Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
3.
Infect Immun ; 82(2): 683-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478083

ABSTRACT

Cryptococcus neoformans is an opportunistic fungal pathogen that initiates infection following inhalation. As a result, the pulmonary immune response provides a first line of defense against C. neoformans. Surfactant protein D (SP-D) is an important regulator of pulmonary immune responses and is typically host protective against bacterial and viral respiratory infections. However, SP-D is not protective against C. neoformans. This is evidenced by previous work from our laboratory demonstrating that SP-D-deficient mice infected with C. neoformans have a lower fungal burden and live longer than wild-type (WT) control animals. We hypothesized that SP-D alters susceptibility to C. neoformans by dysregulating the innate pulmonary immune response following infection. Thus, inflammatory cells and cytokines were compared in the bronchoalveolar lavage fluid from WT and SP-D(-/-) mice after C. neoformans infection. Postinfection, mice lacking SP-D have reduced eosinophil infiltration and interleukin-5 (IL-5) in lung lavage fluid. To further explore the interplay of SP-D, eosinophils, and IL-5, mice expressing altered levels of eosinophils and/or IL-5 were infected with C. neoformans to assess the role of these innate immune mediators. IL-5-overexpressing mice have increased pulmonary eosinophilia and are more susceptible to C. neoformans infection than WT mice. Furthermore, susceptibility of SP-D(-/-) mice to C. neoformans infection could be restored to the level of WT mice by increasing IL-5 and eosinophils by crossing the IL-5-overexpressing mice with SP-D(-/-) mice. Together, these studies support the conclusion that SP-D increases susceptibility to C. neoformans infection by promoting C. neoformans-driven pulmonary IL-5 and eosinophil infiltration.


Subject(s)
Cryptococcosis/immunology , Cryptococcosis/pathology , Cryptococcus neoformans/immunology , Eosinophilia/immunology , Interleukin-5/immunology , Pulmonary Surfactant-Associated Protein D/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Female , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Surfactant-Associated Protein D/deficiency
4.
J Biol Chem ; 287(44): 37406-19, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22948158

ABSTRACT

The innate immune system protects the host from bacterial and viral invasion. Surfactant protein A (SPA), a lung-specific collectin, stimulates macrophage chemotaxis. However, the mechanisms regulating this function are unknown. Hyaluronan (HA) and its receptors RHAMM (receptor for HA-mediated motility, CD168) and CD44 also regulate cell migration and inflammation. We therefore examined the role of HA, RHAMM, and CD44 in SPA-stimulated macrophage chemotaxis. Using antibody blockade and murine macrophages, SPA-stimulated macrophage chemotaxis was dependent on TLR2 but not the other SPA receptors examined. Anti-TLR2 blocked SPA-induced production of TGFß. In turn, TGFß1-stimulated chemotaxis was inhibited by HA-binding peptide and anti-RHAMM antibody but not anti-TLR2 antibody. Macrophages from TLR2(-/-) mice failed to migrate in response to SPA but responded normally to TGFß1 and HA, effects that were blocked by anti-RHAMM antibody. Macrophages from WT and CD44(-/-) mice had similar responses to SPA, whereas those from RHAMM(-/-) mice had decreased chemotaxis to SPA, TGFß1, and HA. In primary macrophages, SPA-stimulated TGFß production was dependent on TLR2, JNK, and ERK but not p38. Pam3Cys, a specific TLR2 agonist, stimulated phosphorylation of JNK, ERK, and p38, but only JNK and ERK inhibition blocked Pam3Cys-stimulated chemotaxis. We have uncovered a novel pathway for SPA-stimulated macrophage chemotaxis where SPA stimulation via TLR2 drives JNK- and ERK-dependent TGFß production. TGFß1, in turn, stimulates macrophage chemotaxis in a RHAMM and HA-dependent manner. These findings are highly relevant to the regulation of innate immune responses by SPA with key roles for specific components of the extracellular matrix.


Subject(s)
Chemotaxis , Extracellular Matrix Proteins/metabolism , Hyaluronan Receptors/metabolism , Hyaluronic Acid/physiology , Macrophages/physiology , Pulmonary Surfactant-Associated Protein A/physiology , Toll-Like Receptor 2/metabolism , Transforming Growth Factor beta1/physiology , Animals , Cell Line , Cytoskeleton/metabolism , Extracellular Matrix Proteins/genetics , Gene Knockout Techniques , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , Lipoproteins/pharmacology , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Mink , Mitogen-Activated Protein Kinases/metabolism , Pseudopodia/metabolism , Pseudopodia/physiology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/genetics , Transforming Growth Factor beta1/metabolism
5.
Infect Immun ; 80(7): 2444-53, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22547543

ABSTRACT

Concurrent with the global escalation of the AIDS pandemic, cryptococcal infections are increasing and are of significant medical importance. Furthermore, Cryptococcus neoformans has become a primary human pathogen, causing infection in seemingly healthy individuals. Although numerous studies have elucidated the virulence properties of C. neoformans, less is understood regarding lung host immune factors during early stages of fungal infection. Based on our previous studies documenting that pulmonary surfactant protein D (SP-D) protects C. neoformans cells against macrophage-mediated defense mechanisms in vitro (S. Geunes-Boyer et al., Infect. Immun. 77:2783-2794, 2009), we postulated that SP-D would facilitate fungal infection in vivo. To test this hypothesis, we examined the role of SP-D in response to C. neoformans using SP-D⁻/⁻ mice. Here, we demonstrate that mice lacking SP-D were partially protected during C. neoformans infection; they displayed a longer mean time to death and decreased fungal burden at several time points postinfection than wild-type mice. This effect was reversed by the administration of exogenous SP-D. Furthermore, we show that SP-D bound to the surface of the yeast cells and protected the pathogenic microbes against macrophage-mediated defense mechanisms and hydrogen peroxide (H2O2)-induced oxidative stress in vitro and in vivo. These findings indicate that C. neoformans is capable of coopting host SP-D to increase host susceptibility to the yeast. This study establishes a new paradigm for the role played by SP-D during host responses to C. neoformans and consequently imparts insight into potential future preventive and/or treatment strategies for cryptococcosis.


Subject(s)
Cryptococcosis/microbiology , Cryptococcosis/pathology , Cryptococcus neoformans/pathogenicity , Host-Pathogen Interactions , Pulmonary Surfactant-Associated Protein D/metabolism , Animals , Colony Count, Microbial , Cryptococcosis/immunology , Cryptococcus neoformans/immunology , Female , Lung/microbiology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Pulmonary Surfactant-Associated Protein D/deficiency , Survival Analysis , Virulence
6.
J Immunol ; 188(9): 4376-84, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22474025

ABSTRACT

TCR signaling plays a critical role in regulatory T cell (Treg) development. However, the mechanism for tissue-specific induction of Tregs in the periphery remains unclear. We observed that surfactant protein A (SP-A)-deficient mice have impaired expression of Foxp3 and fewer CD25(+)Foxp3(+) Tregs after ex vivo stimulation and after stimulation with LPS in vivo. The addition of exogenous SP-A completely reversed this phenotype. Although SP-A is known to inhibit T cell proliferation under certain activation conditions, both IL-2 levels as well as active TGF-ß levels increase on extended culture with exogenous SP-A, providing a key mechanism for the maintenance and induction of Tregs. In addition, kinetic suppression assays demonstrate that SP-A enhances the frequency of functional Foxp3(+) Tregs in responder T cell populations in a TGF-ß-dependent manner. In mice treated with LPS in vivo, Tregs increased ∼160% in wild-type mice compared with only a 50% increase in LPS-treated SP-A(-/-) mice 8 d after exposure. Taken together, these findings support the hypothesis that SP-A affects T cell immune function by the induction of Tregs during activation.


Subject(s)
Lymphocyte Activation/physiology , Pulmonary Surfactant-Associated Protein A/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Pulmonary Surfactant-Associated Protein A/biosynthesis , Pulmonary Surfactant-Associated Protein A/genetics , T-Lymphocytes, Regulatory/metabolism
7.
J Allergy Clin Immunol ; 130(1): 205-14.e2, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22502799

ABSTRACT

BACKGROUND: Mycoplasma pneumoniae (Mp) frequently colonizes the airways of patients with chronic asthma and likely contributes to asthma exacerbations. We previously reported that mice lacking surfactant protein A (SP-A) have increased airway hyperresponsiveness (AHR) during M pneumoniae infection versus wild-type mice mediated by TNF-α. Mast cells (MCs) have been implicated in AHR in asthma models and produce and respond to TNF-α. OBJECTIVE: Determine the contribution of MC/TNF interactions to AHR in airways lacking functional SP-A during Mp infection. METHODS: Bronchoalveolar lavage fluid was collected from healthy and asthmatic subjects to examine TNF-α levels and M pneumoniae positivity. To determine how SP-A interactions with MCs regulate airway homeostasis, we generated mice lacking both SP-A and MCs (SP-A(-/-)Kit(W-sh/W-sh)) and infected them with M pneumoniae. RESULTS: Our findings indicate that high TNF-α levels correlate with M pneumoniae positivity in human asthmatic patients and that human SP-A inhibits M pneumoniae-stimulated transcription and release of TNF-α by MCs, implicating a protective role for SP-A. MC numbers increase in M pneumoniae-infected lungs, and airway reactivity is dramatically attenuated when MCs are absent. Using SP-A(-/-)Kit(W-sh/W-sh) mice engrafted with TNF-α(-/-) or TNF receptor (TNF-R)(-/-) MCs, we found that TNF-α activation of MCs through the TNF-R, but not MC-derived TNF-α, leads to augmented AHR during M pneumoniae infection when SP-A is absent. Additionally, M pneumoniae-infected SP-A(-/-)Kit(W-sh/W-sh) mice engrafted with TNF-α(-/-) or TNF-R(-/-) MCs have decreased mucus production compared with that seen in mice engrafted with wild-type MCs, whereas burden was unaffected. CONCLUSION: Our data highlight a previously unappreciated but vital role for MCs as secondary responders to TNF-α during the host response to pathogen infection.


Subject(s)
Mast Cells/metabolism , Mycoplasma pneumoniae/pathogenicity , Pneumonia, Mycoplasma/immunology , Pulmonary Surfactant-Associated Protein A/deficiency , Receptors, Tumor Necrosis Factor/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchoalveolar Lavage Fluid , Cells, Cultured , Humans , Lung/metabolism , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycoplasma pneumoniae/immunology , Pneumonia, Mycoplasma/physiopathology , Pulmonary Surfactant-Associated Protein A/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
J Immunol ; 188(10): 4897-905, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22508928

ABSTRACT

Graft-versus-host disease (GVHD) is a severe and frequent complication of allogeneic bone marrow transplantation (BMT) that involves the gastrointestinal (GI) tract and lungs. The pathobiology of GVHD is complex and involves immune cell recognition of host Ags as foreign. We hypothesize a central role for the collectin surfactant protein A (SP-A) in regulating the development of GVHD after allogeneic BMT. C57BL/6 (H2b; WT) and SP-A-deficient mice on a C57BL/6 background (H2b; SP-A(-/-)) mice underwent allogeneic or syngeneic BMT with cells from either C3HeB/FeJ (H2k; SP-A-deficient recipient mice that have undergone an allogeneic BMT [SP-A(-/-)alloBMT] or SP-A-sufficient recipient mice that have undergone an allogeneic BMT) or C57BL/6 (H2b; SP-A-deficient recipient mice that have undergone a syngeneic BMT or SP-A-sufficient recipient mice that have undergone a syngeneic BMT) mice. Five weeks post-BMT, mice were necropsied, and lung and GI tissue were analyzed. SP-A(-/-) alloBMT or SP-A-sufficient recipient mice that have undergone an allogeneic BMT had no significant differences in lung pathology; however, SP-A(-/-)alloBMT mice developed marked features of GI GVHD, including decreased body weight, increased tissue inflammation, and lymphocytic infiltration. SP-A(-/-)alloBMT mice also had increased colon expression of IL-1ß, IL-6, TNF-α, and IFN-γ and as well as increased Th17 cells and diminished regulatory T cells. Our results demonstrate the first evidence, to our knowledge, of a critical role for SP-A in modulating GI GVHD. In these studies, we demonstrate that mice deficient in SP-A that have undergone an allogeneic BMT have a greater incidence of GI GVHD that is associated with increased Th17 cells and decreased regulatory T cells. The results of these studies demonstrate that SP-A protects against the development of GI GVHD and establishes a role for SP-A in regulating the immune response in the GI tract.


Subject(s)
Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Pulmonary Surfactant-Associated Protein A/physiology , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Gastrointestinal Diseases/genetics , Graft vs Host Disease/genetics , Lymphocyte Count , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Surfactant-Associated Protein A/deficiency , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology
9.
PLoS One ; 7(2): e32436, 2012.
Article in English | MEDLINE | ID: mdl-22384248

ABSTRACT

Surfactant protein-A (SP-A) has well-established functions in reducing bacterial and viral infections but its role in chronic lung diseases such as asthma is unclear. Mycoplasma pneumoniae (Mp) frequently colonizes the airways of chronic asthmatics and is thought to contribute to exacerbations of asthma. Our lab has previously reported that during Mp infection of non-allergic airways, SP-A aides in maintaining airway homeostasis by inhibiting an overzealous TNF-alpha mediated response and, in allergic mice, SP-A regulates eosinophilic infiltration and inflammation of the airway. In the current study, we used an in vivo model with wild type (WT) and SP-A(-/-) allergic mice challenged with the model antigen ovalbumin (Ova) that were concurrently infected with Mp (Ova+Mp) to test the hypothesis that SP-A ameliorates Mp-induced stimulation of eosinophils. Thus, SP-A could protect allergic airways from injury due to release of eosinophil inflammatory products. SP-A deficient mice exhibit significant increases in inflammatory cells, mucus production and lung damage during concurrent allergic airway disease and infection (Ova+Mp) as compared to the WT mice of the same treatment group. In contrast, SP-A deficient mice have significantly decreased Mp burden compared to WT mice. The eosinophil specific factor, eosinophil peroxidase (EPO), which has been implicated in pathogen killing and also in epithelial dysfunction due to oxidative damage of resident lung proteins, is enhanced in samples from allergic/infected SP-A(-/-) mice as compared to WT mice. In vitro experiments using purified eosinophils and human SP-A suggest that SP-A limits the release of EPO from Mp-stimulated eosinophils thereby reducing their killing capacity. These findings are the first to demonstrate that although SP-A interferes with eosinophil-mediated biologic clearance of Mp by mediating the interaction of Mp with eosinophils, SP-A simultaneously benefits the airway by limiting inflammation and damage.


Subject(s)
Eosinophils/metabolism , Gene Expression Regulation , Hypersensitivity/immunology , Lung/immunology , Mycoplasma pneumoniae/metabolism , Pneumonia, Mycoplasma/metabolism , Pneumonia, Mycoplasma/microbiology , Pulmonary Surfactant-Associated Protein A/metabolism , Albumins/metabolism , Animals , Calcium/metabolism , Eosinophils/immunology , Flow Cytometry , Humans , Hypersensitivity/metabolism , Inflammation , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological
10.
J Immunol ; 188(7): 3371-81, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22371396

ABSTRACT

Asthma is a chronic inflammatory disease in which airway epithelial cells are the first line of defense against exposure of the airway to infectious agents. Src homology protein (SHP)-1, a protein tyrosine phosphatase, is a negative regulator of signaling pathways that are critical to the development of asthma and host defense. We hypothesize that SHP-1 function is defective in asthma, contributing to the increased inflammatory response induced by Mycoplasma pneumoniae, a pathogen known to exacerbate asthma. M. pneumoniae significantly activated SHP-1 in airway epithelial cells collected from nonasthmatic subjects by bronchoscopy with airway brushing but not in cells from asthmatic subjects. In asthmatic airway epithelial cells, M. pneumoniae induced significant PI3K/Akt phosphorylation, NF-κB activation, and IL-8 production compared with nonasthmatic cells, which were reversed by SHP-1 overexpression. Conversely, SHP-1 knockdown significantly increased IL-8 production and PI3K/Akt and NF-κB activation in the setting of M. pneumoniae infection in nonasthmatic cells, but it did not exacerbate these three parameters already activated in asthmatic cells. Thus, SHP-1 plays a critical role in abrogating M. pneumoniae-induced IL-8 production in nonasthmatic airway epithelial cells through inhibition of PI3K/Akt and NF-κB activity, but it is defective in asthma, resulting in an enhanced inflammatory response to infection.


Subject(s)
Asthma/enzymology , Epithelial Cells/immunology , Mycoplasma pneumoniae/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Adult , Asthma/immunology , Asthma/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Cell Nucleus/enzymology , Cells, Cultured/enzymology , Cells, Cultured/immunology , Epithelial Cells/enzymology , Female , Humans , In Vitro Techniques , Inflammation , Interleukin-8/biosynthesis , Interleukin-8/genetics , Male , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Transcription, Genetic , Young Adult
11.
J Immunol ; 188(3): 957-67, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22219327

ABSTRACT

Pulmonary surfactant lipoproteins lower the surface tension at the alveolar-airway interface of the lung and participate in host defense. Previous studies reported that surfactant protein A (SP-A) inhibits lymphocyte proliferation. We hypothesized that SP-A-mediated modulation of T cell activation depends upon the strength, duration, and type of lymphocyte activating signals. Modulation of T cell signal strength imparted by different activating agents ex vivo and in vivo in different mouse models and in vitro with human T cells shows a strong correlation between strength of signal (SoS) and functional effects of SP-A interactions. T cell proliferation is enhanced in the presence of SP-A at low SoS imparted by exogenous mitogens, specific Abs, APCs, or in homeostatic proliferation. Proliferation is inhibited at higher SoS imparted by different doses of the same T cell mitogens or indirect stimuli such as LPS. Importantly, reconstitution with exogenous SP-A into the lungs of SP-A(-/-) mice stimulated with a strong signal also resulted in suppression of T cell proliferation while elevating baseline proliferation in unstimulated T cells. These signal strength and SP-A-dependent effects are mediated by changes in intracellular Ca(2+) levels over time, involving extrinsic Ca(2+)-activated channels late during activation. These effects are intrinsic to the global T cell population and are manifested in vivo in naive as well as memory phenotype T cells. Thus, SP-A appears to integrate signal thresholds to control T cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Lymphocyte Activation/drug effects , Pulmonary Surfactant-Associated Protein A/pharmacology , T-Lymphocytes/drug effects , Animals , Calcium/metabolism , Humans , Lipopolysaccharides/pharmacology , Lung/metabolism , Mice , Mice, Knockout , Mitogens/pharmacology , Pulmonary Surfactant-Associated Protein A/deficiency , Signal Transduction , T-Lymphocytes/immunology
12.
Am J Respir Crit Care Med ; 185(5): 525-36, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22198976

ABSTRACT

RATIONALE: Surfactant protein (SP)-D and SP-A have been implicated in immunomodulation in the lung. It has been reported that patients with idiopathic pulmonary fibrosis (IPF) often have elevated serum levels of SP-A and SP-D, although their role in the disease is not known. OBJECTIVES: The goal of this study was to test the hypothesis that SP-D plays an important role in lung fibrosis using a mouse model of fibrosis induced by bleomycin (BLM). METHODS: Triple transgenic inducible SP-D mice (iSP-D mice), in which rat SP-D is expressed in response to doxycycline (Dox) treatment, were administered BLM (100 U/kg) or saline subcutaneously using miniosmotic pumps. MEASUREMENTS AND MAIN RESULTS: BLM-treated iSP-D mice off Dox (SP-D off) had increased lung fibrosis compared with mice on Dox (SP-D on). SP-D deficiency also increased macrophage-dominant cell infiltration and the expression of profibrotic cytokines (transforming growth factor [TGF]-ß1, platelet-derived growth factor-AA). Alveolar macrophages isolated from BLM-treated iSP-D mice off Dox (SP-D off) secreted more TGF-ß1. Fibrocytes, which are bone marrow-derived mesenchymal progenitor cells, were increased to a greater extent in the lungs of the BLM-treated iSP-D mice off Dox (SP-D off). Fibrocytes isolated from BLM-treated iSP-D mice off Dox (SP-D off) expressed more of the profibrotic cytokine TGF-ß1 and more CXCR4, a chemokine receptor that is important in fibrocyte migration into the lungs. Exogenous SP-D administered intratracheally attenuated BLM-induced lung fibrosis in SP-D(-/-) mice. CONCLUSIONS: These data suggest that alveolar SP-D regulates numbers of macrophages and fibrocytes in the lungs, profibrotic cytokine expression, and fibrotic lung remodeling in response to BLM injury.


Subject(s)
Bleomycin/toxicity , Idiopathic Pulmonary Fibrosis/physiopathology , Pulmonary Surfactant-Associated Protein D/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/physiopathology , Airway Remodeling/physiology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cytokines/physiology , Disease Models, Animal , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pulmonary Surfactant-Associated Protein A/analysis , Pulmonary Surfactant-Associated Protein A/physiology , Pulmonary Surfactant-Associated Protein D/analysis , Rats
13.
J Allergy Ther ; Suppl 1(4)2012 Jan 25.
Article in English | MEDLINE | ID: mdl-24273688

ABSTRACT

BACKGROUND: Caveolin-1, the hallmark protein of caveolae, is highly expressed within the lung in the epithelium, endothelium, and in immune cells. In addition to its classical roles in cholesterol metabolism and endocytosis, caveolin-1 has also been shown to be important in inflammatory signaling pathways. In particular, caveolin-1 is known to associate with the nitric oxide synthase enzymes, downregulating their activity. Endotoxins, which are are composed mainly of lipopolysaccharide (LPS), are found ubiquitously in the environment and can lead to the development of airway inflammation and increased airway hyperresponsiveness (AHR). METHODS: We compared the acute responses of wild-type and caveolin-1 deficient mice after LPS aerosol, a well-accepted mode of endotoxin exposure, to investigate the role of caveolin-1 in the development of environmental lung injury. RESULTS: Although the caveolin-1 deficient mice had greater lung inflammatory indices compared to wild-type mice, they exhibited reduced AHR following LPS exposure. The uncoupling of inflammation and AHR led us to investigate the role of caveolin-1 in the production of nitric oxide, which is known to act as a bronchodilator. The absence of caveolin-1 resulted in increased nitrite levels in the lavage fluid in both sham and LPS treated mice. Additionally, inducible nitric oxide synthase expression was increased in the lung tissue of caveolin-1 deficient mice following LPS exposure and administration of the potent and specific inhibitor 1400W increased AHR to levels comparable to wild-type mice. CONCLUSIONS: We attribute the relative airway hyporesponsiveness in the caveolin-1 deficient mice after LPS exposure to the specific role of caveolin-1 in mediating nitric oxide production.

14.
Am J Physiol Lung Cell Mol Physiol ; 301(4): L598-606, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21784968

ABSTRACT

Surfactant protein A (SP-A) regulates a variety of immune cell functions. We determined the ability of SP-A derived from normal and asthmatic subjects to modulate the inflammatory response elicited by Mycoplasma pneumoniae, a pathogen known to exacerbate asthma. Fourteen asthmatic and 10 normal control subjects underwent bronchoscopy with airway brushing and bronchoalveolar lavage (BAL). Total SP-A was extracted from BAL. The ratio of SP-A1 to total SP-A (SP-A1/SP-A) and the binding of total SP-A to M. pneumoniae membranes were determined. Airway epithelial cells from subjects were exposed to either normal or asthmatic SP-A before exposure to M. pneumoniae. IL-8 protein and MUC5AC mRNA were measured. Total BAL SP-A concentration did not differ between groups, but the percentage SP-A1 was significantly increased in BAL of asthmatic compared with normal subjects. SP-A1/SP-A significantly correlated with maximum binding of total SP-A to M. pneumoniae, but only in asthma. SP-A derived from asthmatic subjects did not significantly attenuate IL-8 and MUC5AC in the setting of M. pneumoniae infection compared with SP-A derived from normal subjects. We conclude that SP-A derived from asthmatic subjects does not abrogate inflammation effectively, and this dysfunction may be modulated by SP-A1/SP-A.


Subject(s)
Asthma/metabolism , Epithelial Cells/metabolism , Inflammation/metabolism , Mycoplasma pneumoniae/metabolism , Pulmonary Surfactant-Associated Protein A/metabolism , Recombinant Proteins/metabolism , Adult , Asthma/complications , Asthma/immunology , Asthma/microbiology , Asthma/physiopathology , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/chemistry , Bronchoscopy , Case-Control Studies , Cell Membrane/metabolism , Cells, Cultured , Epithelial Cells/cytology , Female , HEK293 Cells , Humans , Inflammation/complications , Inflammation/immunology , Inflammation/microbiology , Inflammation/physiopathology , Interleukin-8/biosynthesis , Male , Mucin 5AC/biosynthesis , Mycoplasma pneumoniae/immunology , Plasmids , Polymerase Chain Reaction , Protein Binding , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , RNA, Messenger/analysis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Transfection
15.
J Clin Endocrinol Metab ; 96(4): E624-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21270323

ABSTRACT

CONTEXT: Labor is characterized by "decidual activation" with production of inflammatory mediators. Recent data suggest that surfactant protein-A (SP-A) may be critical to the onset of labor in mice. Whether this is also true in humans is unclear. OBJECTIVES: The aim was to investigate: 1) the expression of SP-A at the maternal-fetal interface; 2) the effect of SP-A on the production of inflammatory mediators by human decidua; and 3) the association between single nucleotide polymorphisms in maternal SP-A genes and spontaneous preterm birth. RESEARCH DESIGN AND METHODS: In situ expression of SP-A was investigated by immunohistochemistry and quantitative RT-PCR. Term decidual stromal cells were isolated, purified, and treated with/without SP-A (1-100 µg/ml), IL-1ß, and/or thrombin. Levels of inflammatory mediators [IL-6, IL-8, TNFα, matrix metalloproteinase-3, monocyte chemotactic protein-1, IL-1ß, PGE(2), prostaglandin F(2α) (PGF(2α))] and angiogenic factors (soluble fms-like tyrosine kinase-1, vascular endothelial growth factor) were measured in conditioned supernatant by ELISA and corrected for protein content. The effect of SP-A on eicosanoid gene expression was measured by quantitative RT-PCR. RESULTS: SP-A localized to endometrium/decidua. High-dose SP-A (100 µg/ml) inhibited PGF(2α) by term decidual stromal cells without affecting the production of other inflammatory mediators, and this effect occurred at a posttranscriptional level. Decidual SP-A expression decreased significantly with labor. Single nucleotide polymorphisms in the SP-A genes do not appear to be associated with preterm birth. CONCLUSIONS: SP-A is produced by human endometrium/decidua, where it significantly and selectively inhibits PGF(2α) production. Its expression decreases with labor. These novel observations suggest that decidual SP-A likely plays a critical role in regulating prostaglandin production within the uterus, culminating at term in decidual activation and the onset of labor.


Subject(s)
Decidua/drug effects , Dinoprost/metabolism , Labor Onset/physiology , Pulmonary Surfactant-Associated Protein A/pharmacology , Term Birth , Case-Control Studies , Cell Culture Techniques , Cells, Cultured , Decidua/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Fetal Membranes, Premature Rupture/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , Labor Onset/drug effects , Labor Onset/genetics , Labor Onset/metabolism , Polymorphism, Single Nucleotide , Pregnancy , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/physiology , Term Birth/drug effects , Term Birth/genetics , Term Birth/metabolism
16.
J Immunol ; 186(5): 2842-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21257967

ABSTRACT

Although many studies have shown that pulmonary surfactant protein (SP)-A functions in innate immunity, fewer studies have addressed its role in adaptive immunity and allergic hypersensitivity. We hypothesized that SP-A modulates the phenotype and prevalence of dendritic cells (DCs) and CD4(+) T cells to inhibit Th2-associated inflammatory indices associated with allergen-induced inflammation. In an OVA model of allergic hypersensitivity, SP-A(-/-) mice had greater eosinophilia, Th2-associated cytokine levels, and IgE levels compared with wild-type counterparts. Although both OVA-exposed groups had similar proportions of CD86(+) DCs and Foxp3(+) T regulatory cells, the SP-A(-/-) mice had elevated proportions of CD4(+) activated and effector memory T cells in their lungs compared with wild-type mice. Ex vivo recall stimulation of CD4(+) T cell pools demonstrated that cells from the SP-A(-/-) OVA mice had the greatest proliferative and IL-4-producing capacity, and this capability was attenuated with exogenous SP-A treatment. Additionally, tracking proliferation in vivo demonstrated that CD4(+) activated and effector memory T cells expanded to the greatest extent in the lungs of SP-A(-/-) OVA mice. Taken together, our data suggested that SP-A influences the prevalence, types, and functions of CD4(+) T cells in the lungs during allergic inflammation and that SP deficiency modifies the severity of inflammation in allergic hypersensitivity conditions like asthma.


Subject(s)
Allergens/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Immunologic Memory , Lung/immunology , Lung/pathology , Lymphocyte Activation/immunology , Pulmonary Surfactant-Associated Protein A/deficiency , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Immunologic Memory/genetics , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Lung/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/administration & dosage , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/physiology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/prevention & control , Severity of Illness Index , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology
17.
Am J Respir Cell Mol Biol ; 44(2): 175-84, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20348208

ABSTRACT

Surfactant protein A (SP-A) mediates innate immune cell responses to LPS, a cell wall component of gram-negative bacteria that is found ubiquitously in the environment and is associated with adverse health effects. Inhaled LPS induces lung inflammation and increases airway responsiveness (AR). However, the role of SP-A in mediating LPS-induced AR is not well-defined. Nitric oxide (NO) is described as a potent bronchodilator, and previous studies showed that SP-A modulates the LPS-induced production of NO. Hence, we tested the hypothesis that increased AR, observed in response to aerosolized LPS exposure, would be significantly reduced in an SP-A-deficient condition. Wild-type (WT) and SP-A null (SP-A(-/-)) mice were challenged with aerosolized LPS. Results indicate that despite similar inflammatory indices, LPS-treated SP-A(-/-) mice had attenuated AR after methacholine challenge, compared with WT mice. The attenuated AR could not be attributed to inherent differences in SP-D concentrations or airway smooth muscle contractile and relaxation properties, because these measures were similar between WT and SP-A(-/-) mice. LPS-treated SP-A(-/-) mice, however, had elevated nitrite concentrations, inducible nitric oxide synthase (iNOS) expression, and NOS activity in their lungs. Moreover, the administration of the iNOS-specific inhibitor 1400W completely abrogated the attenuated AR. Thus, when exposed to aerosolized LPS, SP-A(-/-) mice demonstrate a relative airway hyporesponsiveness that appears to be mediated at least partly via an iNOS-dependent mechanism. These findings may have clinical significance, because recent studies reported associations between surfactant protein polymorphisms and a variety of lung diseases.


Subject(s)
Lipopolysaccharides/pharmacology , Lung/immunology , Lung/physiopathology , Nitric Oxide/physiology , Pulmonary Surfactant-Associated Protein A/deficiency , Animals , Immunity, Innate , Lung/drug effects , Methacholine Chloride/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type II/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/immunology , Pulmonary Surfactant-Associated Protein A/physiology , Pulmonary Surfactant-Associated Protein D/metabolism
18.
J Immunol ; 185(7): 3884-94, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20810986

ABSTRACT

During pulmonary infections, a careful balance between activation of protective host defense mechanisms and potentially injurious inflammatory processes must be maintained. Surfactant protein A (SP-A) is an immune modulator that increases pathogen uptake and clearance by phagocytes while minimizing lung inflammation by limiting dendritic cell (DC) and T cell activation. Recent publications have shown that SP-A binds to and is bacteriostatic for Mycoplasma pneumoniae in vitro. In vivo, SP-A aids in maintenance of airway homeostasis during M. pneumoniae pulmonary infection by preventing an overzealous proinflammatory response mediated by TNF-α. Although SP-A was shown to inhibit maturation of DCs in vitro, the consequence of DC/SP-A interactions in vivo has not been elucidated. In this article, we show that the absence of SP-A during M. pneumoniae infection leads to increased numbers of mature DCs in the lung and draining lymph nodes during the acute phase of infection and, consequently, increased numbers of activated T and B cells during the course of infection. The findings that glycyrrhizin, a specific inhibitor of extracellular high-mobility group box-1 (HMGB-1) abrogated this effect and that SP-A inhibits HMGB-1 release from immune cells suggest that SP-A inhibits M. pneumoniae-induced DC maturation by regulating HMGB-1 cytokine activity.


Subject(s)
Dendritic Cells/immunology , HMGB1 Protein/immunology , Mycoplasma Infections/immunology , Pulmonary Surfactant-Associated Protein A/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Dendritic Cells/cytology , Dendritic Cells/metabolism , Flow Cytometry , HMGB1 Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mycoplasma Infections/metabolism , Mycoplasma pneumoniae/immunology , Pulmonary Surfactant-Associated Protein A/metabolism
19.
Innate Immun ; 16(3): 183-90, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20418258

ABSTRACT

Although the lipoprotein complex of pulmonary surfactant has long been recognized as essential for reducing lung surface tension, its role in lung immune host defense has only relatively recently been elucidated. Surfactant-associated proteins A (SP-A) and D (SP-D) can attenuate bacterial and viral infection and inflammation by acting as opsonins and by regulating innate immune cell functions. Surfactant-associated protein A and D also interact with antigen-presenting cells and T cells, thereby linking the innate and adaptive immune systems. A recent study from our laboratory demonstrated that mice deficient in SP-A have enhanced susceptibility to airway hyper-responsiveness and lung inflammation induced by Mycoplasma pneumonia, an atypical bacterium present in the airways of approximately 50% of asthmatics experiencing their first episode, and further supports an important role for SP-A in the host response to allergic airway disease. Animal and human studies suggest that alterations in the functions or levels of SP-A and SP-D are associated with both infectious and non-infectious chronic lung diseases such as asthma. Future studies are needed to elucidate whether alterations in SP-A and SP-D are a consequence and/or cause of allergic airway disease.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Asthma/immunology , Collectins/immunology , Adaptive Immunity , Alveolitis, Extrinsic Allergic , Animals , Asthma/genetics , Asthma/metabolism , Bronchial Hyperreactivity , Disease Susceptibility , Humans , Immunity, Innate , Inflammation , Mice , Mice, Knockout , Pneumonia, Mycoplasma , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism
20.
Med Mycol ; 48(4): 580-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20141481

ABSTRACT

Surfactant protein D (SP-D) plays a central role in pulmonary innate immune responses to microbes and allergens, often enhancing clearance of inhaled material. Although SP-D functions during bacterial and viral infections are well established, much less is known about its possible roles during invasive fungal infections. Aspergillus fumigatus is a prominent fungal pathogen in immunocompromised individuals, and can cause allergic or invasive aspergillosis. SP-D has been shown to be protective against both of these disease modalities. The moieties present on the fungal surface responsible for SP-D binding remain largely unclear, although cell wall 1,3-beta-D-glucan is bound by SP-D in other fungal species. There is little information regarding the interaction of SP-D with A. fumigatus hyphae which are responsible for the invasive form of disease. Here, we show that SP-D binding to A. fumigatus hyphae is sensitive to the activity of the calcium-activated protein phosphatase calcineurin. Deletion of the catalytic subunit calcineurin A (DeltacnaA) or pharmacologic inhibition of calcineurin through FK506 abrogated SP-D binding. In contrast, SP-D binding to Cruptococcus neoformans was calcineurin-independent. Pharmacologic inhibition of A. fumigatus cell wall components by caspofungin (inhibits 1,3-beta-D-glucan synthesis) and nikkomycin Z (inhibits chitin synthesis) increased SP-D binding to the wild-type strain. In contrast, SP-D binding increased in the DeltacnaA strain only after nikkomycin Z treatment. We conclude that SP-D binding to A. fumigatus hyphae is calcineurin-sensitive, presumably as a consequence of calcineurin's role in regulating production of key cell wall binding partners, such as 1,3-beta-D-glucan. Elucidation of the interaction between lung innate immune factors and A. fumigatus could lead to the development of novel therapeutic interventions.


Subject(s)
Aspergillus fumigatus/metabolism , Calcineurin/metabolism , Hyphae/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Aminoglycosides/pharmacology , Animals , Aspergillosis/immunology , Aspergillosis/microbiology , CHO Cells , Cricetinae , Cricetulus , Cryptococcus neoformans/metabolism , Flow Cytometry , Humans , Lung/immunology , Lung/microbiology , Tacrolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...