected wild-type (TLR9(+/+)) and TLR-deficient (TLR9(-/-)) mice. TLR9 deletion diminished 1) accumulation and activation of CD11b(+) dendritic cells (DCs), 2) the induction of IFN-γ and CCR2 chemokines CCL7, CCL12, but not CCL2, at week 1, and 3) pulmonary accumulation and activation of the major effector cells CD4(+) and CD8(+) T cells, CD11b(+) lung DCs, and exudate macrophages at week 3. The significance of CCL7 induction downstream of TLR9 signaling was investigated by determining whether CCL7 reconstitution would improve immunological parameters in C. neoformans-infected TLR9(-/-) mice. Early reconstitution with CCL7 1) improved accumulation and activation of CD11b(+) DCs at week 1, 2) restored early IFN-γ production in the lungs, and 3) restored the accumulation of major effector cell subsets. CCL7 administration abolished the difference in lung fungal burdens between TLR9(+/+) and TLR9(-/-) mice at week 3; however, significant reduction of fungal burdens between PBS- and CCL7-treated mice has not been observed, suggesting that additional mechanism(s) apart from early CCL7 induction contribute to optimal fungal clearance in TLR9(+/+) mice. Collectively, we show that TLR9 signaling during the afferent phase contributes to the development of protective immunity by promoting the early induction of CCL7 and IFN-γ and the subsequent early recruitment and activation of DCs and additional effector cells in mice with cryptococcal pneumonia.


Subject(s)
Chemokine CCL7/immunology , Cryptococcosis/immunology , Cryptococcus neoformans , Lung/immunology , Pneumonia/immunology , Animals , CD11b Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Colony Count, Microbial , Cryptococcosis/complications , Cryptococcosis/microbiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Interferon-gamma/immunology , Lung/metabolism , Lung/microbiology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Monocyte Chemoattractant Proteins/immunology , Pneumonia/complications , Pneumonia/microbiology , Signal Transduction , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
10.
Am J Physiol Lung Cell Mol Physiol ; 302(5): L447-54, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22160309

ABSTRACT

Toll-like receptors (TLRs) are required for protective host defense against bacterial pathogens. However, the role of TLRs in regulating lung injury during Gram-negative bacterial pneumonia has not been thoroughly investigated. In this study, experiments were performed to evaluate the role of TLR4 in pulmonary responses against Klebsiella pneumoniae (Kp). Compared with wild-type (WT) (Balb/c) mice, mice with defective TLR4 signaling (TLR4(lps-d) mice) had substantially higher lung bacterial colony-forming units after intratracheal challenge with Kp, which was associated with considerably greater lung permeability and lung cell death. Reduced expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) mRNA and protein was noted in lungs and bronchoalveolar lavage fluid of TLR4 mutant mice postintratracheal Kp compared with WT mice, and primary alveolar epithelial cells (AEC) harvested from TLR4(lps-d) mice produced significantly less GM-CSF in vitro in response to heat-killed Kp compared with WT AEC. TLR4(lps-d) AEC underwent significantly more apoptosis in response to heat-killed Kp in vitro, and treatment with GM-CSF protected these cells from apoptosis in response to Kp. Finally, intratracheal administration of GM-CSF in TLR4(lps-d) mice significantly decreased albumin leak, lung cell apoptosis, and bacteremia in Kp-infected mice. Based on these observations, we conclude that TLR4 plays a protective role on lung epithelium during Gram-negative bacterial pneumonia, an effect that is partially mediated by GM-CSF.


Subject(s)
Acute Lung Injury/microbiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Klebsiella Infections/microbiology , Klebsiella pneumoniae , Pneumonia, Bacterial/microbiology , Toll-Like Receptor 4/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/prevention & control , Animals , Apoptosis , Bacterial Load , Bronchoalveolar Lavage Fluid/microbiology , Cells, Cultured , Cytoprotection , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Klebsiella Infections/drug therapy , Klebsiella Infections/metabolism , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiopathology , Time-Lapse Imaging , Toll-Like Receptor 4/genetics
11.
Am J Pathol ; 179(6): 2779-87, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21982832

ABSTRACT

Hypersensitivity pneumonitis (HP), an inflammatory lung disease, develops after repeated exposure to inhaled particulate antigen and is characterized by a vigorous T helper type 1-mediated immune response, resulting in the release of IL-12 and interferon (IFN)-γ. These T helper type 1 cytokines may participate in the pathogenesis of HP. Stachybotrys chartarum (SC) is a dimorphic fungus implicated in a number of respiratory illnesses, including HP. Here, we have developed a murine model of SC-induced HP that reproduces pathology observed in human HP and hypothesized that toll receptor-like 9 (TLR9)-mediated dendritic cell responses are required for the generation of granulomatous inflammation induced by inhaled SC. Mice sensitized and challenged with 10(6) SC spores develop granulomatous inflammation with multinucleate giant cells, accompanied by increased accumulation of neutrophils and CD4(+) and CD8(+) T cells. SC sensitization and challenge resulted in robust pulmonary expression of tumor necrosis factor-α, IL-12, and IFN-γ. SC-mediated granulomatous inflammation required IFN-γ and was TLR9 dependent, because TLR9(-/-) mice displayed reduced peribronchial inflammation, decreased accumulation and/or activation of polymorphonuclear (PMN) and CD4(+) and CD8(+) T cells, and reduced lung expression of type 1 cytokines and chemokines. T-cell production of IFN-γ was IL-12 dependent. Our studies suggest that TLR9 is critical for dendritic cell-mediated development of a type 1 granulomatous inflammation in the lung in response to SC.


Subject(s)
Alveolitis, Extrinsic Allergic/microbiology , Cytokines/metabolism , Lung Diseases, Fungal/immunology , Stachybotrys/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 9/immunology , Alveolitis, Extrinsic Allergic/immunology , Animals , Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/immunology , Granuloma, Respiratory Tract/immunology , Immunization/methods , Interferon-gamma/metabolism , Interleukin-12/biosynthesis , Lymphocyte Activation/immunology , Macrophages, Alveolar/immunology , Mice , Mice, Inbred BALB C , Signal Transduction , T-Lymphocytes/metabolism
12.
Fibrogenesis Tissue Repair ; 4: 18, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21810214

ABSTRACT

BACKGROUND: We have shown previously that murine gammaherpesvirus 68 (γHV68) infection exacerbates established pulmonary fibrosis. Because Toll-like receptor (TLR)-9 may be important in controlling the immune response to γHV68 infection, we examined how TLR-9 signaling effects exacerbation of fibrosis in response to viral infection, using models of bleomycin- and fluorescein isothiocyanate-induced pulmonary fibrosis in wild-type (Balb/c) and TLR-9-/- mice. RESULTS: We found that in the absence of TLR-9 signaling, there was a significant increase in collagen deposition following viral exacerbation of fibrosis. This was not associated with increased viral load in TLR-9-/- mice or with major alterations in T helper (Th)1 and Th2 cytokines. We examined alveolar epithelial-cell apoptosis in both strains, but this could not explain the altered fibrotic outcomes. As expected, TLR-9-/- mice had a defect in the production of interferon (IFN)-ß after viral infection. Balb/c fibroblasts infected with γHV68 in vitro produced more IFN-ß than did infected TLR-9-/- fibroblasts. Accordingly, in vitro infection of Balb/c fibroblasts resulted in reduced proliferation rates whereas infection of TLR-9-/- fibroblasts did not. Finally, therapeutic administration of CpG oligodeoxynucleotides ameliorated bleomycin-induced fibrosis in wild-type mice. CONCLUSIONS: These results show a protective role for TLR-9 signaling in murine models of lung fibrosis, and highlight differences in the biology of TLR-9 between mice and humans.

13.
Am J Pathol ; 177(2): 754-65, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20581055

ABSTRACT

To determine whether TLR9 signaling contributes to the development of the adaptive immune response to cryptococcal infection, wild-type (TLR9+/+) and TLR9 knockout (TLR9-/-) BALB/c mice were infected intratracheally with 10(4) C. neoformans 52D. We evaluated 1) organ microbial burdens, 2) pulmonary leukocyte recruitment, 3) pulmonary and systemic cytokine induction, and 4) macrophage activation profiles. TLR9 deletion did not affect pulmonary growth during the innate phase, but profoundly impaired pulmonary clearance during the adaptive phase of the immune response (a 1000-fold difference at week 6). The impaired clearance in TLR9-/- mice was associated with: 1) significantly reduced CD4(+), CD8+ T cell, and CD19+ B cell recruitment into the lungs; 2) defects in Th polarization indicated by altered cytokine responses in the lungs, lymphonodes, and spleen; and 3) diminished macrophage accumulation and altered activation profile, including robust up-regulation of Arg1 and FIZZ1 (indicators of alternative activation) and diminished induction of inducible nitric oxide synthase (an indicator of classical activation). Histological analysis revealed defects in granuloma formation and increased numbers of intracellular yeast residing within macrophages in the lungs of TLR9-/- mice. We conclude that TLR9 signaling plays an important role in the development of robust protective immunity, proper recruitment and function of effector cells (lymphocytes and macrophages), and, ultimately, effective cryptococcal clearance from the infected lungs.


Subject(s)
Adaptive Immunity/immunology , Cryptococcosis/immunology , Cryptococcus neoformans/immunology , Lung Diseases , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , Animals , Cryptococcus neoformans/pathogenicity , Cytokines/immunology , Female , Humans , Lung Diseases/immunology , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Toll-Like Receptor 9/genetics
14.
PLoS One ; 5(6): e11145, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20585389

ABSTRACT

Sepsis results in a profound state of immunosuppression, which is temporally associated with impaired leukocyte function. The mechanism of leukocyte reprogramming in sepsis is incompletely understood. In this study, we explored mechanisms contributing to dysregulated inflammatory cytokine expression by pulmonary macrophages during experimental sepsis. Pulmonary macrophages (PM) recovered from the lungs of mice undergoing cecal ligation and puncture (CLP) display transiently reduced expression of some, but not all innate genes in response to LPS. Impaired expression of TNF-alpha and iNOS was associated with reduced acetylation and methylation of specific histones (AcH4 and H3K4me3) and reduced binding of RNA polymerase II to the promoters of these genes. Transient impairment in LPS-induced cytokine responses in septic PM temporally correlated with induction of IRAK-M mRNA and protein, which occurred in a MyD88-dependent fashion. PM isolated from IRAK-M(-/-) mice were largely refractory to CLP-induced impairment in cytokine expression, chromatin remodeling, recruitment of RNA polymerase II, and induction of histone deacetylase-2 observed during sepsis. Our findings indicate that systemic sepsis induces epigenetic silencing of cytokine gene expression in lung macrophages, and IRAK-M appears to be a critical mediator of this response.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Interleukin-1 Receptor-Associated Kinases/physiology , Lung/pathology , Macrophages/pathology , Sepsis/physiopathology , Animals , Base Sequence , Chromatin Immunoprecipitation , DNA Primers , Interleukin-1 Receptor-Associated Kinases/genetics , Mice , Mice, Knockout , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/pathology
15.
PLoS One ; 5(3): e9896, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20360853

ABSTRACT

Toll like receptors play an important role in lung host defense against bacterial pathogens. In this study, we investigated independent and cooperative functions of TLR4 and TLR9 in microbial clearance and systemic dissemination during Gram-negative bacterial pneumonia. To access these responses, wildtype Balb/c mice, mice with defective TLR4 signaling (TLR4(lps-d)), mice deficient in TLR9 (TLR9(-/-)) and TLR4/9 double mutant mice (TLR4(lps-d)/TLR9(-/-)) were challenged with K. pneumoniae, then time-dependent lung bacterial clearance and systemic dissemination determined. We found impaired lung bacterial clearance in TLR4 and TLR9 single mutant mice, whereas the greatest impairment in clearance was observed in TLR4(lps-d)/TLR9(-/-) double mutant mice. Early lung expression of TNF-alpha, IL-12, and chemokines was TLR4 dependent, while IFN-gamma production and the later expression of TNF-alpha and IL-12 was dependent on TLR9. Classical activation of lung macrophages and maximal induction of IL-23 and IL-17 required both TLR4 and TLR9. Finally, the i.t. instillation of IL-17 partially restored anti-bacterial immunity in TLR4(lps-d)/TLR9(-/-) double mutant mice. In conclusion, our studies indicate that TLR4 and TLR9 have both non-redundant and cooperative roles in lung innate responses during Gram-negative bacterial pneumonia and are both critical for IL-17 driven antibacterial host response.


Subject(s)
Gram-Negative Bacteria/genetics , Interleukin-17/genetics , Interleukin-23/genetics , Pneumonia, Bacterial/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 9/genetics , Animals , Bronchoalveolar Lavage Fluid , Female , Interleukin-12/biosynthesis , Interleukin-17/metabolism , Klebsiella pneumoniae/genetics , Lung/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Pneumonia, Bacterial/microbiology , Tumor Necrosis Factor-alpha/biosynthesis
16.
J Immunol ; 184(3): 1410-8, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042589

ABSTRACT

Influenza virus is a common cause of respiratory infection and morbidity, which is often due to deleterious host immune responses directed against the pathogen. We investigated the role of IL-1 receptor-associated kinase-M (IRAK-M), an inhibitor of MyD88-dependent TLR signaling, in modulating the innate inflammatory response during influenza pneumonia using a murine model. The intranasal administration of influenza resulted in the upregulation of IRAK-M mRNA and protein levels in the lungs within 2 d after infectious challenge. Pulmonary influenza infection in mice deficient in IRAK-M (IRAK-M(-/-)) resulted in substantially increased mortality compared with similarly treated wild-type animals. Increased mortality in IRAK-M(-/-) mice was associated with enhanced early influx of neutrophils, high permeability edema, apoptosis of lung epithelial cells, markedly increased expression of inflammatory cytokines/chemokines, and release of neutrophil-derived enzymes, including myeloperoxidase and neutrophil elastase. Early viral clearance was not different in mutant mice, whereas viral titers in lungs and blood were significantly higher in IRAK-M(-/-) mice compared with wild-type animals. Increased lethality observed in IRAK-M(-/-) mice after influenza challenge was abrogated by Ab-mediated blockade of CXCR2. Collectively, our findings indicate that IRAK-M is critical to preventing deleterious neutrophil-dependent lung injury during influenza infection of the respiratory tract.


Subject(s)
Inflammation Mediators/metabolism , Interleukin-1 Receptor-Associated Kinases/physiology , Orthomyxoviridae Infections/enzymology , Orthomyxoviridae Infections/immunology , Pneumonia, Viral/enzymology , Pneumonia, Viral/immunology , Receptors, Interleukin-8B/physiology , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Animals , Antibodies/administration & dosage , Antibodies/therapeutic use , Cell Line , Disease Models, Animal , Dogs , Inflammation Mediators/physiology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/pathogenicity , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/therapy , Pneumonia, Viral/pathology , Pneumonia, Viral/therapy , Receptors, Interleukin-8B/immunology
17.
Infect Immun ; 76(7): 2895-904, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18426877

ABSTRACT

Experiments were performed to determine the contribution of TLR9 to the generation of protective immunity against the intracellular respiratory bacterial pathogen Legionella pneumophila. In initial studies, we found that the intratracheal (i.t.) administration of L. pneumophila to mice deficient in TLR9 (TLR9(-/-)) resulted in significantly increased mortality, which was associated with an approximately 10-fold increase in the number of lung CFU compared to that of wild-type BALB/c mice. Intrapulmonary bacterial challenge in TLR9(-/-) mice resulted in the reduced accumulation of myeloid dendritic cells (DC) and activated CD4(+) T cells. Lung macrophages isolated from Legionella-infected TLR9(-/-) mice displayed the impaired internalization of bacteria and evidence of alternative rather than classical activation, as manifested by the markedly reduced expression of nitric oxide and type 1 cytokines, whereas the expression of Fizz-1 and arginase-1 was enhanced. The adoptive transfer of bone marrow-derived DC from syngeneic wild-type, but not TLR9(-/-), mice administered i.t. reconstituted anti-legionella immunity and restored the macrophage phenotype in TLR9(-/-) mice. Finally, the i.t., but not intraperitoneal, administration of the TLR9 agonist molecule CpG oligodeoxynucleotide stimulated protective immunity in Legionella-infected mice. In total, our findings indicate that TLR9 is required for effective innate immune responses against the intracellular bacterial pathogen L. pneumophila, and approaches to maximize TLR9-mediated responses may serve as a means to augment antibacterial immunity in pneumonia.


Subject(s)
Legionella pneumophila/pathogenicity , Legionnaires' Disease/immunology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/immunology , Toll-Like Receptor 9/metabolism , Animals , CpG Islands , Cytokines/metabolism , Female , Humans , Legionnaires' Disease/microbiology , Legionnaires' Disease/mortality , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred BALB C , Phenotype , Toll-Like Receptor 9/genetics
18.
Expert Rev Respir Med ; 2(6): 739-52, 2008 Dec.
Article in English | MEDLINE | ID: mdl-20477236

ABSTRACT

The generation of an innate immune response is essential for rapid clearance of microbes from the respiratory tract, whereas acquired immunity is required for the generation of cellular immunity necessary for the killing of certain intracellular pathogens and the development of immunological memory. Cytokines play an integral role in host defense by serving as leukocyte chemoattractants, leukocyte-activating factors or afferent signals in the induction or regulation of other effector molecules. This review assesses the contribution of cytokine networks to the generation of antimicrobial host defenses in the lung, with an emphasis on cytokines/cytokine networks that are instrumental in innate antibacterial responses, including mucosal immunity, and also introduces networks that instruct the development of adaptive immunity.

19.
J Immunol ; 179(6): 3937-46, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785831

ABSTRACT

In this study, experiments were performed to determine the contribution of TLR9 to the generation of protective innate immunity against virulent bacterial pathogens of the lung. In initial studies, we found that the intratracheal administration of Klebsiella pneumoniae in wild-type (WT) BALB/c mice resulted in the rapid accumulation of dendritic cells (DC) expressing TLR9. As compared with WT mice, animals deficient in TLR9 (TLR9-/-) displayed significantly increased mortality that was associated with a >50-fold increase in lung CFU and a >400-fold increase in K. pneumoniae CFU in blood and spleen, respectively. Intrapulmonary bacterial challenge in TLR9-/- mice resulted in reduced lung DC accumulation and maturation as well as impaired activation of lung macrophages, NK cells, and alphabeta and gammadelta T cells. Mice deficient in TLR9 failed to generate an effective Th1 cytokine response following bacterial administration. The adoptive transfer of bone marrow-derived DC from syngeneic WT but not TLR9-/- mice administered intratracheally reconstituted antibacterial immunity in TLR9-/- mice. Collectively, our findings indicate that TLR9 is required for effective innate immune responses against Gram-negative bacterial pathogens and that approaches to maximize TLR9-mediated DC responses may serve as a means to augment antibacterial immunity in pneumonia.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Pneumonia, Bacterial/immunology , Toll-Like Receptor 9/physiology , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Cell Movement/immunology , Cells, Cultured , Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Immunity, Innate/genetics , Intubation, Intratracheal , Klebsiella Infections/microbiology , Klebsiella Infections/therapy , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred BALB C , Mice, Knockout , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/therapy , Signal Transduction/immunology , Toll-Like Receptor 9/biosynthesis , Toll-Like Receptor 9/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...