scholarly journals Mycoplasma pneumoniae CARDS toxin elicits a functional IgE response in Balb/c mice

PLoS ONE ◽  
2017 ◽  
Vol 12 (2) ◽  
pp. e0172447 ◽  
Author(s):  
Jorge L. Medina ◽  
Edward G. Brooks ◽  
Adriana Chaparro ◽  
Peter H. Dube
mBio ◽  
2014 ◽  
Vol 5 (6) ◽  
Author(s):  
Santanu Bose ◽  
Jesus A. Segovia ◽  
Sudha R. Somarajan ◽  
Te-Hung Chang ◽  
T. R. Kannan ◽  
...  

ABSTRACTThe inflammasome is a major regulator of inflammation through its activation of procaspase-1, which cleaves prointerleukin-1β (pro-IL-1β) into its mature form. IL-1β 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 byMycoplasma pneumoniaeand 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-1β and associated pathologies.IMPORTANCEInflammation 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.


mBio ◽  
2018 ◽  
Vol 9 (1) ◽  
Author(s):  
Kumaraguruparan Ramasamy ◽  
Sowmya Balasubramanian ◽  
Krishnan Manickam ◽  
Lavanya Pandranki ◽  
Alexander B. Taylor ◽  
...  

ABSTRACTMycoplasma pneumoniaeis 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. pneumoniaeexpresses a unique ADP-ribosylating and vacuolating cytotoxin designatedcommunity-acquiredrespiratorydistresssyndrome (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, designatedcommunity-acquiredrespiratorydistresssyndrome (CARDS) toxin. CARDS toxin alone is sufficient to induce and mimic major inflammatory and histopathological phenotypes associated withM. pneumoniaeinfection 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 eliminateM. pneumoniae-associated airway and extrapulmonary pathologies.


PLoS ONE ◽  
2013 ◽  
Vol 8 (5) ◽  
pp. e62706 ◽  
Author(s):  
Manickam Krishnan ◽  
T. R. Kannan ◽  
Joel B. Baseman

2019 ◽  
Vol 25 (1) ◽  
Author(s):  
Gang Li ◽  
Liping Fan ◽  
Yuqing Wang ◽  
Li Huang ◽  
Meijuan Wang ◽  
...  

PLoS ONE ◽  
2009 ◽  
Vol 4 (10) ◽  
pp. e7562 ◽  
Author(s):  
R. Doug Hardy ◽  
Jacqueline J. Coalson ◽  
Jay Peters ◽  
Adriana Chaparro ◽  
Chonnamet Techasaensiri ◽  
...  

PLoS ONE ◽  
2011 ◽  
Vol 6 (7) ◽  
pp. e22877 ◽  
Author(s):  
Coreen Johnson ◽  
T. R. Kannan ◽  
Joel B. Baseman

2015 ◽  
Vol 135 (2) ◽  
pp. AB153
Author(s):  
Jesus A. Segovia ◽  
Santanu Bose ◽  
Sudha R. Somarajan ◽  
Te-Hung Chang ◽  
Thirumalai Kannan ◽  
...  

CHEST Journal ◽  
2012 ◽  
Vol 142 (4) ◽  
pp. 192A
Author(s):  
Diego Maselli ◽  
Jay Peters ◽  
Jorge Medina ◽  
Oriol Sibila ◽  
Jacqueline Coalson ◽  
...  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Kumaraguruparan Ramasamy ◽  
Sowmya Balasubramanian ◽  
Alejandra Kirkpatrick ◽  
Daniel Szabo ◽  
Lavanya Pandranki ◽  
...  

AbstractMycoplasma 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.


2021 ◽  
Vol 9 (3) ◽  
Author(s):  
Shigeyuki Tamiya ◽  
Eisuke Yoshikawa ◽  
Monami Ogura ◽  
Etsushi Kuroda ◽  
Koichiro Suzuki ◽  
...  

Although Mp-induced pneumonia is usually a self-limiting disease, refractory life-threatening pneumonia is often induced. In addition, the development of alternative therapeutic strategies for Mp is expected because of the emergence of antibiotic-resistant Mp.


Sign in / Sign up

Export Citation Format

Share Document