Pseudomonas aeruginosa can induce infections that lead to a rapid loss of visual function. The current therapies include antibiotic treatment which reduces the bacterial burden, but fails to control tissue damage that occurs as a result of acute inflammation. The long-term objective is to develop novel therapeutic approaches that harness inflammatory responses during an acute bacterial infection, while maintaining the bactericidal activities of the host. Despite the promising studies conducted during my current R21 funding period which showed that macrophage migration inhibitory factor (MIF) has an unappreciated importance in regulating innate immune responses to P. aeruginosa in the eye, today we know very little of the cellular sources of MIF, mechanisms of MIF release, and MIF-dependent pathways. Our experiments led to the hypothesis that P. aeruginosa-induced infection triggers rapid release of homotrimeric MIF. When the immune response fails to control the infection, the persistence of MIF trimers induces sustained inflammation via CD74-dependent pathways and should be targeted for therapy to reduce tissue-damaging consequences. We propose to pursue this hypothesis by:
Aim 1. Define the cellular types and mechanisms of MIF release during bacterial keratitis. Bone marrow chimeric mice will be infected with invasive or cytotoxic P. aeruginosa strains and the kinetics, magnitude, and oligomeric state of MIF release characterized. We will investigate the molecular mechanisms of MIF protein export by examining the role of the inflammasome assembly in this process.
Aim 2. Define the biological functions of MIF trimers in response to P. aeruginosa infection. MIF deficient mice will be reconstituted with purified rMIF trimers during infection, and disease development will be characterized. The studies will be complemented by analysis of the potency of corneal epithelial responses and PMN bactericidal activities during infection.
Aim 3. Define the contribution of CD74, a cell surface MIF receptor, in regulating sensitivity to P. aeruginosa infection. We will characterize the role of MIF-CD74 interaction in regulating epithelial responses to infection by monitoring the kinetics and distribution of MIF-CD74 complexes in vitro. We will further expand these studies to determine the significance of MIF-CD74 interaction in regulating P. aeruginosa-induced pathology. The expected impact of the proposed project is to open critical opportunities for intervention during an acute P. aeruginosa-induced corneal infection aimed at curbing pathogenic consequences of excessive inflammation, while preserving key bactericidal properties of the host. Regulating the activities of these novel therapeutic targets will result in """"""""controlled"""""""" inflammatory responses during infection, limit tissue damage, and facilitate an efficient healing process.

Public Health Relevance

Bacterial keratitis is a sight-threatening complication of contact lens wear and eye trauma and Pseudomonas aeruginosa is a commonly isolated pathogen that can damage the eye integrity during keratitis. We have identified an important molecule-macrophage migration inhibitory factor (MIF)-that is involved in the pathogenesis of P.aeruginosa-induced infection and established that inhibition of MIF is therapeutically beneficial during acute P. aeruginosa-induced keratitis. The project investigates the molecular pathways, cellular types, kinetics for MIF production, and the molecular mechanisms of MIF activities.

Agency
National Institute of Health (NIH)
Institute
National Eye Institute (NEI)
Type
Research Project (R01)
Project #
5R01EY022054-02
Application #
8511664
Study Section
Special Emphasis Panel (DPVS)
Program Officer
Mckie, George Ann
Project Start
2012-08-01
Project End
2017-07-31
Budget Start
2013-08-01
Budget End
2014-07-31
Support Year
2
Fiscal Year
2013
Total Cost
$405,294
Indirect Cost
$167,794
Name
Brigham and Women's Hospital
Department
Type
DUNS #
030811269
City
Boston
State
MA
Country
United States
Zip Code
02115
Kugadas, Abirami; Wright, Quentin; Geddes-McAlister, Jennifer et al. (2017) Role of Microbiota in Strengthening Ocular Mucosal Barrier Function Through Secretory IgA. Invest Ophthalmol Vis Sci 58:4593-4600
St Leger, Anthony J; Desai, Jigar V; Drummond, Rebecca A et al. (2017) An Ocular Commensal Protects against Corneal Infection by Driving an Interleukin-17 Response from Mucosal ?? T Cells. Immunity 47:148-158.e5
Pandya, Hardik J; Kanakasabapathy, Manoj Kumar; Verma, Saloni et al. (2017) Label-free electrical sensing of bacteria in eye wash samples: A step towards point-of-care detection of pathogens in patients with infectious keratitis. Biosens Bioelectron 91:32-39
Christiansen, Stig Hill; Murphy, Ronan A; Juul-Madsen, Kristian et al. (2017) The Immunomodulatory Drug Glatiramer Acetate is Also an Effective Antimicrobial Agent that Kills Gram-negative Bacteria. Sci Rep 7:15653
Roux, Damien; Weatherholt, Molly; Clark, Bradley et al. (2017) Immune Recognition of the Epidemic Cystic Fibrosis Pathogen Burkholderia dolosa. Infect Immun 85:
Kugadas, Abirami; Gadjeva, Mihaela (2016) Impact of Microbiome on Ocular Health. Ocul Surf 14:342-9
Gauguet, Stefanie; D'Ortona, Samantha; Ahnger-Pier, Kathryn et al. (2015) Intestinal Microbiota of Mice Influences Resistance to Staphylococcus aureus Pneumonia. Infect Immun 83:4003-14
Chen, Li; Zhou, Xia; Fan, Lucy X et al. (2015) Macrophage migration inhibitory factor promotes cyst growth in polycystic kidney disease. J Clin Invest 125:2399-412
Terzulli, Marielle; Contreras-Ruiz, Laura; Ruiz, Laura Contreras et al. (2015) TSP-1 Deficiency Alters Ocular Microbiota: Implications for Sjögren's Syndrome Pathogenesis. J Ocul Pharmacol Ther 31:413-8
Shan, Qiang; Dwyer, Markryan; Rahman, Samir et al. (2014) Distinct susceptibilities of corneal Pseudomonas aeruginosa clinical isolates to neutrophil extracellular trap-mediated immunity. Infect Immun 82:4135-43

Showing the most recent 10 out of 13 publications