S. aureus blocks efferocytosis of neutrophils by macrophages through the activity of its virulence factor alpha toxin

Bacterial pneumonia, such as those caused by Staphylococcus aureus, is associated with an influx of inflammatory neutrophils into the lung tissue and airways. Regulation and clearance of recruited neutrophils is essential for preventing tissue damage by “friendly fire”, a responsibility of macrophages in a process called efferocytosis. We hypothesized that S. aureus impairs efferocytosis by alveolar macrophages (AMs) through the activity of the secreted virulence factor alpha toxin (AT), which has been implicated in altering the antimicrobial function of AMs. Infection of mice lacking AMs resulted in significantly increased numbers of neutrophils in the lung, while clearance of neutrophils delivered intranasally into uninfected mice was reduced in AM depleted animals. In vitro, sublytic levels of AT impaired uptake of apoptotic neutrophils by purified AMs. In vivo, the presence of AT reduced uptake of neutrophils by AMs. Differential uptake of neutrophils was not due to changes in either the CD47/CD172 axis or CD36 levels. AT significantly reduced lung expression of CCN1 and altered AM surface localization of DD1α, two proteins known to influence efferocytosis. We conclude that AT may contribute to tissue damage during S. aureus pneumonia by inhibiting the ability of AM to clear neutrophils at the site of infection.

[1]  Sam W. Lee,et al.  Control of signaling-mediated clearance of apoptotic cells by the tumor suppressor p53 , 2015, Science.

[2]  J. Suzich,et al.  Staphylococcus aureus α toxin potentiates opportunistic bacterial lung infections , 2016, Science Translational Medicine.

[3]  K. Ravichandran,et al.  Phagocytosis of apoptotic cells in homeostasis , 2015, Nature Immunology.

[4]  P. Buckley,et al.  Antibody-Based Biologics and Their Promise to Combat Staphylococcus aureus Infections. , 2016, Trends in pharmacological sciences.

[5]  L. Lau,et al.  Matricellular Protein CCN1 Activates a Proinflammatory Genetic Program in Murine Macrophages , 2010, The Journal of Immunology.

[6]  J. Cinatl,et al.  Mcl-1-Mediated Impairment of the Intrinsic Apoptosis Pathway in Circulating Neutrophils from Critically Ill Patients Can Be Overcome by Fas Stimulation1 , 2009, The Journal of Immunology.

[7]  Paul M. Orwin,et al.  Exotoxins of Staphylococcus aureus. , 2000, Clinical microbiology reviews.

[8]  N. Palaniyar,et al.  NET balancing: a problem in inflammatory lung diseases , 2013, Front. Immun..

[9]  A. Prince,et al.  Toxin-Induced Necroptosis Is a Major Mechanism of Staphylococcus aureus Lung Damage , 2015, PLoS pathogens.

[10]  Xiaodong Wang,et al.  RIP3-mediated necrotic cell death accelerates systematic inflammation and mortality , 2015, Proceedings of the National Academy of Sciences.

[11]  Yoon-Keun Kim,et al.  CCN1 Secretion Induced by Cigarette Smoking Extracts Augments IL-8 Release from Bronchial Epithelial Cells , 2013, PloS one.

[12]  Y. Tesfaigzi,et al.  CCN1 secretion and cleavage regulate the lung epithelial cell functions after cigarette smoke. , 2014, American journal of physiology. Lung cellular and molecular physiology.

[13]  A. Prince,et al.  Activation of inflammasome signaling mediates pathology of acute P. aeruginosa pneumonia. , 2013, The Journal of clinical investigation.

[14]  A. Prince,et al.  Cystic fibrosis: a mucosal immunodeficiency syndrome , 2012, Nature Medicine.

[15]  T. V. Kolesnikova,et al.  CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[16]  Ki-Hyun Kim,et al.  The Matricellular Protein CCN1 Mediates Neutrophil Efferocytosis in Cutaneous Wound Healing , 2015, Nature Communications.

[17]  Naftali Kaminski,et al.  Comprehensive gene expression profiles reveal pathways related to the pathogenesis of chronic obstructive pulmonary disease. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[18]  K. Dalhoff,et al.  Impaired neutrophil exocytosis in patients with severe pneumonia , 1999, Intensive Care Medicine.

[19]  Robin R. Chamberland,et al.  Staphylococcus aureus α-hemolysin mediates virulence in a murine model of severe pneumonia through activation of the NLRP3 inflammasome. , 2012, The Journal of infectious diseases.

[20]  Jun Xu,et al.  Extracellular histones are major mediators of death in sepsis , 2009, Nature Medicine.

[21]  A. Prince,et al.  Bacterial Pathogens Activate a Common Inflammatory Pathway through IFNλ Regulation of PDCD4 , 2013, PLoS pathogens.

[22]  Mallary C Greenlee-Wacker,et al.  Phagocytosis of Staphylococcus aureus by Human Neutrophils Prevents Macrophage Efferocytosis and Induces Programmed Necrosis , 2014, The Journal of Immunology.

[23]  Seamus J. Martin,et al.  Inflammatory outcomes of apoptosis, necrosis and necroptosis , 2014, Biological chemistry.

[24]  T. Standiford,et al.  Extracellular histones are essential effectors of C5aR‐ and C5L2‐mediated tissue damage and inflammation in acute lung injury , 2013, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[25]  K. Moore,et al.  Response to Staphylococcus aureus requires CD36-mediated phagocytosis triggered by the COOH-terminal cytoplasmic domain , 2005, The Journal of cell biology.

[26]  A. Prince,et al.  CD4+ T cells promote the pathogenesis of Staphylococcus aureus pneumonia. , 2015, The Journal of infectious diseases.

[27]  S. Fortune,et al.  Efferocytosis is an innate antibacterial mechanism. , 2012, Cell host & microbe.

[28]  Simon C Watkins,et al.  CYR61 (CCN1) Is Essential for Placental Development and Vascular Integrity , 2002, Molecular and Cellular Biology.

[29]  M. Matthay,et al.  The acute respiratory distress syndrome. , 1996, The New England journal of medicine.

[30]  J. Hudcová,et al.  α-Hemolysin activity of methicillin-susceptible Staphylococcus aureus predicts ventilator-associated pneumonia. , 2014, American journal of respiratory and critical care medicine.

[31]  L. Lau,et al.  Cyr61, a product of a growth factor-inducible immediate-early gene, promotes cell proliferation, migration, and adhesion , 1996, Molecular and cellular biology.

[32]  K. Preissner,et al.  Neutrophil Extracellular Traps Directly Induce Epithelial and Endothelial Cell Death: A Predominant Role of Histones , 2012, PloS one.

[33]  M. Febbraio,et al.  CD36 Is Essential for Regulation of the Host Innate Response to Staphylococcus aureus α-Toxin–Mediated Dermonecrosis , 2015, The Journal of Immunology.

[34]  A. Condliffe,et al.  The neutrophil in chronic obstructive pulmonary disease. , 2013, American journal of respiratory cell and molecular biology.

[35]  I. Autenrieth,et al.  Bacteria induce CTGF and CYR61 expression in epithelial cells in a lysophosphatidic acid receptor-dependent manner. , 2008, International journal of medical microbiology : IJMM.

[36]  I. Inoshima,et al.  A Staphylococcus aureus Pore-Forming Toxin Subverts the Activity of ADAM10 to Cause Lethal Infection , 2011, Nature Medicine.

[37]  J. Suzich,et al.  Assessment of an Anti-Alpha-Toxin Monoclonal Antibody for Prevention and Treatment of Staphylococcus aureus-Induced Pneumonia , 2013, Antimicrobial Agents and Chemotherapy.

[38]  B. Wilson,et al.  Survival of Staphylococcus aureus Inside Neutrophils Contributes to Infection1 , 2000, The Journal of Immunology.