Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 4(1): e4281, 2009.
Article in English | MEDLINE | ID: mdl-19169353

ABSTRACT

The cell wall of M. tuberculosis is central to its success as a pathogen. Mycolic acids are key components of this cell wall. The genes involved in joining the alpha and mero mycolates are located in a cluster, beginning with Rv3799c and extending at least until Rv3804c. The role of each enzyme encoded by these five genes is fairly well understood, except for Rv3802c. Rv3802 is one of seven putative cutinases encoded by the genome of M. tuberculosis. In phytopathogens, cutinases hydrolyze the waxy layer of plants, cutin. In a strictly mammalian pathogen, such as M. tuberculosis, it is likely that these proteins perform a different function. Of the seven, we chose to focus on Rv3802c because of its location in a mycolic acid synthesis gene cluster, its putative essentiality, its ubiquitous presence in actinomycetes, and its conservation in the minimal genome of Mycobacterium leprae. We expressed Rv3802 in Escherichia coli and purified the enzymatically active form. We probed its activities and inhibitors characterizing those relevant to its possible role in mycolic acid biosynthesis. In addition to its reported phospholipase A activity, Rv3802 has significant thioesterase activity, and it is inhibited by tetrahydrolipstatin (THL). THL is a described anti-tuberculous compound with an unknown mechanism, but it reportedly targets cell wall synthesis. Taken together, these data circumstantially support a role for Rv3802 in mycolic acid synthesis and, as the cell wall is integral to M. tuberculosis pathogenesis, identification of a novel cell wall enzyme and its inhibition has therapeutic and diagnostic implications.


Subject(s)
Bacterial Proteins/physiology , Gene Expression Regulation, Bacterial , Lactones/pharmacology , Mycobacterium tuberculosis/metabolism , Phospholipases/metabolism , Phospholipases/physiology , Thiolester Hydrolases/physiology , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Cell Wall/metabolism , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Hydrolysis , Molecular Sequence Data , Mycolic Acids/metabolism , Orlistat , Phospholipases/antagonists & inhibitors , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Thiolester Hydrolases/antagonists & inhibitors
2.
Am J Respir Crit Care Med ; 177(7): 720-9, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18202345

ABSTRACT

RATIONALE: Respiratory Mycoplasma pneumoniae (Mp) infection is involved in asthma pathobiology, but whether the established allergic airway inflammation compromises lung innate immunity and subsequently predisposes patients with asthma to Mp infection remains unknown. OBJECTIVES: To test whether the established allergic airway inflammation compromises host innate immunity (e.g., Toll-like receptor 2 [TLR2]) to hinder the elimination of Mp from the lungs. METHODS: We used mouse models of ovalbumin (OVA)-induced allergic airway inflammation with an ensuing Mp infection, and cultures of mouse primary lung dendritic cells (DCs) and bone marrow-derived DCs. MEASUREMENTS AND MAIN RESULTS: Lung Mp clearance in allergic mice and TLR2 and IL-6 levels in lung cells, including DCs as well as cultured primary lung DCs and bone marrow-derived DCs, were assessed. The established OVA-induced allergic airway inflammation, or the prominent Th2 cytokines IL-4 and IL-13, inhibited TLR2 expression and IL-6 production in lung cells, including lung DCs, and eventually led to impaired host defense against Mp. Studies in IL-6 knockout mice indicated that IL-6 directly promoted Mp clearance from the lungs. IL-4- and IL-13-induced suppression of TLR2 was mediated by inhibiting nuclear factor-kappaB activation through signal transducer and activator of transcription 6 (STAT6) signaling pathway. CONCLUSIONS: The established OVA-induced allergic airway inflammation impairs TLR2 expression and host defense cytokine (e.g., IL-6) production, and subsequently delays lung bacterial clearance. This could offer novel therapeutic strategies to reinstate TLR2 activation by using TLR2 ligands and/or blocking IL-4 and IL-13 to ameliorate persisting respiratory bacterial infections in allergic lungs.


Subject(s)
Asthma/immunology , Asthma/microbiology , Down-Regulation , Immunity, Innate , Pneumonia, Mycoplasma/immunology , Toll-Like Receptor 2/metabolism , Animals , Colony Count, Microbial , Disease Susceptibility/immunology , Female , Interleukin-13/immunology , Interleukin-4/immunology , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mycoplasma pneumoniae/immunology , Ovalbumin
3.
J Immunol ; 179(6): 3995-4002, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785838

ABSTRACT

Respiratory infections, including Mycoplasma pneumoniae (Mp), contribute to asthma pathobiology. To date, the mechanisms underlying the increased susceptibility of asthmatics to airway Mp infection remain unclear. Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein is a recently described large airway epithelial cell-derived molecule that was predicted to exert host defense activities. However, SPLUNC1 function and regulation in an infectious or allergic milieu are still unknown. We determined host defense and anti-inflammatory functions of SPLUNC1 protein in Mp infection and the regulation of SPLUNC1 by Mp and allergic inflammation (e.g., IL-13). SPLUNC1 function was examined in Mp or human airway epithelial cell cultures by using SPLUNC1 recombinant protein, overexpression and RNA interference. Human and mouse bronchial epithelial SPLUNC1 was examined using immunostaining, Western blotting, ELISA, laser capture microdissection, and real-time PCR. Mouse models of Mp infection and allergic inflammation and air-liquid interface cultures of normal human primary bronchial epithelial cells were used to study SPLUNC1 regulation by Mp and IL-13. We found that: 1) SPLUNC1 protein decreased Mp levels and inhibited epithelial IL-8 production induced by Mp-derived lipoproteins; 2) normal human and mouse large airway epithelial cells expressed high levels of SPLUNC1; and 3) although Mp infection increased SPLUNC1, IL-13 significantly decreased SPLUNC1 expression and Mp clearance. Our results suggest that SPLUNC1 serves as a novel host defense protein against Mp and that an allergic setting markedly reduces SPLUNC1 expression, which may in part contribute to the persistent nature of bacterial infections in allergic airways.


Subject(s)
Glycoproteins/physiology , Inflammation Mediators/physiology , Mycoplasma pneumoniae/growth & development , Mycoplasma pneumoniae/immunology , Phosphoproteins/physiology , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/pathology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Animals , Base Sequence , Cell Line , Disease Models, Animal , Female , Glycoproteins/biosynthesis , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Immunity, Innate/genetics , Inflammation Mediators/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/biosynthesis , Lipoproteins/antagonists & inhibitors , Lipoproteins/physiology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pneumonia, Mycoplasma/metabolism , RNA Interference/immunology , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/microbiology , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Up-Regulation/genetics
4.
Microbes Infect ; 9(1): 78-86, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17198762

ABSTRACT

IL-23 induces IL-17 production in activated CD4+ T cells and participates in host defense against many encapsulated bacteria. However, whether the IL-23/IL-17 axis contributes to a Mycoplasma pneumoniae (Mp)-induced lung inflammation (e.g., neutrophils) has not been addressed. Using an acute respiratory Mp infection murine model, we found significantly up-regulated lung IL-23p19 mRNA in the early phase of infection (4h), and alveolar macrophages were an important cell source of Mp-induced IL-23. We further showed that Mp significantly increased IL-17 protein levels in bronchoalveolar lavage (BAL). Lung gene expression of IL-17, IL-17C and IL-17F was also markedly up-regulated by Mp in vivo. IL-17 and IL-17F were found to be derived mainly from lung CD4+ T cells, and were increased upon IL-23 stimulation in vitro. In vivo blocking of IL-23p19 alone or in combination with IL-23/IL-12p40 resulted in a significant reduction of Mp-induced IL-17 protein and IL-17/IL-17F mRNA expression, which was accompanied by a trend toward reduced lung neutrophil recruitment, BAL neutrophil activity, and Mp clearance. However, IL-23 neutralization had no effect on Mp-induced lung IL-17C mRNA expression. These results demonstrate that IL-17/IL-17F production is IL-23-dependent in an acute Mp infection, and contributes to neutrophil recruitment and activity in the lung defense against the infection.


Subject(s)
Interleukin-17/biosynthesis , Interleukin-23/immunology , Macrophages, Alveolar/immunology , Mycoplasma pneumoniae/immunology , Neutrophils/immunology , Pneumonia, Mycoplasma/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Female , Interleukin-12 Subunit p40/immunology , Interleukin-17/immunology , Interleukin-23 Subunit p19/immunology , Lung/immunology , Lung/microbiology , Macrophages, Alveolar/microbiology , Mice , Mice, Inbred BALB C , Neutrophils/microbiology , Pneumonia, Mycoplasma/microbiology
5.
Am J Physiol Lung Cell Mol Physiol ; 292(5): L1064-72, 2007 May.
Article in English | MEDLINE | ID: mdl-17194718

ABSTRACT

The original hygiene hypothesis suggests that early childhood respiratory infections preceding allergen exposure may decrease the prevalence of allergic diseases. We have recently demonstrated that Mycoplasma pneumoniae infection preceding allergen exposure reduced allergic responses in mice. However, the molecular mechanisms underlying the protective role of M. pneumoniae in allergic responses, particularly airway mucin production, remain unclear. Wild-type and Toll-like receptor 2 (TLR2)-deficient mice with a respiratory M. pneumoniae infection preceding allergen (ovalbumin) challenge were utilized to determine the regulatory role of TLR2-IFN-gamma signaling pathway in airway mucin expression. Furthermore, air-liquid interface cultures of mouse primary tracheal epithelial cells were performed to examine the effects of IFN-gamma on mucin expression. In wild-type mice, M. pneumoniae infection preceding allergen challenge significantly reduced airway mucins but increased IFN-gamma. In sharp contrast, in TLR2-deficient mice, M. pneumoniae preceding allergen challenge resulted in increased mucin protein without a noticeable change of IFN-gamma. In cultured mouse primary tracheal epithelial cells, IFN-gamma was shown to directly inhibit mucin expression in a dose-dependent manner. Our study demonstrates for the first time that a respiratory M. pneumoniae infection preceding allergen challenge reduces airway epithelial mucin expression in part through TLR2-IFN-gamma signaling pathway. A bacterial infection in asthmatic subjects with weakened TLR2-IFN-gamma signaling may result in an exaggerated airway mucin production.


Subject(s)
Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/physiopathology , Toll-Like Receptor 2/deficiency , Allergens , Animals , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Hypersensitivity , Interferon-gamma/genetics , Mice , Mucin 5AC , Mucins/biosynthesis , Mucins/genetics , Mycoplasma pneumoniae , Promoter Regions, Genetic , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/physiology , Trachea
6.
Microbes Infect ; 8(7): 1764-72, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16713727

ABSTRACT

Respiratory Mycoplasma pneumoniae (Mp) infection is involved in several acute and chronic lung diseases including community-acquired pneumonia, asthma and chronic obstructive pulmonary disease. In the chronic disease process, recurrent respiratory bacterial infections could occur, which may result in varying degrees of symptoms and lung inflammation among patients. However, the lung immunologic differences of host responses to repeated bacterial (i.e., Mp) infections remain to be determined. In the present study, we examined cellular and humoral responses to multiple (up to 3) Mp infections in two genetically different strains of mice (BALB/c and C57BL/6). Mice were intranasally inoculated with one Mp infection, two or three Mp infections (4 weeks apart), and sacrificed on days 3, 7 and 14 after the last Mp infection. Overall, compared to C57BL/6 mice, BALB/c mice demonstrated a significantly higher degree of lung tissue inflammatory cell infiltrate, BAL cellularity, and release of pro-inflammatory cytokines (TNF-alpha, keratinocyte-derived chemokine (KC, a mouse homolog of human chemokine Gro-alpha [CXCL1], and IFN-gamma). In addition, BALB/c mice presented higher levels of serum Mp-specific IgG and IgM, but not IgA. Consistently with lung and serum data, Mp load in BAL and lung specimens was significantly higher in BALB/c mice than C57BL/6 mice. Moreover, repeated Mp infections in BALB/c, but not C57BL/6 mice, produced a greater inflammatory response than did a single Mp infection. Our results suggest that hosts with different genetic background may have different susceptibility to repeated respiratory Mp infections along with inflammatory responses.


Subject(s)
Genetic Predisposition to Disease , Pneumonia, Mycoplasma/genetics , Pneumonia, Mycoplasma/immunology , Animals , Antigens, Bacterial/blood , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Colony Count, Microbial , Cytokines/analysis , Disease Models, Animal , Female , Histocytochemistry , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Leukocyte Count , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mycoplasma pneumoniae/growth & development , Mycoplasma pneumoniae/immunology , Mycoplasma pneumoniae/isolation & purification , Recurrence , T-Lymphocytes/immunology
7.
J Immunol ; 174(9): 5713-9, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15843573

ABSTRACT

Excessive airway mucin production contributes to airway obstruction in lung diseases such as asthma and chronic obstructive pulmonary disease. Respiratory infections, such as atypical bacterium Mycoplasma pneumoniae (Mp), have been proposed to worsen asthma and chronic obstructive pulmonary disease in part through increasing mucin. However, the molecular mechanisms involved in infection-induced airway mucin overexpression remain to be determined. TLRs have been recently shown to be a critical component in host innate immune response to infections. TLR2 signaling has been proposed to be involved in inflammatory cell activation by mycoplasma-derived lipoproteins. In this study, we show that TLR2 signaling is critical in Mp-induced airway mucin expression in mice and human lung epithelial cells. Respiratory Mp infection in BALB/c mice activated TLR2 signaling and increased airway mucin. A TLR2-neutralizing Ab significantly reduced mucin expression in Mp-infected BALB/c mice. Furthermore, Mp-induced airway mucin was abolished in TLR2 gene-deficient C57BL/6 mice. Additionally, Mp was shown to increase human lung A549 epithelial cell mucin expression, which was inhibited by the overexpression of a human TLR2 dominant-negative mutant. These results clearly demonstrate that respiratory Mp infection increases airway mucin expression, which is dependent on the activation of TLR2 signaling.


Subject(s)
Lung/immunology , Lung/metabolism , Mucins/biosynthesis , Mycoplasma pneumoniae/immunology , Receptors, Immunologic/physiology , Signal Transduction/immunology , Animals , Bacterial Proteins/immunology , Bacterial Proteins/isolation & purification , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Humans , Immune Sera/pharmacology , Lipoproteins/immunology , Lipoproteins/isolation & purification , Lung/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mycoplasma pneumoniae/chemistry , Pneumonia, Mycoplasma/genetics , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/deficiency , Receptors, Immunologic/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Signal Transduction/genetics , Toll-Like Receptor 2 , Up-Regulation/genetics , Up-Regulation/immunology
8.
Am J Physiol Lung Cell Mol Physiol ; 289(1): L125-33, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15778249

ABSTRACT

Mycoplasma pneumoniae (Mp) has been linked to chronic asthma. Airway remodeling (e.g., airway collagen deposition or fibrosis) is one of the pathological features of chronic asthma. However, the effects of respiratory Mp infection on airway fibrosis in asthma remain unclear. In the present study, we hypothesized that respiratory Mp infection may increase the airway collagen deposition in a murine model of allergic airway inflammation in part through upregulation of transforming growth factor (TGF)-beta1. Double (2 wk apart) inoculations of Mp or saline (control) were given to mice with or without previous allergen (ovalbumin) challenges. On days 14 and 42 after the last Mp or saline, lung tissue and bronchoalveolar lavage (BAL) fluid were collected for analyses of collagen and TGF-beta1 at protein and mRNA levels. In allergen-naïve mice, Mp did not alter airway wall collagen. In allergen-challenged mice, Mp infections did not change airway wall collagen deposition on day 14 but increased the airway collagen on day 42; this increase was accompanied by increased TGF-beta1 protein in the airway wall and reduced TGF-beta1 protein release from the lung tissue into BAL fluid. Our results suggest that Mp infections could modulate airway collagen deposition in a murine model of allergic airway inflammation with TGF-beta1 involved in the collagen deposition process.


Subject(s)
Collagen/metabolism , Lung/pathology , Mycoplasma pneumoniae , Pneumonia, Mycoplasma/metabolism , Pneumonia, Mycoplasma/pathology , Transforming Growth Factor beta/biosynthesis , Animals , Bronchoalveolar Lavage Fluid/microbiology , Disease Models, Animal , Fibrosis , Lung/metabolism , Mice , Mice, Inbred BALB C , Pneumonia, Mycoplasma/microbiology , Respiratory System , Transforming Growth Factor beta1
9.
J Infect Dis ; 189(6): 1119-27, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14999617

ABSTRACT

BACKGROUND: Mycoplasma pneumoniae has been shown to induce airway inflammation and bronchial hyperresponsiveness (BHR) in mice. Inhaled corticosteroids are the mainstay of asthma treatment, but their effects on M. pneumoniae and associated airway inflammation and BHR are poorly understood. METHODS: Four groups of mice were studied to determine whether inhaled fluticasone propionate (FP) could attenuate airway inflammation and BHR by reducing or eliminating M. pneumoniae in lungs. The active group received aerosolized FP once daily for 5 days. Control mice received aerosolized sham solution plus M. pneumoniae, sham solution alone, or FP alone. RESULTS: Mice treated with sham solution or FP alone did not develop airway inflammation or BHR. Mice infected with M. pneumoniae (no FP) developed significant lung inflammation and BHR. FP treatment of infected mice reduced neutrophils in bronchoalveolar lavage fluid (BALF), lung inflammation, and BHR. Expression of Toll-like receptor 2 in lung tissue tended to be down-regulated (P=.18) by FP in infected mice. FP reduced M. pneumoniae by up to 20-fold in lung tissue but not in BALF. CONCLUSION: Inhaled FP suppresses airway inflammation and BHR, which may be caused, in part, by its ability to reduce concentrations of M. pneumoniae in lung tissue.


Subject(s)
Androstadienes/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Bronchial Hyperreactivity/drug therapy , Lung/drug effects , Mycoplasma pneumoniae/drug effects , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/cytology , Chemokines, CXC/analysis , Female , Fluticasone , Lung/microbiology , Membrane Glycoproteins/analysis , Mice , Mice, Inbred BALB C , Mycoplasma pneumoniae/growth & development , Polymerase Chain Reaction , Receptors, Cell Surface/analysis , Toll-Like Receptor 2 , Toll-Like Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...