Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Sci Rep ; 11(1): 11571, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34078958

ABSTRACT

Mycoplasma pneumoniae is the leading cause of bacterial community-acquired pneumonia among hospitalized children in the United States. It is also responsible for a spectrum of other respiratory tract disorders and extrapulmonary manifestations in children and adults. The main virulence factor of M. pneumoniae is a 591 amino acid multifunctional protein called Community Acquired Respiratory Distress Syndrome (CARDS) toxin. The amino terminal region of CARDS toxin (N-CARDS) retains ADP-ribosylating activity and the carboxy region (C-CARDS) contains the receptor binding and vacuolating activities. After internalization, CARDS toxin is transported in a retrograde manner from endosome through the Golgi complex into the endoplasmic reticulum. However, the mechanisms and criteria by which internalized CARDS toxin is transported and activated to execute its cytotoxic effects remain unknown. In this study, we used full-length CARDS toxin and its mutant and truncated derivatives to analyze how pharmacological drugs that alter pH of intracellular vesicles and electrical potential across vesicular membranes affect translocation of CARDS toxin in mammalian cells. Our results indicate that an acidic environment is essential for CARDS toxin retrograde transport to endoplasmic reticulum. Moreover, retrograde transport facilitates toxin clipping and is required to induce vacuole formation. Additionally, toxin-mediated cell vacuolation is strictly dependent on the function of vacuolar type-ATPase.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Endosomes/metabolism , Hydrogen-Ion Concentration , Mycoplasma pneumoniae/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuoles/metabolism
2.
Cell Microbiol ; 21(8): e13032, 2019 08.
Article in English | MEDLINE | ID: mdl-30977272

ABSTRACT

Mycoplasma pneumoniae is the leading cause of bacterial community-acquired pneumonia among hospitalised children in United States and worldwide. Community-acquired respiratory distress syndrome (CARDS) toxin is a key virulence determinant of M. pneumoniae. The N-terminus of CARDS toxin exhibits ADP-ribosyltransferase (ADPRT) activity, and the C-terminus possesses binding and vacuolating activities. Thiol-trapping experiments of wild-type (WT) and cysteine-to-serine-mutated CARDS toxins with alkylating agents identified disulfide bond formation at the amino terminal cysteine residues C230 and C247. Compared with WT and other mutant toxins, C247S was unstable and unusable for comparative studies. Although there were no significant variations in binding, entry, and retrograde trafficking patterns of WT and mutated toxins, C230S did not elicit vacuole formation in intoxicated cells. In addition, the ADPRT domain of C230S was more sensitive to all tested proteases when compared with WT toxin. Despite its in vitro ADPRT activity, the reduction of C230S CARDS toxin-mediated ADPRT activity-associated IL-1ß production in U937 cells and the recovery of vacuolating activity in the protease-released carboxy region of C230S indicated that the disulfide bond was essential not only to maintain the conformational stability of CARDS toxin but also to properly execute its cytopathic effects.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Disulfides/chemistry , Host-Pathogen Interactions/genetics , Macrophages/microbiology , Mycoplasma pneumoniae/genetics , Vacuoles/microbiology , ADP-Ribosylation , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Binding Sites , CHO Cells , Cell Line, Tumor , Cricetulus , Disulfides/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Humans , Interleukin-1beta/biosynthesis , Macrophages/pathology , Models, Molecular , Mutation , Mycoplasma pneumoniae/metabolism , Mycoplasma pneumoniae/pathogenicity , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Vacuoles/metabolism , Vacuoles/ultrastructure , Virulence
3.
mBio ; 9(1)2018 01 23.
Article in English | MEDLINE | ID: mdl-29362229

ABSTRACT

Mycoplasma pneumoniae is an atypical bacterium that causes respiratory illnesses in humans, including pharyngitis, tracheobronchitis, and community-acquired pneumonia (CAP). It has also been directly linked to reactive airway disease, asthma, and extrapulmonary pathologies. During its colonization, M. pneumoniae expresses a unique ADP-ribosylating and vacuolating cytotoxin designated community-acquired respiratory distress syndrome (CARDS) toxin. CARDS toxin persists and localizes in the airway in CAP patients, asthmatics, and trauma patients with ventilator-associated pneumonia. Although CARDS toxin binds to specific cellular receptors, is internalized, and induces hyperinflammation, histopathology, mucus hyperplasia, and other airway injury, the intracellular trafficking of CARDS toxin remains unclear. Here, we show that CARDS toxin translocates through early and late endosomes and the Golgi complex and concentrates at the perinuclear region to reach the endoplasmic reticulum (ER). Using ER-targeted SNAP-tag, we confirmed the association of CARDS toxin with the ER and determined that CARDS toxin follows the retrograde pathway. In addition, we identified a novel CARDS toxin amino acid fingerprint, KELED, that is required for toxin transport to the ER and subsequent toxin-mediated cytotoxicity.IMPORTANCEMycoplasma pneumoniae, a leading cause of bacterial community-acquired pneumonia (CAP) among children and adults in the United States, synthesizes a 591-amino-acid ADP-ribosylating and vacuolating protein, designated community-acquired respiratory distress syndrome (CARDS) toxin. CARDS toxin alone is sufficient to induce and mimic major inflammatory and histopathological phenotypes associated with M. pneumoniae infection in rodents and primates. In order to elicit its ADP-ribosylating and vacuolating activities, CARDS toxin must bind to host cell receptors, be internalized via clathrin-mediated pathways, and subsequently be transported to specific intracellular organelles. Here, we demonstrate how CARDS toxin utilizes its unique KELED sequence to exploit the retrograde pathway machinery to reach the endoplasmic reticulum (ER) and fulfill its cytopathic potential. The knowledge generated from these studies may provide important clues to understand the mode of action of CARDS toxin and develop interventions that reduce or eliminate M. pneumoniae-associated airway and extrapulmonary pathologies.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Mycoplasma pneumoniae/metabolism , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Humans , Protein Transport
4.
Am J Respir Cell Mol Biol ; 58(2): 253-260, 2018 02.
Article in English | MEDLINE | ID: mdl-28915064

ABSTRACT

Mycoplasma pneumoniae infection has been linked to poor asthma outcomes. M. pneumoniae produces an ADP-ribosylating and vacuolating toxin called community-acquired respiratory distress syndrome (CARDS) toxin that has a major role in inflammation and airway dysfunction. The objective was to evaluate the immunopathological effects in primates exposed to M. pneumoniae or CARDS toxin. A total of 13 baboons were exposed to M. pneumoniae or CARDS toxin. At Days 7 and 14, BAL fluid was collected and analyzed for cell count, percent of each type of cell, CARDS toxin by PCR, CARDS toxin by antigen capture, eosinophilic cationic protein, and cytokine profiles. Serum IgM, IgG, and IgE responses to CARDS toxin were measured. All animals had a necropsy for analysis of the histopathological changes on lungs. No animal developed signs of infection. The serological responses to CARDS toxin were variable. At Day 14, four of seven animals exposed to M. pneumoniae and all four animals exposed to CARDS toxin developed histological "asthma-like" changes. T cell intracellular cytokine analysis revealed an increasing ratio of IL-4/IFN-γ over time. Both M. pneumoniae and CARDS toxin exposure resulted in similar histopathological pulmonary changes, suggesting that CARDS toxin plays a major role in the inflammatory response.


Subject(s)
Asthma/immunology , Asthma/pathology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Lung/immunology , Lung/pathology , Mycoplasma pneumoniae/pathogenicity , Animals , CD4-Positive T-Lymphocytes/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Interleukin-13/immunology , Interleukin-4/immunology , Lung/microbiology , Mice , Mycoplasma pneumoniae/immunology , Papio
5.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29061706

ABSTRACT

Mycoplasma pneumoniae is an atypical bacterial respiratory pathogen known to cause a range of airway inflammation and lung and extrapulmonary pathologies. We recently reported that an M. pneumoniae-derived ADP-ribosylating and vacuolating toxin called community-acquired respiratory distress syndrome (CARDS) toxin is capable of triggering NLRP3 (NLR-family, leucine-rich repeat protein 3) inflammasome activation and interleukin-1ß (IL-1ß) secretion in macrophages. However, it is unclear whether the NLRP3 inflammasome is important for the immune response during M. pneumoniae acute infection. In the current study, we utilized in vitro and in vivo models of M. pneumoniae infection to characterize the role of the NLRP3 inflammasome during acute infection. M. pneumoniae-infected macrophages deficient for inflammasome components NLRP3, ASC (apoptosis speck-like protein containing a caspase activation and recruitment domain), or caspase-1 failed to process and secrete IL-1ß. The MyD88/NF-κB signaling pathway was found to be critical for proinflammatory gene expression in macrophages infected with M. pneumoniae C57BL/6 mice deficient for NLRP3 expression were unable to produce IL-1ß in the airways during acute infection, and lack of this inflammatory response led to deficient immune cell activation and delayed bacterial clearance. These findings are the first to report the importance of the NLRP3 inflammasome in regulating the inflammatory response and influencing the progression of M. pneumoniae during acute infection.


Subject(s)
Immunity, Innate/immunology , Inflammation/metabolism , Mycoplasma pneumoniae/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/metabolism , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/immunology , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/immunology , Caspase 1/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/microbiology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Pneumonia, Mycoplasma/microbiology , Signal Transduction/immunology
6.
Ann Allergy Asthma Immunol ; 119(2): 146-152.e2, 2017 08.
Article in English | MEDLINE | ID: mdl-28634021

ABSTRACT

BACKGROUND: Acute infections with Mycoplasma pneumoniae (Mp) have been associated with worsening asthma in children. Mp can be present in the respiratory tract for extended periods; it is unknown whether the long-term persistence of Mp in the respiratory tract affects long-term asthma control. OBJECTIVE: To determine the effect of Mp on asthma control. METHODS: We enrolled 31 pediatric subjects 3 to 10 years of age with persistent asthma who completed up to 8 visits over a 24-month period. We detected Mp by antigen capture and polymerase chain reaction. Primary outcome measurements included symptom scores, quality of life, medication scores, oral corticosteroid use, health care usage, school absences, and exhaled breath condensate pH. RESULTS: Low levels of Mp community-acquired respiratory distress syndrome toxin were detected in 20 subjects (64.5%) at enrollment. Subjects with Mp positivity at a given visit had a .579 probability of remaining Mp positive at the subsequent visit, whereas those with Mp negativity had a .348 probability of becoming Mp positive at the following visit. The incidence of Mp overall was higher in the spring and summer months. Overall, we found no significant relation between the detection of Mp and worse outcome measurements at the same visit or at subsequent visits. CONCLUSION: The long-term persistence of Mp in the respiratory tract is common in children with asthma. However, the detection of Mp was not associated significantly with worse asthma symptoms, quality of life, health care usage, school absences, or exhaled breath condensate pH in this pediatric asthma cohort.


Subject(s)
Asthma/immunology , Asthma/microbiology , Health Status , Mycoplasma pneumoniae/isolation & purification , Quality of Life , Respiratory System/microbiology , Child , Child, Preschool , Female , Humans , Male , Mycoplasma pneumoniae/immunology , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/microbiology , Prospective Studies , Seasons
7.
J Immunol ; 195(9): 4426-37, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26385519

ABSTRACT

Recognition of viral dsRNA by endosomal TLR3 activates innate immune response during virus infection. Trafficking of TLR3 to the endolysosomal compartment arising from fusion of late endosome (LE) with lysosome is required for recognition and detection of pathogen associated molecular patterns, which results in activation of the TLR3-dependent signaling cascade. Existing knowledge about the mechanism(s) and cellular factor(s) governing TLR3 trafficking is limited. In the current study, we identified intracellular S100A9 protein as a critical regulator of TLR3 trafficking. S100A9 was required for maturation of TLR3 containing early endosome (EE) into LE, the compartment that fuses with lysosome to form the endolysosomal compartment. A drastic reduction in cytokine production was observed in S100A9-knockout (KO) primary macrophages following RNA virus infection and treatment of cells with polyinosinic-polycytidylic acid (polyIC; a dsRNA mimetic that acts as a TLR3 agonist). Mechanistic studies revealed colocalization and interaction of S100A9 with TLR3 following polyIC treatment. S100A9-TLR3 interaction was critical for maturation of TLR3 containing EE into LE because TLR3 could not be detected in the LE of polyIC-treated S100A9-KO macrophages. Subsequently, TLR3 failed to colocalize with its agonist (i.e., biotin-labeled polyIC) in S100A9-deficient macrophages. The in vivo physiological role of S100A9 was evident from loss of cytokine production in polyIC-treated S100A9-KO mice. Thus, we identified intracellular S100A9 as a regulator of TLR3 signaling and demonstrated that S100A9 functions during pre-TLR3 activation stages by facilitating maturation of TLR3 containing EE into LE.


Subject(s)
Calgranulin B/immunology , Macrophages/immunology , RNA Viruses/immunology , Toll-Like Receptor 3/immunology , Animals , Blotting, Western , Calgranulin B/genetics , Calgranulin B/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Macrophages/metabolism , Macrophages/virology , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Poly I-C/immunology , Poly I-C/pharmacology , Protein Transport/drug effects , Protein Transport/immunology , RNA Interference , RNA Viruses/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3/metabolism
8.
Proc Natl Acad Sci U S A ; 112(16): 5165-70, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25848012

ABSTRACT

Mycoplasma pneumoniae (Mp) infections cause tracheobronchitis and "walking" pneumonia, and are linked to asthma and other reactive airway diseases. As part of the infectious process, the bacterium expresses a 591-aa virulence factor with both mono-ADP ribosyltransferase (mART) and vacuolating activities known as Community-Acquired Respiratory Distress Syndrome Toxin (CARDS TX). CARDS TX binds to human surfactant protein A and annexin A2 on airway epithelial cells and is internalized, leading to a range of pathogenetic events. Here we present the structure of CARDS TX, a triangular molecule in which N-terminal mART and C-terminal tandem ß-trefoil domains associate to form an overall architecture distinct from other well-recognized ADP-ribosylating bacterial toxins. We demonstrate that CARDS TX binds phosphatidylcholine and sphingomyelin specifically over other membrane lipids, and that cell surface binding and internalization activities are housed within the C-terminal ß-trefoil domain. The results enhance our understanding of Mp pathogenicity and suggest a novel avenue for the development of therapies to treat Mp-associated asthma and other acute and chronic airway diseases.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Cytotoxins/chemistry , Mycoplasma pneumoniae/metabolism , Vacuoles/metabolism , 1,2-Dipalmitoylphosphatidylcholine/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , Adenosine Diphosphate Ribose/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Catalytic Domain , Cytotoxins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Phosphatidylcholines/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sphingomyelins/metabolism , Structure-Activity Relationship
9.
mBio ; 5(6)2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25538194

ABSTRACT

UNLABELLED: The inflammasome is a major regulator of inflammation through its activation of procaspase-1, which cleaves prointerleukin-1ß (pro-IL-1ß) into its mature form. IL-1ß is a critical proinflammatory cytokine that dictates the severity of inflammation associated with a wide spectrum of inflammatory diseases. NLRP3 is a key component of the inflammasome complex, and multiple signals and stimuli trigger formation of the NLRP3 inflammasome complex. In the current study, we uncovered a yet unknown mechanism of NLRP3 inflammasome activation by a pathogen-derived factor. We show that the unique bacterial ADP-ribosylating and vacuolating toxin produced by Mycoplasma pneumoniae and designated community-acquired respiratory distress syndrome (CARDS) toxin activates the NLRP3 inflammasome by colocalizing with the NLRP3 inflammasome and catalyzing the ADP-ribosylation of NLRP3. Mutant full-length CARDS toxin lacking ADP-ribosyltransferase (ADPRT) activity and truncated CARDS toxins unable to bind to macrophages and be internalized failed to activate the NLRP3 inflammasome. These studies demonstrate that CARDS toxin-mediated ADP-ribosylation constitutes an important posttranslational modification of NLRP3, that ADPRT activity of CARDS toxin is essential for NLRP3 inflammasome activation, and that posttranslational ADPRT-mediated modification of the inflammasome is a newly discovered mechanism for inflammasome activation with subsequent release of IL-1ß and associated pathologies. IMPORTANCE: Inflammation is a fundamental innate immune response to environmental factors, including infections. The inflammasome represents a multiprotein complex that regulates inflammation via its ability to activate specific proinflammatory cytokines, resulting in an effective host protective response. However, excessive release of proinflammatory cytokines can occur following infection that skews the host response to "hyperinflammation" with exaggerated tissue damage. Mycoplasma pneumoniae, a common bacterial airway pathogen, possesses a unique protein toxin with ADP-ribosyltransferase and vacuolating properties capable of reproducing the robust inflammation and cytopathology associated with mycoplasma infection. Here, we show that the toxin uniquely activates the NLRP3 inflammasome by colocalizing with and ADP-ribosylating NLRP3, possibly leading to "hyperinflammation" and thus uncovering a novel target for therapeutic intervention.


Subject(s)
Adenosine Diphosphate/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Carrier Proteins/metabolism , Host-Pathogen Interactions , Inflammasomes/metabolism , Mycoplasma pneumoniae/physiology , Protein Processing, Post-Translational , Animals , Cells, Cultured , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Mycoplasma pneumoniae/pathogenicity , NLR Family, Pyrin Domain-Containing 3 Protein
10.
mBio ; 5(4)2014 Aug 19.
Article in English | MEDLINE | ID: mdl-25139904

ABSTRACT

UNLABELLED: Mycoplasma pneumoniae synthesizes a novel human surfactant protein A (SP-A)-binding cytotoxin, designated community-acquired respiratory distress syndrome (CARDS) toxin, that exhibits ADP-ribosylating and vacuolating activities in mammalian cells and is directly linked to a range of acute and chronic airway diseases, including asthma. In our attempt to detect additional CARDS toxin-binding proteins, we subjected the membrane fraction of human A549 airway cells to affinity chromatography using recombinant CARDS toxin as bait. A 36-kDa A549 cell membrane protein bound to CARDS toxin and was identified by time of flight (TOF) mass spectroscopy as annexin A2 (AnxA2) and verified by immunoblotting with anti-AnxA2 monoclonal antibody. Dose-dependent binding of CARDS toxin to recombinant AnxA2 reinforced the specificity of the interaction, and further studies revealed that the carboxy terminus of CARDS toxin mediated binding to AnxA2. In addition, pretreatment of viable A549 cells with anti-AnxA2 monoclonal antibody or AnxA2 small interfering RNA (siRNA) reduced toxin binding and internalization. Immunofluorescence analysis of CARDS toxin-treated A549 cells demonstrated the colocalization of CARDS toxin with cell surface-associated AnxA2 upon initial binding and with intracellular AnxA2 following toxin internalization. HepG2 cells, which express low levels of AnxA2, were transfected with a plasmid expressing AnxA2 protein, resulting in enhanced binding of CARDS toxin and increased vacuolization. In addition, NCI-H441 cells, which express both AnxA2 and SP-A, upon AnxA2 siRNA transfection, showed decreased binding and subsequent vacuolization. These results indicate that CARDS toxin recognizes AnxA2 as a functional receptor, leading to CARDS toxin-induced changes in mammalian cells. IMPORTANCE: Host cell susceptibility to bacterial toxins is usually determined by the presence and abundance of appropriate receptors, which provides a molecular basis for toxin target cell specificities. To perform its ADP-ribosylating and vacuolating activities, community-acquired respiratory distress syndrome (CARDS) toxin must bind to host cell surfaces via receptor-mediated events in order to be internalized and trafficked effectively. Earlier, we reported the binding of CARDS toxin to surfactant protein A (SP-A), and here we show how CARDS toxin uses an alternative receptor to execute its pathogenic properties. CARDS toxin binds selectively to annexin A2 (AnxA2), which exists both on the cell surface and intracellularly. Since AnxA2 regulates membrane dynamics at early stages of endocytosis and trafficking, it serves as a distinct receptor for CARDS toxin binding and internalization and enhances CARDS toxin-induced vacuolization in mammalian cells.


Subject(s)
Annexin A2/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Mycoplasma pneumoniae/metabolism , Animals , Annexin A2/genetics , Annexin A2/immunology , Antibodies, Monoclonal , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Cell Line, Tumor , Endocytosis , Fluorescent Antibody Technique , Hep G2 Cells , Humans , Protein Binding , RNA, Small Interfering/genetics , Respiratory Distress Syndrome , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vacuoles/ultrastructure
11.
PLoS One ; 9(7): e102613, 2014.
Article in English | MEDLINE | ID: mdl-25058417

ABSTRACT

Mycoplasma pneumoniae causes a range of airway and extrapulmonary pathologies in humans. Clinically, M. pneumoniae is associated with acute exacerbations of human asthma and a worsening of experimentally induced asthma in mice. Recently, we demonstrated that Community Acquired Respiratory Distress Syndrome (CARDS) toxin, an ADP-ribosylating and vacuolating toxin synthesized by M. pneumoniae, is sufficient to induce an asthma-like disease in BALB/cJ mice. To test the potential of CARDS toxin to exacerbate preexisting asthma, we examined inflammatory responses to recombinant CARDS toxin in an ovalbumin (OVA) murine model of asthma. Differences in pulmonary inflammatory responses between treatment groups were analyzed by histology, cell differentials and changes in cytokine and chemokine concentrations. Additionally, assessments of airway hyperreactivity were evaluated through direct pulmonary function measurements. Analysis of histology revealed exaggerated cellular inflammation with a strong eosinophilic component in the CARDS toxin-treated group. Heightened T-helper type-2 inflammatory responses were evidenced by increased expression of IL-4, IL-13, CCL17 and CCL22 corresponding with increased airway hyperreactivity in the CARDS toxin-treated mice. These data demonstrate that CARDS toxin can be a causal factor in the worsening of experimental allergic asthma, highlighting the potential importance of CARDS toxin in the etiology and exacerbation of human asthma.


Subject(s)
Asthma/pathology , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Bronchial Hyperreactivity/pathology , Respiratory System/drug effects , Animals , Asthma/chemically induced , Asthma/immunology , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chemokine CCL17/biosynthesis , Chemokine CCL17/immunology , Chemokine CCL22/biosynthesis , Chemokine CCL22/immunology , Eosinophils/immunology , Eosinophils/pathology , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interleukin-13/biosynthesis , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/administration & dosage , Recombinant Proteins/toxicity , Respiratory System/immunology , Respiratory System/pathology , Th2 Cells/immunology , Th2 Cells/pathology
12.
Mol Microbiol ; 93(3): 568-81, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24948331

ABSTRACT

Community-acquired respiratory distress syndrome (CARDS) toxin from Mycoplasma pneumoniae is a 591-amino-acid virulence factor with ADP-ribosyltransferase (ADPRT) and vacuolating activities. It is expressed at low levels during in vitro growth and at high levels during colonization of the lung. Exposure of experimental animals to purified recombinant CARDS toxin alone is sufficient to recapitulate the cytopathology and inflammatory responses associated with M. pneumoniae infection in humans and animals. Here, by molecular modelling, serial truncations and site-directed mutagenesis, we show that the N-terminal region is essential for ADP-ribosylating activity. Also, by systematic truncation and limited proteolysis experiments we identified a portion of the C-terminal region that mediates toxin binding to mammalian cell surfaces and subsequent internalization. In addition, the C-terminal region alone induces vacuolization in a manner similar to full-length toxin. Together, these data suggest that CARDS toxin has a unique architecture with functionally separable N-terminal and C-terminal domains.


Subject(s)
ADP Ribose Transferases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Mycoplasma pneumoniae , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Amino Acid Motifs , Animals , Bacterial Proteins/genetics , Bacterial Toxins/genetics , HeLa Cells , Humans , Models, Molecular , NAD/metabolism , Protein Structure, Tertiary , Proteolysis , Virulence Factors/chemistry , Virulence Factors/metabolism
13.
J Bacteriol ; 196(5): 1012-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24363346

ABSTRACT

Mycoplasma genitalium is the smallest self-replicating bacterium and an important human pathogen responsible for a range of urogenital infections and pathologies. Due to its limited genome size, many genes conserved in other bacteria are missing in M. genitalium. Genes encoding catalase and superoxide dismutase are absent, and how this pathogen overcomes oxidative stress remains poorly understood. In this study, we characterized MG_427, a homolog of the conserved osmC, which encodes hydroperoxide peroxidase, shown to protect bacteria against oxidative stress. We found that recombinant MG_427 protein reduced organic and inorganic peroxide substrates. Also, we showed that a deletion mutant of MG_427 was highly sensitive to killing by tert-butyl hydroperoxide and H2O2 compared to the sensitivity of the wild type. Further, the fully complemented mutant strain reversed its oxidative sensitivity. Examination of the expression pattern of MG_427 during osmotic shock, oxidative stress, and other stress conditions revealed its lack of induction, distinguishing MG_427 from other previously characterized osmC genes.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Mycoplasma genitalium/metabolism , Bacterial Proteins/genetics , Gene Deletion , Mycoplasma genitalium/drug effects , Mycoplasma genitalium/genetics , Osmosis , Oxidative Stress , Peroxides/pharmacology , Protein Transport
14.
PLoS One ; 8(5): e62706, 2013.
Article in English | MEDLINE | ID: mdl-23667510

ABSTRACT

Bacterial toxins possess specific mechanisms of binding and uptake by mammalian cells. Mycoplasma pneumoniae CARDS (Community Acquired Respiratory Distress Syndrome) toxin is a 68 kDa protein, which demonstrates high binding affinity to human surfactant protein-A and exhibits specific biological activities including mono-ADP ribosylation and vacuolization. These properties lead to inflammatory processes in the airway and a range of cytopathologies including ciliostasis, loss of tissue integrity and injury, and cell death. However, the process by which CARDS toxin enters target cells is unknown. In this study, we show that CARDS toxin binds to mammalian cell surfaces and is internalized rapidly in a dose and time-dependent manner using a clathrin-mediated pathway, as indicated by inhibition of toxin internalization by monodansylcadaverine but not by methyl-ß-cyclodextrin or filipin. Furthermore, the internalization of CARDS toxin was markedly inhibited in clathrin-depleted cells.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clathrin/metabolism , Endocytosis , Pneumonia, Mycoplasma , Cell Line , Humans , Recombinant Proteins/metabolism , Time Factors
15.
Ann Allergy Asthma Immunol ; 110(5): 328-334.e1, 2013 May.
Article in English | MEDLINE | ID: mdl-23622002

ABSTRACT

BACKGROUND: The presence of Mycoplasma pneumoniae has been associated with worsening asthma in children. Sensitive assays have been developed to detect M pneumoniae-derived community-acquired respiratory distress syndrome (CARDS) toxin. OBJECTIVES: To identify the frequency and persistence of M pneumoniae detection in respiratory secretions of children with and without asthma and to evaluate antibody responses to M pneumoniae and the impact of M pneumoniae on biological markers, asthma control, and quality of life. METHODS: We enrolled 143 pediatric patients (53 patients with acute asthma, 26 patients with refractory asthma, and 64 healthy controls; age range, 5-17 years) during a 20-month period with 2 to 5 follow-up visits. We detected M pneumoniae using CARDS toxin antigen capture and polymerase chain reaction and P1 adhesin polymerase chain reaction. Immune responses to M pneumoniae were determined by IgG and IgM levels directed against CARDS toxin and P1 adhesin. pH was measured in exhaled breath condensates, and asthma control and quality of life were assessed using the Asthma Control Test and Pediatric Asthma Quality of Life Questionnaire. RESULTS: M pneumoniae was detected in 64% of patients with acute asthma, 65% with refractory asthma, and 56% of healthy controls. Children with asthma had lower antibody levels to M pneumoniae compared with healthy controls. Exhaled breath condensate pHs and asthma control and quality of life scores were lower in M pneumoniae-positive patients with asthma. CONCLUSION: The results suggest that M pneumoniae detection is common in children, M pneumoniae detection is associated with worsening asthma, and children with asthma may have poor humoral immune responses to M pneumoniae.


Subject(s)
Asthma/microbiology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Mycoplasma pneumoniae/immunology , Adolescent , Asthma/immunology , Breath Tests , Child , Child, Preschool , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Mycoplasma pneumoniae/metabolism , Prospective Studies , Quality of Life
16.
PLoS One ; 7(10): e47625, 2012.
Article in English | MEDLINE | ID: mdl-23091634

ABSTRACT

Mycoplasma pneumoniae is the smallest self-replicating bacterium with a streamlined genome of 0.81 Mb. Complete genome analysis revealed the presence of multiple copies of four large repetitive elements (designated RepMP1, RepMP2/3, RepMP4 and RepMP5) that are implicated in creating sequence variations among individual strains. Recently, we described RepMP1-associated sequence variations between reference strain M129 and clinical isolate S1 that involved three RepMP1-genes (i.e. mpn130, mpn137 and mpn138). Using PCR and sequencing we analyze 28 additional M. pneumoniae strains and demonstrate the existence of S1-like sequence variants in nine strains and M129-like variants in the remaining nineteen strains. We propose a series of recombination steps that facilitates transition from M129- to S1-like sequence variants. Next we examined the remaining RepMP1-genes and observed no other rearrangements related to the repeat element. The only other detected difference was varying numbers of the 21-nucleotide tandem repeats within mpn127, mpn137, mpn501 and mpn524. Furthermore, typing of strains through analysis of large RepMPs localized within the adhesin P1 operon revealed that sequence divergence involving RepMP1-genes mpn130, mpn137 and mpn138 is strictly type-specific. Once more our analysis confirmed existence of two highly conserved groups of M. pneumoniae strains.


Subject(s)
DNA, Bacterial/genetics , Genes, Bacterial , Mycoplasma pneumoniae/genetics , Repetitive Sequences, Nucleic Acid , Chromosomes, Bacterial , Gene Deletion , Gene Order , Genetic Variation , Recombination, Genetic , Tandem Repeat Sequences
17.
Am J Respir Cell Mol Biol ; 46(6): 815-22, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22281984

ABSTRACT

Mycoplasma pneumoniae causes acute and chronic lung infections in humans, leading to a variety of pulmonary and extrapulmonary sequelae. Of the airway complications of M. pneumoniae infection, M. pneumoniae-associated exacerbation of asthma and pediatric wheezing are emerging as significant sources of human morbidity. However, M. pneumoniae products capable of promoting allergic inflammation are unknown. Recently, we reported that M. pneumoniae produces an ADP-ribosylating and vacuolating toxin termed the community-acquired respiratory distress syndrome (CARDS) toxin. Here we report that naive mice exposed to a single dose of recombinant CARDS (rCARDS) toxin respond with a robust inflammatory response consistent with allergic disease. rCARDS toxin induced 30-fold increased expression of the Th-2 cytokines IL-4 and IL-13 and 70- to 80-fold increased expression of the Th-2 chemokines CCL17 and CCL22, corresponding to a mixed cellular inflammatory response comprised of a robust eosinophilia, accumulation of T cells and B cells, and mucus metaplasia. The inflammatory responses correlate temporally with toxin-dependent increases in airway hyperreactivity characterized by increases in airway restriction and decreases in lung compliance. Furthermore, CARDS toxin-mediated changes in lung function and histopathology are dependent on CD4(+) T cells. Altogether, the data suggest that rCARDS toxin is capable of inducing allergic-type inflammation in naive animals and may represent a causal factor in M. pneumoniae-associated asthma.


Subject(s)
Bacterial Toxins/toxicity , Eosinophils/cytology , Lung/drug effects , Lymphocytes/cytology , Mycoplasma pneumoniae/physiology , Animals , Bronchoalveolar Lavage Fluid , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lung/cytology , Mice , Real-Time Polymerase Chain Reaction
18.
J Infect Dis ; 204(10): 1596-604, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21957154

ABSTRACT

Mice were infected with Mycoplasma pneumoniae and monitored for the synthesis and distribution of the unique adenosine diphosphate-ribosylating and vacuolating Community Acquired Respiratory Distress Syndrome (CARDS) toxin in bronchiolar lavage fluid (BALF) and lung. We noted direct relationships between the concentration of CARDS toxin and numbers of mycoplasma genomes in BALF and the degree of histologic pulmonary inflammation. Immunostaining of lungs revealed extensive colonization by mycoplasmas, including the detection of CARDS toxin in the corresponding inflamed airways. Lung lesion scores were higher during the early stages of infection, decreased gradually by day 14 postinfection, and reached substantially lower values at day 35. Infected mouse immunoglobulin (Ig) M and IgG titers were positive for CARDS toxin as well as for the major adhesin P1 of M. pneumoniae. These data reinforce the proposed pathogenic role of CARDS toxin in M. pneumoniae-mediated pathologies.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Mycoplasma pneumoniae/metabolism , Pneumonia, Mycoplasma/microbiology , Adhesins, Bacterial/metabolism , Animals , Antibodies, Bacterial/blood , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/microbiology , Disease Models, Animal , Female , Lung/chemistry , Lung/microbiology , Mice , Mice, Inbred BALB C , Mycoplasma pneumoniae/immunology , Mycoplasma pneumoniae/isolation & purification , Mycoplasma pneumoniae/pathogenicity , Pneumonia, Mycoplasma/blood
19.
PLoS One ; 6(7): e22877, 2011.
Article in English | MEDLINE | ID: mdl-21829543

ABSTRACT

Recently, we identified an ADP-ribosylating and vacuolating cytotoxin in Mycoplasma pneumoniae designated Community Acquired Respiratory Distress Syndrome (CARDS) toxin. In this study we show that vacuoles induced by recombinant CARDS (rCARDS) toxin are acidic and derive from the endocytic pathway as determined by the uptake of neutral red and the fluid-phase marker, Lucifer yellow, respectively. Also, we demonstrate that the formation of rCARDS toxin-associated cytoplasmic vacuoles is inhibited by the vacuolar ATPase inhibitor, bafilomycin A1, and the ionophore, monensin. To examine the ontogeny of these vacuoles, we analyzed the distribution of endosomal and lysosomal membrane markers during vacuole formation and observed the enrichment of the late endosomal GTPase, Rab9, around rCARDS toxin-induced vacuoles. Immunogold-labeled Rab9 and overexpression of green fluorescent-tagged Rab9 further confirmed vacuolar association. The late endosomal- and lysosomal-associated membrane proteins, LAMP1 and LAMP2, also localized to the vacuolar membranes, while the late endosomal protein, Rab7, and early endosomal markers, Rab5 and EEA1, were excluded. HeLa cells expressing dominant-negative (DN) Rab9 exhibited markedly reduced vacuole formation in the presence of rCARDS toxin, in contrast to cells expressing DN-Rab7, highlighting the importance of Rab9 function in rCARDS toxin-induced vacuolation. Our findings reveal the unique Rab9-association with rCARDS toxin-induced vacuoles and its possible relationship to the characteristic histopathology that accompanies M. pneumoniae infection.


Subject(s)
Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Endosomes/metabolism , Lysosomes/metabolism , Vacuoles/drug effects , Vacuoles/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Antifungal Agents/pharmacology , Biomarkers/metabolism , Cells, Cultured , Chlorocebus aethiops , Cytoplasm/metabolism , Endosomes/drug effects , Genes, Dominant , HeLa Cells , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lysosomes/drug effects , Macrolides/pharmacology , Microscopy, Immunoelectron , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Vero Cells , rab GTP-Binding Proteins/genetics
20.
Mol Microbiol ; 81(2): 327-39, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21692875

ABSTRACT

Transcriptional regulation remains poorly understood in Mycoplasma genitalium, the smallest self-replicating cell and the causative agent of a spectrum of urogenital diseases. Previously, we reported that MG_149, a lipoprotein-encoding gene, was highly induced under physiological hyperosmolarity conditions. In this study we further analysed MG_149 transcription with a focus on the identification of promoter elements and regulatory mechanisms. We established MG_149 as a genuine osmoinducible gene that exhibited the highest transcript abundance compared with other lipoprotein genes. Using genetic approaches, we demonstrated that the -10 region of the MG_149 promoter was essential for osmoinduction. Moreover, we showed that MG_149 osmoinduction was regulated by DNA supercoiling, as the presence of novobiocin decreased MG_149 expression in a dose-dependent manner. Taken together, these results indicate that DNA supercoiling participates in controlling MG_149 expression during in vivo-like conditions.


Subject(s)
DNA, Bacterial/metabolism , DNA, Superhelical/metabolism , Gene Expression Regulation, Bacterial , Lipoproteins/biosynthesis , Mycoplasma genitalium/physiology , Transcription, Genetic , Anti-Bacterial Agents/metabolism , Bacterial Proteins/biosynthesis , Base Sequence , DNA Gyrase/metabolism , Molecular Sequence Data , Mycoplasma genitalium/drug effects , Mycoplasma genitalium/genetics , Mycoplasma genitalium/metabolism , Novobiocin/metabolism , Osmotic Pressure , Promoter Regions, Genetic , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...