Your browser doesn't support javascript.
loading
Microbial glycoside hydrolases as antibiofilm agents with cross-kingdom activity.
Snarr, Brendan D; Baker, Perrin; Bamford, Natalie C; Sato, Yukiko; Liu, Hong; Lehoux, Mélanie; Gravelat, Fabrice N; Ostapska, Hanna; Baistrocchi, Shane R; Cerone, Robert P; Filler, Elan E; Parsek, Matthew R; Filler, Scott G; Howell, P Lynne; Sheppard, Donald C.
Affiliation
  • Snarr BD; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada.
  • Baker P; Department of Medicine, Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
  • Bamford NC; Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
  • Sato Y; Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada.
  • Liu H; Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.
  • Lehoux M; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada.
  • Gravelat FN; Department of Medicine, Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
  • Ostapska H; Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90502.
  • Baistrocchi SR; Department of Medicine, Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
  • Cerone RP; Department of Medicine, Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
  • Filler EE; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada.
  • Parsek MR; Department of Medicine, Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
  • Filler SG; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada.
  • Howell PL; Department of Medicine, Infectious Diseases and Immunity in Global Health Program, Centre for Translational Biology, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada.
  • Sheppard DC; Department of Microbiology and Immunology, McGill University, Montreal, QC, H3A 2B4, Canada.
Proc Natl Acad Sci U S A ; 114(27): 7124-7129, 2017 07 03.
Article in En | MEDLINE | ID: mdl-28634301
ABSTRACT
Galactosaminogalactan and Pel are cationic heteropolysaccharides produced by the opportunistic pathogens Aspergillus fumigatus and Pseudomonas aeruginosa, respectively. These exopolysaccharides both contain 1,4-linked N-acetyl-d-galactosamine and play an important role in biofilm formation by these organisms. Proteins containing glycoside hydrolase domains have recently been identified within the biosynthetic pathway of each exopolysaccharide. Recombinant hydrolase domains from these proteins (Sph3h from A. fumigatus and PelAh from P. aeruginosa) were found to degrade their respective polysaccharides in vitro. We therefore hypothesized that these glycoside hydrolases could exhibit antibiofilm activity and, further, given the chemical similarity between galactosaminogalactan and Pel, that they might display cross-species activity. Treatment of A. fumigatus with Sph3h disrupted A. fumigatus biofilms with an EC50 of 0.4 nM. PelAh treatment also disrupted preformed A. fumigatus biofilms with EC50 values similar to those obtained for Sph3h In contrast, Sph3h was unable to disrupt P. aeruginosa Pel-based biofilms, despite being able to bind to the exopolysaccharide. Treatment of A. fumigatus hyphae with either Sph3h or PelAh significantly enhanced the activity of the antifungals posaconazole, amphotericin B, and caspofungin, likely through increasing antifungal penetration of hyphae. Both enzymes were noncytotoxic and protected A549 pulmonary epithelial cells from A. fumigatus-induced cell damage for up to 24 h. Intratracheal administration of Sph3h was well tolerated and reduced pulmonary fungal burden in a neutropenic mouse model of invasive aspergillosis. These findings suggest that glycoside hydrolases can exhibit activity against diverse microorganisms and may be useful as therapeutic agents by degrading biofilms and attenuating virulence.
Subject(s)
Key words

Full text: 1 Database: MEDLINE Main subject: Pseudomonas aeruginosa / Aspergillosis / Aspergillus fumigatus / Bacterial Proteins / Biofilms / Glycoside Hydrolases Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Year: 2017 Type: Article

Full text: 1 Database: MEDLINE Main subject: Pseudomonas aeruginosa / Aspergillosis / Aspergillus fumigatus / Bacterial Proteins / Biofilms / Glycoside Hydrolases Type of study: Prognostic_studies Limits: Animals / Female / Humans Language: En Year: 2017 Type: Article