Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SOLUBLE BACTERIAL AND FUNGAL PROTEINS AND METHODS AND USES THEREOF IN INHIBITING AND DISPERSING BIOFILM
Document Type and Number:
WIPO Patent Application WO/2015/184526
Kind Code:
A1
Abstract:
The present disclosure relates to methods of treating or preventing a biofilm-related infection and methods of preventing and treating biofilm formation on indwelling medical devices, implants, and non-medical surfaces comprising administering at least one soluble microbial protein that is encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, wherein the protein comprises a glycosyl hydrolase domain. The present disclosure further provides particular soluble glycosyl hydrolases and compositions thereof.

Inventors:
HOWELL LYNNE (CA)
BAKER PERRIN (CA)
ALNABELSEYA NOOR (US)
BAMFORD NATALIE (CA)
LITTLE DUSTIN (CA)
SHEPPARD DONALD (CA)
SNARR BRENDAN (CA)
LEE MARK JAE (US)
Application Number:
PCT/CA2015/000361
Publication Date:
December 10, 2015
Filing Date:
June 05, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HOSPITAL FOR SICK CHILDREN (CA)
UNIV MCGILL (CA)
International Classes:
C12N9/24; A01N63/50; A01P1/00; A01P3/00; A61K38/16; A61L2/16; A61L27/22; A61L27/54; A61L31/00; A61L31/10; A61L31/16; C12N7/01; C12N15/56; C12N15/63; C12N15/80
Foreign References:
US7989604B22011-08-02
US7989604B22011-08-02
US5264558A1993-11-23
US20050186666A12005-08-25
Other References:
CHEN, M. ET AL.: "Novel strategies for the prevention and treatment of biofilm related infections", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 14, no. 9, 6 September 2013 (2013-09-06), pages 18488 - 18501, XP055241209, ISSN: 1422-0067
GRAVELAT, F. ET AL.: "Identification of a gene cluster mediating the biosynthesis of the Aspergillus fumigatus cell wall and secreted polysaccharide, galactosaminogalactan", THE TENTH INTERNATIONAL ASPERGILLUS MEETING, March 2013 (2013-03-01), XP055362528, Retrieved from the Internet
LOUSSERT, C. ET AL.: "In vivo biofilm composition of Aspergillus fumigatus", CELLULAR MICROBIOLOGY, vol. 12, no. 3, 2010, pages 405 - 410, XP009169676, ISSN: 1462-5814
"GenBank", Database accession no. EAL92787.1
KROGH ALARSSON BVON HEIJNE GSONNHAMMER EL.: "Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes", JOURNAL OF MOLECULAR BIOLOGY, vol. 305, 2001, pages 567 - 80, XP004469188, DOI: 10.1006/jmbi.2000.4315
AUSUBEL ET AL.: "Current Protocols in Molecular Biology", 1992, GREENE PUBLISHING ASSOCIATES
KARLINALTSCHUL, PROC. NATL. ACAD. SCI. U.S.A., vol. 87, 1990, pages 2264 - 2268
KARLINALTSCHUL, PROC. NATL. ACAD. SCI. U.S.A., vol. 90, 1993, pages 5873 - 5877
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
MYERSMILLER, CABIOS, vol. 4, 1988, pages 11 - 17
DAVIES D.: "Understanding biofilm resistance to antibacterial agents", NATURE REVIEWS. DRUG DISCOVERY, vol. 2, 2003, pages 114 - 22, XP002637700
COSTERTON JWSTEWART PSGREENBERG EP: "Bacterial biofilms: a common cause of persistent infections", SCIENCE, vol. 284, 1999, pages 1318 - 22, XP002954870, DOI: 10.1126/science.284.5418.1318
GOEDDEL: "Gene Expression Technology: Methods in Enzymology", vol. 185, 1991, ACADEMIC PRESS
ZHANG ET AL., SCIENCE, vol. 303, no. 5656, 2004, pages 371 - 3
ADAMS PDAFONINE PVBUNKOCZI GCHEN VBDAVIS IW ET AL.: "PHENIX: a comprehensive Python-based system for macromolecular structure solution", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 66, 2010, pages 213 - 21
AFONINE PVMUSTYAKIMOV MGROSSE-KUNSTLEVE RWMORIARTY NWLANGAN PADAMS PD: "Joint X-ray and neutron refinement with phenix.refine", ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY, vol. 66, 2010, pages 1153 - 63
ALHEDE MKRAGH KNQVORTRUP KALLESEN-HOLM MVAN GENNIP M ET AL.: "Phenotypes of non-attached Pseudomonas aeruginosa aggregates resemble surface attached biofilm", PLOS ONE, vol. 6, 2011, pages e27943
ALTSCHUL SFGISH WMILLER WMYERS EWLIPMAN DJ.: "Basic local alignment search tool", J MOL BIOL, vol. 215, 1990, pages 403 - 10, XP002949123, DOI: 10.1006/jmbi.1990.9999
ANTHON GEBARRETT DM.: "Determination of reducing sugars with 3-methyl-2-benzothiazolinonehydrazone", ANALYTICAL BIOCHEMISTRY, vol. 305, 2002, pages 287 - 9, XP027227033
ASHKENAZY HEREZ EMARTZ EPUPKO TBEN-TAL N.: "ConSurf 2010: calculating evolutionary conservation in sequence and structure of proteins and nucleic acids", NUCLEIC ACIDS RES, vol. 38, 2010, pages W529 - 33
BAGOS PGNIKOLAOU EPLIAKOPOULOS TDTSIRIGOS KD: "Combined prediction of Tat and Sec signal peptides with hidden Markov models", BIOINFORMATICS, vol. 26, 2010, pages 2811 - 7
BAKKEVIG KSLETTA HGIMMESTAD MAUNE RERTESVAG H ET AL.: "Role of the Pseudomonas fluorescens alginate lyase (AlgL) in clearing the periplasm of alginates not exported to the extracellular environment", J BACTERIOL, vol. 187, 2005, pages 8375 - 84
BARDAS GAVELOUKAS TKOUTITA OKARAOGLANIDIS GS.: "Multiple resistance of Botrytis cinerea from kiwifruit to SDHIs, Qols and fungicides of other chemical groups", PEST MANAGEMENT SCIENCE, vol. 66, 2010, pages 967 - 73
BILLINGS NMILLAN MCALDARA MRUSCONI RTARASOVA Y ET AL.: "The extracellular matrix Component Psl provides fast-acting antibiotic defense in Pseudomonas aeruginosa biofilms", PLOS PATHOG, vol. 9, 2013, pages e1003526
BJARNSHOLT T: "The role of bacterial biofilms in chronic infections", APMIS, 2013
BJARNSHOLT TCIOFU OMOLIN SGIVSKOV MHOIBY N: "Applying insights from biofilm biology to drug development - can a new approach be developed?", NATURE REVIEWS. DRUG DISCOVERY, vol. 12, 2013, pages 791 - 808, XP055381449, DOI: 10.1038/nrd4000
BRAGONZI AFARULLA IPARONI MTWOMEY KBPIRONE L ET AL.: "Modelling co-infection of the cystic fibrosis lung by Pseudomonas aeruginosa and Burkholderia cenocepacia reveals influences on biofilm formation and host response", PLOS ONE, vol. 7, 2012, pages e52330
BRANDA SSVIK SFRIEDMAN LKOLTER R., TRENDS IN MICROBIOLOGY, vol. 13, 2005, pages 20 - 6
BYRD MSPANG BHONG WWALIGORA EAJUNEAU RA ET AL.: "Direct evaluation of Pseudomonas aeruginosa biofilm mediators in a chronic infection model", INFECTION AND IMMUNITY, vol. 79, 2011, pages 3087 - 95
BYRD MSPANG BMISHRA MSWORDS WEWOZNIAK DJ.: "The Pseudomonas aeruginosa exopolysaccharide Psl facilitates surface adherence and NF-kappaB activation in A549 cells", MBIO 1, 2010
BYRD MSSADOVSKAYA IVINOGRADOV ELU HSPRINKLE AB ET AL.: "Genetic and biochemical analyses of the Pseudomonas aeruginosa Psl exopolysaccharide reveal overlapping roles for polysaccharide synthesis enzymes in Psl and LPS production", MOL MICROBIOL, vol. 73, 2009, pages 622 - 38, XP055163264, DOI: 10.1111/j.1365-2958.2009.06795.x
CERCA NJEFFERSON KKMAIRA-LITRAN TPIER DBKELLY-QUINTOS C ET AL.: "Molecular basis for preferential protective efficacy of antibodies directed to the poorly acetylated form of staphylococcal poly-N-acetyl-beta-(1-6)-glucosaminev", INFECT IMMUN, vol. 75, 2007, pages 3406 - 13
CHOI AHSLAMTI LAVCI FYPIER GBMAIRA-LITRAN T.: "The pgaABCD locus of Acinetobacter baumannii encodes the production of poly-beta-1-6-N-acetylglucosamine, which is critical for biofilm formation", J BACTERIOL, vol. 191, 2009, pages 5953 - 63
COLVIN KMALNABELSEYA NBAKER PWHITNEY JCHOWELL PLPARSEK MR: "PelA deacetylase activity is required for Pel polysaccharide synthesis in Pseudomonas aeruginosa", J BACTERIOL, vol. 195, 2013, pages 2329 - 39
COLVIN KMGORDON VDMURAKAMI KBORLEE BRWOZNIAK DJ ET AL.: "The pel polysaccharide can serve a structural and protective role in the biofilm matrix of Pseudomonas aeruginosa", PLOS PATHOGENS 7, pages e1001264
COLVIN KMIRIE YTART CSURBANO RWHITNEY JC ET AL.: "The Pel and Psl polysaccharides provide Pseudomonas aeruginosa structural redundancy within the biofilm matrix", ENVIRONMENTAL MICROBIOLOGY, vol. 14, 2012, pages 1913 - 28, XP055295391, DOI: 10.1111/j.1462-2920.2011.02657.x
CONOVER MSSLOAN GPLOVE CFSUKUMAR NDEORA R.: "The Bps polysaccharide of Bordetella pertussis promotes colonization and biofilm formation in the nose by functioning as an adhesin", MOL MICROBIOL, vol. 77, 2010, pages 1439 - 55, XP055346089, DOI: 10.1111/j.1365-2958.2010.07297.x
COSTERTON JWCHENG KJGEESEY GGLADD TINICKEL JC ET AL.: "Bacterial biofilms in nature and disease", ANNUAL REVIEW OF MICROBIOLOGY, vol. 41, 1987, pages 435 - 64
CYWES-BENTLEY CSKURNIK DZAIDI TROUX DDEOLIVEIRA RB ET AL.: "Antibody to a conserved antigenic target is protective against diverse prokaryotic and eukaryotic pathogens", PROC NATL ACAD SCI USA, vol. 110, 2013, pages E2209 - 18, XP055222532, DOI: 10.1073/pnas.1303573110
DAROUICHE ROMANSOURI MDGAWANDE PVMADHYASTHA S.: "Antimicrobial and antibiofilm efficacy of triclosan and DispersinB combination", THE JOURNAL OF ANTIMICROBIAL CHEMOTHERAPY, vol. 64, 2009, pages 88 - 93, XP055419589, DOI: 10.1093/jac/dkp158
DAVIES GHENRISSAT B.: "Structures and mechanisms of glycosyl hydrolases", STRUCTURE, vol. 3, 1995, pages 853 - 9, XP004587916, DOI: 10.1016/S0969-2126(01)00220-9
DEAN RVAN KAN JAPRETORIUS ZAHAMMOND-KOSACK KEDI PIETRO A ET AL.: "The Top 10 fungal pathogens in molecular plant pathology", MOLECULAR PLANT PATHOLOGY, vol. 13, 2012, pages 414 - 30, XP055218173, DOI: 10.1111/j.1364-3703.2011.00783.x
DIGIANDOMENICO AWARRENER PHAMILTON MGUILLARD SRAVN P ET AL.: "Identification of broadly protective human antibodies to Pseudomonas aeruginosa exopolysaccharide Psl by phenotypic screening", THE JOURNAL OF EXPERIMENTAL MEDICINE, vol. 209, 2012, pages 1273 - 87, XP055278355, DOI: 10.1084/jem.20120033
DOLINSKY TJCZODROWSKI PLI HNIELSEN JEJENSEN JH ET AL.: "PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulations", NUCLEIC ACIDS RES, vol. 35, 2007, pages W522 - 5
DONELLI GFRANCOLINI IROMOLI DGUAGLIANONE EPIOZZI A ET AL.: "Synergistic activity of dispersin B and cefamandole nafate in inhibition of staphylococcal biofilm growth on polyurethanes", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 51, 2007, pages 2733 - 40
ELLIS MRICHARDSON MDE PAUW B. 2000: "Epidemiology", HOSPITAL MEDICINE, vol. 61, pages 605 - 9
EMSLEY PCOWTAN K.: "Coot: model-building tools for molecular graphics", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 60, 2004, pages 2126 - 32
ESCOBAR-CHAVEZ JJLOPEZ-CERVANTES MNAIK AKALIA YNQUINTANAR-GUERRERO DGANEM-QUINTANAR A: "Applications of thermo-reversible pluronic F-127 gels in pharmaceutical formulations", JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES, vol. 9, 2006, pages 339 - 58, XP002415851
FLEMMING HCWINGENDER J.: "The biofilm matrix", NATURE REVIEWS. MICROBIOLOGY, vol. 8, 2010, pages 623 - 33
FONTAINE TDELANGLE ASIMENEL CCODDEVILLE BVAN VLIET SJ ET AL.: "Galactosaminogalactan, a new immunosuppressive polysaccharide of Aspergillus fumigatus", PLOS PATHOGENS, vol. 7, 2011, pages e1002372
FRANKLIN MJNIVENS DEWEADGE JTHOWELL PL: "Biosynthesis of the Pseudomonas aeruginosa Extracellular Polysaccharides, Alginate, Pel, and Psl", FRONT MICROBIOL, vol. 2, 2011, pages 167
FRIEDMAN LKOLTER R.: "Genes involved in matrix formation in Pseudomonas aeruginosa PA14 biofilms", MOL MICROBIOL, vol. 51, 2004, pages 675 - 90
FRIEDMAN LKOLTER R.: "Two genetic loci produce distinct carbohydrate-rich structural components of the Pseudomonas aeruginosa biofilm matrix", J BACTERIOL, vol. 186, 2004, pages 4457 - 65
GAWANDE PVLEUNG KPMADHYASTHA S.: "Antibiofilm and Antimicrobial Efficacy of DispersinB-KSL-W Peptide-Based Wound Gel Against Chronic Wound Infection Associated Bacteria", CURRENT MICROBIOLOGY, 2014
GEISER DMKLICH MAFRISVAD JCPETERSON SWVARGA JSAMSON RA: "The current status of species recognition and identification in Aspergillus", STUDIES IN MYCOLOGY, vol. 59, 2007, pages 1 - 10
GRABKE AFERNANDEZ-ORTUNO DAMIRI ALI XPERES NA ET AL.: "Characterization of iprodione resistance in Botrytis cinerea from strawberry and blackberry", PHYTOPATHOLOGY, vol. 104, 2014, pages 396 - 402
GRAVELAT FNBEAUVAIS ALIU HLEE MJSNARR BD ET AL.: "Aspergillus galactosaminogalactan mediates adherence to host constituents and conceals hyphal beta-glucan from the immune system", PLOS PATHOGENS, vol. 9, 2013, pages e1003575
GRAVELAT FNDOEDT TCHIANG LYLIU HFILLER SG ET AL.: "In vivo analysis of Aspergillus fumigatus developmental gene expression determined by real-time reverse transcription-PCR", INFECT. IMMUN., vol. 76, 2008, pages 3632 - 39
HALL-STOODLEY LCOSTERTON JWSTOODLEY P.: "Bacterial biofilms: from the natural environment to infectious diseases", NAT REV MICROBIOL, vol. 2, 2004, pages 95 - 108, XP002438975
HARE NJSOLIS NHARMER CMARZOOK NBROSE B ET AL.: "Proteomic profiling of Pseudomonas aeruginosa AES-1 R, PA01 and PA14 reveals potential virulence determinants associated with a transmissible cystic fibrosis-associated strain", BMC MICROBIOLOGY, vol. 12, 2012, pages 16, XP021117886, DOI: 10.1186/1471-2180-12-16
HENRISSAT BBAIROCH A: "Updating the sequence-based classification of glycosyl hydrolases", THE BIOCHEMICAL JOURNAL, vol. 316, no. 2, 1996, pages 695 - 6, XP001176681
HOFFMAN LRD'ARGENIO DAMACCOSS MJZHANG ZJONES RAMILLER SI: "Aminoglycoside antibiotics induce bacterial biofilm formation", NATURE, vol. 436, 2005, pages 1171 - 5
HOIBY NBJARNSHOLT TGIVSKOV MMOLIN SCIOFU O.: "Antibiotic resistance of bacterial biofilms", INT J ANTIMICROB AGENTS, vol. 35, 2010, pages 322 - 32, XP026915279, DOI: 10.1016/j.ijantimicag.2009.12.011
HOIBY NCIOFU OJOHANSEN HKSONG ZJMOSER C ET AL.: "The clinical impact of bacterial biofilms", INTERNATIONAL JOURNAL OF ORAL SCIENCE, vol. 3, pages 55 - 65
HOLM LKAARIAINEN SROSENSTROM PSCHENKEL A.: "Searching protein structure databases with DaliLite v.3.", BIOINFORMATICS, vol. 24, 2008, pages 2780 - 1
HUSE HKKWON TZLOSNIK JESPEERT DPMARCOTTE EMWHITELEY M.: "Pseudomonas aeruginosa Enhances Production of a Non-Alginate Exopolysaccharide during Long-Term Colonization of the Cystic Fibrosis Lung", PLOS ONE, vol. 8, 2013, pages e82621
IRIE YBORLEE BRO'CONNOR JRHILL PJHARWOOD CS ET AL.: "Self-produced exopolysaccharide is a signal that stimulates biofilm formation in Pseudomonas aeruginosa", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 109, 2012, pages 20632 - 6
ITOH YRICE JDGOLLER CPANNURI ATAYLOR J ET AL.: "Roles of pgaABCD genes in synthesis, modification, and export of the Escherichia coli biofilm adhesin poly-beta-1,6-N-acetyl-D-glucosamine", JOURNAL OF BACTERIOLOGY, vol. 190, 2008, pages 3670 - 80, XP055346263, DOI: 10.1128/JB.01920-07
ITOH YWANG XHINNEBUSCH BJPRESTON JF3RD, ROMEO T.: "Depolymerization of beta-1,6-N-acetyl-D-glucosamine disrupts the integrity of diverse bacterial biofilms", JOURNAL OF BACTERIOLOGY, vol. 187, 2005, pages 382 - 7
IZANO EASADOVSKAYA IVINOGRADOV EMULKS MHVELLIYAGOUNDER K ET AL.: "Poly-N-acetylglucosamine mediates biofilm formation and antibiotic resistance in Actinobacillus pleuropneumoniae", MICROBIAL PATHOGENESIS, vol. 43, 2007, pages 1 - 9, XP022065093, DOI: 10.1016/j.micpath.2007.02.004
IZANO EASADOVSKAYA IWANG HVINOGRADOV ERAGUNATH C ET AL.: "Poly-N-acetylglucosamine mediates biofilm formation and detergent resistance in Aggregatibacter actinomycetemcomitans", MICROBIAL PATHOGENESIS, vol. 44, 2008, pages 52 - 60, XP022388638, DOI: 10.1016/j.micpath.2007.08.004
JACKSON KDSTARKEY MKREMER SPARSEK MRWOZNIAK DJ.: "Identification of psl, a locus encoding a potential exopolysaccharide that is essential for Pseudomonas aeruginosa PA01 biofilm formation", JOURNAL OF BACTERIOLOGY, vol. 186, 2004, pages 4466 - 75
JAIN SOHMAN DE: "Deletion of algK in mucoid Pseudomonas aeruginosa blocks alginate polymer formation and results in uronic acid secretion", JOURNAL OF BACTERIOLOGY, vol. 180, 1998, pages 634 - 41
JARRETT CODEAK EISHERWOOD KEOYSTON PCFISCHER ER ET AL., TRANSMISSION OF YERSINIA PESTIS FROM AN INFECTIOUS BIOFILM IN THE FLEA VECTOR. J INFECT DIS, vol. 190, 2004, pages 783 - 92
KAIL LKROGH ASONNHAMMER EL.: "A combined transmembrane topology and signal peptide prediction method", JOURNAL OF MOLECULAR BIOLOGY, vol. 338, 2004, pages 1027 - 36, XP004504149, DOI: 10.1016/j.jmb.2004.03.016
KAPLAN JBVELLIYAGOUNDER KRAGUNATH CROHDE HMACK D ET AL.: "Genes involved in the synthesis and degradation of matrix polysaccharide in Actinobacillus actinomycetemcomitans and Actinobacillus pleuropneumoniae biofilms", JOURNAL OF BACTERIOLOGY, vol. 186, 2004, pages 8213 - 20, XP055568931, DOI: 10.1128/JB.186.24.8213-8220.2004
KELLEY LASTERNBERG MJ.: "Protein structure prediction on the Web: a case study using the Phyre server", NATURE PROTOCOLS, vol. 4, 2009, pages 363 - 71
KIM JHAHN JSFRANKLIN MJSTEWART PSYOON J.: "Tolerance of dormant and active cells in Pseudomonas aeruginosa PA01 biofilm to antimicrobial agents", THE JOURNAL OF ANTIMICROBIAL CHEMOTHERAPY, vol. 63, 2009, pages 129 - 35
KUKAVICA-IBRULJ IBRAGONZI APARONI MWINSTANLEY CSANSCHAGRIN F ET AL.: "In vivo growth of Pseudomonas aeruginosa strains PA01 and PA14 and the hypervirulent strain LESB58 in a rat model of chronic lung infection", JOURNAL OF BACTERIOLOGY, vol. 190, 2008, pages 2804 - 13
KUMAR CGANAND SK: "Significance of microbial biofilms in food industry: a review", INTERNATIONAL JOURNAL OF FOOD MICROBIOLOGY, vol. 42, 1998, pages 9 - 27, XP002647868, DOI: 10.1016/S0168-1605(98)00060-9
LEE DGURBACH JMWU GLIBERATI NTFEINBAUM RL ET AL.: "Genomic analysis reveals that Pseudomonas aeruginosa virulence is combinatorial", GENOME BIOLOGY, vol. 7, 2006, pages R90, XP021027293, DOI: 10.1186/gb-2006-7-10-r90
LEE JECORNELL KARISCOE MKHOWELL PL: "Structure of E. coli 5'-methylthioadenosine/S-adenosylhomocysteine nucleosidase reveals similarity to the purine nucleoside phosphorylases", STRUCTURE, vol. 9, 2001, pages 941 - 53
LEROCH MPLESKEN CWEBER RWKAUFF FSCALLIET GHAHN M.: "Gray mold populations in german strawberry fields are resistant to multiple fungicides and dominated by a novel clade closely related to Botrytis cinerea", APPL ENVIRON MICROBIOL, vol. 79, 2013, pages 159 - 67
LIN SJSCHRANZ JTEUTSCH SM: "Aspergillosis case-fatality rate: systematic review of the literature", CLINICAL INFECTIOUS DISEASES : AN OFFICIAL PUBLICATION OF THE INFECTIOUS DISEASES SOCIETY OF AMERICA, vol. 32, 2001, pages 358 - 66
LITTLE DJLI GING CDIFRANCESCO BRBAMFORD NC ET AL.: "Modification and periplasmic translocation of the biofilm exopolysaccharide poiy-p-1,6-N-acetyl-D-glucosamine", PROC NATL ACAD SCI U S A SUBMITTED, 2014
LITTLE DJPOLOCZEK JWHITNEY JCROBINSON HNITZ MHOWELL PL: "The structure- and metal-dependent activity of Escherichia coli PgaB provides insight into the partial de-N-acetylation of poly-beta-1,6-N-acetyl-D-glucosamine", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 287, 2012, pages 31126 - 37
LITTLE DJWHITNEY JCROBINSON HYIP PNITZ MHOWELL PL.: "Combining in situ proteolysis and mass spectrometry to crystallize Escherichia coli PgaB", ACTA CRYSTALLOGRAPHICA. SECTION F, STRUCTURAL BIOLOGY AND CRYSTALLIZATION COMMUNICATIONS, vol. 68, 2012, pages 842 - 5
LOMBARD VGOLACONDA RAMULU HDRULA ECOUTINHO PMHENRISSAT B.: "The carbohydrate-active enzymes database (CAZy", NUCLEIC ACIDS RESEARCH, vol. 42, 2013, pages D490 - 5, XP055519748, DOI: 10.1093/nar/gkt1178
LOUSSERT CSCHMITT CPREVOST MCBALLOY V, FADEL E ET AL.: "In vivo biofilm composition of Aspergillus fumigatus", CELLULAR MICROBIOLOGY, vol. 12, 2010, pages 405 - 10, XP009169676, DOI: 10.1111/j.1462-5822.2009.01409.x
MA LCONOVER MLU HPARSEK MRBAYLES KWOZNIAK DJ.: "Assembly and development of the Pseudomonas aeruginosa biofilm matrix", PLOS PATHOGENS, vol. 5, 2009, pages e1000354, XP055163270, DOI: 10.1371/journal.ppat.1000354
MA LJACKSON KDLANDRY RMPARSEK MRWOZNIAK DJ: "Analysis of Pseudomonas aeruginosa conditional psl variants reveals roles for the psl polysaccharide in adhesion and maintaining biofilm structure postattachment", JOURNAL OF BACTERIOLOGY, vol. 188, 2006, pages 8213 - 21, XP055163311, DOI: 10.1128/JB.01202-06
MA LWANG SWANG DPARSEK MRWOZNIAK DJ: "The roles of biofilm matrix polysaccharide Psl in mucoid Pseudomonas aeruginosa biofilms", FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY, vol. 65, 2012, pages 377 - 80
MACK DFISCHER WKROKOTSCH ALEOPOLD KHARTMANN R ET AL.: "The intercellular adhesin involved in biofilm accumulation of Staphylococcus epidermidis is a linear beta-1,6-linked glucosaminoglycan: purification and structural analysis", J BACTERIOL, vol. 178, 1996, pages 175 - 83, XP000867576
MAH TFPITTS BPELLOCK BWALKER GCSTEWART PSO'TOOLE GA: "A genetic basis for Pseudomonas aeruginosa biofilm antibiotic resistance", NATURE, vol. 426, 2003, pages 306 - 10, XP001176614, DOI: 10.1038/nature02122
MAHARAJ RMAY TBWANG SKCHAKRABARTY AM: "Gene", vol. 136, 1993, article "Sequence of the alg8 and alg44 genes involved in the synthesis of alginate by Pseudomonas aeruginosa", pages: 267 - 9
MANN EEWOZNIAK DJ.: "Pseudomonas biofilm matrix composition and niche biology", FEMS MICROBIOLOGY REVIEWS, vol. 36, 2012, pages 893 - 916
MANUEL SGRAGUNATH CSAIT HBIZANO EAKAPLAN JBRAMASUBBU N.: "Role of active-site residues of dispersin B, a biofilm-releasing beta-hexosaminidase from a periodontal pathogen, in substrate hydrolysis", THE FEBS JOURNAL, vol. 274, 2007, pages 5987 - 99, XP055589742, DOI: 10.1111/j.1742-4658.2007.06121.x
MATSUKAWA MGREENBERG EP: "Putative exopolysaccharide synthesis genes influence Pseudomonas aeruginosa biofilm development", JOURNAL OF BACTERIOLOGY, vol. 186, 2004, pages 4449 - 56
MCKENNEY DPOULIOT KLWANG YMURTHY VULRICH M ET AL.: "Broadly protective vaccine for Staphylococcus aureus based on an in vivo-expressed antigen", SCIENCE, vol. 284, 1999, pages 1523 - 7, XP002129165, DOI: 10.1126/science.284.5419.1523
MERRITT JHKADOURI DE: "Current protocols in microbiology", article "O'Toole GA. 2005. Growing and analyzing static biofilms"
MICHIELSE CBREP M.: "Pathogen profile update: Fusarium oxysporum", MOLECULAR PLANT PATHOLOGY, vol. 10, 2009, pages 311 - 24
MISHRA MBYRD MSSERGEANT SAZAD AKPARSEK MR ET AL.: "Pseudomonas aeruginosa Psl polysaccharide reduces neutrophil phagocytosis and the oxidative response by limiting complement-mediated opsonization", CELLULAR MICROBIOLOGY, vol. 14, 2012, pages 95 - 106
MONDAY SRSCHILLER NL.: "Alginate synthesis in Pseudomonas aeruginosa: the role of AlgL (alginate lyase) and AlgX", JOURNAL OF BACTERIOLOGY, vol. 178, 1996, pages 625 - 32
NIEMAN CEWONG AWHE SCLARKE LHOPWOOD JJWITHERS SG: "Family 39 alpha-l-iduronidases and beta-D-xylosidases react through similar glycosyl-enzyme intermediates: identification of the human iduronidase nucleophile", BIOCHEMISTRY, vol. 42, 2003, pages 8054 - 65
OTWINOWSKI ZMINOR W.: "Processing of X-ray diffraction data collected in oscillation mode", 1997, ELSEVIER, pages: 307 - 26
PAINTER JMERRITT EA: "Optimal description of a protein structure in terms of multiple groups undergoing TLS motion", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 62, 2006, pages 439 - 50
PARISE GMISHRA MITOH YROMEO TDEORA R.: "Role of a putative polysaccharide locus in Bordetella biofilm development", J BACTERIOL, vol. 189, 2007, pages 750 - 60, XP055345700, DOI: 10.1128/JB.00953-06
PARK BHKARPINETS TVSYED MHLEUZE MRUBERBACHER EC: "CAZymes Analysis Toolkit (CAT): web service for searching and analyzing carbohydrate-active enzymes in a newly sequenced organism using CAZy database", GLYCOBIOLOGY, vol. 20, 2010, pages 1574 - 84
PETERSEN TNBRUNAK SVON HEIJNE GNIELSEN H.: "SignalP 4.0: discriminating signal peptides from transmembrane regions", NATURE METHODS, vol. 8, 2011, pages 785 - 6
POKROVSKAYA VPOLOCZEK JLITTLE DJGRIFFITHS HHOWELL PLNITZ M.: "Functional characterization of Staphylococcus epidermidis IcaB, a de-N-acetylase important for biofilm formation", BIOCHEMISTRY, vol. 52, 2013, pages 5463 - 71
RAHME LGSTEVENS EJWOLFORT SFSHAO JTOMPKINS RGAUSUBEL FM: "Common virulence factors for bacterial pathogenicity in plants and animals", SCIENCE, vol. 268, 1995, pages 1899 - 902
RODRIGUEZ AACOSTA ARODRIGUEZ C: "Fungicide resistance of Botrytis cinerea in tomato greenhouses in the Canary Islands and effectiveness of non-chemical treatments against gray mold", WORLD JOURNAL OF MICROBIOLOGY & BIOTECHNOLOGY, vol. 30, 2014, pages 2397 - 406
RYBTKE MTJENSEN POHOIBY NGIVSKOV MTOLKER-NIELSEN TBJARNSHOLT T: "The implication of Pseudomonas aeruginosa biofilms in infections", INFLAMMATION & ALLERGY DRUG TARGETS, vol. 10, 2011, pages 141 - 57, XP008175555, DOI: 10.2174/187152811794776222
SANG HHULVEY JPOPKO JTJR., LOPES JSWAMINATHAN A ET AL.: "A pleiotropic drug resistance transporter is involved in reduced sensitivity to multiple fungicide classes in Sclerotinia homoeocarpa (F.T. Bennett", MOLECULAR PLANT PATHOLOGY, vol. 16, 2015, pages 251 - 61, XP055474995, DOI: 10.1111/mpp.12174
SCHNEIDMAN-DUHOVNY DINBAR YNUSSINOV RWOLFSON HJ: "PatchDock and SymmDock: servers for rigid and symmetric docking", NUCLEIC ACIDS RESEARCH, vol. 33, 2005, pages W363 - 7
SINGH N.: "The current management of infectious diseases in the liver transplant recipient", CLINICS IN LIVER DISEASE, vol. 4, 2000, pages 657 - 73
SKURNIK DROUX DASCHARD HCATTOIR VYODER-HIMES D ET AL.: "A comprehensive analysis of in vitro and in vivo genetic fitness of Pseudomonas aeruginosa using high-throughput sequencing of transposon libraries", PLOS PATHOGENS, vol. 9, 2013, pages e1003582
SLOAN GPLOVE CFSUKUMAR NMISHRA MDEORA R.: "The Bordetella Bps polysaccharide is critical for biofilm development in the mouse respiratory tract", J BACTERIOL, vol. 189, 2007, pages 8270 - 6
ST JOHN FJGONZALEZ JMPOZHARSKI E: "Consolidation of glycosyl hydrolase family 30: a dual domain 4/7 hydrolase family consisting of two structurally distinct groups", FEBS LETTERS, vol. 584, 2010, pages 4435 - 41, XP027446887, DOI: 10.1016/j.febslet.2010.09.051
STARKEY MHICKMAN JHMA LZHANG NDE LONG S ET AL.: "Pseudomonas aeruginosa rugose small-colony variants have adaptations that likely promote persistence in the cystic fibrosis lung", J BACTERIOL, vol. 191, 2009, pages 3492 - 503
STEWART PS.: "New ways to stop biofilm infections", LANCET, vol. 361, 2003, pages 97, XP027155232, DOI: 10.1016/S0140-6736(03)12245-3
STEWART PSCOSTERTON JW.: "Antibiotic resistance of bacteria in biofilms", LANCET, vol. 358, 2001, pages 135 - 8, XP004805434, DOI: 10.1016/S0140-6736(01)05321-1
STOVER CKPHAM XQERWIN ALMIZOGUCHI SDWARRENER P ET AL.: "Complete genome sequence of Pseudomonas aeruginosa PA01, an opportunistic pathogen", NATURE, vol. 406, 2000, pages 959 - 64, XP055125439, DOI: 10.1038/35023079
SUTHERLAND I: "Biofilm exopolysaccharides: a strong and sticky framework", MICROBIOLOGY, vol. 147, 2001, pages 3 - 9
SUTHERLAND IW.: "The biofilm matrix-an immobilized but dynamic microbial environment", TRENDS MICROBIOL, vol. 9, 2001, pages 222 - 7
TERWILLIGER TCBERENDZEN J.: "Automated MAD and MIR structure solution", ACTA CRYSTALLOGR D BIOL CRYSTALLOGR, vol. 55, 1999, pages 849 - 61, XP008060283, DOI: 10.1107/S0907444999000839
TIAN LXU SHUTCHINS WCYANG CHLI J.: "Impact of the exopolysaccharides Pel and Psl on the initial adhesion of Pseudomonas aeruginosa to sand", BIOFOULING, vol. 30, 2014, pages 213 - 22
TONG KBLAU CJMURTAGH KLAYTON AJSEIFELDIN R.: "The economic impact of aspergillosis: analysis of hospital expenditures across patient subgroups", INTERNATIONAL JOURNAL OF INFECTIOUS DISEASES : IJID : OFFICIAL PUBLICATION OF THE INTERNATIONAL SOCIETY FOR INFECTIOUS DISEASES, vol. 13, 2009, pages 24 - 36, XP025838906, DOI: 10.1016/j.ijid.2008.02.013
TURK RSINGH AROUSSEAU JWEESE JS.: "In vitro evaluation of DispersinB on methicillin-resistant Staphylococcus pseudintermedius biofilm", VETERINARY MICROBIOLOGY, vol. 166, 2013, pages 576 - 9, XP055419584, DOI: 10.1016/j.vetmic.2013.07.011
VAN HOUDT RMICHIELS CW: "Biofilm formation and the food industry, a focus on the bacterial outer surface", JOURNAL OF APPLIED MICROBIOLOGY, vol. 109, 2010, pages 1117 - 31, XP055474364, DOI: 10.1111/j.1365-2672.2010.04756.x
VOCADLO DJMACKENZIE LFHE SZEIKUS GJWITHERS SG: "Identification of glu-277 as the catalytic nucleophile of Thermoanaerobacterium saccharolyticum beta-xylosidase using electrospray MS", THE BIOCHEMICAL JOURNAL, vol. 335, no. 2, 1998, pages 449 - 55
VOCADLO DJWICKI JRUPITZ KWITHERS SG: "A case for reverse protonation: identification of Glu160 as an acid/base catalyst in Thermoanaerobacterium saccharolyticum beta-xylosidase and detailed kinetic analysis of a site-directed mutant", BIOCHEMISTRY, vol. 41, 2002, pages 9736 - 46
VU BCHEN MCRAWFORD RJIVANOVA EP: "Bacterial extracellular polysaccharides involved in biofilm formation", MOLECULES, vol. 14, 2009, pages 2535 - 54
VUONG CKOCIANOVA SVOYICH JMYAO YFFISCHER ER ET AL.: "A crucial role for exopolysaccharide modification in bacterial biofilm formation, immune evasion, and virulence", J BIOL CHEM, vol. 279, 2004, pages 54881 - 86
WANG SPARSEK MRWOZNIAK DJMA LZ. 2013: "A spider web strategy of type IV pili-mediated migration to build a fibre-like Psl polysaccharide matrix in Pseudomonas aeruginosa biofilms", ENVIRON MICROBIOL, vol. 15, pages 2238 - 53
WANG XPRESTON JFROMEO T.: "The pgaABCD locus of Escherichia coli promotes the synthesis of a polysaccharide adhesin required for biofilm formation", J BACTERIOL, vol. 186, 2004, pages 2724 - 34, XP055568590, DOI: 10.1128/JB.186.9.2724-2734.2004
WASYLNKA JAHISSEN AHWAN ANMOORE MM.: "Intracellular and extracellular growth of Aspergillus fumigatus", MEDICAL MYCOLOGY, vol. 43, no. 1, 2005, pages 27 - 30
WASYLNKA JAMOORE MM.: "Adhesion of Aspergillus species to extracellular matrix proteins: evidence for involvement of negatively charged carbohydrates on the conidial surface", INFECTION AND IMMUNITY, vol. 68, 2000, pages 3377 - 84
WASYLNKA JAMOORE MM: "Uptake of Aspergillus fumigatus Conidia by phagocytic and nonphagocytic cells in vitro: quantitation using strains expressing green fluorescent protein", INFECTION AND IMMUNITY, vol. 70, 2002, pages 3156 - 63, XP055685981, DOI: 10.1128/IAI.70.6.3156-3163.2002
WASYLNKA JAMOORE MM.: "Aspergillus fumigatus conidia survive and germinate in acidic organelles of A549 epithelial cells", JOURNAL OF CELL SCIENCE, vol. 116, 2003, pages 1579 - 87
WIEHLMANN LWAGNER GCRAMER NSIEBERT BGUDOWIUS P ET AL.: "Population structure of Pseudomonas aeruginosa", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 104, 2007, pages 8101 - 6
WIERENGA RK.: "The TIM-barrel fold: a versatile framework for efficient enzymes", FEBS LETTERS, vol. 492, 2001, pages 193 - 8, XP004257362, DOI: 10.1016/S0014-5793(01)02236-0
WOLCOTT RCOSTERTON JWRAOULT DCUTLER SJ.: "The polymicrobial nature of biofilm infection", CLINICAL MICROBIOLOGY AND INFECTION : THE OFFICIAL PUBLICATION OF THE EUROPEAN SOCIETY OF CLINICAL MICROBIOLOGY AND INFECTIOUS DISEASES, vol. 19, 2013, pages 107 - 12
WOLFGANG MCKULASEKARA BRLIANG XBOYD DWU K ET AL.: "Conservation of genome content and virulence determinants among clinical and environmental isolates of Pseudomonas aeruginosa", PROC NATL ACAD SCI U S A, vol. 100, 2003, pages 8484 - 9, XP002329865, DOI: 10.1073/pnas.0832438100
YAKANDAWALA NGAWANDE PVLOVETRI KCARDONA STROMEO T ET AL.: "Characterization of the poly-beta-1,6-N-acetylglucosamine polysaccharide component of Burkholderia biofilms", APPL ENVIRON MICROBIOL, vol. 77, 2011, pages 8303 - 9, XP055346124, DOI: 10.1128/AEM.05814-11
ZEGANS MEWOZNIAK DGRIFFIN ETOUTAIN-KIDD CMHAMMOND JH ET AL.: "Pseudomonas aeruginosa exopolysaccharide Psl promotes resistance to the biofilm inhibitor polysorbate 80", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 56, 2012, pages 4112 - 22
ZHAO KTSENG BSBECKERMAN BJIN FGIBIANSKY ML ET AL.: "Psl trails guide exploration and microcolony formation in Pseudomonas aeruginosa biofilms", NATURE, vol. 497, 2013, pages 388 - 91
See also references of EP 3152303A4
Attorney, Agent or Firm:
BERESKIN & PARR LLP/S.E.N.C.R.L., S.R.L. (40 King Street West40th Floo, Toronto Ontario M5H 3Y2, CA)
Download PDF:
Claims:
CLAIMS:

1. A method of treating or preventing a biofilm-related infection comprising administering at least one, at least two of, at least three of, at least four of, at least five of, or all of: (i) a soluble protein comprising a Sph3 GH domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, to an animal or plant in need thereof. 2. The method of claim 1 , wherein the soluble protein comprising a Sph3 GH domain comprises amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 or a glycosyl hydrolase variant thereof.

3. The method of claim 1 , wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 54 to 304 of the Sph3 sequence deposited into Genbank under accession no. EAW09379.1 or a glycosyl hydrolase variant thereof.

4. The method of claim 1 , wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 43 to 299 of the Sph sequence deposited into Genbank under accession no. EAA63523.1 or a glycosyl hydrolase variant thereof.

5. The method of claim 1 , wherein the soluble protein comprising a PelA GH domain comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 or glycosyl hydrolase variants thereof.

6. The method of claim 1 , wherein the protein comprising a PelA GH domain ortholog comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 or glycosyl hydrolase variants thereof.

7. The method of claim 1 , wherein the soluble protein comprising a BpsB GH domain comprises amino acids 318 to 670 or amino acids 27 to 701 of the

BpsB sequence deposited into GenBank under accession no. CAE32265.1 or glycosyl hydrolase variants thereof.

8. The method of claim 1 , wherein the soluble protein comprising a PgaB GH domain comprises amino acids 310 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 or glycosyl hydrolase variants thereof.

9. The method of claim 1 , wherein the soluble protein comprising a PsIG GH domain comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 or a glycosyl hydrolase variant thereof.

10. The method of claim 1 , wherein the soluble protein comprising an Ega3 GH domain comprises amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 or a glycosyl hydrolase variant thereof. 1 1. The method of any one of claims 1 to 10, wherein the biofilm-related infection is the result of a wound, burn infection, keratitis, bioprosthetic or indwelling medical device infection in the animal.

12. The method of any one of claims 1 to 10, wherein the biofilm-related infection is in the lung of the animal and wherein the animal has chronic pulmonary disease or lung infection.

13. The method of any one of claims 1 to 10, wherein the biofilm-related infection is on the surface of the plant or a plant part.

14. The method of any one of claims 1 to 13, wherein the biofilm of the biofilm-related infection is Pel-dependent, PsI-dependent, PNAG-dependent and/or GAG-dependent biofilm.

15. The method of any one of claims 1 to 13, wherein the biofilm-related infection is caused by P. aeruginosa, S. aureus, E. coli, Candida spp.,

Aspergillus spp., Acinetobacter spp., T. asahii, B. cineria and Fusarium spp.

16. The method of any one of claims 1 to 15 further comprising coadministering an antimicrobial agent to the animal or plant in need thereof.

17. The method of claim 16, wherein the antimicrobial agent is an antibiotic or antifungal agent.

18. The method of any one of claims 1 to 17, wherein the at least one soluble protein is expressed by a vector and the method comprises administering the vector to the animal or plant in need thereof.

19. The method of claim 18, wherein the vector is a phage vector that is able to invade bacteria of the biofilm.

20. The method of claim 18, wherein the vector is a mycoviral vector that is able to invade fungi of the biofilm.

21. A method of preventing biofilm formation on an indwelling medical device or implant comprising coating the device with at least one of, at least two of, at least three of, at least four of, at least five of or all of: (i) a soluble protein comprising a Sph3 GH domain (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, prior to use in an animal in need thereof.

22. The method of claim 21 , wherein the soluble protein comprising a Sph3 GH domain comprises amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 or a glycosyl hydrolase variant thereof. 23. The method of claim 21 , wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 54 to 304 of the Sph3 sequence deposited into Genbank under accession no. EAW09379.1 or a glycosyl hydrolase variant thereof.

24. The method of claim 21 , wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 43 to 299 of the Sph sequence deposited into Genbank under accession no. EAA63523.1 or a glycosyl hydrolase variant thereof.

25. The method of claim 21 , wherein the soluble protein comprising a PelA GH domain comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35 to 291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 or glycosyl hydrolase variants thereof.

26. The method of claim 21 , wherein the soluble protein comprising a PelA GH domain ortholog comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 or glycosyl hydrolase variants thereof.

27. The method of claim 21 , wherein the soluble protein comprising a BpsB GH domain comprises amino acids 3 8 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 or glycosyl hydrolase variants thereof.

28. The method of claim 21 , wherein the soluble protein comprising a

PgaB GH domain comprises amino acids 3 0 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 or glycosyl hydrolase variants thereof.

29. The method of claim 21 , wherein the soluble protein comprising a PsIG GH domain comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 or a glycosyl hydrolase variant thereof.

30. The method of claim 21 , wherein the soluble protein comprising an Ega3 GH domain comprises amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 or a glycosyl hydrolase variant thereof.

31. The method of any one of claims 21 to 30, wherein the indwelling medical device or implant is a catheter or intravenous tube.

32. The method of any one of claims 21 to 30, wherein the indwelling medical device or implant is a prosthetic joint, or bioprosthetic. 33. The method of any one of claims 21 to 32 wherein the biofilm is Pel- dependent, Psl-dependent, PNAG-dependent and/or GAG-dependent biofilm.

34. The method of any one of claims 21 to 32, wherein the biofilm is caused by P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp., Acinetobacter spp. and Fusarium spp.

35. The method of any one of claims 21 to 34, further comprising coating the indwelling medical device or implant with an antimicrobial agent.

36. The method of claim 35, wherein the antimicrobial agent is an antibiotic or antifungal agent. 37. A method of treating or preventing biofilm formation on a non-medical surface that is susceptible to biofilm formation comprising applying to the surface or coating the surface with at least one of, at least two of, at least three of, at least four of, at least five of or all of: (i) a soluble protein comprising a Sph3 GH domain, (ii) a soluble protein comprising a Pe!A GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, prior to use in an animal in need thereof.

38. The method of claim 37, wherein the soluble protein comprising a Sph3 GH domain comprises amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 or a glycosyl hydrolase variant thereof.

39. The method of claim 37, wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 54 to 304 of the Sph3 sequence deposited into Genbank under accession no. EAW09379.1 or a glycosyl hydrolase variant thereof.

40. The method of claim 37, wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 43 to 299 of the Sph sequence deposited into Genbank under accession no. EAA63523.1 or a glycosyl hydrolase variant thereof.

41. The method of claim 37, wherein the soluble protein comprising a PelA GH domain comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 or glycosyl hydrolase variants thereof.

42. The method of claim 37, wherein the soluble protein comprising a PelA GH domain ortholog comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 or glycosyl hydrolase variants thereof.

43. The method of claim 37, wherein the soluble protein comprising a BpsB GH domain comprises amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 or glycosyl hydrolase variants thereof.

44. The method of claim 37, wherein the soluble protein comprising a PgaB GH domain comprises amino acids 310 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 or glycosyl hydrolase variants thereof.

45. The method of claim 37, wherein the soluble protein comprising a PsIG GH domain comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 or a glycosyl hydrolase variant thereof.. 46. The method of claim 37, wherein the soluble protein comprising an Ega3 GH domain comprises amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 or a glycosyl hydrolase variant thereof.

47. The method of any one of claims 37 to 46, wherein the biofilm of the biofilm-related infection is Pel-dependent, PsI-dependent, PNAG-dependent and/or GAG-dependent biofilm.

48. The method of any one of claims 37 to 46, wherein the biofilm is caused by P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp., Acinetobacter spp. and/or Fusarium spp.

49. The method of any one of claims 37 to 48, further comprising co- applying or coating with an antimicrobial agent.

50. The method of claim 49, wherein the antimicrobial agent is an antibiotic or an antifungal agent.

51. An indwelling medical device or implant coated with at least one of, at least two of, at least three of, at least four of, at least five of or all of: (i) a soluble protein comprising a Sph3 GH domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof. 52. The indwelling medical device or implant of claim 51 , wherein the soluble protein comprising a Sph3 GH domain comprises amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 or a glycosyl hydrolase variant thereof.

53. The indwelling medical device or implant of claim 51 , wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 54 to 304 of the Sph3 sequence deposited into Genbank under accession no. EAW09379.1 or a glycosyl hydrolase variant thereof.

54. The indwelling medical device or implant of claim 51 , wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 43 to 299 of the Sph sequence deposited into Genbank under accession no. EAA63523.1 or a glycosyl hydrolase variant thereof.

55. The indwelling medical device or implant of claim 51 , wherein the protein comprising a PelA GH domain comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 or glycosyl hydrolase variants thereof.

56. The indwelling medical device or implant of claim 51 , wherein the soluble protein comprising a PelA GH domain ortholog comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 or glycosyl hydrolase variants thereof. 57. The indwelling medical device or implant of claim 51 , wherein the protein comprising a BpsB GH domain comprises amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 or glycosyl hydrolase variants thereof.

58. The indwelling medical device or implant of claim 51 , wherein the protein comprising a PgaB GH domain comprises amino acids 310 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 or glycosyl hydrolase variants thereof.

59. The indwelling medical device or implant of claim 51 , wherein the protein comprising a PsIG GH domain comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 or a glycosyl hydrolase variant thereof.

60. The indwelling medical device or implant of claim 51 , wherein the soluble protein comprising an Ega3 GH domain comprises amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 or a glycosyl hydrolase variant thereof.

61. The indwelling medical device or implant of claim 51 , wherein the device or implant is further coated with an antimicrobial agent.

62. The indwelling medical device or implant of claim 61 , wherein the antimicrobial agent is an antibiotic or an antifungal agent. 63. The indwelling medical device or implant of any one of claims 51 to 62, wherein the indwelling medical device is a catheter or intravenous tube.

64. The indwelling medical device or implant of any one of claims 51 to 62, wherein the indwelling medical device is a prosthetic joint or bioprosthetic.

65. The indwelling medical device or implant of any one of claims 51 to 64, wherein the biofilm is Pel-dependent, Psl-dependent, PNAG-dependent and/or GAG-dependent biofilm.

66. The indwelling medical device or implant of any one of claims 51 to 64, wherein the biofilm is caused P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp., Acinetobacter spp. and/or Fusarium spp. 67. A vector encoding (i) a soluble protein comprising a Sph3 GH domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, or (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, or combinations thereof.

68. The vector of claim 67, wherein the soluble protein comprising a Sph3 GH domain comprises amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 or a glycosyl hydrolase variant thereof. 69. The vector of claim 67, wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 54 to 304 of the Sph3 sequence deposited into Genbank under accession no. EAW09379.1 or a glycosyl hydrolase variant thereof.

70. The vector of claim 67, wherein the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 43 to 299 of the Sph sequence deposited into Genbank under accession no. EAA63523.1 or a glycosyl hydrolase variant thereof.

71. The vector of claim 67, wherein the soluble protein comprising a PelA GH domain comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 or glycosyl hydrolase variants thereof.

72. The vector of claim 67, wherein the soluble protein comprising a PelA GH domain ortholog comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 or glycosyl hydrolase variants thereof.

73. The vector of claim 67, wherein the soluble protein comprising a BpsB GH domain comprises amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 or glycosyl hydrolase variants thereof. 74. The vector of claim 67, wherein the soluble protein comprising a PgaB GH domain comprises amino acids 310 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 or glycosyl hydrolase variants thereof.

75. The vector of claim 67, wherein the soluble protein comprising a PsIG GH domain comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 or a glycosyl hydrolase variant thereof.

76. The vector of claim 67, wherein the soluble protein comprising an Ega3 GH domain comprises amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 or a glycosyl hydrolase variant thereof.

77. The vector of any one of claims 67 to 77, wherein the vector is a lytic phage.

78. The vector of any one of claims 67 to 77, wherein the vector is a mycovirus.

Description:
TITLE: SOLUBLE BACTERIAL AND FUNGAL PROTEINS AND METHODS AND USES THEREOF IN INHIBITING AND DISPERSING BIOFILM

RELATED APPLICATION

[0001] This application claims the benefit of priority to United States Provisional application No. 62/008,836 filed June 6, 2014, the contents of which are incorporated herein by reference in their entirety.

FIELD

[0002] The present disclosure relates to glycosyl hydrolase-containing soluble microbial proteins. In particular, the present disclosure relates to compositions and methods and uses thereof in treating and preventing microbial biofilms.

BACKGROUND

Microbial Biofilms

[0003] The composition of microbial biofilms varies between strains/species and environmental conditions, but generally contains proteinaceous adhesins, nucleic acids, and exopolysaccharides as the main components (Branda et al 2005, Sutherland 2001 a, Vu et al 2009). Exopolysaccharides are the predominant biofilm matrix component of many microbial organisms, contributing to biofilm adhesion, architecture, and resistance (Colvin et al 201 1 , Colvin et al 2012, Ma et al 2009, Ma et al 2006, Mah et al 2003, Matsukawa & Greenberg 2004). Biofilms form on biotic surfaces, such as lung epithelial cells or other organs, and abiotic surfaces including, but not limited to, medical devices, and implants, and are responsible for biofouling in industrial and commercial settings including, but not limited to; pipes and drains, water filtration apparatuses and food-contact surfaces (Bjarnsholt et al 2013, Kumar & Anand 1998). The hallmark of chronic, biofilm-related infections is an extreme resistance to antibiotics and the ability to evade the host immune system (Bjarnsholt 2013, Hoiby et al 2010, Kim et al 2009, Mishra et al 2012, Rybtke et al 201 1 , Stewart 2003, Stewart & Costerton 2001 ). The tolerance of bacterial biofilms to antibiotics and detergents is often 1 ,000 times higher compared to their planktonic counterparts (Alhede et al 201 1 , Costerton et al 1987, Costerton et al 1999, Davies 2003). Mature biofilms have a highly complex structure that allows for the movement of nutrients and waste products (Hall-Stoodley et al 2004, Sutherland 2001 b). Biofilms act to sequester a large microbial population, including many pathogenic bacterial species, which symbiotically utilize the strengths of each organism for survival in the host (Wolcott et al 2013). Therefore, strategies to inhibit biofilms may have implications for reducing the total microbial biomass (Bragonzi et al 2012).

P. aeruginosa and Biofilm Formation.

[0004] During infection, the bacterium P. aeruginosa undergoes a lifestyle change from a free-swimming state to a surface attached, matrix embedded biofilm state (Figure 1). Upon establishment of a biofilm, the infection becomes chronic and untreatable, as the biofilm is highly resistant to antibiotics and aids in the bacterial adaptation to changing environments (Colvin et al 201 1 , Mishra et al 2012, Zegans et al 2012). In CF patients this biofilm allows the bacteria to persist within the lungs for decades (Ma et al 2012, Mann & Wozniak 2012, Starkey et al 2009), while in wounds it allows for initial colonization and protection. P. aeruginosa biofilms are mainly composed of the exopolysaccharides; Psl, Pel and alginate. All three polysaccharides are important virulence factors and aid in genetic fitness of the bacterium (Skurnik et al 2013). Two common clinical biofilm forming strains P. aeruginosa PA 4 and PA01 have been extensively studied (Hare et al 2012, Kukavica-lbrulj et al 2008). PA 4, the most abundant strain worldwide (Wiehlmann et al 2007), was originally identified in burn wound isolates (Rahme et al 1995). This strain is highly virulent and exclusively utilizes Pel (Colvin et al 2012), while the second clinical strain PA01 is a moderately virulent strain (Lee et al 2006) that primarily utilizes Psl for biofilm production (Colvin et al 2012). Under duress, PA01 is able to utilize Pel if production of Psl is compromised (Colvin et al 2012), allowing it to maintain infection in the host (Byrd et al 201 1 ). Several recent studies indicate that Pel and Psl are critical during early biofilm formation. In addition, PsI has been demonstrated to be important for continued maintenance in established biofilms as demonstrated in CF patients (Billings et al 2013, Huse et al 2013, Irie et al 2012, Wang et al 2013, Zhao et al 2013).

PsI Polysaccharide

[0005] PsI is composed of a pentasaccharide repeating unit of D- mannose, D-glucose and L-rhamnose, and is distinct from other known polysaccharides (Figure 2). The psl operon, discovered in 2004 (Jackson et al 2004), consists of 15 open reading frames (ORFs) psIABCDEFGHIJKLMNO, encoding putative proteins required for the biosynthesis of the exopolysaccharide. Four of these ORFs, psIB, psIM, psIN, psIO are not required for Psl biosynthesis (Byrd et al 2009). The psl operon is present in numerous Pseudomonas strains (Figure 3). The initial steps in Psl biosynthesis are proposed to occur in the cytoplasm and are then transferred to the periplasm prior to secretion through the outer membrane (Franklin et al 201 1 ).

[0006] Psl functions as a "molecular glue" for bacterial adhesion and is important for initial adherence of planktonic or "free-swimming" bacteria to abiotic and biotic surfaces (Byrd et al 2010, Byrd et al 2009, Ma et al 2006). Psl also aids in the structural stability, and maintenance of the architecture of the mature biofilm (Ma et al 2009, Ma et al 2012). The polysaccharide provides protection against the immune system (Mishra et al 2012) and is a first line of defense during the initial stages of biofilm development, especially towards attack by antibiotics with diverse targets and biochemical properties (Billings et al 2013). For example, Psl-producing biofilms have a -35-50% increase in resistance to Polymyxin B, Tobramycin and Ciprofloxacin which are standard antibiotics used in the clinic to treat P. aeruginosa infections, and a 75% increase in resistance to Coiistin, one of the last-resort antibiotics for multi-drug resistant P. aeruginosa (Billings et al 2013). Non-Psl producing, antibiotic-sensitive P. aeruginosa, Escherichia coli, and Staphylococcus aureus, gain antibiotic tolerance by integrating into Psl-containing biofilms. Sub-inhibitory concentrations of these antibiotics induce further biofilm formation (Hoffman et al 2005) exacerbating the infection. While alginate is the major exopolysaccharide in chronic CF lung infection, recent evidence suggests that Psl is just as important during long-term colonization (~40,000 bacterial generations) (Huse et al 2013). Patients recovering from P. aeruginosa infections have specific antibodies against Psl demonstrating that Psl is clinically relevant during infection (Digiandomenico et al 2012).

Pel Polysaccharide

[0007] Biofilms that form at the air-liquid interface are referred to as pellicles. Pellicles formed by the clinical isolate P. aeruginosa PA14 are encoded on the seven-gene operon pelABCDEFG, all of which are necessary for Pel-dependent biofilm formation (Figure 4) (Colvin et al 201 1 , Friedman & Kolter 2004a). The pel operon was initially discovered in a transposon library in 2004 (Friedman & Kolter 2004b). The chemical composition of Pel and linkage is currently unknown. The biosynthesis is believed to start in the cytoplasm and the polymer is transported across the inner membrane for translocation through the periplasm. Recent work suggests that the Pel polysaccharide must be deacetylated or partially deacetylated by the multi- domain periplasmic protein PelA for biofilm formation to occur (Colvin et al 2013). P. aeruginosa PA14 largely relies on the Pel polysaccharide for adhesion and cell-to-cell interaction compared to other strains. The polysaccharide is crucial for maintaining cell-to-cell interactions and forms a structural scaffold for the biofilm community (Colvin et al 201 1 ). Pel also serves a protective role against commonly used antibiotics tobramycin and ciprofloxacin (Colvin et al 20 1 ).

PNAG Biofilms in Gram-positive and Gram-negative bacteria

[0008] The polysaccharide PNAG (Figure 5) has been found in the biofilms of Staphylococcus epidermidis (Mack et al 1996), S. aureus (McKenney et al 1999), E. coli (Wang et al 2004), Pseudomonas fluorescens (Itoh et al 2005), Acinetobacter baumannii (Choi et al 2009), Actinobacillus pleuropneumoniae (Izano et al 2007), Yersinia pestis (Jarrett et al 2004), Aggregatibacter actinomycetemcomitans (Izano et al 2008), Bordetella bronchiseptica (Sloan et al 2007), Bordetella pertussis (Conover et al 2010), and Burkholderia spp. (Yakandawala et al 201 1 ). The biosynthesis of PNAG requires the icaADBC and pgaABCD operons in Gram-positive and Gram- negative bacteria, respectively. Interestingly, recent immunogenic studies probing the cellular surface of a wide variety of prokaryotic and eukaryotic pathogens that do not contain the canonical genetic loci for the biosynthesis of PNAG revealed the presence of surface-associated PNAG (Cywes-Bentley et al 2013). This included Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus dysgalactiae, Enterococcus faecalis, Listeria monocytogenes, Clostridium difficile, Mycobacterium tuberculosis, Mycobacterium smegmatis, Neisseria meningitidis, Neisseria gonorrheae, nontypable Haemophilus influenzae, Haemophilus ducreyi, Helicobacter pylori, Campylobacter jejuni, Citrobacter rodentium, Salmonella enterica, Candida albicans, Aspergillus flavus, Fusarium solani, Cryptococcus neoformans, Trichomonas vaginalis, Plasmodium berghei, and Plasmodium falciparum (Cywes-Bentley et al 2013). Furthermore, administration of monoclonal antibodies that bind to PNAG were able to mediate complement- dependent opsonic or bactericidal killing that protected mice against local or systemic infections from a number of the pathogens (Cywes-Bentley et al 2013).

PNAG polysaccharide

[0009] PNAG is a homopolymer of repeating -acetyl-D-glucosamine units, similar to chitin, however it is synthesized with a β(1 ,6) linkage. The mature form of PNAG is partially deacetylated, is commonly referred to as dPNAG, and is required for the formation of the biofilm in a large number bacteria (Figure 6). Given the differences in cell wall architecture between Gram-positive and Gram-negative bacteria, the sequence homology between the protein products of the two genetic loci is limited to IcaA and PgaC, and IcaB and the N-terminal domain of PgaB. PgaB, a two-domain outer- membrane lipoprotein, and IcaB, an extracellular single-domain protein, are required for the partial de-A/-acetylation of PNAG (Little et al 2012a, Pokrovskaya et al 2013). Impairment of de-N-acetylation of PNAG has shown to prevent biofilm formation in E. coli, S. aureus, and S. epidermidis (Cerca et al 2007, Itoh et al 2008, Vuong et al 2004).

Aspergillus fumigatus and biofilm formation

[0010] Aspergillus fumigatus is a fungal pathogen and is one of the most common Aspergillus species to cause disease in immuno-compromised individuals (Geiser et al 2007). Aspergillus species are the second most common cause of fungal infection in the healthcare setting after C. albicans (Ellis et al 2000). The average person inhales several hundred A. fumigatus conidia (fungal spores) daily, which pass through the airways and are deposited in the alveoli of the lungs. Importantly, while these conidia account for less than 0.1 % of all airborne fungal conidia, A. fumigatus conidia accounts for >80% of invasive infection in humans. In healthy individuals with normal lung function, conidia are removed by the mucociliary elevator or rapidly phagocytosed and killed by alveolar macrophages to prevent infection. However, in immune-compromised individuals, these conidia are not killed but instead adhere to pulmonary epithelial cells and macrophages before being internalized and germinating within host cells (Wasylnka et al 2005, Wasylnka & Moore 2002, Wasylnka & Moore 2003). Following germination, the newly formed hyphae remain intimately associated with host epithelial, endothelial and immune cells that can result in inflammation and tissue injury (Gravelat et al 2013, Wasylnka & Moore 2000). In patients with chronic lung disease whose immune system is intact, Aspergillus spores can germinate to produce hyphae that colonize the airways or pre-existing lung cavities but do not invade into tissues. These forms of Aspergillus infection are associated with persistent airway inflammation, allergic responses and declining lung function. In both invasive and chronic pulmonary aspergillosis infection, hyphae of the fungus are found within a dense extracellular matrix (Loussert et al 2010). It has been shown that biofilm formation and the adherence of hyphae to epithelial cells is mediated by the exopolysaccharide galactosaminogalactan (GAG) (Gravelat et al 2013). Aspergillus is also a cause of wound and corneal infections. Other Aspergillus species and some non-Aspergillus fungi, including the fungal pathogen Fusarium, as well as a number of plant pathogenic fungi have the genetic capability to produce GAG.

Galactosaminogalactan (GAG)

[0011] Galactosaminogalactan (GAG) is a heterogeneous, linear extracellular polysaccharide that is composed of 1-4 linked galactose and ot1-4 linked ^-acetylgalactosamine (Fontaine et al 201 1 ). The exopolysaccharide is secreted by A. fumigatus to form a biofilm, encapsulating the hyphae and allowing adherence to both biotic (epithelial cells & fibronectin) and abiotic (glass & plastic) surfaces (Gravelat et al 2013). Additionally, GAG is a component of the A. fumigatus cell wall, constituting -2% of the total polysaccharides from cell wall of static and aerial hyphae (Loussert et al 2010). The functions of GAG in A. fumigatus pathogenesis are as follows (Gravelat et al 2013); mediate adherence of A. fumigatus hyphae to biotic and abiotic surfaces (Gravelat et al 2013), act as an immunosuppressor (Fontaine et al 201 1 ), modulate host immune responses through cloaking β- glucans and other pathogen-associated molecular pattern molecules on the surface of the hyphae (Gravelat et al 2013).

Hvdrolvtic enzymes involved in exopolysaccharide biosynthesis

[0012] Many exopolysaccharide biosynthetic machinery systems, including those of cellulose, alginate, Psl, Pel and GAG polysaccharide, encode a putative or functionally characterized glycosyl hydrolase. These enzymes may be required for biofilm formation to occur but the exact biological role in this process remains undetermined.

Putative glycosyl hydrolase PsIG (Psl polysaccharide system) [0013] Although PsIG is essential for polymer production (Byrd et al 2009), the biological role of PsIG in the biosynthesis of PsI is not fully understood. Glycosyl hydrolases (GHs) are grouped into families based on amino acid sequence (Davies & Henrissat 1995, Henrissat & Bairoch 1996). Based on amino acid sequence identity, the Carbohydrate-active enzymes database (CAZy) (Lombard et al 2014) categorizes PsIG as a member of the GH39 family. Interestingly, all current bacterial members of this family are currently annotated as β-xylosidases while human members are annotated as a-L-iduronidase. All structures are composed of a N-terminal (β/α)β TIM-barrel containing a conserved active site, and a C-terminal β-sandwich domain. The catalytic reaction occurs via a retaining mechanism with two catalytic glutamate residues responsible for catalysis (Vocadlo et al 2002).

The multi-domain protein PelA (Pel polysaccharide system)

[0014] Bioinformatics analysis of PelA indicates that it is a multi-domain periplasmic protein with three potential catalytic activities. The Phyre 2 (Kelley & Sternberg 2009) server suggests that the protein may have upwards of five distinct domains (Figure 7) (Colvin et al 2013) including a predicted TIM- barrel domain composing residues 47-303, a reductase domain from residues 304-409, a deacetylase domain from residues 520-800 and a β-jelly roll fold from residues 840-927 (Colvin et al 2013). It was previously demonstrated that site specific mutation of putative catalytic residues in the deacetylase domain abrogated biofilm formation (Colvin et al 2013). It is unknown whether the proposed catalytic function of the hydrolase domain is required for Pel biosynthesis.

PqaB/BpsB is a PNAG deacetylase and putative glycosyl hydrolase

[0015] Bioinformatics analysis indicates that PgaB is the only periplasmic protein in the pga biosynthetic operon with a catalytic de-N- acetylation domain and a predicted carbohydrate-binding domain. Although known to be essential for PNAG de-A/-acetylation, the biological role of PgaB C-terminal domain (PgaB 31 0- 672) in the de-/V-acetylation of PNAG has only recently been determined (Little et al 2014). PgaB 31 0 -672 was shown to bind PNAG oligomers and molecular dynamics simulations combined with structures in complex with A/-acetylglucosamine (GlcNAc), glucosamine (GlcN), and a PNAG hexamer suggest the domain preferentially binds dPNAG. In Bordetella spp. the pgaABCD operon is known as bpsABCD (Figure 6) (Parise et al 2007). BpsB from B. bronchiseptica is predicted to contain a deacetylase domain that shares 40% and 24% sequence identity to PgaB and IcaB, respectively (Parise et al 2007).

[0016] Based on amino acid sequence identity, the CAZy database (Lombard et al 2014) recently categorized both BpsB and PgaB C-terminal domains as members of the GH13 family. This GH superfamily contains a functional (β/α)β TIM-barrel catalytic domain (Little et al 2014, Lombard et al 2014). Structural prediction servers suggest that the proteins have high structural similarity to families GH18 and GH20. However, previous and ongoing efforts to show PgaB 3 -| 0 -672 hydrolase activity with PNAG oligomers and artificial para-n itrophenyl (pNP) glycoside substrates have proven unsuccessful (Little et al 2012a). Interestingly, sequence and structural comparison of PgaB 3 i 0- 672 to GH13, GH18 and GH20 family members reveals the absence of the catalytic consensus sequences AED, DXXDXDXE, or GGDE, respectively.

Clinical and Economic Significance of Microbial Biofilms.

[0017] Biofilm-related infections account for between 65-80% of all chronic, persistent bacterial infections and the number of infections and conditions involving biofilms continues to grow (Bjamsholt et al 2013, Flemming & Wingender 2010). P. aeruginosa and the formation of highly- resistant biofilms are a dominant bacterial species in three specific areas; burn wound victims, chronic wound infections and cystic fibrosis (CF). Other bacteria such as, but not limited to; S. epidermis, S. aureus and E. coli are also significant in biofilm formation, especially in relation to medical-device related infections. In industry, biofouling - the accumulation of microorganisms embedded in a biofilm on abiotic surfaces - is a significant problem due to biotransfer potential between abiotic and biotic surfaces (Van Houdt & Michiels 2010), and reduced flow rates in pipes and other objects due to increased drag (Tian et al 2014).

[0018] Aspergillus fumigatus is a fungal pathogen and is one of the most common Aspergillus species to cause disease in immuno-compromised individuals. Importantly, while A. fumigatus conidia account for less than 0.1 % of all airborne fungal conidia, A. fumigatus conidia accounts for >80% of invasive infection in humans. Invasive aspergillosis is associated with higher morbidity and mortality, particularity in immunocompromised patients (Tong et al 2009). Studies have reported mortality rates of over 80% in bone marrow transplantation, 90% in liver transplant recipients and 49% with patients with leukemia or lymphoma (Lin et al 2001 , Singh 2000). A. fumigatus is the second most common cause of fungal infection found in hospitalized patients/healthcare settings after C. albicans (Ellis et al 2000).

[0019] Exopolysaccharide producing fungi are important plant pathogens. Botrytis c/nerea affects over 200 species of flowering and bulb- producing plants including grapes, onions, strawberries, and ornamental cut flowers (Dean et al 2012). Crop losses due to Botrytis infections and the cost of fungicides to combat these costs are a significant economic problem. In 2001 an estimated€540 million was spent on the control of Botrytis infections alone (Dean et al 2012). Further, resistance to fungicides continues to increase (Bardas et al 2010, Grabke et al 2014, Leroch et al 2013, Rodriguez et al 2014). Other pathogens account for important economic losses: Blumeria graminis is an important pathogen of wheat and barley (Dean et al 2012); while Fusarium oxysporum can infect over 100 different plant hosts and is associated with important losses of crops such as tomato cotton, melon and banana (Michielse & Rep 2009). Treatments against bacterial and fungal biofilms

[0020] The ability of microorganisms within biofilms to withstand the actions of antimicrobial agents and host defenses presents a substantial challenge. Apart from surgical intervention (when appropriate), antimicrobial agents remain the only treatment option and are administered for extended durations in an attempt to penetrate the protective biofilm barrier and eradicate the infection. This methodology is partly to blame for the increase in anti-microbial resistance by bacteria and fungi. The continued presence of non-lethal doses of antimicrobial agents leads to desensitization, thereby inducing selection for resistant mutants (Hoiby et al 201 1 ). Evolutionary mutation rates in biofilms have been shown, both in vitro and in vivo, to occur more rapidly. Furthermore, it has been demonstrated that sub-inhibitory concentrations of antibiotics used to treat P. aeruginosa infections induce biofilm formation, exacerbating the inability to manage these bacterial infections (Hoffman et al 2005).

[0021] There is precedence that enzyme therapy is a viable treatment option for bacterial biofilms. One example is the glycosyl hydrolase DspB or DispersinB® (US 7,989,604 B2) (Kaplan et al 2004, Manuel et al 2007). Initially isolated from A. actinomycetemcomitans, the enzyme degrades the biofilm exopolysaccharide poly^-1 ,6-/v-acetylglucosamine (PNAG) used by several pathogenic organisms including but not limited to; E. coli, S. aureus and S. epidermidis (Darouiche et al 2009, Donelli et al 2007, Gawande et al 2014, Kaplan et al 2004, Turk et al 2013). DspB nor homologs of the enzyme are found within the PNAG biosynthetic operon of A. actinomycetemcomitans. DispersinB® does not have the capability of inhibiting and dispersing P. aeruginosa or A. fumigatus biofilms as these bacteria do not have the genetic capacity to produce PNAG.

SUMMARY

[0022] The inability of microorganisms to produce a biofilm diminishes attachment to biotic and abiotic surfaces, and increases susceptibility of microorganisms to antimicrobial agents (Davies 2003) and the host immune system (Bakkevig et al 2005, Jain & Ohman 1998, Maharaj et al 1993, Monday & Schiller 1996). The present inventors have demonstrated that exogenously applying soluble forms of glycosyl-hydrolase containing bacterial or fungal proteins: PelA, PsIG, BpsB, PgaB, Sph3, Ega3 and their orthologs, results in prevention and/or inhibition of microbial biofilms. The inventors thus proposed the use any soluble glycosyl-hydrolase protein located in any exopolysaccharide biosynthetic operon or functional gene cluster in any microbial species for the inhibition and dispersal of the formed microbial biofilm resulting from the use of the biosynthetic operon or functional gene cluster in the formation of the biofilm.

[0023] Accordingly, in one aspect, the present disclosure provides a method of treating or preventing a biofilm-related infection comprising administering at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain. Also provided herein is use of at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain for treating or preventing a biofilm-related infection. Further provided is use of at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain in the manufacture of a medicament for treating or preventing a biofilm-related infection. Even further provided is at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain for use in treating or preventing a biofilm-related infection.

[0024] In one embodiment, the present disclosure provides a method of treating or preventing a biofilm-related infection comprising administering at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, to an animal or plant in need thereof. Also provided is use of at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, for treating or preventing a biofilm related infection in an animal or plant in need thereof. Further provided is use of at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, in the preparation of a medicament for treating or preventing a biofilm-related infection in an animal or plant in need thereof. Even further provided is at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, for use in treating or preventing a biofilm-related infection in an animal or plant in need thereof.

[0025] In an embodiment, the methods or uses disclosed herein comprise at least two of, at least three of, at least four of, at least five of, or all of the soluble proteins.

[0026] In an embodiment, the methods or uses disclosed herein further comprise administering other soluble proteins that degrade other components of biofilm, such as alginate and/or cellulose. [0027] In one embodiment, the biofilm-related infection may be the result of a wound or burn infection in the animal.

[0028] In one embodiment, the biofilm-related infection may be the result of keratitis in the animal.

[0029] In another embodiment, the biofilm-related infection may be a lung infection in the animal, wherein the animal has chronic pulmonary disease.

[0030] In another embodiment, the biofilm-related infection is a result of microbial contamination on or of medical devices or implants in the animal.

[0031] In another embodiment, the biofilm-related infection may be a lung infection in the animal, which includes but is not limited to invasive aspergillosis, an acute disease of the immunocompromised host or chronic aspergillus infection that occurs in immunocompetent individuals with compromised lung function.

[0032] In an embodiment, at least one soluble protein comprising a glycosyl hydrolase potentiates neutrophil killing of the microorganism. In a particular embodiment, the soluble protein is a PelA protein disclosed herein.

[0033] In yet another embodiment, the biofilm-related infection is mediated by a fungus producing an exopolysaccharide sheath in an animal or a plant. In an embodiment, the plant includes plant materials, such as fruit or flowers.

[0034] In yet another embodiment, the biofilm-related infection may be caused by any microorganism or group of microorganisms some or all of which have the genetic capacity to synthesize the exopolysaccharides, Pel, Psl, PNAG and/or GAG and combinations thereof. These organisms include, but are not limited to; P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp., Acinetobacter spp., T. asahii, B. cineria and Fusarium spp. In another embodiment, the biofilm may be dependent on the secretion of any exopolysaccharide that is able to be degraded by the soluble glycosyl hydrolases disclosed herein.

[0035] In yet another embodiment, the methods or uses disclosed herein further comprise co-administering an antimicrobial agent to the animal or plant in need thereof. In one embodiment, the antimicrobial agent is an antibiotic. In another embodiment, the antimicrobial agent is an antifungal agent. In yet another embodiment, the antimicrobial agent is a fungicide for use on plants.

[0036] In a further embodiment, the at least one soluble protein may be expressed by a vector and the methods or uses disclosed herein comprise use of or administration of the vector to the animal or plant in need thereof. In an embodiment, the vector is a lytic phage that is able to invade bacteria of the biofilm. In another embodiment, the vector is a mycovirus.

[0037] In another aspect, the present disclosure provides a method of preventing biofilm formation on an indwelling medical device or implant comprising coating the device with at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain.

[0038] In one embodiment, the present disclosure provides a method of preventing biofilm formation on an indwelling medical device or implant comprising coating the device with at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, prior to use in an animal in need thereof. In an embodiment, the method comprises at least two of, at least three of, at least four of, or all of the soluble proteins. [0039] The indwelling medical device or implant can be any device or implant that is inserted into the body of the animal and that is susceptible to biofilm formation. In an embodiment, the indwelling medical device or implant is a catheter, an intravenous tube, a bioprosthetic including, but not limited to a heart valve or a prosthetic joint.

[0040] In an embodiment, the biofilm is caused by any microorganism or microorganisms that have the genetic capacity to synthesize the exopolysaccharides, Pel, Psl, PNAG and/or GAG and combinations thereof. These organisms include, but are not limited to; P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp. Aspergillus spp, Botrytis spp., Trichosporon spp., Acinetobacter spp. and Fusarium spp. In another embodiment, the biofilm may be dependent on the secretion of any exopolysaccharide that is able to be degraded by the soluble glycosyl hydrolases disclosed herein.

[0041] In yet another embodiment, the methods disclosed herein further comprise coating an antimicrobial agent on the indwelling medical device or implant. In one embodiment, the antimicrobial agent is an antibiotic. In another embodiment, the antimicrobial agent is an antifungal agent.

[0042] In yet a further aspect, provided herein is a method of preventing or treating biofilm on a non-medical surface that is susceptible to biofilm comprising coating with or applying to the surface at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain.

[0043] In one embodiment, the present disclosure provides a method of preventing or treating biofilm on a non-medical surface that is susceptible to biofilm formation comprising coating with or applying to the surface with at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain (v) a soluble protein comprising a Sph3 GH domain, and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof. In an embodiment, the method comprises at least two of, at least three of, at least four of, at least five of, or all of the soluble proteins.

[0044] In yet another embodiment, the methods disclosed herein further comprise coating or applying an antimicrobial agent on the nonmedical surface. In one embodiment, the antimicrobial agent is an antibiotic. In another embodiment, the antimicrobial agent is an antifungal agent.

[0045] Also provided herein is an indwelling medical device or implant prepared by the methods disclosed herein.

[0046] In an embodiment, the soluble protein comprising a PsIG GH domain comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 or a glycosyl hydrolase variant thereof.

[0047] In an embodiment, the soluble protein comprising a PelA GH domain comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 or glycosyl hydrolase variants thereof.

[0048] In an embodiment, the soluble protein comprising a PelA GH domain ortholog comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 or glycosyl hydrolase variants thereof.

[0049] In an embodiment, the soluble protein comprising a BpsB GH domain comprises amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 or glycosyl hydrolase variants thereof. [0050] In an embodiment, the soluble protein comprising a PgaB GH domain comprises amino acids 310 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 or glycosyl hydrolase variants thereof.

[0051] In an embodiment, the soluble protein comprising a Sph3 GH domain comprises amino acids 52 to 298 of the Sph3 sequence from Aspergillus fumigatus deposited into GenBank under accession no. EAL92786.1 or a glycosyl hydrolase variant thereof.

[0052] In an embodiment, the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 54 to 304 of the Sph3 A c sequence from Aspergillus clavatus NRRL 1 deposited into GenBank under accession no. EAW09379.1 or a glycosyl hydrolase variant thereof.

[0053] In an embodiment, the soluble protein comprising a Sph3 GH domain ortholog comprises amino acids 43 to 299 of the Sph3 A N sequence from Aspergillus nidulans FGSC A4 deposited into GenBank under accession no. EAA63523.1 or a glycosyl hydrolase variant thereof.

[0054] In an embodiment, the soluble protein comprising an Ega3 GH domain comprises amino acids 46 to 318 of the Ega3 sequence from Aspergillus fumigatus deposited into GenBank under accession no. EAL92787.1 or a glycosyl hydrolase variant thereof.

[0055] In yet further aspects, provided herein is an isolated protein consisting of amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 , an isolated protein consisting of amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2, an isolated protein consisting of amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201.1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 , an isolated protein consisting of amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 , an isolated protein consisting of amino acids 310 to 672 of the PgaB deposited into GenBank under accession no. AAC74108.1 , an isolated protein consisting of amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 , an isolated protein consisting of amino acids 54 to 304 of the Sph3 A c sequence from Aspergillus clavatus NRRL 1 deposited into GenBank under accession no. EAW09379.1 , an isolated protein consisting of amino acids 43 to 299 of the Sph3 AN sequence from Aspergillus nidulans FGSC A4 deposited into GenBank under accession no. EAA63523.1 , and/or an isolated protein consisting of amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1.

[0056] In yet another aspect, the present disclosure provides a vector encoding at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain. In one embodiment, the vector is able to invade the microbial organism. In an embodiment, the vector is a lytic phage. In another embodiment, the vector is a mycovirus.

[0057] In one embodiment, the present disclosure provides a vector encoding (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, such as a PsIG GH soluble protein or ortholog described herein, (ii) a soluble protein comprising a PelA GH domain, such as a PelA GH soluble protein or ortholog described herein, (iii) a soluble protein comprising a BpsB GH domain, such as a BpsB GH soluble protein or ortholog described herein, (iv) a soluble protein comprising a PgaB GH domain, such as a PgaB GH soluble protein or ortholog described herein, (v) a soluble protein comprising a Sph3 GH domain, such as a Sph3 GH soluble protein or ortholog described herein, or (vi) a soluble protein comprising an Ega3 GH domain, such as an Ega3 GH soluble protein or ortholog described herein, or orthologs thereof, or combinations thereof. In an embodiment, the vector is a lytic phage. In another embodiment, the vector is a mycovirus.

[0058] In yet another aspect, the present disclosure provides pharmaceutical compositions comprising at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein comprising a glycosyl hydrolase domain; and a pharmaceutically acceptable carrier. In an embodiment, the pharmaceutically acceptable carrier is a gel, such as Poloxamer.

[0059] In one embodiment, the present disclosure provides pharmaceutical compositions comprising at least one, at least two, at least three, at least four of, at least five of, or all of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, such as a PsIG GH soluble protein or ortholog described herein, (ii) a soluble protein comprising a PelA GH domain, such as a PelA GH soluble protein or ortholog described herein, (iii) a soluble protein comprising a BpsB GH domain, such as a BpsB GH soluble protein or ortholog described herein, (iv) a soluble protein comprising a PgaB GH domain, such as a PgaB GH soluble protein or ortholog described herein; (v) a soluble protein comprising a Sph3 GH domain, such as a Sph3 GH soluble protein or ortholog described herein, and (vi) a soluble protein comprising an Ega3 GH domain, such as a Ega3 GH soluble protein or ortholog described herein, or orthologs thereof, and a pharmaceutically acceptable carrier.

[0060] Other features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples while indicating embodiments of the disclosure are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description. BRIEF DESCRIPTION OF THE DRAWINGS

[0061] The disclosure will now be described in relation to the drawings in which:

[0062] Figure 1 shows a depiction and composition of a microbial biofilm. The formation of a biofilm can be divided into four distinct phases. Planktonic cells attach to a biotic surface such as lung epithelial cells. Here, the bacteria begin secreting the exopolysaccharides allowing attachment to biotic or abiotic surfaces, thereby initiating the biofilm.

[0063] Figure 2 shows the chemical structure of Psl. Psl is composed of a pentasaccharide-repeating unit of D-mannose, D-glucose and L- rhamnose, and is chemically distinct from other known polysaccharides.

[0064] Figure 3 shows Psl operons in several P. aeruginosa species. The psl operon is found in numerous Pseudomonas species (not all are shown), however it is unknown whether all species produce the exopolysaccharide. Due to a mutation in P. aeruginosa PA14, the strain has been demonstrated to be unable to synthesize the Psl polysaccharide. The location of PsIG is located in the same position in the operon across species and its amino acid sequence identity relative to PsIG from P. aeruginosa PA01 is shown. The sequence identity refers to the amino acid sequence identity to PsIG from P. aeruginosa PA01.

[0065] Figure 4 shows Pel operons in several bacterial species. The pel operon is found in a number of bacterial species including those of Geobacter metallireducens and Ralstonia solanacearum (not all species that contain the operon are depicted in this figure). PelA is located at the beginning of each operon.

[0066] Figure 5 shows the chemical structure of poly- (1 ,6)-A/-acetyl-D- glucosamine (PNAG). PNAG is a homopolymer of repeating /V-acetyl-D- glucosamine units, similar to chitin, however it is synthesized with a β( 1 ,6) linkage. PNAG present in the biofilm is often partially deacetylated. This form of the polymer is commonly referred to as dPNAG, and is the medically relevant form of the polymer.

[0067] Figure 6 shows Pga operons in several Gram-negative species. The pga operon is found in numerous bacterial species.

[0068] Figure 7 shows the domain boundaries of PelA. The approximate boundaries for each domain are indicated on the diagram with the relative sizes proportional to the number of residues in each predicted region. The following domains represent those of PelA as predicted by Phyre 2 (Kelley & Sternberg 2009); GH (hydrolase), reductase, carbohydrate deacetylase and a region with no predicted function, the β-jelly roll. The small grey rectangles represent regions of the PelA 4 7-303 protein for which Phyre 2 was unable to make high confidence predictions, or for which the entire domain could not be modeled.

[0069] Figure 8 shows sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel of PslG 3 i -4 42. Analysis from SDS-PAGE gel indicates that the protein is >95% pure and has a molecular weight of ~48 kDa which is consistent with the expected molecular weight of the purified protein.

[0070] Figure 9 shows the X-ray crystal structure of PslG 3 i.4 4 2. (A) The enzyme is a two-domain protein composed of a TIM-barrel domain and a β- sandwich domain (B) The putative active site groove, containing the proposed catalytic residues is -40 A in length and runs equatorially across the TIM- barrel domain.

[0071] Figure 10 shows inhibition of PsI Biofilm from P. aeruginosa PA01 pBADps/ strain. Titration of PslG3i- 44 2 into static P. aeruginosa cultures with inducible PsI production indicates that addition of >10 nM of PSIG31-442 is sufficient to inhibit PsI biofilm formation. Titration of PslG 3 i-442E165Q/E276Q (EC50 = 466.5 ± 1.1 nM) also results in biofilm inhibition but requires significantly higher concentrations of protein relative to the WT enzyme (EC50 = 4.1 ± 1.1 nM). [0072] Figure 1 1 shows the dispersion of pre-formed Psl-biofilms. (A) Addition of 86 nM of PSIG3- 42 to pre-formed Psl biofilms resulted in a significant reduction of the biofiim after 20 min and complete abolishment of the biofiim after 35 min. (B) Addition of 58 nM of PSIG31-442 was able to disperse the biofiim in 30 minutes. When PslG 3 i. 442 E165Q/E276Q double catalytic variant was added in 100-fold excess compared to the non-variant enzyme (5 μΜ), no significant difference was observed compared to that of the untreated biofiim. Single enzyme catalytic variants were also impaired in their ability to disrupt the biofiim.

[0073] Figure 12 shows sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel of PelA 47- 303. Analysis from SDS-PAGE gel indicates that the protein is >95% pure and has a molecular weight of -28 kDa which is consistent with the expected molecular weight of the purified protein.

[0074] Figure 13 shows the X-ray crystal structure of PelA 47.303 . The core structure represents a β 8 7 TIM-barrel fold with the β-sheets in light grey and the a-helices in dark grey. Extra loop insertions within the core backbone structure are illustrated in light grey. (A) Top view of the cartoon representation of PelA 47 .3 03 . (B) Side view cartoon representation of PelA 47.303 showing the extra loops clustering in two main groups. (C) Topological representation showing in detail the eight β-sheets and seven a-helices of the core structure of the TIM- barrel. There are four loops inserted within the core structure labeled loop 1 — loop 4. The light grey segment represents the C- terminus that connects to the following putative reductase domain. The N- and C-termini are indicated at either end by N and C, respectively.

[0075] Figure 14 shows conserved residues lining the putative binding cleft in PelA 47 . 303 . (A) The conservation profile of PelA 47.303 as represented by the ConSurf server (Ashkenazy et al 2010) with the conservation bar shown. (B) A close up of the highly conserved region in PelA 47 . 303 . Highly conserved acidic residues line the putative binding cleft of the protein. Spatial separation was measured between the carboxyl oxygens of the residues and indicated in angstroms (A).

[0076] Figure 15 shows inhibition of Pel biofilm formation by PelA 4 7-303. Purified PelA 47 . 303 and variants at increasing concentrations of 0.5, 1 , 2, 4, 8 and 16 μΜ. Arabinose at 0.5% (w/v) was added to induce Pel polysaccharide production. The first bar in each graph contained buffer D and represents the positive control. The results were normalized against the buffer control in each plate. Triplicate reactions were incubated for 48 h at ambient temperature. Error bars represent the standard error of the mean.

[0077] Figure 16 shows inhibition of Pel biofilm by PelA 35- 29i from P. protogens Pf-5. Titration of PelA 3 5 -2 9i from P. protogens Pf-5 in static wild-type P. aeruginosa PA01 wspFApslP BAD pel culture. The addition of PelA 3 5 -2 gi from P. protogens Pf-5 prevented biofilm formation at >70 nM and had an EC 5 o of 69.3 ± 1.2 nM. PslG3i-4 4 2 was unable at preventing biofilm formation indicating that this inhibition is enzyme specific.

[0078] Figure 17 shows dispersion of pre-formed Pel-biofilms. (A) Addition of PelA 7-3 03 from P. aeruginosa PA01 to pre-formed Pel biofilms resulted in biofilm dispersal after 2 hours while putative catalytic variants retained the majority of the biofilm. (B) Titration of wild-type PelA 35-2 9i from P. protogens Pf-5 into pre-formed Pel biofilms was able to disperse the biofilm in as little as 30 min.

[0079] Figure 18 shows biofilm inhibition through the coating of PslG 3 i. 442 and PelA 47- 303 to polystyrene plastic. (A) Treatment of polystyrene plates with 40 μg/mL of PelA 47 - 3 o3 or BSA at 4 °C overnight in 1 x PBS buffer. The wells were washed prior to bacterial inoculation. (B) Treatment of polystyrene slides with 40 pg/mL of adsorbed PslG 3 i -44 2 prevented bacterial cell attachment and biofilm formation as visualized by the lack of SYTOX green staining using confocal microscopy. (C) Covalent attachment of PslG3i -4 42 to glass was also effective at preventing biofilm formation whereas a BSA (negative control) was unable to prevent biofilm formation. (D) Covalent attachment of PslG 3 i - 4 2 to glass was effective at preventing cell attachment and biofilm formation for at least 8 days as visualized by a lack of SYTOX green staining.

[0080] Figure 19 shows sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel of BpsB constructs. Analysis from SDS- PAGE gel indicates that each protein is >95% pure and has a molecular weight of -79 kDa for BpsB27-7oi and -42 kDa for BpsB 3 i 8- 67o which are consistent with the expected molecular weights of each purified protein.

[0081] Figure 20 shows the crystal structure of BpsB 3 8-6 7o. The enzyme is a (β/α) 8 TIM-barrel fold with an electronegative groove -41 A long and 1 1 A wide.

[0082] Figure 21 shows sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel of PgaB 310 -672- Analysis from SDS-PAGE gel indicates that the protein is >95% pure and has a molecular weight of -42 kDa for PgaB 3 io-672 which is consistent with the expected molecular weights of the purified protein.

[0083] Figure 22 shows inhibition of PNAG-dependent biofilms. (A) Titration curves of BpsB 27 -7o-i , BpsB 3 i 8-67o, PgaB 22 -672, and PgaB 3 i o-672 added prior to inoculation for the inhibition of E. coli biofilms. (B) Testing of, BpsB 3 -| 8 . 670, on the inhibition of Staphylococcus carnosus biofilm formation. Background staining represents S. carnosus treated with gentamycin.

[0084] Figure 23 shows dispersion of pre-formed PNAG-dependent biofilms. (A) Titration curves of BpsB 27 -7oi , BpsB 318-6 7o, PgaB 2 2-672 > and PgaB 3 io-672 to preformed E. coli biofilms after 60 min incubation. (B) Testing of different 96-well plates for BpsB-mediated E. coli biofilm dispersal. (C) Comparing BpsB-mediated E. coli biofilm dispersal against other known biofilm degrading enzymes, PelA 47-3 o 3 and PslG 3 i - 4 2.

[0085] Figure 24 shows a reducing sugar assay on dPNAG. BpsB 2 7-7oi , BpsB 318-670 , PgaB 22 -672, and PgaB 31 0-672 hydrolyze dPNAG. BPSB27-701 and BpsB3i8-67o show ~4 times higher rates of dPNAG hydrolysis using the reducing sugar assay than PgaB22-672 PgaB3i 0-672■

[0086] Figure 25 shows the inhibition of GAG-dependent biofilm formation by PelA 47- 303. (A) Purified PelA 47- 303 and variants were added to A. fumigatus cultures prior to biofilm formation. Cultures were grown for 20 hours in Brian media. (B) Purified P. protogens PelA 35- 29i was added in the same manner and was also shown to prevent GAG-dependent biofilm formation. The amount of GAG biofilm present following growth was assessed using crystal violet staining.

[0087] Figure 26 shows the results of a reducing sugar assay when purified GAG, from A. fumigatus was treated with the glcosyl hydrolases PelA 47-3 o3 and its inactive catalytic variant E218A and Sph3 52 -298 and its inactive variant D166A. An increase in reducing ends indicates that the enzymes are capable of hydrolyzing the glycosidic bonds of the GAG polysaccharide. Under assay conditions, PelA 47- 303 lead to the production of ~2-fold more reducing ends than Sph3s2-298- PelA refers to PelA 47-3 o3 and Sph3 refers to Sph3s2-298-

[0088] Figure 27 shows the dispersion of pre-formed GAG biofilms using PelA 7-30 3 and P. protogens PelA 35 -29i . Exogenous addition of PelA 7 . 303 and P. protogens PelA 35 . 2 9i to pre-formed A. fumigatus GAG biofilms resulted in the elimination of the GAG biofilms as detected through the crystal violet assay. In comparison, PelA 47-303 variants D160A and E218A as well as a BSA control did not disperse the biofilm.

[0089] Figure 28 shows the dispersion of pre-formed GAG biofilms using RagA 6 -317 and G/T7PelA 2 3-277- Addition of RagA 6 i-3i7 to pre-formed A. fumigatus GAG biofilms resulted in the elimination of the GAG biofilms as detected through the crystal violet assay. The putative glycosyl hydrolase PelA 47- 303 was used as a positive control while the PelA 47-303 variant E218A acts as negative control. GmPelA 2 3-277 was less effective at eliminating GAG biofilms as detected through the crystal violet assay than PelA and RagA. [0090] Figure 29 show that PelA 4 7-303 protects epithelial cells from damage caused by A. fumigatus infection. The exogenous addition of wild- type PelA 4 7-303 blocked the ability of A. fumigatus to induce pulmonary epithelial cell injury as measured by a chromium release assay over a period of 16 h while PelA 47-3 o3 E218A did not prevent epithelial cell damage. Cell damage results in Chromium release and thus less damage is visualized by a lower percentage of chromium release.

[0091] Figure 30 depicts the enhancement of the bacteriocidal properties of the HL60 neutrophil-like cell line in the presence of PelA 47-3 o3. Biofilms of P. aeruginosa over-expressing the pel operon were grown for 20 h and incubated with neutrophil-like differentiated HL60 cells for 2 h. P. aeruginosa killing was assessed by quantitative CFU plating. The presence of PelA 47-30 3 with neutrophils lead to ~2-fold more bacterial killing than neutrophils alone.

[0092] Figure 31 shows the results of encapsulation of PelA 47 .303 into the thermoreversible gel PF-127. Addition of 1 *PBS or 20% PF-127 did not result in dispersal of the Pel-dependent biofilm however addition of PelA 47 - 30 3 at three different concentrations was able to disperse the biofilm in a 1 hour period.

[0093] Figure 32 shows the result of biofilm dispersal of clinical and environmental isolates of P. aeruginosa by PelA 47- 3 0 3, PslG3i -44 2 and when these enzymes used in combination. A combination of PelA 47- 303 and PslG 3 i- 442 resulted in≥90% reduction in biofilm biomass as detected through crystal violet staining. Class I and Class II strains are solely dependent on Pel and Psl production, respectively. Class III strains are redundant exopolysaccharide matrix producers as mutations in both the pel and psl operons are necessary to eliminate the biofilm production in the microtiter dish assay. Finally, Class IV strains are matrix over-producers that form biofilms characterized by Pel and Psl overproduction. [0094] Figure 33 shows a diagram of genes required for the biosynthesis of GAG on chromosome 3 of A. fumigatus.

[0095] Figure 34 shows the construct generation of Sph3s2-298- The TMHMM server indicates that amino acids 20-42 compose a transmembrane domain. A construct composed of residues 52-298 was used to generate a soluble protein.

[0096] Figure 35 shows a sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel of Sph3 52 -298- Analysis from SDS-PAGE gel indicates that the protein is >95% pure and has a molecular weight of -30 kDa which is consistent with the expected molecular weight of the purified protein.

[0097] Figure 36 shows the dispersion of pre-formed GAG biofilms using Sph3. Addition of Sph3 52 -298 to pre-formed A. fumigatus GAG biofilms resulted in the elimination of the GAG biofilms as detected through the crystal violet assay. The putative glycosyl hydrolase PelA 47- 303 was used as a positive control while the PelA 47-30 3 variant E218A acts as negative controls.

[0098] Figure 37 shows that orthologs Sph3 5 2 -2 98 and Sph3AC(54-304 ) from different Aspergillus species are capable of disrupting GAG-dependent biofilms. The mutation of putative catalytic residues abrogates biofiim disruption by the enzyme. Sph3Ac refers to Sph3AC(54-304).

[0099] Figure 38 shows dose-dependent activity of recombinant Sph352-298 against preformed biofilms of multiple clinical isolates. Biofilms of the indicated A. fumigatus strains were grown for 24 hours, and then incubated for 1 hour with the indicated concentration of Sph3 5 2-298- Biofiim disruption was measured by crystal violet staining of the residual biofiim mass after gentle washing.

[00100] Figure 39 demonstrates that the opportunistic fungal pathogen Trichosporon asahii produces an exopolysaccharide which is recognized by GalNAc-specific lectin indicating the presence of GAG on the surface of T. ashaii. This exopolysccharide can be degraded by treatment with 0.5 μΜ Sph3AC(54-304) for 3 hours results in a complete loss of detectable surface GalNAc (top right). Fungi were counterstained with DRAQ5 (bottom panels). Sph3 A c refers to Sph3 A c(54-304).

[00101] Figure 40 demonstrates that recombinant hydrolases protect airway epithelial cells from pathogen-induced damage. (A) Chromium-loaded airway epithelial cell line A549 was incubated with Af2943 conidia and recombinant hydrolases. Chromium released into supernatant was measured at indicated time points. The presence of active hydrolase resulted in less chromium release while enzyme variants failed to protect the airway cells, resulting in a loss of chromium equal to that of the control. (B) Epithelial cell damage by P. aeruginosa was measured by determining the fraction of lactate dehydrogenase (LDH) in the presence and absence of Sph3A (43-299)- This result demonstrates that Sph3 A N(43-299) is effective at preventing bacterial damage.

[00102] Figure 41 indicates that hydrolases have synergistic effects in combination with antifungal drugs on the metabolic activity of A. fumigatus. A. fumigatus conidia were grown in the presence of indicated antifungal drugs and hydrolases at various concentrations for 20 h at 37°C, 5% C0 2 , and their metabolic activity measured via XTT assay. MIC50 defined as the drug concentration resulting in 50% of the metabolic activity of the sample not exposed to drug. Sph3Ac refers to Sph3AC(54-304), Sph3 A N refers to Sph3 A N(43- 299) and Sph3AF refers to Sph3 52- 298. N.D.: Not Determined.

[00103] Figure 42 demonstrates that the glycosyl hydrolases PslG 3 i_ 4 42 and PelA 47-3 o3 are able to potentiate the antibiotic colistin dosed at a final concentration of 100 g/mL. No growth (NG) was observed on LB agar plates indicating that in the presence of the glcoysyl hydrolases, colistin resulted in >100-fold more bacterial killing than in the absence of the enzyme.

[00104] Figure 43 demonstrates the degradation of the GAG exopolysaccharide coating A. fumigatus by Sph3 A c(5 4- 3o 4 ) increases the activity of antifungals through enhancing their ability to penetrate fungal cells. Sph3Ac refers to Sph3AC(54-304).

[00105] Figure 44 shows the taxonomic relationship of fungal classes containing the GAG biosynthetic cluster as identified through bioinformatics analysis.

[00106] Figure 45 demonstrates that staining Botrytis cinerea with a GalNAc-specific lectin detects the presence of GAG on the surface of B. cinerea. Treatment of hyphae of B. cinerea with recombinant A. clavatus Sph3AC(54-304) resulted in a complete loss of this exopolysaccharide indicating that Sph3AC(54-304) is able to hydrolyze the GAG on the surface of the hyphae. Sph3Ac refers to Sph3AC(54-304).

[00107] Figure 46 shows a sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel of Ega3 4 6-3i s- To determine whether the apparent mass of Ega3 4 6-3is was larger due to glycosylation a sample of protein was treated with endoglycosidase H (EndoH). The EndoH treated Ega3 4 6-318 produced a band near the predicted mass of the unglycosylated protein at -31 kDa.

[00108] Figure 47 shows the ability of Ega346 -3 is to inhibit and disperse A. fumigatus biofilms. (A) The presence of Ega3 4 6-3is prevented biofilm formation in a microtiter dish and also allowed for disruption of the biofilm. (B) While untreated hyphae display extensive staining with the GalNAc specific fluorescent lectin SBA-FITC, treatment of the hyphae with 0.5 μΜ Ega3 46- 318 for 3 hours resulted in a complete loss of detectable GalNAc on the surface.

DETAILED DESCRIPTION

[00109] The present inventors have demonstrated that the exogenous application of putative hydrolases involved in the biosynthesis of the exopolysaccharides: Psl, Pel, poly-βθ ,6)- V-acetyl-D-glucosamine (PNAG) and galactosaminogalactan (GAG) may be utilized to inhibit and disperse microbial biofilms that employ these sugar polymers in biofilm formation. Definitions

[00110] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. The following definitions supplement those in the art and are directed to the present disclosure and are not to be imputed to any related or unrelated case. Generally, nomenclatures used in connection with, and techniques of, molecular biology, immunology, microbiology, genetics, protein and nucleic acid chemistry and hybridization described herein are those well known and commonly used in the art. Methods and techniques employed in the present disclosure are generally performed according to conventional methods known in the art and as described, for example, in general references such as Sambrook et al, Molecular Cloning: A Laboratory Manual, 2 nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989) and Ausubel et al, Current Protocols in Molecular Biology, Greene Publishing Associates (1992). Although any methods and materials similar or equivalent to those described herein can be used in the practice of the invention, particular materials and methods are described herein.

[00111] As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise.

[00112] As used herein, the words "comprising" (and any form of comprising, such as "comprise" and "comprises"), "having" (and any form of having, such as "have" and "has"), "including" (and any form of including, such as "includes" and "include") or "containing" (and any form of containing, such as "contains" and "contain") are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.

[00113] The term "administering" means to provide or give an agent (e.g. therapeutic agent) to an animal or plant by an effective route. [00114] The term "biofilm" as used herein refers to a composition or matrix of microorganism strains/species formed on either a biotic or abiotic surface. These microorganisms are encapsulated in a matrix generally containing proteins, nucleic acids and exopolysaccharides as the main components.

[00115] The phrase "biofilm-related infection" as used herein refers to an infection related to the formation of biofilm on a biotic surface, including, without limitation, epithelial cells of skin, eye and organs, such as the lung and respiratory system as well as surfaces and tissues of plants.

[00116] The term "lung infection" as used herein refers to respiratory infections or disease, which includes but is not limited to invasive aspergiiliosis, an acute disease of the immuno-compromised host or chronic aspergillus infection that occurs in immuno-competent individuals with compromised lung function.

[00117] The term "chronic pulmonary disease" as used herein refers to respiratory infections or disease, which includes but is not limited to cystic fibrosis and pneumonia.

[00118] The term "encoded by an exopolysaccharide biosynthetic operon or functional gene cluster" as used herein refers to a nucleic acid sequence of a microbial operon or functional gene cluster that encodes a protein involved in the production of an exopolysaccharide by the microbial organism.

[001 9] The term "glycosyl hydrolase domain" or "GH domain" as used herein refers to a protein domain that encodes a putative glycosyl hydrolase, which may be identified by comparison to known glycosyl hydrolase family members. A glycosyl hydrolase enzyme is able to hydrolyze glycosidic bonds.

[00120] The term "isolated" refers to a nucleic acid or protein substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors, or other chemicals when chemically synthesized.

[00121] The term "Psl" as used herein refers to the exopolysaccharide composed of a pentassaccharide repeating unit of D-mannose, D-glucose and L-rhamnose and is distinct from other known polysaccharides (Figure 2).

[00122] The term "ps/G" as used herein refers to the 7 th open reading frame of the psl operon that encodes for proteins required for the biosynthesis of Psl. The psl operon is found in several Pseudomonas species. The term "PsIG" as used herein refers to the encoded protein as will be clear in context.

[00123] The term "Pel" as used herein refers to the exopolysaccharide that is one of the main components of biofilm that forms at the air-liquid interface and is referred to as pellicles.

[00124] The term "pelA" as used herein refers to the 1 st open reading frame of the pel operon that encodes for proteins required for biosynthesis of Pel. The pel operon is found in numerous Pseudomonas species. pelA orthologs include, without limitation, ragA from Ralstonia solanacearum and pelA from Geobacter metallireducens. The term "PelA" or "RagA" as used herein refers to the encoded proteins as will be clear in context.

[00125] The term "PNAG" as used herein refers to the homopolymer of repeating N-acetyl-D-glucosamine units, similar to chitin, however it is synthesized with a β(1 ,6) linkage. The partially deacetylated form is called dPNAG.

[00126] The term "bpsB" as used herein refers to the 2 nd open reading frame of the bps operon that encodes for proteins required for PNAG biosynthesis. The bps operon is found in Bordetella species. The term "BpsB" as used herein refers to the encoded protein as will be clear in context.

[00127] The term "pgaB" as used herein refers to the 2 nd open reading frame of the pga operon that encodes for proteins required for PNAG biosynthesis. The pga operon is found in E. coli and numerous Gram-negative species and is sometimes annotated as the hms operon. The term "PgaB" as used herein refers to the encoded protein as will be clear in context.

[00128] The term "GAG" as used herein refers to a heterogeneous, linear extracellular polymer that is composed of a1-4 linked galactose and a1- 4 linked partially deacetylated /V-acetylgalactosamine.

[00129] The term u sph3" as used herein refers to the exon coding sequence with exon chromosomal coordinates 1 ,999,871 to 1 ,999,654 and 1 ,999,541 to 1 ,999, 184 and 1 ,998,991 to 1 ,998,671 located in a functional gene cluster on chromosome 3 in A. fumigatus Af293, which encodes for a putative glycosyl hydrolase. sph3 orthologs include, without limitation, sph3kc from Aspergillus clavatus, and sp/?3 A N from Aspergillus nidulans. The terms "Sph3", "Sph3 A c" or "Sph3 A N" as used herein refer to the respective encoded protein as will be clear in context.

[00130] The term "ega3" as used herein refers to the exon coding sequence with exon chromosomal coordinates 1 ,995,843 to 1 ,996,799 located in a functional gene cluster on chromosome 3 in A. fumigatus Af293, which encodes for a putative glycosyl hydrolase. The term "Ega3" as used herein refers to the encoded protein as will be clear in context.

[00131] The term "soluble protein" as used herein refers to a protein lacking a signal sequence or transmembrane domain(s) and if expressed from a nucleotide sequence in a cell, is not attached/associated with the membrane or other non-soluble components.

[00132] The term "antimicrobial agent" as used herein refers to any substance that kills microorganisms or inhibits their growth. These may include but are not limited to; antibiotics, antimicrobial peptides, chemotherapeutic agents, antifungals, fungicides, chemical disinfectants such as, but not limited to alcohols, aldehydes and silver, antimicrobial peptides, biocides such as benzyalkonium chloride (BAC), cetylpyridinium chloride (CPC) and chlorhexidine (CHX), or any natural or recombinant agents that demonstrate antimicrobial activity. [00133] The term "coating" refers to the immobilization of soluble proteins to a solid abiotic support, either non-specifically through protein absorption and chemical cross-linking or specifically through recombinant or chemical means including but not limited to; the Staudinger ligation reaction, "click" chemistry, expressed protein ligation, chemoenzymatic methods or through surface modification as practiced in the art.

[00134] The phrase "surface susceptible to biofilm" as used herein refers to any biotic or abiotic surface that is prone to bacterial colonization, growth and biofilm formation.

[00135] The term "biofouling" refers to the adhesion and accumulation of microorganisms on an abiotic surface through the use of a biofilm.

[00136] The term "administering" in the context of administering to an animal is defined as any conventional route for administering an agent to an animal for use, for example, in reducing or preventing biofilm, as is known to one skilled in the art. This may include, for example, administration via the parenteral (i.e. subcutaneous, intradermal, intramuscular, etc.), the mucosal surface route, or through aerosolization, for example, through the use of a nebulizer for administration into the airways of animals. In other embodiments this may include oral administration to the animal. The dose of the agent may vary according to factors such as the health, age, weight and sex of the animal. The dosage regime may be adjusted to provide the optimum dose. One skilled in the art will appreciate that the dosage regime can be determined and/or optimized without undue experimentation.

[00137] The term "administering" in the context of a plant is defined as applying to the surface of the plant through spraying. It may also include the insertion of the gene encoding the glycosyl hydrolase into the genome or a plasmid in the plant such that the plant has the ability to produce a functional glycosyl hydrolase that can be secreted.

[00138] To "inhibit" or "suppress" or "lower" or "reduce" an activity, such as biofilm formation, is to reduce the function or activity when compared to otherwise same conditions except for a condition or parameter of interest, or alternatively, as compared to another condition or control.

[00139] To "disperse", such as biofilms, is to reduce, liberate, or degrade biomass and matrix components thereof that are part of or associated with the biofilm when compared to another condition or control.

[00140] The term "animal" as used herein includes all members of the animal kingdom including mammals, suitably humans.

[00141] The term "plant" as used herein includes all members of the plant kingdom, such as flowering and bulb-forming plants, and includes whole plants, and plant parts, such as fruit, that are susceptible to the formation of biofilm from plant pathogens.

[00142] The term "treatment or treating" as used herein means an approach for obtaining beneficial or desired results, including clinical results. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e. not worsening) state of disease, preventing spread of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.

[00143] The term "treating a biofilm-related infection" as used herein refers to penetrating or dispersing biofilm such that the biofilm biomass is reduced or compromised thereby causing microorganisms to become exposed and vulnerable to attack, either by the immune system or by exogenous agents, either chemical or biological in nature, such as antimicrobials.

[00144] The term a "therapeutically effective amount", "effective amount" or a "sufficient amount" of a compound of the present disclosure is a quantity sufficient to, when administered to the animal, including a mammal, for example a human, or plant, effect beneficial or desired results, including clinical results, and, as such, an "effective amount" or synonym thereto depends upon the context in which it is being applied. For example, in the context of inhibiting biofilm formation or dispersing biofilm, it is an amount of the agent sufficient to achieve such an inhibition or dispersal as compared to the response obtained without administration of the agent. The amount of a given agent that will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical formulation, the route of administration, the type of condition, disease or disorder, the identity of the animal/plant or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art. As defined herein, a therapeutically effective amount of an agent may be readily determined by one of ordinary skill by routine methods known in the art.

[00145] Moreover, a "treatment" or "prevention" regime with a therapeutically effective amount of an agent may consist of a single administration, or alternatively comprise a series of applications. For example, the agent may be administered at least once a week. However, in another embodiment, the agent may be administered from about one time per week to about once or more daily for a given treatment. The length of the treatment period depends on a variety of factors, such as the severity of the disease, the age of the animal or plant, the concentration and the activity of the agent, or a combination thereof. It will also be appreciated that the effective dosage of the agent used for the treatment or prophylaxis may increase or decrease over the course of a particular treatment or prophylaxis regime. Changes in dosage may result and become apparent by standard diagnostic assays known in the art. In some instances, chronic administration may be required.

[00146] The term "nucleic acid" as used herein refers to a sequence of nucleotide or nucleoside monomers consisting of naturally occurring bases, sugars, and inter-sugar (backbone) linkages, and includes single stranded and double stranded molecules, RNA and DNA. The term also includes modified or substituted oligomers comprising non-naturaily occurring monomers or portions thereof, which function similarly, which are referred to herein as "chemical analogues" and/or "oligonucleotide analogues" such as "peptide nucleic acids". Such modified or substituted nucleic acids may be preferred over naturally occurring forms because of properties such as enhanced cellular uptake, or increased stability in the presence of nucleases. The term also includes chimeric nucleic acids that contain two or more chemically distinct regions. For example, chimeric nucleic acids may contain at least one region of modified nucleotides that confer beneficial properties (e.g. increased nuclease resistance, increased uptake into cells), or two or more nucleic acids of the disclosure may be joined to form a chimeric nucleic acid.

[00147] The term "isolated nucleic acid molecule" as used herein refers to a nucleic acid substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors, or other chemicals when chemically synthesized. An isolated nucleic acid is also substantially free of sequences, which naturally flank the nucleic acid (i.e. sequences located at the 5' and 3' ends of the nucleic acid) from which the nucleic acid is derived. The term "nucleic acid" is intended to include DNA and RNA and can be either double stranded or single stranded, and represents the sense or antisense strand.

[00148] The term "variant" as used herein includes modifications, substitutions, additions, derivatives, analogs, fragments or chemical equivalents of the nucleic acid or amino acid sequences disclosed herein that perform substantially the same function in substantially the same way.

[00149] As used herein, the term "glycosyl hydrolase variant thereof" means an amino acid sequence with at least 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 99% sequence identity to the amino acid sequence of interest and which functions to inhibit or disperse microbial biofilms. In some embodiments, certain catalytic residues are maintained while other residues are altered. Residues that are particularly relevant for catalytic function include those residues disclosed in the Examples.

[00150] As used herein, the term "protein" or "polypeptide" refers to a sequence of amino acid residues encoded by a nucleic acid molecule. Within the context of the present disclosure, a polypeptide of the disclosure may in one embodiment include various structural forms of the primary protein. For example, a polypeptide of the disclosure may be in the form of acidic or basic salts or in neutral form. In addition, individual amino acid residues may be modified by oxidation or reduction.

[00151] The proteins and polypeptides of the present disclosure may also include truncations, analogs, homologs and orthologs of the proteins and polypeptides as described herein having substantially the same function as the proteins or polypeptides of the present disclosure, such as the ability to inhibit and/or disperse microbial biofilms and/or prevent biofilm formation.

[00152] Analogs of the proteins described herein, may include, but are not limited to an amino acid sequence containing one or more amino acid substitutions, insertions, and/or deletions. Amino acid substitutions may be of a conserved or non-conserved nature. Conserved amino acid substitutions involve replacing one or more amino acids of the proteins of the disclosure with amino acids of similar charge, size, and/or hydrophobicity characteristics. When only conserved substitutions are made the resulting analog should be functionally equivalent. Non-conserved substitutions involve replacing one or more amino acids of the amino acid sequence with one or more amino acids, which possess dissimilar charge, size, and/or hydrophobicity characteristics.

[00153] Conservative substitutions are described in the patent literature, as for example, in United States Patent No. 5,264,558. It is thus expected, for example, that interchange among non-polar aliphatic neutral amino acids, glycine, alanine, proline, valine and isoleucine, would be possible. Likewise, substitutions among the polar aliphatic neutral amino acids, serine, threonine, methionine, asparagine and glutamine could possibly be made. Substitutions among the charged acidic amino acids, aspartic acid and glutamic acid, could probably be made, as could substitutions among the charged basic amino acids, lysine and arginine. Substitutions among the aromatic amino acids, including phenylalanine, histidine, tryptophan and tyrosine would also likely be possible. Other substitutions might well be possible.

[00154] The term "sequence identity" as used herein refers to the percentage of sequence identity between two polypeptide sequences or two nucleic acid sequences. To determine the percent identity of two amino acid sequences or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino acid or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity=number of identical overlapping positions/total number of positions.times.100%). In one embodiment, the two sequences are the same length. The determination of percent identity between two sequences can also be accomplished using a mathematical algorithm. An optional, non-limiting example of a mathematical algorithm utilized for the comparison of two sequences is the algorithm of Karlin and Altschul, 1990, Proc. Natl. Acad. Sci. U.S.A. 87:2264-2268, modified as in Karlin and Altschul, 1993, Proc. Natl. Acad. Sci. U.S.A. 90:5873-5877. Such an algorithm is incorporated into the N BLAST and XBLAST programs of Altschul et al., 1990, J. Mol. Biol. 215:403. BLAST nucleotide searches can be performed with the NBLAST nucleotide program parameters set, e.g., for score=100, wordlength=12 to obtain nucleotide sequences homologous to a nucleic acid molecules of the present disclosure. BLAST protein searches can be performed with the XBLAST program parameters set, e.g., to score-50, wordlength=3 to obtain amino acid sequences homologous to a protein molecule of the present disclosure. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., 1997, Nucleic Acids Res. 25:3389- 3402. Alternatively, PSI-BLAST can be used to perform an iterated search, which detects distant relationships between molecules (Id.). When utilizing BLAST, Gapped BLAST, and PSI-Blast programs, the default parameters of the respective programs (e.g., of XBLAST and NBLAST) can be used (see, e.g., the NCBI website). Another optional, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, 1988, CABIOS 4:1 1 -17. Such an algorithm is incorporated in the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. The percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically only exact matches are counted.

[00155] The term "pharmaceutically acceptable" means compatible with the treatment of animals, suitably humans.

[00156] The term "a cell" as used herein includes a plurality of cells. Administering a compound to a cell includes in vivo, ex vivo and in vitro treatment. Methods and Uses:

[00157] In one aspect, the present disclosure provides a method of treating or preventing a biofilm-related infection comprising administering at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain. Also provided herein is use of at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain for treating or preventing a biofilm-related infection. Further provided is use of at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain in the manufacture of a medicament for treating or preventing a biofilm-related infection. Even further provided is at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain for use in treating or preventing a biofilm-related infection.

[00158] The present disclosure also provides a method of treating or preventing a biofilm-related infection comprising administering at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, to an animal or plant in need thereof.

[00159] Also provided is use of at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, for treating or preventing a biofilm related infection in an animal or plant in need thereof. Further provided is use of at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, in the preparation of a medicament for treating or preventing a biofilm-related infection in an animal or plant in need thereof. Even further provided is at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, for use in treating or preventing a biofilm- related infection in an animal or plant in need thereof.

[00160] In an embodiment, at least two of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00161] In another embodiment, at least three of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00162] In yet another embodiment, at least four of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00163] In a further embodiment, at least five of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00164] In yet another embodiment, (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00165] In one embodiment, the soluble protein comprising a PsIG GH domain and the soluble protein comprising a PelA GH domain, or orthologs thereof, may be administered or used.

[00166] In another embodiment, the soluble protein comprising a PsIG GH domain and the soluble protein comprising a BpsB and/or PgaB GH domain, or orthologs thereof, may be administered or used.

[00167] In yet another embodiment, the soluble protein comprising a PsIG GH domain and the soluble protein comprising a Sph3 GH domain, or orthologs thereof, may be administered or used.

[00168] In a further embodiment, the soluble protein comprising a PsIG GH domain and the soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00169] In one embodiment, the soluble protein comprising a PelA GH domain or ortholog thereof and the soluble protein comprising a BpsB and/or PgaB GH domain, or orthologs thereof, may be administered or used.

[00170] In yet another embodiment, the soluble protein comprising a PelA GH domain or ortholog thereof and the soluble protein comprising a Sph3 GH domain, or orthologs thereof, may be administered or used. [00171] In a further embodiment, the soluble protein comprising a PelA GH domain or ortholog thereof and the soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00172] In one embodiment, the soluble protein comprising a BpsB and/or PgaB GH domain and the soluble protein comprising a Sph3 GH domain, or orthologs thereof, may be administered or used.

[00173] In another embodiment, the soluble protein comprising a BpsB and/or PgaB GH domain and the soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be administered or used.

[00174] The biofilm-related infection may be any microbial infection in the body that has formed a layer of biofilm on the body surface or medical implant or bioprosthetic device. In one embodiment, the biofilm-related infection may be the result of a wound, burn infection or keratitis. In another embodiment, the biofilm-related infection may be a lung infection, wherein the animal has chronic pulmonary disease. In another embodiment, the biofilm- related infection may be a lung infection wherein the animal has invasion aspergillosis. In another embodiment, the biofilm-related infection may be from chronic pulmonary disease.

[00175] In an embodiment, the at least one soluble protein comprising a glycosyl hydrolase potentiates neutrophil killing of the microorganism. In a particular embodiment, the soluble protein is a PelA protein disclosed herein.

[00176] In another embodiment, the biofilm-related infection may be any microbial infection that has formed a layer of biofilm on the surface of or within a plant or plant part.

[00177] In an embodiment, the biofilm-related infection may be caused by any Pel-dependent, PsI-dependent, PNAG-dependent or GAG-dependent biofilm. In one embodiment, the biofilm-related infection may be caused by any microorganism that has the genetic capacity to synthesize the exopolysaccharides, Pel, Psl, PNAG and/or GAG and combinations thereof. These organisms include, but are not limited to; P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp, Acinetobacter spp., Trichosporon asahii, Saccharata proteae, Zopfia rhizophila, Phaeosphaeria nodorum, Setosphaeria turcica, Botrytis cinerea, Cryphonectria parasitica, Melanconium sp., Verticillium dahlia, Nectria haematococca, Neurospora crassa, Leptosphaeria maculans, Pleomassaria siparia, Cochliobolus heterostrophus, Pyrenophora tritici- repentis, Blumeria graminis, Marssonina brunnea, Sclerotinia sclerotiorum, Taphrina deformans, Cercospora zeae-maydis and Fusarium spp. In another embodiment, the biofilm may be dependent on the secretion of any exopolysaccharide that is able to be degraded by the soluble glycosyl hydrolases disclosed herein.

[00178] The soluble proteins disclosed herein provide for dispersion or degradation of the biofilm, providing an opportunity for other anti-microbial agents to access and treat the microbial infection. Accordingly, in another embodiment, the methods and uses for treating or preventing a biofilm-related infection further comprise co-administering an anti-microbial agent, such as an anti-fungal or anti-bacterial agent, to the animal or plant in need thereof. In one embodiment, the anti-microbial agent is an antibiotic.

[00179] When used in combination with other agents useful in treating microbial infection, the agents disclosed herein are suitably administered contemporaneously with those other agents. As used herein, "contemporaneous administration" or "coadministration" of two substances to an individual animal or plant means providing each of the two substances so that they are both biologically active in the individual at the same time. The exact details of the administration will depend on the pharmacokinetics of the two substances in the presence of each other, and can include administering the two substances within a few hours of each other, or even administering one substance within 24 hours of administration of the other, if the pharmacokinetics are suitable. Design of suitable dosing regimens is routine for one skilled in the art. In particular embodiments, two substances will be administered substantially simultaneously, i.e., within minutes of each other, or in a single composition that contains both substances.

[00180] In an embodiment, the at least one soluble protein is expressed by a vector and the vector is administered to the animal or plant in need thereof.

[00181] In one embodiment, the vector is a lytic phage that is able to invade bacteria of the biofilm.

[00182] In one embodiment, the vector is a mycovirus that is able to invade fungal biofilms.

[00183] In an embodiment, the methods or uses disclosed herein further comprise administering other soluble proteins that degrade other components of biofilm, such as alginate and/or cellulose.

[00184] In yet another aspect, there is provided a method of preventing biofilm formation on an indwelling medical device or implant comprising coating the device with at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain.

[00185] In an embodiment, there is provided a method of preventing biofilm formation on an indwelling medical device or implant comprising coating the device with at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, prior to use in an animal in need thereof. Particular combinations of soluble proteins as described above may be coated on the device or implant.

[00186] In an embodiment, at least two of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be coated on the device or implant.

[00187] In another embodiment, at least three of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be coated on the device or implant.

[00188] In yet another embodiment, at least four of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be coated on the device or implant.

[00189] In a further embodiment, at least five of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be coated on the device or implant.

[00190] In yet another embodiment, (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be coated on the device or implant.

[00191] In another embodiment, the device or implant is further coated with other soluble proteins that degrade other components of biofilm, such as alginate and/or cellulose.

[00192] The indwelling medical device or implant may be any device or implant that is inserted into the body of the animal and whose surface would thus be susceptible to biofilm formation. In an embodiment, the indwelling medical device or implant may be a catheter, intravenous tube, prosthetic joint or bioprosthetic.

[00193] In an embodiment, the biofilm may be caused by any Pel- dependent, Psl-dependent, PNAG-dependent and/or GAG-dependent biofilm or any combinations thereof. In one embodiment, the biofilm may be caused by any microorganism or group of microorganisms that have the genetic capacity to synthesize the exopolysaccharides, Pel, Psl, PNAG and/or GAG and combinations thereof. These organisms include, but are not limited to; P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp., Acinetobacter spp. and Fusarium spp. In another embodiment, the biofilm may be dependent on the secretion of any exopolysaccharide that is able to be degraded by the soluble glycosyl hydrolases disclosed herein.

[00194] In yet another embodiment, the methods disclosed herein further comprise coating an antimicrobial agent on the indwelling medical device or implant. In one embodiment, the antimicrobial agent is an antibiotic. In another embodiment, the antimicrobial agent is an antifungal agent.

[00195] In yet another aspect, there is provided a method of treating or preventing biofilm formation on a non-medical surface comprising coating with or applying to the surface at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain. [00196] Also provided herein is a method of treating or preventing biofilm formation on a non-medical surface that is susceptible to biofilm formation comprising coating with or applying to the surface at least one of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthoiogs thereof, prior to use in an animal in need thereof. Particular combinations of soluble proteins as described above may be applied or coated on the non-medical surface.

[00197] In an embodiment, at least two of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthoiogs thereof, may be applied or coated on the non-medical surface.

[00198] In another embodiment, at least three of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthoiogs thereof, may be applied or coated on the non-medical surface.

[00199] In yet another embodiment, at least four of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be applied or coated on the non-medical surface.

[00200] In a further embodiment, at least five of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (vi) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be applied or coated on the non-medical surface.

[00201] In yet another embodiment, (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, (ii) a soluble protein comprising a PelA GH domain, (iii) a soluble protein comprising a BpsB GH domain, (iv) a soluble protein comprising a PgaB GH domain, (v) a soluble protein comprising a Sph3 GH domain and (iv) a soluble protein comprising an Ega3 GH domain, or orthologs thereof, may be applied or coated on the nonmedical surface.

[00202] In another embodiment, the non-medical surface is further coated with other soluble proteins that degrade other components of biofilm, such as alginate and/or cellulose.

[00203] The above methods for coating on non-medical surfaces may be used to prevent or disrupt biofouling. Such abiotic surfaces include, but are not limited to; faucets, drains, pipes, devices related to water filtration and food-contact surfaces related to the manufacturing, preparation and serving of food for the consumption by members of the animal kingdom including mammals, suitably humans.

[00204] In an embodiment, the biofilm is caused by microorganisms or group of microorganisms with the genetic capacity to produce one or more of the following exopolysaccharides; PsI, Pel, PNAG and GAG with organisms which include but are not limited to; P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkolderia spp., Candida spp., Aspergillus spp, Acinetobacter spp. and Fusarium spp. In another embodiment, the biofilm may be caused by a microorganism or group of microorganisms with the genetic capacity to produce any exopolysaccharide that is able to be degraded by the soluble glycosyl hydrolases disclosed herein.

[00205] In yet another embodiment, the methods disclosed herein further comprise coating an antimicrobial agent on the non-medical surface. In one embodiment, the antimicrobial agent is an antibiotic. In another embodiment, the antimicrobial agent is an antifungal agent.

[00206] In one embodiment, the soluble protein comprising a PsIG GH domain disclosed herein comprises amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 (or as shown in SEQ ID NO: 1 1 ) or a glycosyl hydrolase variant thereof.

[00207] In one embodiment, the soluble protein comprising a PelA GH domain disclosed herein comprises amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 (or as shown in SEQ ID NO: 12) or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2 (or as shown in SEQ ID NO:13) or glycosyl hydrolase variants thereof.

[00208] In one embodiment, the soluble protein comprising a PelA GH domain ortholog disclosed herein comprises amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201 .1 (or as shown in SEQ ID NO: 15) or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 (or as shown in SEQ ID NO: 14) or glycosyl hydrolase variants thereof.

[00209] In one embodiment, the soluble protein comprising a BpsB GH domain disclosed herein comprises amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 (or as shown in SEQ ID NO: 19 or 18, respectively) or glycosyl hydrolase variants thereof. [00210] In one embodiment, the soluble protein comprising a PgaB GH domain disclosed herein comprises amino acids 310 to 672 or amino acids 22 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 (or as shown in SEQ ID NO:17 or 16, respectively) or glycosyl hydrolase variants thereof.

[00211] In one embodiment, the soluble protein comprising a Sph3 GH domain disclosed herein comprises amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 (or as shown in SEQ ID NO:20) or a glycosyl hydrolase variant thereof.

[00212] In an embodiment, the soluble protein comprising a Sph3 GH domain ortholog disclosed herein comprises amino acids 54 to 304 of the Sph3 A c sequence from Aspergillus clavatus NRRL 1 deposited into GenBank under accession no. EAW09379.1 (or as shown in SEQ ID NO:22) or a glycosyl hydrolase variant thereof.

[00213] In an embodiment, the soluble protein comprising a Sph3 GH domain ortholog disclosed herein comprises amino acids 43 to 299 of the Sph3 A N sequence from Aspergillus nidulans FGSC A4 deposited into GenBank under accession no. EAA63523.1 (or as shown in SEQ ID NO:23) or a glycosyl hydrolase variant thereof.

[00214] In one embodiment, the soluble protein comprising an Ega3 GH domain disclosed herein comprises amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 (or as shown in SEQ ID NO:21 ) or a glycosyl hydrolase variant thereof.

[00215] As described further herein, the soluble proteins are referred to using the GenBank accession numbers noted above but are the same as those shown in the Table of Sequences, SEQ ID NOs:1 1 -23 and as noted above. Devices

[00216] The present inventors have demonstrated that pre-coating a plastic surface with the soluble proteins disclosed herein reduces the ability of microbes to form a biofilm layer on the surface.

[00217] Accordingly, provided herein is an indwelling medical device or implant coated with at least one soluble microbial protein encoded by an exopolysaccharide biosynthetic operon or functional gene cluster, such as a bacterial or fungal protein, comprising a glycosyl hydrolase domain.

[00218] Also provided herein is an indwelling medical device or implant coated with at least one, at least two of, at least three of, at least four of, at least five of, or all of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, such as a PsIG GH soluble protein or ortholog described herein, (ii) a soluble protein comprising a PelA GH domain, such as a PelA GH soluble protein or ortholog described herein, (iii) a soluble protein comprising a BpsB GH domain, such as a BpsB GH soluble protein or ortholog described herein, (iv) a soluble protein comprising a PgaB GH domain, such as a PgaB GH soluble protein or ortholog described herein, (v) a soluble protein comprising a Sph3 GH domain, such as a Sph3 GH soluble protein or ortholog described herein, and (vi) a soluble protein comprising an Ega3 GH domain, such as a Ega3 GH soluble protein or ortholog described herein, or orthologs thereof. Particular combinations of soluble proteins as described above may be coated on the device or implant.

[00219] A person skilled in the art will appreciate that the soluble proteins disclosed herein may be immobilized to solid supports through non- specific protein absorption or chemical cross-linking. Additionally, soluble proteins may be modified through recombinant or chemical means to allow for attachment to solid supports including but not limited to; the Staudinger ligation reaction, "click" chemistry, expressed protein ligation, chemoenzymatic methods or through surface modification as practiced in the art. [00220] A person skilled in the art will appreciate that the soluble proteins disclosed herein may be encapsulated or delivered to the skin or surface wounds using topical delivery methods such as synthetic polymers such as poloxamer, hydrogels of various compositions of matter which include but are not limited to; protein-based hydrogels, polysaccharide-based hydrogels and DNA-based hydrogels. Other delivery methods include nanoparticles, ointments, petroleum jelly and other aqueous solutions compatible with delivery. Additionally, soluble proteins may be modified through recombinant or chemical means to allow for increased stability, penetrability and compatibility with the delivery compound.

[00221] In another embodiment, the device or implant is further coated with other soluble proteins that degrade other components of biofilm, such as alginate and/or cellulose.

[00222] The indwelling medical device or implant may be any medical device that may be introduced into the body that will have a surface susceptible to biofilm formation. In one embodiment, the indwelling medical device or implant is a catheter or intravenous tube.

[00223] In another embodiment, the indwelling medical device or implant is a prosthetic joint or a bioprosthetic, including but not limited to a heart valve.

[00224] The biofilm that may be formed on the surface may be caused by any microorganism or group of microorganisms that forms Pel-dependent, Psl-dependent, PNAG-dependent biofilm or GAG-dependent, including without limitation, P. aeruginosa, S. aureus, E. coli, S. epidermidis, Y. pestis, B. pertussis, Burkholderia spp., Candida spp., Aspergillus spp., Acinetobacter spp. and Fusarium spp. In another embodiment, the biofilm may be caused by build up of any exopolysacchahde produced by a microorganism or group of microorganisms that is able to be degraded by the soluble glycosyl hydrolases disclosed herein. [00225] In yet another embodiment, the indwelling medical device or implant further comprise an antimicrobial agent coated on the device or implant. In one embodiment, the antimicrobial agent is an antibiotic. In another embodiment, the antimicrobial agent is an antifungal agent.

Compositions

[00226] Also provided herein is an isolated protein consisting of amino acids 31 to 442 of the PsIG sequence deposited into GenBank under accession no. AAG05625.1 , an isolated protein consisting of amino acids 47 to 303 of the PelA sequence deposited into GenBank under accession no. AAG06452.1 or amino acids 35-291 of the PelA sequence deposited into GenBank under accession no. AAY92244.2, an isolated protein consisting of amino acids 61 to 317 of the RagA sequence deposited into GenBank under accession no. CAQ62201 .1 or amino acids 23 to 277 of the PelA sequence deposited into GenBank under accession no. ABB32191.1 , an isolated protein consisting of amino acids 318 to 670 or amino acids 27 to 701 of the BpsB sequence deposited into GenBank under accession no. CAE32265.1 , an isolated protein consisting of amino acids 310 to 672 of the PgaB sequence deposited into GenBank under accession no. AAC74108.1 , an isolated protein consisting of amino acids 52 to 298 of the Sph3 sequence deposited into GenBank under accession no. EAL92786.1 , an isolated protein consisting of amino acids 54 to 304 of the Sph3Ac sequence from Aspergillus clavatus NRRL 1 deposited into GenBank under accession no. EAW09379.1 , an isolated protein consisting of amino acids 43 to 299 of the Sph3AN sequence from Aspergillus nidulans FGSC A4 deposited into GenBank under accession no. EAA63523.1 , and/or an isolated protein consisting of amino acids 46 to 318 of the Ega3 sequence deposited into GenBank under accession no. EAL92787.1 .

[00227] In yet another aspect, the present disclosure provides compositions, such as pharmaceutical compositions, comprising at least one, at least two, at least three of, at least four of, at least five of or all six of: (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, such as a PsIG GH soluble protein or ortholog described herein, (ii) a soluble protein comprising a PelA GH domain, such as a PelA GH soluble protein or ortholog described herein, (iii) a soluble protein comprising a BpsB GH domain, such as a BpsB GH soluble protein or ortholog described herein, (iv) a soluble protein comprising a PgaB GH domain, such as a PgaB GH soluble protein or ortholog described herein, (v) a soluble protein comprising a Sph3 GH domain, such as a Sph3 GH soluble protein or ortholog described herein, and (vi) a soluble protein comprising an Ega3 GH domain, such as an Ega3 GH soluble protein or ortholog described herein, or orthologs thereof; and a pharmaceutically acceptable carrier. Particular combinations of soluble proteins as described above may be included in the compositions.

[00228] The compositions containing the agents can be prepared by known methods for the preparation of pharmaceutically acceptable compositions which can be administered to animals, optionally humans, such that an effective quantity of the active agent is combined in a mixture with a pharmaceutically acceptable vehicle. Suitable vehicles are described, for example, in Remington's Pharmaceutical Sciences (2003 - 20th edition) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19) published in 1999. On this basis, the compositions include, albeit not exclusively, solutions of the agents in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.

[00229] In accordance with the methods and uses of the disclosure, the disclosed agents, salts or solvates thereof may be administered to an animal, optionally a human in a variety of forms depending on the selected route of administration, as will be understood by those skilled in the art. The compositions may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal (topical) administration and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, transepithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.

[00230] In one embodiment, the pharmaceutically acceptable carrier is a gel, such as Poloxamer.

[00231] Compositions suitable for plants containing the agents can be prepared by known methods for the preparation of acceptable compositions which can be administered to plants or plant parts.

[00232] The agents may be administered to an animal or plant alone or in combination with pharmaceutically acceptable carriers, as noted above, and/or with other pharmaceutically active agents such as antibiotics, the proportion of which is determined by the solubility and chemical nature of the agents, chosen route of administration and standard pharmaceutical practice.

[00233] The dosage of the agents and/or compositions can vary depending on many factors such as the pharmacodynamic properties of the agent, the mode of administration, the age, health and weight of the recipient animal or plant, the nature and extent of the symptoms, the frequency of the treatment and the type of concurrent treatment, if any, and the clearance rate of the compound in the animal or plant to be treated. One of skill in the art can determine the appropriate dosage based on the above factors. The agents may be administered initially in a suitable dosage that may be adjusted as required, depending on the clinical response. For ex vivo treatment of animal cells over a short period, for example for 30 minutes to 1 hour or longer, higher doses of agent may be used than for long term in vivo therapy in animals.

[00234] A person skilled in the art will appreciate that the soluble proteins disclosed herein may be prepared in any of several ways, including, without limitation, by using recombinant methods. [00235] Accordingly, nucleic acid molecules encoding the proteins of the disclosure may be incorporated in a known manner into an appropriate expression vector which ensures good expression of the proteins. Possible expression vectors include but are not limited to cosmids, plasmids, or modified viruses (e.g. replication defective retroviruses, adenoviruses and adeno-associated viruses), so long as the vector is compatible with the host cell used. The expression vectors are "suitable for transformation of a host cell", which means that the expression vectors contain a nucleic acid molecule of the application and regulatory sequences selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid molecule. Operatively linked is intended to mean that the nucleic acid is linked to regulatory sequences in a manner that allows expression of the nucleic acid.

[00236] The disclosure therefore contemplates a recombinant expression vector containing a nucleic acid molecule disclosed herein, and the necessary regulatory sequences for the transcription and translation of the inserted protein-sequence.

[00237] Suitable regulatory sequences may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, or insect genes (For example, see the regulatory sequences described in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990)). Selection of appropriate regulatory sequences is dependent on the host cell chosen as discussed below, and may be readily accomplished by one of ordinary skill in the art. Examples of such regulatory sequences include: a transcriptional promoter and enhancer or RNA polymerase binding sequence, a ribosomal binding sequence, including a translation initiation signal. Additionally, depending on the host cell chosen and the vector employed, other sequences, such as an origin of replication, additional DNA restriction sites, enhancers, and sequences conferring inducibility of transcription may be incorporated into the expression vector. [00238] The recombinant expression vectors of the disclosure may also contain a selectable marker gene which facilitates the selection of host cells transformed or transfected with a recombinant molecule of the application. Examples of selectable marker genes are genes encoding a protein such as G418 and hygromycin, which confer resistance to certain drugs, β- galactosidase, chloramphenicol acetyltransferase, firefly luciferase, or an immunoglobulin or portion thereof such as the Fc portion of an immunoglobulin, optionally IgG. Transcription of the selectable marker gene is monitored by changes in the concentration of the selectable marker protein such as β-galactosidase, chloramphenicol acetyltransferase, or firefly luciferase. If the selectable marker gene encodes a protein conferring antibiotic resistance such as neomycin resistance transformant cells can be selected with G418. Cells that have incorporated the selectable marker gene will survive, while the other cells die. This makes it possible to visualize and assay for expression of recombinant expression vectors of the application and in particular to determine the effect of a mutation on expression and phenotype. It will be appreciated that selectable markers can be introduced on a separate vector from the nucleic acid of interest.

[00239] The recombinant expression vectors may also contain genes which encode a fusion moiety which provides increased expression of the recombinant protein; increased solubility of the recombinant protein; and aid in the purification of the target recombinant protein by acting as a ligand in affinity purification. For example, a proteolytic cleavage site may be added to the target recombinant protein to allow separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Typical fusion expression vectors include pGEX (Amrad Corp., Melbourne, Australia), pMal (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the recombinant protein. [00240] Recombinant expression vectors can be introduced into host cells to produce a transformed host cell. The terms "transformed with", "transfected with", "transformation" and "transfection" are intended to encompass introduction of nucleic acid (e.g. a vector) into a cell by one of many possible techniques known in the art. The term "transformed host cell" as used herein is intended to also include cells capable of glycosylation that have been transformed with a recombinant expression vector of the disclosure. Prokaryotic cells can be transformed with nucleic acid by, for example, electroporation or calcium chloride-mediated transformation. For example, nucleic acid can be introduced into mammalian cells via conventional techniques such as calcium phosphate or calcium chloride co- precipitation, DEAE-dextran mediated transfection, lipofectin, electroporation or microinjection. Suitable methods for transforming and transfecting host cells can be found in Sambrook et al. (Molecular Cloning: A Laboratory Manual, 3rd Edition, Cold Spring Harbor Laboratory Press, 2001 ), and other laboratory textbooks.

[00241] Suitable host cells include a wide variety of eukaryotic host cells and prokaryotic cells. For example, the proteins of the disclosure may be expressed in yeast cells or mammalian cells. Other suitable host cells can be found in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1991 ). In addition, the proteins of the disclosure may be expressed in prokaryotic cells, such as Escherichia coli (Zhang et al., Science 303(5656): 371-3 (2004)). In addition, a Pseudomonas- based expression system such as P. fluorescens can be used (US Patent Application Publication No. US 2005/0186666, Schneider, Jane C et al.).

[00242] Accordingly, also provided herein is a host cell comprising a nucleic acid molecule of the disclosure.

[00243] The nucleic acid molecules encoding the proteins of the disclosure may also be incorporated in a known manner into the genome of a phage that is capable of infecting bacteria found in biofilm. Lytic phages include those which target bacteria which have the genetic capacity to produce one or more of the exopolysaccharides; Psl, Pel, PNAG and GAG. A person skilled in the art will readily identify and use a lytic phage specific to the target bacteria.

[00244] The nucleic acid molecules encoding the proteins of the disclosure may also be incorporated in a known manner into the genome of a mycovirus that is capable of infecting the fungus found in GAG biofilm. A person skilled in the art will readily identify and use a mycovirus specific to the target bacteria.

[00245] Accordingly, provided herein is a lytic phage or mycovirus encoding (i) a soluble protein comprising a PsIG glycosyl hydrolase (GH) domain, such as a PsIG GH soluble protein or ortholog described herein, (ii) a soluble protein comprising a PelA GH domain, such as a PelA GH soluble protein or ortholog described herein, (iii) a soluble protein comprising a BpsB GH domain, such as a BpsB GH soluble protein or ortholog described herein, (iv) a soluble protein comprising a PgaB GH domain, such as a PgaB GH soluble protein or ortholog described herein, (v) a soluble protein comprising a Sph3 GH domain, such as a Sph3 GH soluble protein or ortholog described herein, or (vi) a soluble protein comprising an Ega3 GH domain, such as an Ega3 GH soluble protein or ortholog described herein, or orthologs thereof, or combinations thereof.

[00246] The above disclosure generally describes the present application. A more complete understanding can be obtained by reference to the following specific examples. These examples are described solely for the purpose of illustration and are not intended to limit the scope of the disclosure. Changes in form and substitution of equivalents are contemplated as circumstances might suggest or render expedient. Although specific terms have been employed herein, such terms are intended in a descriptive sense and not for purposes of limitation. [00247] The following non-limiting examples are illustrative of the present disclosure:

EXAMPLES

Example 1 - PslGai-442 from P. aeruginosa PAQ1 is a soluble protein

Method:

[00248] The amino acid sequence of ps/G from P. aeruginosa PA01 was deposited into GenBank under accession no. AAG05625.1 and released in August 2000 (Stover et al 2000). The TMHMM server v2.0 (Krogh et al 2001 ) indicates that PsIG from Pseudomonas aeruginosa PA01 possesses a transmembrane helix from residues 5-24 that tethers the periplasmic catalytic domain to the cytoplasmic membrane. To obtain a soluble protein construct from the ps/G gene from P. aeruginosa genomic DNA was amplified by PCR using the primers G G G C AT ATG GAGATCCAGG T ACTG AAG (SEQ ID NO:1 ) and GGGAAGCTTTCACTCCCAGACCAGCA (SEQ ID NO:2).

[00249] E. coli BL21 (DE3) cells were transformed with the protein expression vector and grown in 1 L Luria-Bertani (LB) broth containing 50 pg/mL kanamycin at 37 °C. When the OD 600 of the cell culture reached 0.4- 0.5, the temperature was reduced to 18 °C for 20-30 min and protein expression was induced by the addition of isopropyl β-D- - thiogalactopyranoside (IPTG) to a final concentration of 1 mM. The cultures were incubated post-induction overnight at 18 °C with shaking then harvested by centrifugation at 5,000 * g for 20 min at 4 °C.

[00250] Cell pellets were resuspended in 40 mL of buffer A (20 mM imidazole, 50 mM Tris pH 7.5, 300 mM NaCI, 2% (v/v) glycerol and one SIGMA AS7™ Protease Inhibitor Tablet) and the cells were lysed by at least three passes through an Emulsiflex C3 at 100 MPa (Avestin Inc). The resulting cell debris was separated from soluble protein by centrifugation at

35,000 x g for 30 min. The supernatant was applied to a 5 mL Ni-NTA

Superflow gravity column (Qiagen) pre-equilibrated with buffer A. The column was washed with 3 column volumes (CV) of buffer A and the expressed protein was eluted with buffer A supplemented with 250 mM imidazole. The eluted fractions were pooled and dialyzed against 4 L of buffer B (50 mM Tris pH 7.5, 150 mM NaCI 2% (v/v) glycerol) overnight at 4 °C. The His-tag was removed by incubating the protein at 25 °C for 3 h with one unit of thrombin (Novagen) per 4 mg protein. Untagged protein was separated from tagged protein by purification on a 5 rtiL Ni-NTA Superflow gravity column pre- equilibrated with buffer A. The untagged protein was collected and buffer exchanged into buffer B by size-exclusion chromatography using a HiLoad 16/60 Superdex 200 gel-filtration column (GE Healthcare).

Result:

[00251] A soluble PsIG construct encompassing residues 31-442 was expressed and purified. The expressed protein produces ~7 mg per liter of bacterial culture with a molecular weight of 46.9 kDa (Figure 8). The purity of protein was judged to be >95% by SDS-PAGE and the protein could be concentrated to 8-10 mg/mL and stored at 4 °C for more than one month without precipitation or degradation.

Example 2 - PsIG 31-44? is a two-domain protein with distinct active site groove Method:

[00252] Initial crystallization trials were performed with 8 mg/mL PslG 31 . 442 using a Gryphon robot (Art Robbins) with 96-well Art Robbins Instruments Intelli-Plates ® (Hampton Research) and the MCSG1-4 sparse-matrix screen from Microlytic. Protein (1 μΙ_) was mixed with precipitant in a 1 :1 ratio and equilibrated against 60 μΙ_ of precipitant using the sitting-drop vapour diffusion method at 20 °C. Optimal crystals were grown in 48-well VDX plates (Hampton Research) using 1 μΙ_ protein with equal volume of precipitant (1 mM CdCI 2 , 0.1 M HEPES pH 7.0 and 5% (w/v) PEG3350) and equilibrated against 130 μΙ_ precipitant at 20 °C. PSIG31.442 crystals were cryoprotected for 10 s in precipitant solution supplemented with 25% (v/v) ethylene glycol prior to vitrification in liquid nitrogen. Diffraction data were collected at 100 K with a wavelength of 1.075 A on beamline X29, National Synchrotron Light Source (NSLS). A 0.16 mm collimator was used to collect a high redundancy, 360° dataset with a total of 720 images with 0.5° oscillation on an ADSC Quantum- 315 detector with a 250 mm crystal-to-detector distance and an exposure time of 0.3 s per image. The beam was attenuated and 90 images with 2° oscillation were collected over 180°. The combined data were integrated, reduced and scaled using HKL2000 (Otwinowski & Minor 1997). For PslG 31- 442 Autosolve (Terwilliger & Berendzen 1999) was used to generate experimental phases using the cadmium single wavelength dispersion (SAD) technique. A total of four cadmium-binding sites were found and subsequently used to generate a density-modified map. The resulting electron density of each map was of high quality and enabled PHENIX AutoBuild to build >95% of the protein. The remaining residues were built manually in COOT (Adams et al 2010, Emsley & Cowtan 2004). Refinement was carried out using PHENIX. REFINE (Afonine et al 2010). TLS groups were added to the refinement in PHENIX through the use of the TLSMD server (Painter & Merritt 2006). Structure figures were generated using the PyMOL molecular graphics system (DeLano Scientific) (Dolinsky et al 2007).

Result:

[00253] PslG 3 i-442 crystallized in the presence of divalent metal ions including: Ni, Cu, Co, Zn and Cd and diffraction data were collected to 2.0 A on a crystal grown in the presence of 1 mM CdCI 2 . The protein bound four cadmium ions, allowing the structure to be solved using the cadmium SAD technique. Refinement produced a final model with good geometry and R factors. PsIG crystallized in space-group P4i2i2 with one protomer in the asymmetric unit. This is consistent with a calibrated analytical size exclusion column that indicates that PsIG is a monomer in solution. The enzyme contains two domains: a TIM-barrel motif and a β-sandwich domain that is composed of one β-strand from the N-terminus and several β-strands from the C-terminal end of the protein (Figure 9A). The TIM-barrel fold is the most common enzyme fold in the Protein Data Bank (PDB) of known protein structures. It is estimated that 10% of all known enzymes contain this fold (Wierenga 2001 ). The putative active site is located in the TIM-barrel domain and runs 40 A equatorially across the domain (Figure 9B). Molecular docking simulations using PatchDock (Schneidman-Duhovny et al 2005) of the Psl polysaccharide suggest that this groove is theoretically able to accommodate between 12-15 sugar units. Consistent with other members of the GH39 family, attempts to isolate the TIM-barrel domain alone were unsuccessful (St John et al 2010). This supports the hypothesis that the β-sandwich is critical for proper protein folding and/or stability of the protein.

[00254] Comparison of the amino acid sequence and crystal structure of PsIG with other GH39 members suggests that two acidic residues, Glu-165 and Glu-276 are highly conserved. Structural alignment using DaliLite (Holm et al 2008) indicates that Glu- 65 and Glu-276 are located in the middle of the putative active site groove, and at equivalent positions to other GH39 family members. Previous characterization of XynB, a GH39 member, suggests that Glu-165 is the acid/base while Glu-276 would function as the nucleophile in the reaction (Nieman et al 2003, Vocadio et al 1998). The amino acids that line the groove are not well conserved in distantly related homologs. Many homologs exhibit activity towards the polysaccharide xylan, which is distinct from Psl. The lack of conservation in the active site suggests that homologs have been evolutionary selected to bind a specific polymer and would not be able to bind and catalyze the hydrolysis of Psl.

Method:

[00255] Psl- arabinose inducible P. aeruginosa PA01 pBADps/ was grown at 37 °C overnight with shaking at 200 rpm. The culture was diluted 1 :100 in LB and arabinose was added to a final concentration of 0.5% (w/v) to induce Psl biosynthesis. 95 μΙ_ of diluted culture was added to sterile 96-well polystyrene microtiter plates (Thermo Scientific Cat No. 243656) and varying concentrations of PelA 4 7. 3 o3 or PslG3i -442 (1 nM - 10 μΜ) were added in 5 μΙ_ aliquots to give a final volume of 100 μ[_. The next day fresh cultures were prepared from the overnight culture using a 1 :100 dilution. 95 pL of diluted culture was added to a sterile 96-well polystyrene round bottom microtiter plate and varying concentrations of PSIG31-442 (2 nM - 5 μΜ) were added in 5 μΙ_ aliquots to give a final volume of 100 μΙ_. The cultures were incubated statically for 24 h at 26 °C to allow for biofilm formation. To eliminate edge- effects, -200 μΙ_ of sterile water was placed in all outside wells and the plate was sealed with parafilm. After incubation, non-adherent cells and media were removed by thoroughly washing the plate with deionized water. The wells were stained with 50 μΙ_ of 0.1 % (w/v) crystal violet for 10 min followed by rinsing with water. The remaining dye was solubilized by addition of 200 pl_ of 95% (v/v) ethanol and left for 10 min after which time the absorbance was measured at 595 nm using a SpectraMax M2 from Molecular Devices (Sunnyvale, CA). The amount of biofilm is proportional to the absorbance from staining with crystal violet (Merritt et al 2005). All reactions were completed in triplicates and purified PelA 4 7-303 and the buffer B were added as negative controls. The addition of 2.5 mg/mL of kanamycin to culture prior to biofilm formation was used as positive control as no cell growth occurs.

Result:

[00256] In the P. aeruginosa PA01 pBADps/ which produces significantly more Psl than the clinical PA01 strain, addition of PSIG31-442 prevented biofilm formation at≥50 nM and had a EC50 of 4.1 ± 1.1 nM (Figure 10). To examine whether the effect was the direct result of PsIG activity a double catalytic variant PSIG31-442 E165Q/E276Q was constructed and tested. Addition of this resulted in a >100-fold increase in the EC50 in the pBADps/ inducible strain (Figure 10). Enzyme concentrations in excess of 10 μΜ, prevented Psl-dependent biofilm formation. The addition of PelA 4 7. 3 o3, another putative glycosyl hydrolase, did not inhibit Psl biofilm formation suggesting that biofilm inhibition is protein specific. In addition, 10 μΜ of PslG3i_442 did not prevent PA01 growth suggesting that the loss of the biofilm is not due to perturbations in bacterial growth. Without wishing to be bound by theory, the enzyme variants may remain capable of binding the polysaccharide, thereby reducing the ability of the bacteria to adhere to the abiotic plate.

Method:

[00257] Bacterial cultures expressing Psl were inoculated as stated above and were grown at 37 °C overnight with shaking at 200 rpm. All cultures were diluted in 1 :100 in LB and arabinose was added to a final concentration of 0.5% (w/v) to induce Psl biosynthesis. Media containing the Psl-producing cultures were supplemented with 200 pg/nriL ampicillin and 100 pg/mL kanamycin. The cultures were incubated statically for 24 h at 26 °C to allow for biofilm formation. After incubation non-adherent cells and media were removed by washing the plate with deionized water three times. The wells were filled with 95 μΙ_ of 100 mM sodium HEPES buffer pH 7.0 followed by 5 μΙ_ of varying concentrations of each hydrolytic enzyme (2 nM - 5 μΜ). Reactions were allowed to proceed for up to 60 min at 25 °C on a rotating nutator at which time, the reaction was quenched by washing the plates with deionized water. The wells were stained with 200 μΙ_ of 0.1 % (w/v) crystal violet for 10 min, and washed with water three times. Crystal violet dye from Psl cultures was solubilized in 100 pL of 95% ethanol for 10 min with rotation, after which time the absorbance was measured at 595 nm using a SpectraMax M2 from Molecular Devices (Sunnyvale, CA). The amount of biofilm is proportional to the absorbance from staining with crystal violet (Merritt et al 2005). All reactions were completed in at least triplicate and 100 mM sodium HEPES buffer pH 7.0 was used as an untreated control. The addition of 2.5 mg/mL kanamycin to culture prior to biofilm formation was used as a positive control as no cell growth occurs.

Result:

[00258] The addition of 86 nM of PslG 3 -i -442 was able to disperse a biofilm in 35 minutes (Figure 11 A). This suggests that very little protein is required to disperse these biofilms under the conditions tested. When PslG 3 i_ 442 E165Q/E276Q double catalytic variant was added in 100-fold excess compared to the wild-type enzyme (5 μΜ), no significant difference was observed compared to that of the untreated biofilm (Figure 11 B). This suggests that these residues are critical in catalysis. It also indicates that biofilm inhibition is not a true indicator of catalytic activity. The addition of the hydrolase domain of PelA 4 7-303 did not result in the loss of the Psl-dependent biofilm suggesting that dispersion is protein specific.

and G/nefPelA?3-?77 are soluble proteins with high yield

Method:

[00259] The protein sequence of pelA from P. aeruginosa PA01 was deposited into GenBank under accession no. AAG06452.1 and released in August 2000 (Stover et al 2000). The PRED-TAT server (Bagos et al 2010) indicates that PelA from Pseudomonas aeruginosa PA01 possesses a TAT signal sequence from residues 1 -45, allowing the protein to be translocated from the cytosol to the periplasm in a folded state. To obtain a soluble protein construct the pe//A 47-30 3, the gene (residues 47-303) was amplified from the genomic DNA of P. aeruginosa POA1 by PCR using the primers CTGCATATGGGCGGGCCGTCCAGCGTGGCG (SEQ ID NO:3) and TTTCTCGAGTCACGGTTGCACCTCGACGTC (SEQ ID NO:4). Constructs encoding residues 61 -317 of RagA from Ralstonia solanacearum were isolated using primers GCGCATATGGCGGACGCACCGAACATTGCC (SEQ ID NO:5) and GGGAAGCTTTCACG GCAGCACCTCGA TGCGCC (SEQ ID NO:6), and residues 23-277 of PelA from Geobacter metallireducens using the primers G G G C AT ATG C AC CTC CTTTAAG CGTGGCCTTG (SEQ ID NO:7) and GCGAAGCTTTCAC GGCATAACCTCCACGCTCCC (SEQ ID NO:8) to remove the signal sequence for protein expression in the cytosol. Introduced Nde\, Hind\ and Xho\ restriction sites are underlined and each gene was ligated into the pET28a (Novagen) expression vector encoding an N-terminal His-tag. Protein expression of PelA 4 7-303 and its orthologs was the same as described in for PslG3i_442 in Example 1.

[00260] Cell pellets were resuspended in 40 mL of buffer C (20 mM imidazole, 50 mM Tris pH 8.0, 300 mM NaCI, 10% (v/v) glycerol and one protease tablet (Simga) and the cells were lysed by at least three passes through an Emulsiflex C3 at 100 MPa (Avestin Inc). The resulting cell debris was separated from soluble protein by centrifugation at 35,000 χ g for 30 min. The supernatant was applied to a 5 mL Ni-NTA Superflow gravity column (Qiagen) pre-equilibrated with buffer C. The column was washed with 3 column volumes of buffer A and the expressed protein was eluted with buffer A supplemented with 250 mM imidazole. The eluted fractions were pooled and dialyzed against 4 L of buffer D (50 mM Tris pH 8.0, 150 mM NaCI, 10% (v/v) glycerol) overnight at 4 °C. Expression of the selenium-methionine derivative of PelA 47 . 3 o3 in minimal medium was carried out using B834 Met " E. coli cells (Novagen) as described previously (Lee et al 2001 ). Purification of (SeMet)-labeled PelA 47- 303 and variants was completed as described for the wild-type enzyme.

Result:

[00261] A PelA47-303 construct encompassing residues 47-303 was expressed and purified. The expressed protein produces -50 mg per liter of bacterial culture with a molecular weight of 28.2 kDa (Figure 12). The purity of protein was judged to be >95% by SDS-PAGE and the protein could be concentrated to 8-10 mg/mL and stored at 4 °C for more than one month without precipitation or degradation. In comparison, a soluble, full-length construct PelA 47- 9 4 8, encompassing amino acid residues 47 to 948 yields only ~1 mg/L of bacterial culture (Colvin et al 2013).

Example 6 - PelA 4 7-3n has a a/a7 TIM-barrel structure

Method: [00262] Purified PelA 47-30 3 was concentrated to 20 mg/mL and screened using commercially available crystallization screens (Microlytic). Native crystals were set up and grown using the popular hanging-drop vapor diffusion method. Crystals were optimized using a 1 :2 protein:well-solution ratio and crystallized in the solution screen composed of 25% (w/v) PEG MME 5K and Bis-Tris pH 7.5 at 20 °C. Selenomethionine (SeMet)-labeled PelA 47- 303 was set up in the same fashion and crystallized in a similar screen condition except this condition contained a slightly higher PEG concentration (26% (v/v)). Crystallization solution supplemented with 15% (v/v) ethylene glycol was used to cryoprotect crystals prior to vitrification in liquid nitrogen. Crystals were sent to NSLS for data collection on beamline X29. The selenium sites in PelA 47- 303 were used for selenium SAD phasing. All model building and refinement was completed as described in Example 2.

Result:

[00263] PelA 47- 303 from P. aeruginosa was expressed, purified and crystallized. The final protein sample was purified to homogeneity as revealed by SDS-PAGE analysis and produced high yields of -40 mg of protein from 1 L Luria-Bretani broth. Diffraction data were collected from the native and SeMet protein crystals to 1.5 A and 1.9 A, respectively. The structure was solved using the SAD technique and refined to a final resolution of 1.5 A with good refinement statistics. PelA 47- 303 crystallized in the orthorhombic space group 2i2-|2 with one protomer in the asymmetric unit. The structure indicates that PelA 47 -303 has a TIM-barrel-like fold with a total of 12 β-sheets and nine a-helices (Figure 13). The putative active site is located in the TIM- barrel domain and is composed of a deep electronegative groove that runs equatorially across the domain (Figure 14). The electronegativity of this groove suggests that Pel is not negatively charged as previously suggested (Colvin et al 201 1 ). Molecular docking simulations using PatchDock (Schneidman-Duhovny et al 2005) of a polymer of glucosamine residues suggest that this groove is theoretically able to accommodate at least 8 sugar units. [00264] The CAZymes Analysis Toolkit (CAT) (Park et al 2010) suggests that the hydrolase domain (residues 47-303) belongs to GH family 1 14. Of significant importance is the high degree of amino acid conservation in the putative catalytic groove. While the catalytic residues are currently unknown, there are several highly conserved acidic residues that could function as the nucleophile and catalytic acid/base in the reaction (Figure 14).

Example 7 - PelA 47 -303 and ortholoqs can prevent Pel biofilm formation in static culture

Method:

[00265] The methodology to examine the inhibition of Pel biofilm formation is identical to that previously described in Example 3 for Psl biofilms with the exception that the strain used is P. aeruginosa PA01 AwspFApslpB AD pel. The chemical composition of Pel is currently unknown.

Result:

[00266] An ex vivo assay was employed to examine whether exogenously added PelA prior to biofilm formation could prevent Pel polysaccharide biofilm formation. A Pel overproducing strain, which is unable to produce the Psl polysaccharide (PA01AwspFAps/P BAD pe/) was utilized for all experiments. This strain allows for inducible over expression of the Pel polysaccharide upon addition of arabinose to the culture media. The addition of 500 nM of PelA 47 . 3 o3 from P. aeruginosa POA1 prevented the formation of Pel-dependent biofilms. In comparison, two putative catalytic variants, D160A and E218A were unable to prevent biofilm formation at this concentration (Figure 15). The addition of PelA 35 - 2 9i from P. protogens Pf-5 prevented biofilm formation at >70 nM and had an EC 50 of 69.3 ± 1.2 nM (Figure 16).

The addition of PslG 31-442 , another putative glycosyl hydrolase, did not inhibit

Pel biofilm formation suggesting that biofilm inhibition is protein specific.

Without wishing to be bound by theory, the enzyme variants may remain capable of binding the polysaccharide, thereby reducing the ability of the bacteria to adhere to the abiotic plate. Example 8 - PelA 47 .3 0 3 and its orthologs can disperse pre-formed Pel biofilms Method:

[00267] The methodology to generate pre-formed Pel biofilms is identical to that of Psl biofilms, described in Example 4 with the exception that the strain used is P. aeruginosa PA01 AwspF pslpBAOpel.

Result:

[00268] PelA 47 _303 and the two putative catalytic variants D160A and E218A were incubated at two concentrations; 1.14 mg/mL (38 μΜ) and 0.57 mg/mL (19 μΜ). After incubation for 2 h, PelA 4 .303 had successfully dispersed the Pel biofilm while the E218A variant retained a similar level of biofilm as a buffer control. The D160A variant retained -50% of the biofilm level as the buffer control (Figure 17A). This suggests that amino acids D160 and E218 are important for catalysis. A similar experiment was completed with PelA 35- 29i from P. protogens Pf-5 which revealed that 1 μΜ of the enzyme was able to disperse the biofilm in as little as 30 min and that a 10-fold dilution was able to disperse nearly all the biofilm in 1 h (Figure 17B). This suggests that both wild-type hydrolase domains are catalytically active and can efficiently be added to disperse biofilms.

Example 9 - Hvdrolvtic enzymes can be pre-coated to plastic to prevent biofilm formation

Method:

[00269] Sterile 96-well polystyrene plates were pre-coated by soaking 100 μΐ_ of 1 *PBS (pH 7.4) containing 40 pg/mL of PelA 47-30 3 or PSIG 3 -M 42 in each well for 12 h at 4 °C. The enzyme solution was removed and the wells were washed by the addition of 100 pL of 1 *PBS (pH 7.4) for 5 min prior to its removal. The plates were dried at 37 °C for 1 h prior to use. Bovine Serum Albumin (BSA) at a final concentration of 40 pg/mL was used as a control. The next day, plates were inoculated with a 1 : 100 dilution of P. aeruginosa Psl or Pel arabinose inducible cultures. Immobilization of PslG 3 i -44 2 or BSA by either adsorption or cross-linking was performed on flat glass slides that are uniformly thin (0.17-0.25 mm) and corrosion-resistant. The glass surface was first activated by immersion in piranha solution (H2SO4: H2O2, 3:1 ) for 30 min to remove any organic matter residuals and hydroxylate the surface, then rinsed several times with double distilled water to remove any chemical residuals. For immobilization by chemical cross-linking, the surface was functionalized with amine (NH 2 ) groups by silanization, in which the glass cover was immersed in 3-aminopropyltrimethoxysilane (APTMS) solution (0.05 g/mL in 80% (v/v) ethanol) for 2 h at room temperature. The surface was washed with 80% (v/v) ethanol three times to remove unreacted APTMS. PslG3i -44 2 was linked onto amino functionalized glass surface through a molecular coupling agent, glutaraldehyde, by immersion of the surface in 4% glutaraldehyde in PBS buffer solution (pH 7.2) for 2 h under gentle stirring and room temperature conditions. The mixture was then rinsed 3 times with 80% ethanol to remove unreacted glutaraldehyde. Activated or modified glass (NH2- glutaraldehyde) was immersed in an enzyme solution (80 pg/mL) and incubated overnight at 4°C. Finally, the surface was washed with PBS buffer several times to remove the unbound enzyme. The next day, plates were inoculated with a 1 :100 dilution of P. aeruginosa Psl or Pel arabinose inducible cultures. The cultures were grown statically for a duration of at least 20 h after which time biofilm formation was measured using the crystal violet method described previously in Example 4 or SYTOX green to visualize bacteria via confocal microscopy.

Results:

[00270] Pre-coating wells with PelA 4 7-303 completely abrogated biofilm under the conditions tested (Figure 18A). Additionally, BSA treated wells did not reduce the biofilm suggesting that coating with PelA 7-3 o3 specifically target and inhibit P. aeruginosa biofilm formation. PslG 3 i.4 4 2 was also able to adsorb to plastic as enzyme treatment prevented cell and biofilm attachment as visualized using confocal microscopy (Figure 18B). PSIG31-442 could also be chemically cross-linked to glass using glutaraldehyde and this treatment also prevented cell attachment and biofilm formation (Figure 18C). Treatment with BSA demonstrated that a non-specific protein coating is insufficient to prevent biofilm formation. Glass surfaces that were covalently linked with PSIG31-442 were able to prevent biofilm formation for at least 8 days (Figure 18D). Without wishing to be bound by theory, it is likely that other enzyme orthologs are capable of adsorbing to plastics and may be used to prevent bacterial attachment and biofilm formation.

Example 10 - BpsB and its isolated C-terminal domain are stable and soluble proteins

Method:

[00271] The protein sequence of bpsB from B. bronchiseptica RB50 was deposited into GenBank under accession no. CAE32265.1 and released in July 2008. The SignalP server v4.0 (Petersen et al 201 1 ) indicates that BpsB from B. bronchiseptica RB50 possesses a periplasmic signal sequence from residues 1-26. For soluble protein generation, the bpsB gene containing residues 27-701 was amplified from genomic DNA by inverse PCR with flanking Λ/del and Hind\\\ endonuclease restriction sites, then digested and ligated into the pET28a (Novagen) expression vector encoding an N-terminal hexahistidine-tag. The resulting plasmid, pET28-BpsB 2 7-7oi was then used as a template for cloning the BpsB C-terminal domain, residues 318-670, using the same procedure as above to yield plasmid pET28-BpsB 3 is-67o- Protein expression of BpsB was the same as described in for PSIG31-442.

[00272] Active site alanine variants were generated using the QuikChange site-directed mutagenesis kit according to the manufacturer's instructions, and expressed and purified as for BpsB mentioned previously.

[00273] Cell pellets were resuspended in 40 mL of buffer E (50 mM

HEPES pH 8.0, 300 mM NaCI, 10 mM imidazole, 5% (v/v) glycerol and one

SIGMAFy4S7 ~ ™ Protease Inhibitor Tablet). The cells were lysed by three passes through an Emulsiflex C3 at 100 MPa (Avestin Inc). Insoluble and cell debris was clarified from soluble protein by centrifugation at 30,000 χ g for 30 min. The supernatant was applied to 4 mL of Ni-NTA Superflow resin (Qiagen) packed into a 30 mL gravity column pre-equilibrated with buffer F (20 mM HEPES pH 8.0, 300 mM NaCI, 10 mM imidazole). The column was washed with 10 column volumes (CV) of buffer F and the expressed protein was eluted with buffer B supplemented with 200 mM imidazole. The eluted fractions were pooled and dialyzed against 1 L of buffer G (20 mM HEPES pH 8.0, 300 mM NaCI) overnight at 4 °C. The His-tag was removed by incubating the protein at 25 °C for 2 h with one unit of thrombin (Novagen) per 4 mg protein. Untagged protein was separated from tagged protein by a second round of Ni-NTA purification with the flow-through and wash fractions being collected, concentrated, and applied to a HiLoad 16/60 Superdex 200 gel- filtration column (GE Healthcare) equilibrated in buffer H (20 mM HEPES pH 7.5, 150 mM NaCI).

Result:

[00274] A full-length BpsB encompassing residues 27-701 (BpsB 2 7-7oi ) and a C-terminal hydrolase domain construct encompassing residues 318- 670 (BpsB3i8-67o) were expressed and purified. BpsB 2 7-70i protein produces -30 mg per liter of bacterial culture with a molecular weight of 78.5 kDa (Figure 19), while BpsB 3 i 8-6 7o produces -50 mg per liter of bacterial culture with a molecular weight of 42.3 kDa. The purity of each protein was judged to be >95% by SDS-PAGE and the proteins could be concentrated to 8-10 mg/mL and stored at 4 °C for more than one month without precipitation or degradation.

Example 11 - BpsB3i»-fi7n adopts a (β/α)κ barrel with a distinct electronegative groove

Method:

[00275] Purified BpsB 3 i 8- 67o was concentrated to 10 mg/mL and screened for crystallization conditions at 20 °C using hanging-drop vapor diffusion in 48-well VDX plates (Hampton Research) and the MCSG 1-4 sparse matrix suites (Microlytic). An initial crystallization hit was obtained in condition #77 from the MCSG-1 suite. Optimized crystals were grown using 8.6 mg/mL BpsB 3 -i 8- 670 with a 3 μΙ_ drop with equal amounts of protein and precipitant equilibrated against 250 μΙ_ precipitant solution (0.1 M bis(2- hydroxyethyl)amino-tris(hydroxymethyl) methane (BIS-TRIS) pH 6.9, and 1.7 M ammonium sulfate). BpsB 3 i8-67o crystals were cryoprotected for 10 s in precipitant solution supplemented with 25% (v/v) ethylene glycol prior to vitrification in liquid nitrogen. Diffraction data were collected at 100 K with a wavelength of 1 .075 A on beamline X29, at NSLS. A 0.16 mm collimator was used to collect a 360° dataset with 1.0° oscillations on an ADSC Quantum- 315r detector with a 220 mm crystal-to-detector distance and an exposure time of 0.4 s per image. The beam was attenuated and 90 images with a 2° oscillation were collected over 180°. AutoMR was used to obtain phases information through molecular replacement with PgaB residues 310-646 (PDB 4F9D).

Result:

[00276] BpsB 3 i 8 .67o crystallized in the space-group P2i and diffraction data were collected to 1 .76 A resolution. The structure was solved using molecular replacement and refinement produced a final model containing two molecules in the asymmetric unit with good geometry and R factors. Analytical size exclusion chromatography indicates that BpsB 3 i 8-67o is a monomer in solution. BpsB 3 i8-67o adopts a (β/α)β barrel fold common to glycosyl hydrolases (Figure 20), and is the most common enzyme fold in the Protein Data Bank (PDB) of known protein structures. The putative active site is located in a groove at the top of the (β/α)β barrel and is 41 A long and 1 1 A wide (Figure 20). Molecular docking simulations using PatchDock (Schneidman-Duhovny et al 2005) of BpsB 3 i 8-67o and PNAG suggest that this groove is theoretically able to accommodate between 10-12 sugar units.

[00277] Comparison of the amino acid sequence and crystal structure of BpsB 3 8- 67o with other GH13, GH18, and GH20 members suggests that it does not contain the canonical catalytic sequence motifs. Structural alignment using DaliLite (Holm et al 2008) indicate that His473 and Asp474 are located in the middle of the putative active site groove, and at equivalent positions to the GH13, GH18, and GH20 family members catalytic Asp and Glu residues. This suggests BpsB and PgaB may exhibit a novel mechanism for dPNAG hydrolysis, or belongs to a unique GH family. Variants that maintain ability to cut the exopolysaccharide will likely require such residues to be maintained. Alternatively, such residues could be further optimized and a person skilled in the art would readily be able to determine if activity is increased or decreased upon mutation.

Example 12 - The C-terminal domain of PgaB is a stable soluble protein Method:

[00278] The plasmid pET28-PgaB 22 - 6 72 (Little et al 2012a, Little et al 2012b) was used as a template and pgaB specific primers were designed to subclone residues 310-672 into a pET28a expression vector (Novagen) using inverse PCR with an Nde\ and Xho\ site flanking the gene fragment. The resulting plasmid pET28-PgaB3i 0- 672 encodes the C-terminal domain of PgaB with a thrombin cleavable hexahistidine tag. PgaB 3 0-6 72 was expressed and purified as described previously (Little et al 2012a, Little et al 2012b), with the following modification: glycerol was only included in the lysis buffer during purification.

Result:

[00279] A method for the production of PgaB encompassing residues 22-672 (PgaB 22 -672) has been described previously (Little et al 2012a, Little et al 2012b). Herein, a C-terminal hydrolase domain construct encompassing residues 310-672 of PgaB (PgaB 31 0 -672) was expressed and purified. A yield of PgaB 3 i 0- 672 of -10 mg/L of bacterial culture was obtained. The purity of the protein was judged to be >95% by SDS-PAGE (Figure 21 ) and the protein could be concentrated to 8-10 mg/mL, however to minimize precipitation and degradation it was stored at 0.5-1.0 mg/mL at 4 °C for two weeks. Example 13 - BpsB ?7- 7m , Β εΒ,^,^ο, PqaB??,^?, and PqaB^g?? can prevent biofilm formation in static culture

Method:

[00280] The methodology to inhibit PNAG biofilms is similar to that of Psl biofilms, described in Example 3. In brief, PNAG-overproducing E. coli or S. carnosus was grown at 37 °C overnight in LB broth supplement with 200 pg/mL ampicillin and 100 pg/mL kanamycin, and 10 pg/mL tetracycline, respectively, with shaking at 200 rpm. Fresh cultures with antibiotic were prepared from the overnight culture using a 1 : 100 dilution, using LB broth for E. coli, and tryptic soy broth for S. carnosus. 95 pL of diluted culture was added to a sterile 96-well polystyrene round bottom microtiter plate and varying concentrations of protein (2 nM - 5 pM) were added in 5 pL aliquots to give a final volume of 100 pL. The cultures were incubated statically for 24 h at 26 °C for E. coli, and 37 °C for S. carnosus, to allow for biofilm formation. To eliminate edge-effects, -200 pL of sterile water was placed in all outside wells and the plate was sealed with parafilm. After incubation non-adherent cells and media were removed by washing the plate with deionized water three times. The wells were stained with 150 pL of 0.1 % (w/v) crystal violet for 10 min, and washed with water three times. The remaining dye was solubilized with 100 pL of 33% (v/v) acetic acid for 10 min with rotation, after which time the absorbance was measured at 595 nm using a SpectraMax M2 from Molecular Devices (Sunnyvale, CA). The amount of biofilm is proportional to the absorbance from staining with crystal violet (Merritt et al 2005). All reactions were completed in at least triplicates and buffer G was used as an untreated control. Either an E. coli pgaABCD knockout strain (DPGA) or S. carnosus treated with gentamycin, was used as a control for background straining.

Result:

[00281] Over-producing PNAG strains from E. coli and S. carnosus that can be turned on with the addition of antibiotics were used. The addition of BpsB 2 7-7oi . BpsB 3 i8-67o, PgaB 22 -672, and PgaB 3 io-67o to E. coli cultures prevented biofilm formation with EC50 values of -40 nM, -60 nM, -90 nM, and -230 nM, respectively (Figure 22A). Preliminary testing of biofilm inhibition for PNAG-dependent S. carnosus biofilms shows BpsB 3 -i8-67o can inhibit biofilm formation at 5 μΜ (Figure 22B).

disperse pre-formed PNAG- dependent biofilms

Method:

[00282] The methodology to form PNAG-dependent biofilms utilizes a similar methodology to that of Psl, described in Example 4, with the exception of the following differences. PNAG-overproducing E. coli was grown at 37 °C overnight in LB broth supplement with 200 ug/mL ampicillin and 100 ug/mL kanamycin with shaking at 200 rpm. An E. coli pgaABCD knockout strain was used as a non-biofilm forming control for background straining. To test different manufacturers plates, experiments were performed using sterile 96- well polystyrene round bottom microtitre plates from both Nunc and Sarstedt.

Result:

[00283] To examine whether BpsB 2 7 -7 oi , BpsB 3 i 8- 67o, PgaB 22- 672, and PgaB3io-672 can degrade pre-formed PNAG biofilms, E. coli biofilms were grown overnight prior to addition of enzyme. The catalytic rate of an enzyme reaction is dependent on both the amount of substrate and enzyme added. It is therefore important to note that the biofilm mass is variable between experiments. The addition of BpsB 27 -7oi , BpsB 3 i 8 -67o > PgaB 22 -672, and PgaB 3 i 0 - 672 , to pre-formed biofilms for 60 min with an average starting OD 595 of 0.5, degraded the biofilm with EC50 values of -170 nM, -90 nM, >1000 nM, and 200 nM, respectively (Figure 23A). Plates from different manufacturers were also tested, which resulted in varying levels of adherence, but similar levels of dispersal (Figure 23B). Lastly, it was shown that dispersal of PNAG- dependent biofilms is specific, as the biofilm degrading enzymes PelA4 7-3 o 3 and PslG 3 i -4 4 2 had no affect (Figure 23C). The following alanine variants of BpsB: D326A, D328A, H473A, D474A, and E585A, were unable to disperse PNAG-dependent biofilms suggesting they play an important role in binding and/or hydrolyzing PNAG. Variants that maintain ability to cut the exopolysaccharide will likely require such residues to be maintained. Alternatively, such residues could be further optimized and a person skilled in the art would readily be able to determine if activity is increased or decreased upon mutation.

hydrolvze dPNAG purified from S. aureus

Method:

[00284] Isolated dPNAG from S. aureus strain MN8m was solubilized in 6 N HCI at - 10 mg/mL. 10 N NaOH was used to titrate the re-suspension until neutrality was reached, ~pH 7-8. In a 50 μΙ_ reaction, 1 μΜ BpsB 3 i 8- 67o was incubated with 1 -5 mg/mL of dPNAG in 100 mM HEPES pH 7.0 for 18 hours. The sample was split into two 20 μΙ_ aliquots and treated with DTT/3- Methyl-2- benzothiazolinone hydrazone hydrochloride hydrate and heated at 80 °C for 15 minutes. A solution of 0.5% (w/w) iron (III) ammonium sulfate dodecahydrate, 0.5% (w/w) sulfamic acid, and 0.25 N hydrochloric acid was added, mixed, and cooled to room temperature. 100 μΙ_ was then transferred to a 96-well clear bottom plate and the absorbance was measured at 620 nm using a SpectraMax M2 from Molecular Devices (Sunnyvale, CA). Protein and dPNAG in buffer H were used as background controls.

Result:

[00285] To test whether the BpsB and PgaB anti-biofilm activity was directly due to the cleavage of the PNAG polysaccharide, reducing sugar assays were conducted. The results indicate a clear signal above background using a 24 h end-point assay indicating that the enzyme is able to hydrolyze the polysaccharide (Figure 24). As dPNAG is estimated to be -5% deacetylated, the average length of the dPNAG polymer (-200 units), and the signal generated in the hydrolysis assay suggests that cleavage may occur at a glucosamine residue. This would generate about 10 additional reducing ends per polymer. Without wishing to be bound by theory, since this result indicates that hydrolysis may occur at a glucosamine residue, having a BpsB or PgaB construct that includes the deacetylase domain should be beneficial in increasing the amount of glucosamine residues present on dPNAG thereby increasing sites for hydrolysis. This in turn should increase the efficiency of biofilm inhibition and degradation. Additionally, BpsB 2 7-7oi and BpsB3i 8- 67o, showed about 4 times the hydrolysis activity compared to PgaB 2 2-672 and PgaB3io-672, suggesting BpsB 2 7-7oi and BpsB 3 i 8 . 6 7o are better hydrolases under the conditions of this assay. This result correlates with the biofilm assays (Figure 23A). The following alanine variants of BpsB: D326A, D328A, H473A, D474A, and E585A, were unable to hydrolyze dPNAG, suggesting they play an important role in binding or hydrolysis of dPNAG. As stated above, variants that maintain ability to cut the exopolysaccharide will likely require such residues to be maintained. Alternatively, such residues could be further optimized and a person skilled in the art would readily be able to determine if activity is increased or decreased upon mutation.

Example 16 - PelA 7 -3oa can prevent A. fumiqatus GAG-dependent biofilms

Method:

[00286] To examine whether PelA 7_ 3 o3, could inhibit the formation of GAG dependent biofilm, 5x10 4 A. fumigatus conidia/well were grown at 37 °C and 5% C0 2 for a duration of 20 h in Brian media supplemented with PelA 47 . 303. To quantify GAG biofilm formation, each well was washed twice with 400 pL of dH 2 0 and stained for 10 min with 300 μΙ_ of 0.1% (w/v) crystal violet (Merritt et al 2005). Following this stain the wells were washed twice and the remaining dye was solubilized by addition of 300 pl_ of 95% (v/v) ethanol and left for 10 min after which time the absorbance was measured at 600 nm.

Result:

[00287] An ex vivo assay was employed to examine whether exogenously added PelA prior to biofilm formation could prevent GAG polysaccharide biofilm formation. The addition of ≥2 μΜ of PelA 47 _3 0 3 was sufficient to prevent GAG biofilm formation as detected through the crystal violet assay (Figure 25A). Two putative catalytic variants D160A and E218A were also shown to inhibit at these concentrations. A similar result was obtained for PelA 35- 29i (Figure 25B). Without wishing to be bound by theory, the enzyme variants may remain capable of binding the polysaccharide, thereby reducing the ability of the A. fumigatus to adhere to the abiotic plate.

Example 17 - PelA 4 7-m can hvdrolvze the GAG polysaccharide from A. fumigatus.

Method:

[00288] Crude GAG was isolated from A. fumigatus biofilms. 200 μ[_ GAG aliquots were centrifuged to pellet the gelatinous fraction. The pellets were washed twice with 350 μΙ_ PBS. The wash procedure included vortexing for 5 min, sonicated in a bath for 3 min, and manual mixing by pipetting to reach homogeneity. The final pellet was resuspended in 200 μΙ_ of PBS. Samples were treated with 10-20 μΜ protein and incubated at 26 °C. Samples were taken at 24 h. GAG hydrolysis was quantified using a reducing sugar assay as described previously (Anthon & Barrett 2002) with slight modifications. Briefly, 20 μΙ_ of enzyme reaction was mixed with 20 μΙ_ of 0.5 M NaOH and 20 μΙ_ of MBTH/DTT solution (1.5 mg/L 3-methyl-2- benzothiazolinone hydrazine (MBTH) and 0.5 mg/L DTT). The samples were incubated at 80 °C for 15 min before the addition of 40 μΙ_ of acidic iron reagent (0.5% (FeNH4(S0 4 ) 2 ) · 12 H 2 0, 0.5% sulfamic acid, and 0.25 N HCI). Samples were diluted two-fold in water, before the absorbance was quantified at 620 nm.

Result:

[00289] To determine whether PelA 47 -303 is capable of hydrolyzing the

GAG polysaccharide, a reducing sugar assay was completed using purified GAG from A. fumigatus strain Af293. The number of reducing ends in solution increased over the 24 h reaction period as compared to an untreated sample (Figure 26). The activity was specific as the release of chitin and chitosan was not significant over background hydrolysis. The single point variant E218A, abolished activity. Variants that maintain ability to cut the exopolysaccharide will likely require such residue to be maintained. Alternatively, such residue could be further optimized and a person skilled in the art would readily be able to determine if activity is increased or decreased upon mutation.

Example 18 - PelA 4 7-m? and orthologs can disperse A. fumigatus GAG- dependent biofilms

Method:

[00290] A total of 5x10 4 A. fumigatus conidia/well were grown at 37 °C and 5% C0 2 for a duration of 20 h in Brian media to allow for GAG production and adherence to sterile 24-well plates. To measure the dispersion of the GAG biofilm, media was aspirated and replaced with media containing PelA 47- 303 or putative catalytic variants at concentrations as low as 0.28 μΜ and allowed to incubate in fresh media for an additional 20 h. To quench the reaction, each well was washed twice with 400 pL of dH 2 0 and stained for 0 min with 300 μΙ_ of 0.1 % (w/v) crystal violet. Following this stain, the wells were washed twice and the remaining dye was solubilized by addition of 300 μΙ_ of 95% (v/v) ethanol and left for 10 min after which time the absorbance was measured at 600 nm.

Result:

[00291] It was observed that PelA 47 - 303 from P. aeruginosa PA01 and PelA 35 . 2 9i from P. protogens resulted in the elimination of the GAG biofilm while the putative catalytic variants and BSA control were unable to disrupt the biofilm (Figure 27). PelA ortholog RagA 6 i-3i 7 was also able to disperse GAG-dependent biofilms, while the PelA ortholog from G. metallireducens (GmefPelA23-2 77 ) was less efficacious (Figure 28) but nonetheless worked. Example 19 - PelA,.?-.^ can prevent epithelial cell damage caused by A. fumigatus infection

Method:

[00292] The immortalized airway epithelial cell line A549 was loaded with chromium-51 by incubating monolayers grown in 24-well tissue culture plates with 3 mCi of 51 Cr at 37 °C in 5% C0 2 for 24 h. Excess chromium was removed by washing with Hank's Balanced Salt Solution (HBSS). The labeled A549 cells were then infected with 5* 10 5 conidia in 1 mL serum free DF12K medium with 0.5 μΜ PelA 47 .303 or the E218A variant. For testing germ tubes, conidia were grown for 7 h in SAB media at 37 °C, then the fungus was collected and resuspended in DF12K media before 0.5 μΜ of hydrolase was added. The germ tubes were incubated with hydrolase for 1 h before being added to the A549 cells. After 16, 20 and 24 h of co-incubation, an aliquot of the medium above the cells was retrieved and replaced with fresh media. The cells were then lysed with 6 N NaOH and the lysate collected. The 51 Cr content of the medium and lysates was then measured in a gamma counter and the extent of epithelial cell damage was calculated as a function of the degree of 51 Cr release. Each strain was tested in triplicate, and all results were corrected for spontaneous chromium release by uninfected epithelial cells.

Result:

[00293] The addition of exogenous PelA 4 7.303 blocked the ability of A. fumigatus to induce pulmonary epithelial cell injury as measured by a chromium release assay over a period of 16 h (Figure 29). In comparison, the exogenous addition of PelA 7-3 o3 E218A, previously shown to be unable to disrupt GAG-dependent biofilm did not prevent epithelial cell damage, confirming that the protective effect observed in the wild-type enzyme is a direct result at inhibiting and dispersing GAG-dependent biofilms. Without wishing to be bound by theory, PelA orthologs mentioned herein likely complete the same function. As stated above, variants that maintain ability to cut the exopolysaccharide will likely require such residue to be maintained. Alternatively, such residue could be further optimized and a person skilled in the art would readily be able to determine if activity is increased or decreased upon mutation.

Example 20 - PelA 47- 30 can enhance human neutrophil killing of P. aeruginosa

Method:

[00294] Overnight cultures of P. aeruginosa PA01 AwspFApslp B ADpel were diluted to an OD 60 o of 0.05 in LB+0.5% arabinose and inoculated in a 96 well tissue culture-treated plate at a final volume of 100pl/well. The plate was incubated statically at 28 °C for 20 h. Supernatants were aspirated and 100 μΙ_ of phenol red-free RPMI+10% Fetal Bovine Serum (FBS) containing 0.5 μΜ of PelA 47 -303 was added. The plate was incubated at room temperature on a Nutator for 1 h. Following pretreatment with hydrolase, 100 μΙ_ RPMI+10% FBS containing 6X10 6 differentiated HL-60 cells were added to the wells, and plate was incubated for 90 min at 37 °C, 5% C0 2 . Wells were aspirated and supernatant was diluted between 1/200000 and 1/400000, and plated (50μΙ) onto LB agar. To aspirated wells, 200 pL of "disruption solution" (PBS containing 2 μΜ of PelA 47 -303 and 2 μΜ PslG 3 i -44 2) was added and plate was incubated at room temperature on the Nutator for 1-1 .5 h. Wells were aspirated, diluted and plated onto LB agar as above.

Result:

[00295] To determine if hydrolase treatment could enhance the susceptibility of microorganisms to immune killing, the ability of PelA 47 . 30 3 to enhance the susceptibility of P. aeruginosa to neutrophils was examined. Treatment of Pel-containing P. aeruginosa biofilms with PelA increased the degree of microbial killing by the HL-60 neutrophil cell line from approximately 22% to 42% (Figure 30). It is predicted that other soluble glycosyl hydrolases will also function in an analogous manner to potentiate neutrophil killing. Example 21 - PelA 47 . 3n3 can be formulated in a gel to disperse Pel-dependent P. aeruginosa biofilms

Method:

[00296] Poloxamer 407 (trade name Pluronic® F-127 or PF-127) is a hydrophilic non-ionic surfactant has been used as a drug delivery system for a number of routes of administration (Escobar-Chavez et al 2006). A 20% gel of PF-127 was prepared by mixing 1 x PBS with Pluronic® F-127 at 4°C until the polymer was dissolved. PelA 47- 303 was added at three concentrations; 100 μg/mL, 200 μg/mL and 500 μg/mL to this solution. Pel-dependent biofilms from strain P. aeruginosa PA01 AwspFAps/pBADpe/ were formed as described in Example 8. The buffer, gel and gels containing various concentrations of PelA 47-30 3 were applied to the biofilm.

Result:

[00297] Treatment of biofilms with either 1 x PBS buffer or 20% Pluronic® F-127 did not result in biofilm dispersal, however Pluronic® F-127 containing the three concentrations of PelA 47- 303 successfully dispersed the biofilm as evident through crystal violet staining following a 1 h treatment. (Figure 31 ) Without wishing to be bound by theory, it is believed that a similar delivery strategy can be utilized for any of the glycosyl hydrolases disclosed herein using agents that can encapsulate the enzymes.

Psl-containing biofilms from clinical and environmental isolates of P. aeruginosa

Method:

[00298] The methodology used for the growth and dispersal of biofilms from clinical and environmental P. aeruginosa isolates is identical to that of laboratory strains as initially described in Example 4. One exception to this methodology is that L-arabinose is not required to induce formation of biofilms in these strains and therefore the ability to form biofilms is strain specific. The diversity of these selected isolates was previously published (Wolfgang et al 2003) and the propensity for these strains to utilize the Pel and Psl polysaccharides was characterized using genetic deletions of the pel and psl operons (Colvin et al 2012).

Result:

[00299] An ex vivo assay was employed to examine whether exogenously added PsiG, PelA and combinations thereof could be utilized to disperse biofilms formed by clinical and environmental P. aeruginosa isolates. The addition of 100-1000 nM of PslG 3 -442 , PelA 47-30 3 or equimolar concentrations of both enzymes together resulted in >90% reducing in biofilm biomass as detected through crystal violet staining in a 1 hour period (Figure 32). This demonstrates that these glycosyl hydrolases can be applied in combination to disperse biofilms in clinically relevant strains.

Example 23 - Sph3s?-7ga is a soluble protein

Method:

[00300] A comparative transcriptomic analysis of transcription factor mutants with impaired GAG production identified a cluster of 5 co-regulated genes predicted to encode proteins required for GAG biosynthesis. Several genes required for GAG biosynthesis in A. fumigatus are encoded on chromosome 3 in the genome (Figure 33). A constructed Asph3 knockout is unable to produce deacetylated GAG. The protein coding sequence of sph3 and ega3 from A. fumigatus Af293 were deposited into GenBank under accession no. EAL92786.1 and EAL92787.1 , respectively. Sph3 is a 298 amino acid protein that is predicted, by the TMHMM server, to contain a transmembrane helix at its N-terminus (aa 20-42) (Krogh et al 2001 ). TMHMM classifies Sph3 as a type II membrane protein with the C-terminal domain on the extracellular surface of the cell. Structural homology searches suggest that Sph3 52 -298 is composed of a (β/α) 8 TIM-barrel fold. Phyre 2 bases these models on glycosyl hydrolases with confidence of up to 96.9 % (Kelley & Sternberg 2009). The nucleotide sequence was used to design primers specific to the gene that would amplify the region encoding residues 52 to 298, which excluded the putative transmembrane helix. For Sph3, the forward primer, 5'-GGGCATATGTCCAAGGTCTTTGTGCCTCTCTATGTG -3' (SEQ ID NO:9) encoded a Nde\ site and the reverse, 5'- GGCTCGAGCTATTTTCCCATCAAATCCACAAACTC -3' (SEQ ID NO:10), contained an Xho\ site (Figure 34). The PCR amplified product was digested using the Nde\ and Xho\ endonucleases and then ligated into a pET28a vector (Novagen). The sequence was confirmed by ACGT DNA Technologies Corporation before use.

[00301] Protein expression plasmids were transformed into E. coli BL21 (DE3) cells and grown in 1.5 L Luria-Bertani (LB) broth, with 50 Mg/mL kanamycin at 37°C to an OD600 of ~ 0.35-0.40. The temperature was reduced to 18°C and at OD600 ~ 0.5-0.6 protein expression was induced using 0.5 mM isopropyl β-D-l-thiogalactopyranoside (IPTG). Cells were grown post-induction overnight and harvested by centrifugation for 20 min at 4000 x g. Cells were re-suspended in Buffer I (50 mM CHES pH 9.0, 300 mM NaCI, 2% (v/v) glycerol, 10 mM imidazole, 2 mM TCEP) with a protease tablet (Sigma), and lysed using an Emulsiflex C3 homogenizer at 15,000 psi. Cellular debris was pelleted by centrifugation at 30,000 x g and the supernatant was applied to a Ni-NTA agarose column (Qiagen). The column was washed with 10 column volumes of Buffer I and 4 column volumes of Buffer A with 20 mM imidazole. The hexa-histidine tagged protein was then eluted using Buffer I with 200 mM imidazole and the resulting fraction was concentrated using an Amicon Ultra filtration device (Millipore) to a volume of 2 ml_. The concentrated protein was further purified using a HiLoad Superdex 200 size exclusion column (GE Healthcare) equilibrated with Buffer J (50 mM CHES pH 9.0, 150 mM NaCI, 1 mM TCEP).

Result:

[00302] A Sph3 52 -298 construct encompassing residues 52-298 was expressed and purified. The expressed protein produces -10 mg per liter of bacterial culture with a molecular weight of 29.6 kDa. The protein was judged to be >95% pure by SDS-PAGE and the protein could be concentrated to 8- 10 mg/mL and stored at 4 °C (Figure 35).

Example 24 - Sph3q?-?g« can hvdrolvze the GAG polysaccharide

Method:

[00303] The methodology to examine hydrolysis of the GAG polysaccharide using Sph352-298 is identical to that previously described in Example 17 for the use PelA 7- 303-

Result:

[00304] To determine whether Sph3 52- 298 can hydrolyze the GAG polysaccharide, a reducing sugar assay was completed using purified GAG. The number of reducing ends in solution increased over the 24 h reaction period as compared to an untreated sample (Figure 26). The single catalytic point variant D166A, abolished activity. Variants that maintain ability to cut the exopolysaccharide will likely require such residue to be maintained. Alternatively, such residue could be further optimized and a person skilled in the art would readily be able to determine if activity is increased or decreased upon mutation.

Example 25 - Sph3 S 7,7qa can prevent and disperse A. fumipatus GAG biofilms including A. fumipatus clinical isolates.

Method:

[00305] The methodology to examine the dispersal of GAG biofilms using Sph3s 2 -298 is identical to that previously described in Example 18 for the use PelA 47- 303. Clinical isolates of A. fumigatus were obtained from clinical mycology lab at McGill University Health Centre.

Result:

[00306] An ex vivo assay was employed to examine whether exogenously added Sph3 52- 298 could disperse pre-formed biofilms. The addition of 280 nM of Sph3 5 2-298 was sufficient at preventing GAG biofilm formation as detected through the crystal violet assay (Figure 36). This suggests that Sph3s2-298 is able to disperse GAG biofilms in a similar manner as PelA 4 7_303 disrupting Pel polysaccharide biofilms from P. aeruginosa and that this enzyme may represent a novel antifungal therapy. Recombinant Sph3 from A. fumigatus, A. nidulans and A. clavatus all inhibit the formation of A. fumigatus biofilms in a dose dependent manner. Similarly, soluble Sph3 from all three Aspergillus species disrupted preformed biofilms of A. fumigatus in a dose dependent manner (Figure 37). Introduction of point mutations in the catalytic domain of these soluble Sph3 proteins was associated with a decrease or loss of activity, suggesting that the anti-biofilm activity of these proteins is mediated by enzymatic activity. Sph3 52- 298 from A. fumigatus was found to be active against multiple strains of A. fumigatus suggesting that these agents will be active against a variety of clinical isolates (Figure 38). Example 26 - Sph3Ac<5 -304^ can disperse the GAG biofilm of the fungal pathogen Trichosporon asahii.

Method:

[00307] To assess the effects of hydrolase treatment on Trichosporon asahii, 1 x10 5 T. asahii yeast cells or A. fumigatus conidia were inoculated in Dulbecco's Modified Eagle Medium (DMEM), and grown for 10 h at 37°C, 5% C0 2 on glass coverslips. Wells were gently washed once with Ham's F-12K (Kaighn's) Medium (F12K) media, and Sph3AC(54-304) was added at a final concentration of 0.5 μΜ in F12K, and incubated for an additional 3 h at 37°C, 5% C0 2 . Young hyphae were washed twice with PBS, and stained with FITC tagged SBA for 1 h. The stained samples were washed with PBS and fixed with 4% PFA for 15 min. Slides were then mounted, sealed, and imaged under confocal microscopy with a 488 nm laser (Zeiss).

Result:

[00308] As shown in Figure 39, the opportunistic fungal pathogen Trichosporon asahii produces a GalNAc-rich exopolysaccharide, which can be degraded by Sph3AC(54-304)- Similar to A. fumigatus, untreated T. asahii displays GalNAc decorations on the cell surface, as determined by SBA-FITC staining (top left and center). However, treatment with 0.5 μΜ Sph3AC(54-304) for 3 h results in a complete loss of detectable surface GalNAc (top right). Fungi were counterstained with DRAQ5 (bottom panels).

Example 27 - SphS^?.?^ can prevent epithelial cell damage caused by A. fumigatus and P. aeruginosa infection- Method:

[00309] The methodology for prevention of epithelial cell damage caused by A. fumigatus using Sph3 5 2-298 is identical to that previously described in Example 19 for the use PelA 47 _ 3 o 3 . For P. aeruginosa- ' mduced damage, A549s were exposed to Pel-producing P. aeruginosa in the presence or absence of Sph3AN(43-299) at 0.5 μΜ for 16 h, and mammalian cell damage was assayed by release of lactose dehydrogenase (LDH) into the supernatant.

Result:

[00310] To determine if recombinant hydrolases could protect host cells from injury by A. fumigatus, a chromium release damage assay was used to assess injury of the A549 pulmonary epithelial cell line by A. fumigatus. Treatment of A. fumigatus with Sph3 from A. clavatus, A. nidulans, or A. fumigatus reduced fungal damage to epithelial cells (Figure 40A). Of note, Sph3 from A. clavatus (Sph3 A c(54-304>) and A. nidulans (Sph3 A N(43-299)) completely protected epithelial cells from damage by A. fumigatus for 24 h. Sph3 AN (43-299) was also capable of reducing damage caused by P. aeruginosa (Figure 40B).

Example 28 - The glycoside hydrolases PslG, -44?, PelA^-ana and Sph3R?-?gs

compounds used to treat P. aeruginosa and A. fumigatus infections. Method:

[00311] The effect of a combination treatment with antifungals and Sph3AC(54-304) or PelA4 7- 303 on the growth of A. fumigatus conidia was determined using a multititer plate assay. Resting A. fumigatus conidia in RPMI 1640 medium (Life Technology) buffered with MOPS (3-(N-Morpholino) Propane-Sulfonic Acid) (Fisher) (RPMI-MOPS) were added to tissue culture- treated multititer plates (BD Falcon). The conidia were incubated for 9 h at 37 °C, 5% CO2. Stock solutions of antifungals were prepared in dimethyl sulfoxide (DMSO) and further diluted in RPMI-MOPS. Stock solutions of hydrolase were prepared and diluted in RPMI-MOPS. The antifungal compounds were two-fold serially diluted across the rows and 0.5 μΜ of Sph3 A c(5 4 -304) or PelA 4 7-303 was added. The plates were then incubated at 37 °C and 5% C0 2 and examined after 15 h under an inverted light microscope (Zeiss, Inc.) and images acquired using an Infinity2 camera (Lumenera, Inc.). To measure fungal viability, XTT [2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)- 2H-tetrazolium-5-carboxanilide] (Sigma) metabolic assay was subsequently performed; Menadione (Sigma) was added to XTT solution, and further diluted with RPMI-MOPS. The solution was added to the hyphae and incubated for 90 min. The OD 4 5o was measured using a spectrophotometer (ASYS UVM 340).

[00312] To examine the effect of PelA 7-3 o3 and PslG 3 i -442 on antibiotic potentiation, Pel-dependent and Psl-dependent biofiims were grown as previously described in Example 4 in either the absence or presence of 1 μΜ PelA 47- 303 or PslG 31 -4 42, respectively. After 24 h, all samples were treated with 100 Mg/mL of colistin and incubated at 25 °C for 24 h. Following this treatment, planktonic cells were removed by pipetting and remaining biofiims were suspended in buffer and subjected to a hydrolase treatment outlined in Example 4. Planktonic and biofilm biomass was pooled and serially diluted on LB agar plates for colony counts. Result:

[00313] To determine if treatment with recombinant hydrolases could enhance the activity of antimicrobial agents, the effects of Sph3AC(54-304) and PelA 47-3 o3 on antifungal susceptibility of A. fumigatus were examined. Treatment of A. fumigatus biofilms with Sph3AF, Sph3AC(54-304), Sph3 A N(43-299), or PelA 4 - 303 resulted in an >50% reduction in the minimal inhibitory concentration of the antifungals posaconazole, amphotericin B and caspofungin (Figure 41 ). Similar reductions in MIC were seen for sensitive and resistant strains of A. fumigatus.

[00314] PelA 47- 303 and PslG3i.4 4 2 were also able to potentiate the antibiotic colistin dosed at a final concentration of 100 pg/mL. The addition of either enzyme lead to a more than 100-fold increase in bacterial killing as no growth was observed on the LB plates (Figure 42). Without wishing to be bound by theory, it is likely that other classes of antibiotics with different mechanisms of action will also be potentiated by any glycosyl hydrolase given the involvement of exopolysaccharides in antibiotic protection.

Example 29 - Treatment of A. fumigatus with Sph3Ar-.ts 4- 304¾ increases antifungal drug penetration- Method:

[00315] Red Fluorescent Protein (RFP)-expressing Af293 hyphae were pretreated with 0.5 μΜ Sph3AC(54-304) hydrolase for 90 min before treatment with Bodipy-conjugated posaconazole (BDP-PCZ). At various time points, samples were fixed and imaged using fluorescent confocal microscopy to determine the kinetics of drug penetration of hyphae.

Result:

[00316] To determine if this increased sensitivity to antifungals in the presence of recombinant glycosyl hydrolases resulted from enhanced cellular penetration in the absence of exopolysaccharide, the rate of uptake of posaconazole by A. fumigatus was examined in the presence or absence of Sph3 A c(54-304)- Treatment of hyphae with Sph3 A c(54-304) increased the speed and degree of uptake of fluorescent-tagged posaconazole as visualized by confocal microscopy (Figure 43). Collectively these data demonstrate the degradation of exopolysaccharide by recombinant hydrolases can increase the activity of antifungals through enhancing their ability to penetrate fungal cells.

Example 30 - Identification of other species that have the genetic capacity to synthesize GAG and form GAG-dependent biofilms.

Method:

[00317] For bioinformatics analyses, amino acid sequences of the genes within the GAG biosynthesis cluster: AFU_3G07860 (Gtb3), AFU_3G7870 (Agd3), AFU_3G7890 (Ega3), AFU_3G7900 (Sph3), and AFU_3G7910 (Uge3), were obtained from the Aspergillus Genome Database (http://www.aspergillusgenome.org) and queried by BLAST on NCBI protein database (http://blast.ncbi.nlm.nih.gov/Blast.cgi). The Conserved Domain Database (CDD) was used to predict functional domains for each putative protein. Amino acid sequences of proteins with similar domain to the cluster genes were aligned by multiple sequence alignment (MSA) using ClustalW (http://www.ebi.ac.uk/Tools/msa/clustalw2/). The resulting phylogenic relationship output was exported, formatted, and drawn for visualization using TreeVector (http://supfam.cs.bris.ac.uk/TreeVector/) for each member of the cluster. For taxonomic analysis, classes of fungi containing the GAG cluster were selected and visualized using PhyloT (http://phylot.biobYte.de).

[00318] GAG production of Trichosporon asahii was assessed using soybean agglutinin (SBA) lectin staining. Briefly, 1x10 5 T. asahii yeast cells or A. fumigatus conidia were inoculated in RPMI 1640, and grown for 9 h at 37 °C, 5% CO2 on glass coverslips. Botrytis cinerea isolate 7b1 (from Drs. Carisse & Tremblay from Agriculture and Agri-Food Canada, St-Jean-sur- Richelieu, Quebec) was inoculated at a concentration of 6x10 3 conidia/mL in 500 μΙ of Brian media on poly-D-lysine-coated coverslips (Corning, Bedford, MA) for 17 h at room temperature, in the dark. Resulting young hyphae were fixed in 4% PFA for 30 min, washed in PBS, and stained with FITC tagged SBA for 2 h. The stained samples were washed with PBS, mounted, and imaged under confocal microscope with a 488 nm laser (Olympus, Inc.).

Result:

[00319] Bioinformatic analyses revealed the presence of the GAG biosynthetic gene cluster within the fungi of many ascomycete fungi, as well as the basidiomycete Trichosporon (Figure 44). These fungi include the plant pathogenic species: Saccharata proteae, Zopfia rhizophila, Phaeosphaeria nodorum, Setosphaeria turcica, Botrytis cinerea, Cryphonectria parasitica, Melanconium sp., Verticillium dahlia, Nectria haematococca, Neurospora crassa, Leptosphaeria maculans, Pleomassaria siparia, Cochliobolus heterostrophus, Pyrenophora tritici-repentis, Blumeria graminis, Marssonina brunnea, Sclerotinia sclerotiorum, Taphrina deformans, Cercospora zeae- maydis and human pathogenic fungi: Fusarium, Trichosporon and Aspergillus species. To confirm that the presence of these clusters in other fungi predicted the production of GAG, hyphae of T. asahii (Figure 39), and B. cinerea (Figure 45) were stained with GalNAc-specific lectins to detect the presence of GAG. Consistent with the presence of the GAG biosynthetic cluster within the genomes of these species, lectin staining demonstrated the production of GAG-like exopolysaccharide by both organisms.

Example 31 - Botrytis cinerea can produce GAG-dependent biofilms which can be digested by S h3AC(54-304)_

Method:

[00320] Botrytis cinerea isolate 7b1 was inoculated at a concentration of

6x10 3 conidia/ml in 500 μΙ of Brian media on poly-D-lysine-coated coverslips

(Corning, Bedford, MA) for 17 h at room temperature, in the dark. Samples were incubated either in the presence or absence of 1 μΜ Sph3AC(54-304)-

Media was then removed and the hyphae were incubated in PBS with or without 1 μΜ Sph3 A c(54-304) for 1 h at room temperature. Samples were washed twice with PBS, and stained with 30 pg/mL of Wisteria fluoribunda (WFL) lectin conjugated to fluoresceine for 2 h at 4°C. Samples were washed twice with PBS, and fixed with 4% paraformaldehyde for 10 min at 4°C. Samples were washed once and stained with a 1 : 1000 dilution of DRAQ5™ (eBioscience, San Diego, CA) for 5 min at room temperature. Samples were washed once with PBS, mounted onto microscope slides using SlowFade® Gold antifade reagent (life Technologies™, Eugene, OR), and sealed with nail polish. Images were acquired on an Olympus Fluoview confocal laser microscope using 488 and 633nm lasers coupled with BA505-525 and BA650IF filters, respectively. Z-stacks of 0.2 pm increments were acquired, and 3D-rendered using ImageJ software (National Institutes of Health).

Result:

[00321] Treatment of hyphae of B. cinerea with Sph3 A c ( 54-304) resulted in a complete loss of this exopolysaccharide (Figure 45). Without wishing to be bound by theory, this data suggests that recombinant hydrolases can digest GAG biofilms from other fungal species.

Example 32 - Ega34e-ai8 is a soluble protein

Method:

[00322] Bioinformatics predictions suggest that Ega3 has a single transmembrane (TM) helix from amino acid 23 to 45. The C-terminal domain, encompassing amino acids 46-318, has been predicted to be extracellular by

TMHMM. The Phyre 2 structural homology recognition server predicts the protein contains an (β/α) 8 TIM-barrel and is aligned with glycosyl hydrolyse family GH1 14. A plasmid containing the ega3 gene codon optimized for expression in E. coli was obtained from GeneArt. The sequence encoding the predicted extracellular domain, amino acids 46-318, were subcloned into a pET28 vector between the Ndel and Hindlll sites. The forward primer, 5'-

GGGAGTCATCGTATGGGCAGCAGCCATCATCATCATC-3' (SEQ ID NO:

24) encoding a Mlyl site and the reverse, 5'- GGGGGTACCTTAGCAATATTCCACCCA-3' (SEQ ID NO:25), contained an Kpnl site were used to amplify the Ega3 4 6-318 construct with the N-terminal hexahistidine tag. The PCR amplified product was digested using the Mlyl and Kpnl endonucleases and then ligation into a pPinka-HC vector pre- digested with the endonucleases Stul and Kpnl (Invitrogen). The sequence was confirmed by ACGT DNA Technologies Corporation before use.

[00323] Protein expression plasmids were transformed into Pichia pastoris (PichiaPink Strain 4) plated on Pichia Adenine Dropout (PAD) selection plates to identify transformed colonies. Colonies were selected and grown in 2 mL buffered complex glycerol medium (BGMY) media at 30 °C. Starters were used to inoculate 500 mL of BMGY and then grown at 26 °C overnight. This larger culture was harvested and then resuspended in 450 mL buffered complex methanol medium (BMMY) media, grown at 26 °C with shaking for protein expression. Cells were harvested after 48 h by centrifugation for 20 minutes at 4000 x g. The culture supernatant was applied to a 3 mL Ni-NTA agarose column (Qiagen). The column was washed with 10 column volumes of Buffer K (50 mM HEPES pH 7.5, 300 mM NaCI, 5 mM imidazole). The hexa-histidine tagged protein was then eluted using Buffer K supplemented with 200 mM imidazole and the resulting fraction was concentrated using an Amicon Ultra filtration device (Millipore) to a volume of 2 mL. The concentrated protein was further purified using a HiLoad Superdex 200 size exclusion column (GE Healthcare) equilibrated with Buffer L (50 mM HEPES pH 7.5, 300 mM NaCI).

Result:

[00324] An Ega3 6- 3is construct encompassing residues 46-318 was expressed and purified. The expressed protein produces ~3 mg per liter of yeast culture with a molecular weight of 31.8 kDa. The protein was judged to be >95% pure by SDS-PAGE (Figure 46). Protein glycosylation is common during recombinant protein expression and secretion in P. pastoris. To determine whether the apparent mass of Ega3 46- 318 was larger due to glycosylation a sample of protein was treated with endoglycosidase H (EndoH). After treatment the sample was run on SDS-PAGE. The EndoH treated Ega3 46 -318 produced a band near the predicted mass of the unglycosylated protein at 31 kDa.

Example 33 - Eqa3 4 fi-31 B can inhibit and disperse GAG biofilms from A. fumigatus.

Method:

[00325] A. fumigatus conidia were inoculated into polystyrene, round- bottom 24-well plates at a concentration of 10 5 conidia/well in DMEM media. To assess the inhibition of biofilm formation, Ega3 4 6-3ie was added at a final concentration of 1 μΜ and the conidia incubated for 18 h at 37 °C, 5% C0 2 . The resulting biofilms were then gently washed twice with distilled water, and stained with 0.1 % crystal violet for 10 min. Samples were washed twice with water and the wells were imaged.

[00326] To determine the effect hydrolase has on a preformed biofilm, 10 5 A fumigatus conidia were inoculated per well in DMEM media as above, but in the absence of hydrolase. Following 18 h incubation at 37 °C, 5% C0 2 , the resulting biofilm was treated with Ega3 4 6-318 at a final concentration of 1 μΜ for 1.5 h at room temperature on a nutator. The biofilms were then washed and stained as mentioned above.

Result:

[00327] While an untreated A. fumigatus biofilm grown for 18 h remains adherent to polystyrene after gentle washing, conidia grown in the presence of 1 μΜ Ega3 46 -318 were found to be completely non-adherent (Figure 47). Also, the addition of Ega3 4 6-3i8 at a concentration of 1 μΜ to a preformed, 18 h A. fumigatus biofilm and incubation for 1.5 h at room temperature resulted in the disruption of the biofilm. Figure 47 suggests that Ega3 4 6-318 is capable of degrading /V-acetylgalactosamine (GalNAc) residues from the surface of A. fumigatus. While untreated hyphae display extensive surface GalNAc decorations when stained with the fluorescent lectin SBA-FITC (top left), treatment of the hyphae with 0.5 μΜ Ega3 46- 318 for 3 h resulted in a complete loss of detectable GalNAc on the surface (top right). Hyphae were counterstained with DRAQ5 (bottom panels). It is important to note that both samples were inoculated with the same density of A. fumigatus conidia, and that treatment with Ega3 4 6-318 resulted in a loss of adherence and therefore hyphae retained.

[00328] While the present disclosure has been described with reference to what are presently considered to be the examples, it is to be understood that the disclosure is not limited to the disclosed examples. To the contrary, the disclosure is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.

[00329] All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

Table of Sequences

PsIGs-i^ ^. aeruginosa PA01 )

EIQVLKAPRAWWKDFLGVNAQFLWFSPERYNKQIDRLQDLGLEWVRLDLH WDRLETAEDQYQLASLDQLVKDLEARQLKSVFYLVGSARFITTAPFYSPFQD QYPPRDPEVFARRMAMLSQRYPSVAAWQVWNEPNLIGFWRPKADPEGYA KLLQASTIALRMVDPEKPVVSAGMAFFSEMPDGRTMFDALGHLGVESLGTI ATYHPYTQLPEGNYPWNLDFVSHANQINRALRNAGVPAIWSTEWGWSAYK GPKELQDIIGVEGQADYVLRRLALMSALDYDRIFLFTLSDLDQRASVRDRDY GLLDLDANPKPVYLALQRFLKVTGPKLRPADPPVTEDLPDGSFSIGWTRED GRN VWLFWSARGGNVRLPKLKEATLHDPLSGKVTPLSGSDGLEVPVKSSL QMLVWE (SEQ ID NO:1 1 )

PelA 47 -303 (P. aeruginosa PA01 )

GGPSSVAFWYAERPPLAELSQFDWWLEAAHLKPADVGYLKEQGSTPFAY LSVGEFDGDAAAIADSGLARGKSAVRNQAWNSQVMDLAAPSWRAHLLKRA AELRKQGYAGLFLDTLDSFQLQAEERREGQRRALASFLAQLHRQEPGLKLF FNRGFEVLPELPGVASAVAVESIHAGWDAAAGQYREVPQDDRDWLKGHLD ALRAQGMPIVAIDYLPPERRDEARALAARLRSEGYVPFVSTPALDYLGVSDV EVQP (SEQ ID NO:12)

PelA 35- 29i (P. protogens Pf-5)

AAPASVGFWYAEQPPLQELAQFEWAWEPGHMASADVATLRKLGSQPFAY LSVGEFDGNRAALAKQALAQGASPVRNKAWDSQVMDIATPAWREHLFKRA KALQDQGYAGLFLDTLDSFQLLPEADREPQRKALASFLRELHSRLPNLKLFF NRGFEVLGELDGVASAVAVESIHAGWDASAKRYRPVSEADRTWLEGELKP LRARNIPLVAIDYLPANRREEARKLVRQLSQEGFIPWTTPDLNALSMSTVEV QP (SEQ ID NO: 13)

Pel A23-2 7 7 (Geobacter metallireducens)

PPLSVALYYGKQPPVNDLHAFDIWIDPDSGLTPSEYGSGRSELFAYVSVGE ADTARSYTKQMPDRWIIGKNPVWKSKIVDVSSEEWKQFFLDDVVEPLWQA GYRGFFLDTLDSYLIAAPTEAHPRMEAGLVSWRAIRQRHPEARLILNRGFEI FDRVKDLVYAVAAESLFQNFNTVSGKYGAVDDKDRSWLTSRLNVIRETGVP VIAIDYVDPGNRPLMRETADKIRSLGFTPWVTDKDLAGLGIGSVEVMPRTVL GLYDGGEGAG (SEQ ID NO:14)

RagA 6 i -3i 7 (Ralstonia solanacearum GMI1000)

ADAPNIAWFYGDKPPVAQLRAFDAWVEPDHGFDPSRAKTPTTQWFAYVS VGEVAPERRWYKELPKAWLAGSNAAWASHVIDQSQPQWPAFYVDRVIAPL WDRGYRGFFLDTLDSYQLVAKDDAARAAQEAGMVRVIRAIKARYPEAKLIFN RGFEILPQVHDLAYAVAFESLYRAWDQGNKQYREVNDADRAWLMGQARKI QDEYHLPVISIDYCPPADRACARETAKRIKAQGLIPYVTDPALSTIGVGRIEVL

P (SEQ ID O:15) PgaB 22 -672 (Escherichia coli K-12 MG1655) ISQSRTSFIPPQDRESLLAEQPWPHNGFVAISWHNVEDEAADQRFMSVRTS ALREQFAWLRENGYQPVSIAQIREAHRGGKPLPEKAWLTFDDGYQSFYTR VFPILQAFQWPAVWAPVGSWVDTPADKQVKFGDELVDREYFATWQQVRE VARSRLVELASHTWNSHYGIQANATGSLLPVYVNRAYFTDHARYETAAEYR ERIRLDAVKMTEYLRTKVEVNPHVFVWPYGEANGIAIEELKKLGYDMFFTLE SGLANASQLDSIPRVLIANNPSLKEFAQQIITVQEKSPQRIMHIDLDYVYDENL QQMDRNIDVLIQRVKDMQISTVYLQAFADPDGDGLVKEVWFPNRLLPMKAD IFSRVAWQLRTRSGVNIYAWMPVLSWDLDPTLTRVKYLPTGEKKAQIHPEQ YHRLSPFDDRVRAQVGMLYEDLAGHAAFDGILFHDDALLSDYEDASAPAITA YQQAGFSGSLSEIRQNPEQFKQWARFKSRALTDFTLELSARVKAIRGPHIKT ARNIFALPVIQPESEAWFAQNYADFLKSYDWTAIMAMPYLEGVAEKSADQW LIQLTNQIKNIPQAKDKSILELQAQNWQKNGQHQAISSQQLAHWMSLLQLNG VKNYGYYPDNFLHNQPEIDLIRPEFSTAWYPKND (SEQ ID N0:16) PgaB 31 0-672 {Escherichia coli K-12 MG1655 )

EKSPQRIMHIDLDYVYDENLQQMDRNIDVLIQRVKDMQISTVYLQAFADPDG DGLVKEVWFPNRLLPMKADIFSRVAWQLRTRSGVNIYAWMPVLSWDLDPT LTRVKYLPTGEKKAQIHPEQYHRLSPFDDRVRAQVGMLYEDLAGHAAFDGI LFHDDALLSDYEDASAPAITAYQQAGFSGSLSEIRQNPEQFKQWARFKSRA LTDFTLELSARVKAIRGPHIKTARNIFALPVIQPESEAWFAQNYADFLKSYDW TAIMAMPYLEGVAEKSADQWLIQLTNQIKNIPQAKDKSILELQAQNWQKNGQ HQAISSQQLAHWMSLLQLNGVKNYGYYPDNFLHNQPEIDLIRPEFSTAWYP KND (SEQ ID N0:17)

BpsB27-7oi {Bordetella bronchiseptica RB50 )

YKVDMLPPPDPDDGLTFRVLCMHDVRDNLRASFADMPDQFAIETRTLTDLF EWIRVKGFNPISMQQIIDSRAGVRPLPPRPILLTFDDGYASTYTKVFPLLKKF NYPAVVAWTSWTDAPAGTKIRLSPKIEVPHDFFMTWAQLREMAQSGLVEL ASHSHNLHRGVLANPQGNEQPAASSRQYLPASGRYENDAEYRARVRQDLK TSADLIREHTGVTIRSIVWPYGAHNRDTDQVAAEVGLNIGLTLQPGPNTPDV ALTQIRRSLVDYEVNVATVARAMREPVSYHGQVRPIERIVQVDLDYIYDPDP EQQNRNLGQLIDRMKDLAPSAVYLQAFADPKGDGDITEVYFPNRHLPMRAD LFNRVAWQLKTRAGVMVYAWLPVLTFSVPPGNPAYGKVVQSTTRKPGERG LGSPTRLSPFHPDAHRVISEIYEDLAKAAHFDGLLFHDDAVLDDTEDSSPEAL ATYQGWGLPPDIAAIRADPKLAQQWSKGKIRYLIDFTMHLRHIVSGYQNDRD MVVARNLYAQPVLDPVSEAWYGQSLPEFLKSYDFVALMAMPNMEGAARPE QWMRQLVAAVARQKGLDRTIFELQARDWRVG PIDTEILRRQMVQLRSLGA INYGYYPDDFIANHPDAEALRDVMSLKSTLEKRRLTKAQELSRQTTLYGSAS QAEPTQR (SEQ ID N0:18)

BpsB 3 i 8-670 (Bordetella bronchiseptica RB50 )

PIERIVQVDLDYIYDPDPEQQNRNLGQLIDRMKDLAPSAVYLQAFADPKGDG DITEVYFPNRHLPMRADLFNRVAWQLKTRAGVMVYAWLPVLTFSVPPGNPA YGKWQSTTRKPGERGLGSPTRLSPFHPDAHRVISEIYEDLAKAAHFDGLLF HDDAVLDDTEDSSPEALATYQGWGLPPDIAAIRADPKLAQQWSKGKIRYLID FTMHLRHIVSGYQNDRDMVVARNLYAQPVLDPVSEAWYGQSLPEFLKSYD FVALMAMPNMEGAARPEQWMRQLVAAVARQKGLDRTIFELQARDWRVGK PIDTEILRRQMVQLRSLGAINYGYYPDDFIANHPDAEALRDVMSLKS (SEQ ID NO:19) Sph3 5 2-298 (Aspergillus fumigatus Af293)

SKVFVPLYVYPAPGAWTPLEDVISKHPDVNFTWINPGSGPGPNALPDGNY TREIPKLASYENVRLLGYVATTYAKRNISLVRRDIETYAAWPTNSSNPALAVR GIFFDETPQQYDEDALAYLQELTDWKNTPGLGPDHYVVHNPGAIPDSRYLS TADSTVVFEATYDTFQERHGAKLFEAIPDSNRSQLCAVIHSVPESVEGSALR SLVKQVRKVADEIFITHLDTDYYASFGRQWPEFVDLMGK (SEQ ID NO:20)

Sph3Ac (54-304) (Aspergillus clavatus NRRL 1 )

MGPKSKVFVPLYVYPAPGAWDPLEDVISKHPDVNFTVVINPGSGPGPE ALPDGNYTREIPKLASYENVRLLGYVATTYAKRNISEVRRDIETYAAWPTQS SNANLAVRGIFFDETPQQ

YDADILAYLRELTDWKGTSGLGPDHYWHNPGAIPDSRYLSTADSTWFEA TYATFQERHGAELFDTIP

DSHRDQLCAVIHSVPTSVEGSDLRGLVKQVRQVADEIFITHLETDYYAGFGG QWSEFVDL AS (SEQ ID NO:22)

Sph3 AN (43-299 ) ( ^sperg/7/us nidulans FGSC A4)

RRKNNNMGPKAKVFVPLYVYPAPGAWDPLVNVITAHPDVNFTVWNPGSG PGPNPLPDRNYTQEIPRLTAHDNVRVLGYVATTYAKRNISSVRNDIETYAAW PTISANPKLAVRGIFFDETPQQYNASDLAYLEELTSWKNTPGLGPDHFVFH NPGWPDPRYLSTADSTVVFEATYDTFQDRDGARLFETIPNSNRSQLCAW HSVPDSVEGSELRKFVKQARRVADEIFVTHLSTNYYASFGDKWDDFVRLMA

Q (SEQ ID NO:23)

Ega3 4 6-318 Aspergillus fumigatus Af293)

GLGGGGGGEGEEGSGGETTPPEGNYTTAKWQPAVGTKWQIELLYALNDT SVDAEIYDIDLFINDKSTIAGLQRAGRKVICYFSAGSYENWRPDKDKFKDSDL GHDLDDWPGEKWLNISSANVRQIMLDRLDMARDKGCDGVDPDNVDGYDN DNGLDLTQADSISFVNFLANAAHARNMSIGLKNAGDIIPSVIKNMQWSVNEQ CAQYNECDTYAVFPQNGKPVFHIEYPKGDKTNNDLSVTASQKNAACDFAG SANFSTVIKNMNLNNWVEYC (SEQ ID N0:21 )

REFERENCES

Adams PD, Afonine PV, Bunkoczi G, Chen VB, Davis IW, et al. 2010. PHENIX: a comprehensive Python-based system for macromolecular structure solution.

Acta Crystallogr D Biol Crystallogr 66: 213-21

Afonine PV, Mustyakimov M, Grosse-Kunstleve RW, Moriarty NW, Langan P, Adams

PD. 2010. Joint X-ray and neutron refinement with phenix. refine. Acta crystallographica. Section D, Biological crystallography 66: 1 153-63

Alhede M, Kragh KN, Qvortrup K, Allesen-Holm M, van Gennip M, et al. 201 1 .

Phenotypes of non-attached Pseudomonas aeruginosa aggregates resemble surface attached biofilm. PloS one 6: e27943

Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. 1990. Basic local alignment search tool. J Mol β/Ό/ 215: 403-10

Anthon GE, Barrett DM. 2002. Determination of reducing sugars with 3-methyl-2- benzothiazolinonehydrazone. Analytical biochemistry 305: 287-9

Ashkenazy H, Erez E, Martz E, Pupko T, Ben-Tal N. 2010. ConSurf 2010: calculating evolutionary conservation in sequence and structure of proteins and nucleic acids. Nucleic Acids Res 38: W529-33

Bagos PG, Nikolaou EP, Liakopoulos TD, Tsirigos KD. 2010. Combined prediction of

Tat and Sec signal peptides with hidden Markov models. Bioinformatics 26: 281 1 -7

Bakkevig K, Sletta H, Gimmestad M, Aune R, Ertesvag H, et al. 2005. Role of the

Pseudomonas fluorescens alginate lyase (AlgL) in clearing the periplasm of alginates not exported to the extracellular environment. J Bacteriol 187: 8375-

84

Bardas GA, Veloukas T, Koutita O, Karaoglanidis GS. 2010. Multiple resistance of

Botrytis cinerea from kiwifruit to SDHIs, Qols and fungicides of other chemical groups. Pest management science 66: 967-73

Billings N, Millan M, Caldara M, Rusconi R, Tarasova Y, et al. 2013. The extracellular matrix Component Psl provides fast-acting antibiotic defense in

Pseudomonas aeruginosa biofilms. PLoS Pathog 9: e1003526

Bjarnsholt T. 2013. The role of bacterial biofilms in chronic infections. APMIS.

Supplementum: 1-51

Bjarnsholt T, Ciofu O, Molin S, Givskov M, Hoiby N. 2013. Applying insights from biofilm biology to drug development - can a new approach be developed? Nature reviews. Drug discovery 12: 791 -808

Bragonzi A, Farulla I, Paroni M, Twomey KB, Pirone L, et al. 2012. Modelling co- infection of the cystic fibrosis lung by Pseudomonas aeruginosa and

Burkholderia cenocepacia reveals influences on biofilm formation and host response. PloS one 7: e52330

Branda SS, Vik S, Friedman L, Kolter R. 2005. Biofilms: the matrix revisited. Trends in microbiology 13: 20-6

Byrd MS, Pang B, Hong W, Waligora EA, Juneau RA, et al. 201 . Direct evaluation of Pseudomonas aeruginosa biofilm mediators in a chronic infection model.

Infection and immunity 79: 3087-95

Byrd MS, Pang B, Mishra M, Swords WE, Wozniak DJ. 2010. The Pseudomonas aeruginosa exopolysaccharide Psl facilitates surface adherence and NF- kappaB activation in A549 cells. mBio 1

Byrd MS, Sadovskaya I, Vinogradov E, Lu H, Sprinkle AB, et al. 2009. Genetic and biochemical analyses of the Pseudomonas aeruginosa Psl exopolysaccharide reveal overlapping roles for polysaccharide synthesis enzymes in Psl and

LPS production. Mol Microbiol 73: 622-38

Cerca N, Jefferson KK, Maira-Litran T, Pier DB, Kelly-Quintos C, et al. 2007.

Molecular basis for preferential protective efficacy of antibodies directed to the poorly acetylated form of staphylococcal poly-N-acetyl-beta-(1 -6)- glucosaminev. Infect Immun 75: 3406-13

Choi AH, Slamti L, Avci FY, Pier GB, Maira-Litran T. 2009. The pgaABCD locus of

Acinetobacter baumannii encodes the production of poly-beta-1 -6-N- acetylglucosamine, which is critical for biofilm formation. J Sacter/o/ 191 : 5953-63

Colvin KM, AInabelseya N, Baker P, Whitney JC, Howell PL, Parsek MR. 2013. PelA deacetylase activity is required for Pel polysaccharide synthesis in

Pseudomonas aeruginosa. J Bacteriol 195: 2329-39

Colvin KM, Gordon VD, Murakami K, Borlee BR, Wozniak DJ, et al. 20 1 . The pel polysaccharide can serve a structural and protective role in the biofilm matrix of Pseudomonas aeruginosa. PLoS pathogens 7: e1001264

Colvin KM, Irie Y, Tart CS, Urbano R, Whitney JC, et al. 2012. The Pel and Psl polysaccharides provide Pseudomonas aeruginosa structural redundancy within the biofilm matrix. Environmental microbiology 14: 1913-28

Conover MS, Sloan GP, Love CF, Sukumar N, Deora R. 2010. The Bps

polysaccharide of Bordetella pertussis promotes colonization and biofilm formation in the nose by functioning as an adhesin. Mol Microbiol 77: 1439-55 Costerton JW, Cheng KJ, Geesey GG, Ladd Tl, Nickel JC, et al. 1987. Bacterial biofilms in nature and disease. Annual review of microbiology 41 : 435-64 Costerton JW, Stewart PS, Greenberg EP. 1999. Bacterial biofilms: a common cause of persistent infections. Science 284: 1318-22

Cywes-Bentley C, Skurnik D, Zaidi T, Roux D, Deoliveira RB, et al. 2013. Antibody to a conserved antigenic target is protective against diverse prokaryotic and eukaryotic pathogens. Proc Natl Acad Sci U S A 'l 'l 0: E2209-18

Darouiche RO, Mansouri MD, Gawande PV, Madhyastha S. 2009. Antimicrobial and antibiofilm efficacy of triclosan and DispersinB combination. The Journal of antimicrobial chemotherapy 64: 88-93

Davies D. 2003. Understanding biofilm resistance to antibacterial agents. Nature reviews. Drug discovery 2: 1 14-22

Davies G, Henrissat B. 1995. Structures and mechanisms of glycosyl hydrolases.

Structure 3: 853-9

Dean R, Van Kan JA, Pretorius ZA, Hammond-Kosack KE, Di Pietro A, et al. 2012.

The Top 10 fungal pathogens in molecular plant pathology. Molecular plant pathology ^: 414-30

Digiandomenico A, Warrener P, Hamilton M, Guillard S, Ravn P, et al. 2012.

Identification of broadly protective human antibodies to Pseudomonas aeruginosa exopolysaccharide Psl by phenotypic screening. The Journal of experimental medicine 209: 1273-87

Dolinsky TJ, Czodrowski P, Li H, Nielsen JE, Jensen JH, et al. 2007. PDB2PQR: expanding and upgrading automated preparation of biomolecular structures for molecular simulations. Nucleic Acids Res 35: W522-5

Donelli G, Francolini I, Romoli D, Guaglianone E, Piozzi A, et al. 2007. Synergistic activity of dispersin B and cefamandole nafate in inhibition of staphylococcal biofilm growth on polyurethanes. Antimicrobial agents and chemotherapy 5 : 2733-40

Ellis M, Richardson M, de Pauw B. 2000. Epidemiology. Hospital medicine 61 : 605-9 Emsley P, Cowtan K. 2004. Coot: model-building tools for molecular graphics. Acta

Crystallogr D Biol Crystallogr 60: 2126-32

Escobar-Chavez JJ, Lopez-Cervantes M, Naik A, Kalia YN, Quintanar-Guerrero D,

Ganem-Quintanar A. 2006. Applications of thermo-reversible pluronic F-127 gels in pharmaceutical formulations. Journal of pharmacy & pharmaceutical sciences : a publication of the Canadian Society for Pharmaceutical

Sciences, Societe canadienne des sciences pharmaceutiques 9: 339-58 Flemming HC, Wingender J. 2010. The biofilm matrix. Nature reviews. Microbiology

8: 623-33

Fontaine T, Delangle A, Simenel C, Coddeville B, van Vliet SJ, et al. 201 1.

Galactosaminogalactan, a new immunosuppressive polysaccharide of Aspergillus fumigatus. PLoS pathogens 7: e1002372

Franklin MJ, Nivens DE, Weadge JT, Howell PL. 201 1 . Biosynthesis of the

Pseudomonas aeruginosa Extracellular Polysaccharides, Alginate, Pel, and Psl . Front Microbiol 2: 167

Friedman L, Kolter R. 2004a. Genes involved in matrix formation in Pseudomonas aeruginosa PA14 biofilms. Mol Microbiol 51 : 675-90

Friedman L, Kolter R. 2004b. Two genetic loci produce distinct carbohydrate-rich structural components of the Pseudomonas aeruginosa biofilm matrix. J Bacteriol 186: 4457-65

Gawande PV, Leung KP, Madhyastha S. 2014. Antibiofilm and Antimicrobial Efficacy of DispersinB-KSL-W Peptide-Based Wound Gel Against Chronic Wound Infection Associated Bacteria. Current microbiology

Geiser DM, Klich MA, Frisvad JC, Peterson SW, Varga J, Samson RA. 2007. The current status of species recognition and identification in Aspergillus. Studies in mycology 59: 1 -10

Grabke A, Fernandez-Ortuno D, Amiri A, Li X, Peres NA, et al. 2014.

Characterization of iprodione resistance in Botrytis cinerea from strawberry and blackberry. Phytopathology 104: 396-402

Gravelat FN, Beauvais A, Liu H, Lee MJ, Snarr BD, et al. 2013. Aspergillus

galactosaminogalactan mediates adherence to host constituents and conceals hyphal beta-glucan from the immune system. PLoS pathogens 9: e1003575

Gravelat FN, Doedt T, Chiang LY, Liu H, Filler SG, et al. 2008. In vivo analysis of Aspergillus fumigatus developmental gene expression determined by realtime reverse transcription-PCR. Infect. Immun. 76: 3632-39

Hall-Stoodley L, Costerton JW, Stoodley P. 2004. Bacterial biofilms: from the natural environment to infectious diseases. Nat Rev Microbiol 2: 95-108

Hare NJ, Solis N, Harmer C, Marzook NB, Rose B, et al. 2012. Proteomic profiling of Pseudomonas aeruginosa AES-1 R, PA01 and PA14 reveals potential virulence determinants associated with a transmissible cystic fibrosis- associated strain. BMC microbiology 12: 16

Henrissat B, Bairoch A. 1996. Updating the sequence-based classification of giycosyl hydrolases. The Biochemical journal 316 ( Pt 2): 695-6

Hoffman LR, D'Argenio DA, MacCoss MJ, Zhang Z, Jones RA, Miller SI. 2005.

Aminoglycoside antibiotics induce bacterial biofilm formation. Nature 436: 1 171 -5

Hoiby N, Bjarnsholt T, Givskov M, Molin S, Ciofu O. 2010. Antibiotic resistance of bacterial biofilms. Int J Antimicrob Agents 35: 322-32

Hoiby N, Ciofu O, Johansen HK, Song ZJ, Moser C, et al. 201 1. The clinical impact of bacterial biofilms. International journal of oral science 3: 55-65 Holm L, Kaariainen S, Rosenstrom P, Schenkel A. 2008. Searching protein structure databases with DaliLite v.3. Bioinformatics 24: 2780-1

Huse HK, Kwon T, Zlosnik JE, Speert DP, Marcotte EM, Whiteley M. 2013.

Pseudomonas aeruginosa Enhances Production of a Non-Alginate

Exopolysaccharide during Long-Term Colonization of the Cystic Fibrosis

Lung. PloS one 8: e82621

Irie Y, Borlee BR, O'Connor JR, Hill PJ, Harwood CS, et al. 2012. Self-produced exopolysaccharide is a signal that stimulates biofilm formation in

Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the United States of America 109: 20632-6

Itoh Y, Rice JD, Goller C, Pannuri A, Taylor J, et al. 2008. Roles of pgaABCD genes in synthesis, modification, and export of the Escherichia coli biofilm adhesin poly-beta-1 ,6-N-acetyl-D-glucosamine. Journal of bacteriology 190: 3670-80 Itoh Y, Wang X, Hinnebusch BJ, Preston JF, 3rd, Romeo T. 2005. Depolymerization of beta-1 ,6-N-acetyl-D-glucosamine disrupts the integrity of diverse bacterial biofilms. Journal of bacteriology 187: 382-7

Izano EA, Sadovskaya I, Vinogradov E, Mulks MH, Velliyagounder K, et al. 2007.

Poly-N-acetylglucosamine mediates biofilm formation and antibiotic resistance in Actinobacillus pleuropneumoniae. Microbial pathogenesis 43: 1 - 9

Izano EA, Sadovskaya I, Wang H, Vinogradov E, Ragunath C, et al. 2008. Poly-N- acetylglucosamine mediates biofilm formation and detergent resistance in Aggregatibacter actinomycetemcomitans. Microbial pathogenesis 44: 52-60 Jackson KD, Starkey M, Kremer S, Parsek MR, Wozniak DJ. 2004. Identification of psl, a locus encoding a potential exopolysaccharide that is essential for

Pseudomonas aeruginosa PA01 biofilm formation. Journal of bacteriology 186: 4466-75

Jain S, Ohman DE. 1998. Deletion of algK in mucoid Pseudomonas aeruginosa blocks alginate polymer formation and results in uronic acid secretion. Journal of bacteriology 180: 634-41

Jarrett CO, Deak E, Isherwood KE, Oyston PC, Fischer ER, et al. 2004.

Transmission of Yersinia pestis from an infectious biofilm in the flea vector. J Infect Dis 90: 783-92

Kali L, Krogh A, Sonnhammer EL. 2004. A combined transmembrane topology and signal peptide prediction method. Journal of molecular biology 338: 1027-36

Kaplan JB, Velliyagounder K, Ragunath C, Rohde H, Mack D, et al. 2004. Genes involved in the synthesis and degradation of matrix polysaccharide in

Actinobacillus actinomycetemcomitans and Actinobacillus pleuropneumoniae biofilms. Journal of bacteriology 186: 8213-20

Kelley LA, Sternberg MJ. 2009. Protein structure prediction on the Web: a case study using the Phyre server. Nature protocols 4: 363-71

Kim J, Hahn JS, Franklin MJ, Stewart PS, Yoon J. 2009. Tolerance of dormant and active cells in Pseudomonas aeruginosa PA01 biofilm to antimicrobial agents. The Journal of antimicrobial chemotherapy 63: 129-35

Krogh A, Larsson B, von Heijne G, Sonnhammer EL. 2001 . Predicting

transmembrane protein topology with a hidden Markov model: application to complete genomes. Journal of molecular biology 305: 567-80

Kukavica-lbrulj I, Bragonzi A, Paroni M, Winstanley C, Sanschagrin F, et al. 2008. In vivo growth of Pseudomonas aeruginosa strains PA01 and PA14 and the hypervirulent strain LESB58 in a rat model of chronic lung infection. Journal of bacteriology 90: 2804-13 Kumar CG, Anand SK. 1998. Significance of microbial biofilms in food industry: a review. International journal of food microbiology 42: 9-27

Lee DG, Urbach JM, Wu G, Liberati NT, Feinbaum RL, et al. 2006. Genomic analysis reveals that Pseudomonas aeruginosa virulence is combinatorial. Genome biology 7: R90

Lee JE, Cornell KA, Riscoe MK, Howell PL. 2001 . Structure of E. coli 5'- methylthioadenosine/S-adenosylhomocysteine nucleosidase reveals similarity to the purine nucleoside phosphorylases. Structure 9: 941 -53

Leroch M, Plesken C, Weber RW, Kauff F, Scalliet G, Hahn M. 2013. Gray mold populations in german strawberry fields are resistant to multiple fungicides and dominated by a novel clade closely related to Botrytis cinerea. Appl

Environ Microbiol 79: 159-67

Lin SJ, Schranz J, Teutsch SM. 2001. Aspergillosis case-fatality rate: systematic review of the literature. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 32: 358-66

Little DJ, Li G, Ing C, DiFrancesco BR, Bamford NC, et al. 2014. Modification and periplasmic translocation of the biofilm exopolysaccharide poly-β-1 ,6-N- acetyl-D-glucosamine. Proc Natl Acad Sci U S A Submitted

Little DJ, Poloczek J, Whitney JC, Robinson H, Nitz M, Howell PL. 2012a. The

structure- and metal-dependent activity of Escherichia coli PgaB provides insight into the partial de-N-acetylation of poly-beta-1 ,6-N-acetyl-D- glucosamine. The Journal of biological chemistry 287: 31 126-37

Little DJ, Whitney JC, Robinson H, Yip P, Nitz M, Howell PL. 2012b. Combining in situ proteolysis and mass spectrometry to crystallize Escherichia coli PgaB. Acta crystallographica. Section F, Structural biology and crystallization communications 68: 842-5

Lombard V, Golaconda Ramulu H, Drula E, Coutinho PM, Henrissat B. 2014. The carbohydrate-active enzymes database (CAZy) in 2013. Nucleic acids research 42: D490-5

Loussert C, Schmitt C, Prevost MC, Balloy V, Fadel E, et al. 2010. In vivo biofilm composition of Aspergillus fumigatus. Cellular microbiology 12: 405-10 Ma L, Conover M, Lu H, Parsek MR, Bayles K, Wozniak DJ. 2009. Assembly and development of the Pseudomonas aeruginosa biofilm matrix. PLoS pathogens 5: e1000354

Ma L, Jackson KD, Landry RM, Parsek MR, Wozniak DJ. 2006. Analysis of

Pseudomonas aeruginosa conditional psl variants reveals roles for the psl polysaccharide in adhesion and maintaining biofilm structure postattachment. Journal of bacteriology 188: 8213-21

Ma L, Wang S, Wang D, Parsek MR, Wozniak DJ. 2012. The roles of biofilm matrix polysaccharide Psl in mucoid Pseudomonas aeruginosa biofilms. FEMS immunology and medical microbiology 65: 377-80

Mack D, Fischer W, Krokotsch A, Leopold K, Hartmann R, et al. 1996. The

intercellular adhesin involved in biofilm accumulation of Staphylococcus epidermidis is a linear beta-1 ,6-linked glucosaminoglycan: purification and structural analysis. J Bacteriol 178: 175-83

Mah TF, Pitts B, Pellock B, Walker GC, Stewart PS, O'Toole GA. 2003. A genetic basis for Pseudomonas aeruginosa biofilm antibiotic resistance. Nature 426: 306-10

Maharaj R, May TB, Wang SK, Chakrabarty AM. 1993. Sequence of the alg8 and alg44 genes involved in the synthesis of alginate by Pseudomonas aeruginosa. Gene 136: 267-9 Mann EE, Wozniak DJ. 2012. Pseudomonas biofilm matrix composition and niche biology. FEMS microbiology reviews 36: 893-9 6

Manuel SG, Ragunath C, Sait HB, Izano EA, Kaplan JB, Ramasubbu N. 2007. Role of active-site residues of dispersin B, a biofilm-releasing beta- hexosaminidase from a periodontal pathogen, in substrate hydrolysis. The

FEBS journal 274: 5987-99

Matsukawa M, Greenberg EP. 2004. Putative exopolysaccharide synthesis genes influence Pseudomonas aeruginosa biofilm development. Journal of bacteriology 186: 4449-56

McKenney D, Pouliot KL, Wang Y, Murthy V, Ulrich M, et al. 1999. Broadly protective vaccine for Staphylococcus aureus based on an in vivo-expressed antigen.

Science 284: 1523-7

Merritt JH, Kadouri DE, O'Toole GA. 2005. Growing and analyzing static biofilms.

Current protocols in microbiology Chapter : Unit 1 B 1

Michielse CB, Rep M. 2009. Pathogen profile update: Fusarium oxysporum.

Molecular plant pathology 10: 31 1 -24

Mishra M, Byrd MS, Sergeant S, Azad AK, Parsek MR, et al. 2012. Pseudomonas aeruginosa Psl polysaccharide reduces neutrophil phagocytosis and the oxidative response by limiting complement-mediated opsonization. Cellular microbiology 14: 95-106

Monday SR, Schiller NL. 1996. Alginate synthesis in Pseudomonas aeruginosa: the role of AlgL (alginate lyase) and AlgX. Journal of bacteriology 178: 625-32 Nieman CE, Wong AW, He S, Clarke L, Hopwood JJ, Withers SG. 2003. Family 39 alpha-l-iduronidases and beta-D-xylosidases react through similar glycosyl- enzyme intermediates: identification of the human iduronidase nucleophile.

Biochemistry 42: 8054-65

Otwinowski Z, Minor W. 1997. Processing of X-ray diffraction data collected in

oscillation mode, pp. 307-26: Elsevier

Painter J, Merritt EA. 2006. Optimal description of a protein structure in terms of multiple groups undergoing TLS motion. Acta Crystallogr D Biol Crystallogr

62: 439-50

Parise G, Mishra M, Itoh Y, Romeo T, Deora R. 2007. Role of a putative

polysaccharide locus in Bordetella biofilm development. J Bacteriol 189: 750- 60

Park BH, Karpinets TV, Syed MH, Leuze MR, Uberbacher EC. 2010. CAZymes

Analysis Toolkit (CAT): web service for searching and analyzing

carbohydrate-active enzymes in a newly sequenced organism using CAZy database. Glycobiology 20: 1574-84

Petersen TN, Brunak S, von Heijne G, Nielsen H. 20 1 . SignalP 4.0: discriminating signal peptides from transmembrane regions. Nature methods 8: 785-6

Pokrovskaya V, Poloczek J, Little DJ, Griffiths H, Howell PL, Nitz M. 2013. Functional characterization of Staphylococcus epidermidis IcaB, a de-N-acetylase important for biofilm formation. Biochemistry 52: 5463-71

Rahme LG, Stevens EJ, Wolfort SF, Shao J, Tompkins RG, Ausubel FM. 1995.

Common virulence factors for bacterial pathogenicity in plants and animals.

Science 268: 1899-902

Rodriguez A, Acosta A, Rodriguez C. 2014. Fungicide resistance of Botrytis cinerea in tomato greenhouses in the Canary Islands and effectiveness of non- chemical treatments against gray mold. World journal of microbiology & biotechnology 30: 2397-406 Rybtke MT, Jensen PO, Hoiby N, Givskov M, Tolker-Nielsen T, Bjarnsholt T. 201 1 .

The implication of Pseudomonas aeruginosa biofilms in infections.

Inflammation & allergy drug targets 10: 141 -57

Sang H, Hulvey J, Popko JT, Jr., Lopes J, Swaminathan A, et al. 2015. A pleiotropic drug resistance transporter is involved in reduced sensitivity to multiple fungicide classes in Sclerotinia homoeocarpa (FT. Bennett). Molecular plant pathology 16: 251 -61

Schneidman-Duhovny D, Inbar Y, Nussinov R, Wolfson HJ. 2005. PatchDock and

SymmDock: servers for rigid and symmetric docking. Nucleic acids research 33: W363-7

Singh N. 2000. The current management of infectious diseases in the liver transplant recipient. Clinics in liver disease 4: 657-73, ix

Skurnik D, Roux D, Aschard H, Cattoir V, Yoder-Himes D, et al. 2013. A

comprehensive analysis of in vitro and in vivo genetic fitness of

Pseudomonas aeruginosa using high-throughput sequencing of transposon libraries. PLoS pathogens 9: e1003582

Sloan GP, Love CF, Sukumar N, Mishra M, Deora R. 2007. The Bordetella Bps polysaccharide is critical for biofilm development in the mouse respiratory tract. J Bacteriol 189: 8270-6

St John FJ, Gonzalez JM, Pozharski E. 2010. Consolidation of glycosyl hydrolase family 30: a dual domain 4/7 hydrolase family consisting of two structurally distinct groups. FEBS letters 584: 4435-41

Starkey M, Hickman JH, Ma L, Zhang N, De Long S, et al. 2009. Pseudomonas aeruginosa rugose small-colony variants have adaptations that likely promote persistence in the cystic fibrosis lung. J Bacteriol 191 : 3492-503

Stewart PS. 2003. New ways to stop biofilm infections. Lancer 361 : 97

Stewart PS, Costerton JW. 2001. Antibiotic resistance of bacteria in biofilms. Lancet

358: 135-8

Stover CK, Pham XQ, Erwin AL, Mizoguchi SD, Warrener P, et al. 2000. Complete genome sequence of Pseudomonas aeruginosa PA01 , an opportunistic pathogen. Nature 406: 959-64

Sutherland I. 2001 a. Biofilm exopolysaccharides: a strong and sticky framework.

Microbiology 147: 3-9

Sutherland IW. 2001 b. The biofilm matrix-an immobilized but dynamic microbial environment. Trends Microbiol 9: 222-7

Terwilliger TC, Berendzen J. 1999. Automated MAD and MIR structure solution. Acta

Crystallogr D Biol Crystallogr 55: 849-61

Tian L, Xu S, Hutchins WC, Yang CH, Li J. 2014. Impact of the exopolysaccharides Pel and Psl on the initial adhesion of Pseudomonas aeruginosa to sand. Biofouling 30: 213-22

Tong KB, Lau CJ, Murtagh K, Layton AJ, Seifeldin R. 2009. The economic impact of aspergillosis: analysis of hospital expenditures across patient subgroups. International journal of infectious diseases : I J ID : official publication of the International Society for Infectious Diseases 13: 24-36

Turk R, Singh A, Rousseau J, Weese JS. 2013. In vitro evaluation of DispersinB on methicillin-resistant Staphylococcus pseudintermedius biofilm. Veterinary microbiology 166: 576-9

Van Houdt R, Michiels CW. 2010. Biofilm formation and the food industry, a focus on the bacterial outer surface. Journal of applied microbiology 109: 1 1 17-31 Vocadlo DJ, MacKenzie LF, He S, Zeikus GJ, Withers SG. 1998. Identification of glu- 277 as the catalytic nucleophile of Thermoanaerobacterium saccharolyticum beta-xylosidase using electrospray MS. The Biochemical journal 335 ( Pt 2): 449-55

Vocadlo DJ, Wicki J, Rupitz K, Withers SG. 2002. A case for reverse protonation: identification of Glu160 as an acid/base catalyst in Thermoanaerobacterium saccharolyticum beta-xylosidase and detailed kinetic analysis of a site- directed mutant. Biochemistry 41 : 9736-46

Vu B, Chen M, Crawford RJ, Ivanova EP. 2009. Bacterial extracellular

polysaccharides involved in biofilm formation. Molecules 14: 2535-54

Vuong C, Kocianova S, Voyich JM, Yao YF, Fischer ER, et al. 2004. A crucial role for exopolysaccharide modification in bacterial biofilm formation, immune evasion, and virulence. J Biol Chem 279: 54881 -86

Wang S, Parsek MR, Wozniak DJ, Ma LZ. 2013. A spider web strategy of type IV pili- mediated migration to build a fibre-like Psl polysaccharide matrix in

Pseudomonas aeruginosa biofilms. Environ Microbiol 15: 2238-53

Wang X, Preston JF, 3rd, Romeo T. 2004. The pgaABCD locus of Escherichia col ' i promotes the synthesis of a polysaccharide adhesin required for biofilm formation. J BacterioH 86: 2724-34

Wasylnka JA, Hissen AH, Wan AN, Moore MM. 2005. Intracellular and extracellular growth of Aspergillus fumigatus. Medical mycology 43 Suppl : S27-30 Wasylnka JA, Moore MM. 2000. Adhesion of Aspergillus species to extracellular matrix proteins: evidence for involvement of negatively charged

carbohydrates on the conidial surface. Infection and immunity 68: 3377-84

Wasylnka JA, Moore MM. 2002. Uptake of Aspergillus fumigatus Conidia by

phagocytic and nonphagocytic cells in vitro: quantitation using strains expressing green fluorescent protein. Infection and immunity 70: 3156-63

Wasylnka JA, Moore MM. 2003. Aspergillus fumigatus conidia survive and germinate in acidic organelles of A549 epithelial cells. Journal of cell science 16: 1579- 87

Wiehlmann L, Wagner G, Cramer N, Siebert B, Gudowius P, et al. 2007. Population structure of Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the United States of America 104: 8101 -6

Wierenga RK. 2001. The TIM-barrel fold: a versatile framework for efficient enzymes.

FEBS letters 492: 193-8

Wolcott R, Costerton JW, Raoult D, Cutler SJ. 2013. The polymicrobial nature of biofilm infection. Clinical microbiology and infection : the official publication of the European Society of Clinical Microbiology and Infectious Diseases 19: 107-12

Wolfgang MC, Kulasekara BR, Liang X, Boyd D, Wu K, et al. 2003. Conservation of genome content and virulence determinants among clinical and

environmental isolates of Pseudomonas aeruginosa. Proc Natl Acad Sci U S

A 100: 8484-9

Yakandawala N, Gawande PV, LoVetri K, Cardona ST, Romeo T, et al. 20 .

Characterization of the poly-beta-1 ,6-N-acetylglucosamine polysaccharide component of Burkholderia biofilms. Appl Environ Microbiol 77: 8303-9 Zegans ME, Wozniak D, Griffin E, Toutain-Kidd CM, Hammond JH, et al. 2012.

Pseudomonas aeruginosa exopolysaccharide Psl promotes resistance to the biofilm inhibitor polysorbate 80. Antimicrobial agents and chemotherapy 56: 41 12-22

Zhao K, Tseng BS, Beckerman B, Jin F, Gibiansky ML, et al. 2013. Psl trails guide exploration and microcolony formation in Pseudomonas aeruginosa biofilms.

Nature 497: 388-91