Cargando…

Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms

Bacterial biofilms present a significant medical challenge because they are recalcitrant to current therapeutic regimes. A key component of biofilm formation in the opportunistic human pathogen Pseudomonas aeruginosa is the biosynthesis of the exopolysaccharides Pel and Psl, which are involved in th...

Descripción completa

Detalles Bibliográficos
Autores principales: Baker, Perrin, Hill, Preston J., Snarr, Brendan D., Alnabelseya, Noor, Pestrak, Matthew J., Lee, Mark J., Jennings, Laura K., Tam, John, Melnyk, Roman A., Parsek, Matthew R., Sheppard, Donald C., Wozniak, Daniel J., Howell, P. Lynne
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Association for the Advancement of Science 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4928890/
https://www.ncbi.nlm.nih.gov/pubmed/27386527
http://dx.doi.org/10.1126/sciadv.1501632
_version_ 1782440515065610240
author Baker, Perrin
Hill, Preston J.
Snarr, Brendan D.
Alnabelseya, Noor
Pestrak, Matthew J.
Lee, Mark J.
Jennings, Laura K.
Tam, John
Melnyk, Roman A.
Parsek, Matthew R.
Sheppard, Donald C.
Wozniak, Daniel J.
Howell, P. Lynne
author_facet Baker, Perrin
Hill, Preston J.
Snarr, Brendan D.
Alnabelseya, Noor
Pestrak, Matthew J.
Lee, Mark J.
Jennings, Laura K.
Tam, John
Melnyk, Roman A.
Parsek, Matthew R.
Sheppard, Donald C.
Wozniak, Daniel J.
Howell, P. Lynne
author_sort Baker, Perrin
collection PubMed
description Bacterial biofilms present a significant medical challenge because they are recalcitrant to current therapeutic regimes. A key component of biofilm formation in the opportunistic human pathogen Pseudomonas aeruginosa is the biosynthesis of the exopolysaccharides Pel and Psl, which are involved in the formation and maintenance of the structural biofilm scaffold and protection against antimicrobials and host defenses. Given that the glycoside hydrolases PelA(h) and PslG(h) encoded in the pel and psl biosynthetic operons, respectively, are utilized for in vivo exopolysaccharide processing, we reasoned that these would provide specificity to target P. aeruginosa biofilms. Evaluating these enzymes as potential therapeutics, we demonstrate that these glycoside hydrolases selectively target and degrade the exopolysaccharide component of the biofilm matrix. PelA(h) and PslG(h) inhibit biofilm formation over a 24-hour period with a half maximal effective concentration (EC(50)) of 69.3 ± 1.2 and 4.1 ± 1.1 nM, respectively, and are capable of disrupting preexisting biofilms in 1 hour with EC(50) of 35.7 ± 1.1 and 12.9 ± 1.1 nM, respectively. This treatment was effective against clinical and environmental P. aeruginosa isolates and reduced biofilm biomass by 58 to 94%. These noncytotoxic enzymes potentiated antibiotics because the addition of either enzyme to a sublethal concentration of colistin reduced viable bacterial counts by 2.5 orders of magnitude when used either prophylactically or on established 24-hour biofilms. In addition, PelA(h) was able to increase neutrophil killing by ~50%. This work illustrates the feasibility and benefits of using bacterial exopolysaccharide biosynthetic glycoside hydrolases to develop novel antibiofilm therapeutics.
format Online
Article
Text
id pubmed-4928890
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher American Association for the Advancement of Science
record_format MEDLINE/PubMed
spelling pubmed-49288902016-07-06 Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms Baker, Perrin Hill, Preston J. Snarr, Brendan D. Alnabelseya, Noor Pestrak, Matthew J. Lee, Mark J. Jennings, Laura K. Tam, John Melnyk, Roman A. Parsek, Matthew R. Sheppard, Donald C. Wozniak, Daniel J. Howell, P. Lynne Sci Adv Research Articles Bacterial biofilms present a significant medical challenge because they are recalcitrant to current therapeutic regimes. A key component of biofilm formation in the opportunistic human pathogen Pseudomonas aeruginosa is the biosynthesis of the exopolysaccharides Pel and Psl, which are involved in the formation and maintenance of the structural biofilm scaffold and protection against antimicrobials and host defenses. Given that the glycoside hydrolases PelA(h) and PslG(h) encoded in the pel and psl biosynthetic operons, respectively, are utilized for in vivo exopolysaccharide processing, we reasoned that these would provide specificity to target P. aeruginosa biofilms. Evaluating these enzymes as potential therapeutics, we demonstrate that these glycoside hydrolases selectively target and degrade the exopolysaccharide component of the biofilm matrix. PelA(h) and PslG(h) inhibit biofilm formation over a 24-hour period with a half maximal effective concentration (EC(50)) of 69.3 ± 1.2 and 4.1 ± 1.1 nM, respectively, and are capable of disrupting preexisting biofilms in 1 hour with EC(50) of 35.7 ± 1.1 and 12.9 ± 1.1 nM, respectively. This treatment was effective against clinical and environmental P. aeruginosa isolates and reduced biofilm biomass by 58 to 94%. These noncytotoxic enzymes potentiated antibiotics because the addition of either enzyme to a sublethal concentration of colistin reduced viable bacterial counts by 2.5 orders of magnitude when used either prophylactically or on established 24-hour biofilms. In addition, PelA(h) was able to increase neutrophil killing by ~50%. This work illustrates the feasibility and benefits of using bacterial exopolysaccharide biosynthetic glycoside hydrolases to develop novel antibiofilm therapeutics. American Association for the Advancement of Science 2016-05-20 /pmc/articles/PMC4928890/ /pubmed/27386527 http://dx.doi.org/10.1126/sciadv.1501632 Text en Copyright © 2016, The Authors http://creativecommons.org/licenses/by-nc/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license (http://creativecommons.org/licenses/by-nc/4.0/) , which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.
spellingShingle Research Articles
Baker, Perrin
Hill, Preston J.
Snarr, Brendan D.
Alnabelseya, Noor
Pestrak, Matthew J.
Lee, Mark J.
Jennings, Laura K.
Tam, John
Melnyk, Roman A.
Parsek, Matthew R.
Sheppard, Donald C.
Wozniak, Daniel J.
Howell, P. Lynne
Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms
title Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms
title_full Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms
title_fullStr Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms
title_full_unstemmed Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms
title_short Exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent Pseudomonas aeruginosa biofilms
title_sort exopolysaccharide biosynthetic glycoside hydrolases can be utilized to disrupt and prevent pseudomonas aeruginosa biofilms
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4928890/
https://www.ncbi.nlm.nih.gov/pubmed/27386527
http://dx.doi.org/10.1126/sciadv.1501632
work_keys_str_mv AT bakerperrin exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT hillprestonj exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT snarrbrendand exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT alnabelseyanoor exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT pestrakmatthewj exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT leemarkj exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT jenningslaurak exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT tamjohn exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT melnykromana exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT parsekmatthewr exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT shepparddonaldc exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT wozniakdanielj exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms
AT howellplynne exopolysaccharidebiosyntheticglycosidehydrolasescanbeutilizedtodisruptandpreventpseudomonasaeruginosabiofilms