Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PEPTIDES FOR INDUCING BACTERIOCIN SYNTHESIS AND METHODS TO IDENTIFY AND/OR SELECT AND/OR OPTIMIZE THE SAME
Document Type and Number:
WIPO Patent Application WO/2020/104662
Kind Code:
A1
Abstract:
Described herein is a peptide or peptidomimetic with a length of at least 6 residues comprising, consisting essentially of, or consisting of the sequence motif Xaa1-Trp-Xaa2-Xaa3-Xaa4-Xaa5 (SEQ ID NO:1), wherein: Xaa1 represents an aromatic residue (Phe, Tyr, Trp, His), Cys or Ser; Xaa2, Xaa3 and Xaa4 represent any residue; and Xaa5 represents Gly, lie or Val.

Inventors:
MIGNOLET JOHANN (BE)
HOLS PASCAL (BE)
LEDESMA GARCÍA LAURA (BE)
Application Number:
PCT/EP2019/082242
Publication Date:
May 28, 2020
Filing Date:
November 22, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SYNGULON S A (BE)
UNIV CATHOLIQUE LOUVAIN (BE)
International Classes:
C07K14/195; C07K7/06; C12P21/02
Domestic Patent References:
WO2015024855A12015-02-26
Foreign References:
US7662592B22010-02-16
US7662592B22010-02-16
US9333227B22016-05-10
Other References:
EDWARD A. SVETOCH ET AL: "Inducer Bacteria, Unique Signal Peptides, and Low-Nutrient Media Stimulate in Vitro Bacteriocin Production by Lactobacillus Spp. and Enterococcus Spp. Strains", JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY, vol. 58, no. 10, 26 May 2010 (2010-05-26), pages 6033 - 6038, XP055000529, ISSN: 0021-8561, DOI: 10.1021/jf902802z
DATABASE UniProt [online] 1 May 2000 (2000-05-01), "SubName: Full=ORF10S {ECO:0000313|EMBL:BAA85070.1}; Flags: Fragment;", XP002790693, retrieved from EBI accession no. UNIPROT:Q9S0U6 Database accession no. Q9S0U6
DATABASE UniProt [online] 30 August 2017 (2017-08-30), "SubName: Full=PH domain-containing protein {ECO:0000313|EMBL:SMQ79867.1};", XP002790691, retrieved from EBI accession no. UNIPROT:A0A1Y6FVS5 Database accession no. A0A1Y6FVS5
DATABASE UniProt [online] 18 July 2018 (2018-07-18), "SubName: Full=TIGR03758 family integrating conjugative element protein {ECO:0000313|EMBL:POV42023.1};", XP002790692, retrieved from EBI accession no. UNIPROT:A0A2S4STJ7 Database accession no. A0A2S4STJ7
DATABASE UniProt [online] 1 April 2015 (2015-04-01), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:KIL79283.1};", XP002790700, retrieved from EBI accession no. UNIPROT:A0A0C2YCN2 Database accession no. A0A0C2YCN2
JOHANN MIGNOLET ET AL: "Circuitry Rewiring Directly Couples Competence to Predation in the Gut Dweller Streptococcus salivarius", CELL REPORTS, vol. 22, no. 7, 1 February 2018 (2018-02-01), US, pages 1627 - 1638, XP055581149, ISSN: 2211-1247, DOI: 10.1016/j.celrep.2018.01.055
FRANZ C M ET AL: "Simple method to identify bacteriocin induction peptides and to auto-induce bacteriocin production at low cell density", FEMS MICROBIOLOGY LETTERS, WILEY-BLACKWELL PUBLISHING LTD, GB, vol. 186, no. 2, 5 May 2000 (2000-05-05), pages 181 - 185, XP027360381, ISSN: 0378-1097, [retrieved on 20000505]
DATABASE UniProt [online] 12 April 2017 (2017-04-12), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:SDC97056.1};", XP002790694, retrieved from EBI accession no. UNIPROT:A0A1G6QZH3 Database accession no. A0A1G6QZH3
DATABASE UniProt [online] 15 March 2017 (2017-03-15), "RecName: Full=Putative fluoride ion transporter CrcB {ECO:0000256|HAMAP-Rule:MF_00454};", XP002790695, retrieved from EBI accession no. UNIPROT:A0A1M5S5D2 Database accession no. A0A1M5S5D2
DATABASE UniProt [online] 7 November 2018 (2018-11-07), "SubName: Full=DUF805 domain-containing protein {ECO:0000313|EMBL:AXS39116.1};", XP002790696, retrieved from EBI accession no. UNIPROT:A0A346QY07 Database accession no. A0A346QY07
DATABASE UniProt [online] 14 May 2014 (2014-05-14), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:GAI68768.1}; Flags: Fragment;", XP002790697, retrieved from EBI accession no. UNIPROT:X1QKI4 Database accession no. X1QKI4
DATABASE UniProt [online] 24 July 2013 (2013-07-24), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:CDC57668.1};", XP002790698, retrieved from EBI accession no. UNIPROT:R6S903 Database accession no. R6S903
DATABASE UniProt [online] 10 October 2018 (2018-10-10), "RecName: Full=3',5'-cyclic adenosine monophosphate phosphodiesterase CpdA {ECO:0000256|HAMAP-Rule:MF_00905}; Short=3',5'-cyclic AMP phosphodiesterase {ECO:0000256|HAMAP-Rule:MF_00905}; Short=cAMP phosphodiesterase {ECO:0000256|HAMAP-Rule:MF_00905}; EC=3.1.4.53 {ECO:0000256|HAMAP-Rule:MF_00905};", XP002790699, retrieved from EBI accession no. UNIPROT:A0A328X1S3 Database accession no. A0A328X1S3
DATABASE UniProt [online] 27 June 2006 (2006-06-27), "SubName: Full=Cytochrome C oxidase subunit IV {ECO:0000313|EMBL:ABF65019.1};", XP002790701, retrieved from EBI accession no. UNIPROT:Q1GE97 Database accession no. Q1GE97
DATABASE UniProt [online] 31 January 2018 (2018-01-31), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:PIE05801.1}; Flags: Fragment;", XP002790702, retrieved from EBI accession no. UNIPROT:A0A2G6I3R1 Database accession no. A0A2G6I3R1
DATABASE UniProt [online] 25 October 2017 (2017-10-25), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:ASV17796.1};", XP002790703, retrieved from EBI accession no. UNIPROT:A0A223U4A6 Database accession no. A0A223U4A6
DATABASE UniProt [online] 15 March 2017 (2017-03-15), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:OJU66760.1};", XP002790704, retrieved from EBI accession no. UNIPROT:A0A1M3AIY8 Database accession no. A0A1M3AIY8
DATABASE UniProt [online] 20 December 2017 (2017-12-20), "SubName: Full=Tellurite resistance protein TehA-like permease {ECO:0000313|EMBL:PFG32060.1};", XP002790705, retrieved from EBI accession no. UNIPROT:A0A2A9E1J4 Database accession no. A0A2A9E1J4
DATABASE UniProt [online] 7 November 2018 (2018-11-07), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:HAZ30760.1}; Flags: Fragment;", XP002790706, retrieved from EBI accession no. UNIPROT:A0A351JWU0 Database accession no. A0A351JWU0
DATABASE UniProt [online] 4 March 2015 (2015-03-04), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:JAD30899.1};", XP002790707, retrieved from EBI accession no. UNIPROT:A0A0A8YUT1 Database accession no. A0A0A8YUT1
DATABASE UniProt [online] 29 May 2013 (2013-05-29), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:CCO29484.1};", XP002790708, retrieved from EBI accession no. UNIPROT:M5BQK2 Database accession no. M5BQK2
DATABASE UniProt [online] 18 January 2017 (2017-01-18), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:ODN65441.1};", XP002790709, retrieved from EBI accession no. UNIPROT:A0A1E3GPU9 Database accession no. A0A1E3GPU9
DATABASE UniProt [online] 7 September 2016 (2016-09-07), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:OAG75443.1};", XP002790710, retrieved from EBI accession no. UNIPROT:A0A177G535 Database accession no. A0A177G535
DATABASE UniProt [online] 31 January 2018 (2018-01-31), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:PIC05503.1};", XP002790711, retrieved from EBI accession no. UNIPROT:A0A2G5RS15 Database accession no. A0A2G5RS15
DATABASE UniProt [online] 14 May 2014 (2014-05-14), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:GAI10111.1};", XP002790712, retrieved from EBI accession no. UNIPROT:X1LWE7 Database accession no. X1LWE7
DATABASE UniProt [online] 25 April 2018 (2018-04-25), "SubName: Full=DoxX-like family protein {ECO:0000313|EMBL:PJZ25793.1};", XP002790713, retrieved from EBI accession no. UNIPROT:A0A2M9XDL6 Database accession no. A0A2M9XDL6
DATABASE UniProt [online] 31 January 2018 (2018-01-31), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:MAR57216.1};", XP002790714, retrieved from EBI accession no. UNIPROT:A0A2E0EP50 Database accession no. A0A2E0EP50
DATABASE UniProt [online] 15 March 2017 (2017-03-15), "SubName: Full=Uncharacterized protein {ECO:0000313|EMBL:SIO19486.1};", XP002790715, retrieved from EBI accession no. UNIPROT:A0A1N6HIC8 Database accession no. A0A1N6HIC8
DATABASE UniProt [online] 16 April 2014 (2014-04-16), "SubName: Full=D-alanine/D-serine/glycine permease {ECO:0000313|EMBL:EUK19008.1};", XP002790716, retrieved from EBI accession no. UNIPROT:W7DXS3 Database accession no. W7DXS3
ANONYMOUS: "DDE-type integrase/transposase/recombinase [Polyangium spumosum] - Protein - NCBI", 18 November 2019 (2019-11-18), XP055660871, Retrieved from the Internet [retrieved on 20200122]
JOHANN MIGNOLET ET AL: "Subtle selectivity in a pheromone sensor triumvirate desynchronizes competence and predation in a human gut commensal", ELIFE, 21 August 2019 (2019-08-21), pages 1 - 23, XP055660908, DOI: . https://doi.org/10.7554/eLife.47139
MERRIFIELD, J. AM. CHEM. SOC., vol. 85, no. 14, 1963, pages 2149 - 2154
ATHERTONSHEPPARD: "Solid Phase Peptide Synthesis: A practical Approach", 1999, IRL PRESS
"Peptidomimetics I and II, Topics in Heterocyclic Chemistry (Book 48", vol. XVI, 2017, SPRINGER, pages: 310
FONTAINE ET AL., INFECTION, GENETICS AND EVOLUTION, vol. 33, 2015, pages 343 - 360
ZACCARIA ET AL., PLOS ONE, vol. 11, no. 5, 2016, pages e0153571
WANG ET AL., NUCLEIC ACIDS RES., vol. 44, 2016, pages D1087 - D1093
COTTER, P.D. ET AL.: "Bacteriocins- a viable alternative to antibiotics", NATURE REVIEWS MICROBIOLOGY, vol. 11, no. 2, pages 95 - 105, XP055306514, DOI: 10.1038/nrmicro2937
ADETUNJIOLAOYE, MALAYSIAN JOURNAL OF MICROBIOLOGY, vol. 9, 2013, pages 130 - 13
SHEKHROY, BMC MICROBIOLOGY, vol. 12, 2012, pages 132
SHALANISRIVASTAVA, THE INTERNET JOURNAL OF MICROBIOLOGY, vol. 5, no. 2, 2008
ZUBER, P ET AL.: "Biochemistry, Physiology, and Molecular Genetics", 1993, AMERICAN SOCIETY FOR MICROBIOLOGY, article "Peptide Antibiotics. In Bacillus subtilis and Other Gram-Positive Bacteria", pages: 897 - 916
SHENIN ET AL., ANTIBIOT KHIMIOTER, vol. 50, 1995, pages 3 - 7
WANG ET AL.: "Genome Mining Demonstrates the Widespread Occurrence of Gene Clusters Encoding Bacteriocins in Cyanobacteria", PLOS ONE, vol. 6, no. 7, 2011, pages e22384
MIGNOLET ET AL., CELL REP, vol. 22, 2018, pages 1627 - 1638
MOUNT D.: "Bioinformatics: Sequence and Genome Analysis", 2004, COLD SPRING HARBOR LABORATORY PRESS
HENIKOFFHENIKOFF, PNAS, vol. 89, 1992, pages 915 - 919
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 10
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, no. 17, 1997, pages 3389 - 3402
GIBSON ET AL.: "Creation of a Bacterial Cell Controlled by a Chemically Synthesized Genome", SCIENCE, vol. 329, 2010, pages 52 - 56, XP055082599, DOI: 10.1126/science.1190719
WRIGHT ET AL.: "Building-in biosafety for synthetic biology", MICROBIOLOGY, vol. 159, 2013, pages 1221 - 1235, XP055155454, DOI: 10.1099/mic.0.066308-0
FONTAINE, L.GOFFIN, P.DUBOUT, H.DELPLACE, B.BAULARD, A.LECAT-GUILLET, N.CHAMBELLON, E.GARDAN, R.HOLS, P.: "Mechanism of competence activation by the ComRS signalling system in streptococci", MOL MICROBIOL, vol. 87, 2013, pages 1113 - 1132
MIGNOLET, J.HOLDEN, S.BERGE, M.PANIS, G.EROGLU, E.THERAULAZ, L.MANLEY, S.VIOLLIER, P.H.: "Functional dichotomy and distinct nanoscale assemblies of a cell cycle-controlled bipolar zinc-finger regulato", ELIFE, vol. 5, 2016, pages e18647
FONTAINE ET AL., MOL MICROBIOL, vol. 87, 2010, pages 1113 - 1132
CHOPIN, A.CHOPIN, M.C.MOILLO-BATT, A.LANGELLA, P.: "Two plasmid-determined restriction and modification systems in", STREPTOCOCCUS LACTIS. PLASMID, vol. 11, 1984, pages 260 - 263, XP024868081, DOI: 10.1016/0147-619X(84)90033-7
FONTAINE, L.DANDOY, D.BOUTRY, C.DELPLACE, B.DE FRAHAN, M.H.FREMAUX, C.HORVATH, P.BOYAVAL, P.HOLS, P.: "Development of a versatile procedure based on natural transformation for marker-free targeted genetic modification in", STREPTOCOCCUS THERMOPHILUS. APPL ENVIRON MICROBIOL, vol. 76, 2010, pages 7870 - 7877, XP055011495, DOI: 10.1128/AEM.01671-10
HAUSTENNE, L.BASTIN, G.HOLS, P.FONTAINE, L.: "Modeling of the ComRS Signaling Pathway Reveals the Limiting Factors Controlling Competence in Streptococcus thermophilus", FRONT MICROBIOL, vol. 6, 2015, pages 1413
LAMBERT, J.M.BONGERS, R.S.KLEEREBEZEM, M.: "Cre-lox-based system for multiple gene deletions and selectable-marker removal in Lactobacillus plantarum", APPL ENVIRON MICROBIOL, vol. 73, 2007, pages 1126 - 1135, XP002457430, DOI: 10.1128/AEM.01473-06
MIGNOLET, J.FONTAINE, L.KLEEREBEZEM, M.HOLS, P.: "Complete Genome Sequence of Streptococcus salivarius HSISS4, a Human Commensal Bacterium Highly Prevalent in the Digestive Tract", GENOME ANNOUNC, vol. 4, 2016, pages e01637 - 01615
MIGNOLET, J.FONTAINE, L.SASS, A.NANNAN, C.MAHILLON, J.COENYE, T.HOLS, P.: "Circuitry Rewiring Directly Couples Competence to Predation in the Gut Dweller", STREPTOCOCCUS SALIVARIUS. CELL REP, vol. 22, 2018, pages 1627 - 1638, XP055581149, DOI: 10.1016/j.celrep.2018.01.055
OVERKAMP, W.BEILHARZ, K.DETERT OUDE WEMER., SOLOPOVA, A.KARSENS, H.KOVACS, A.KOK, J.KUIPERS, O.P.VEENING, J.W.: "Benchmarking various green fluorescent protein variants in Bacillus subtilis, Streptococcus pneumoniae, and Lactococcus lactisfor live cell imaging", APPL ENVIRON MICROBIOL, vol. 79, 2013, pages 6481 - 6490
VAN DEN BOGERT, B.BOEKHORST, J.HERRMANN, R.SMID, E.J.ZOETENDAL, E.G.KLEEREBEZEM, M.: "Comparative genomics analysis of Streptococcus isolates from the human small intestine reveals their adaptation to a highly dynamic ecosystem", PLOS ONE, vol. 8, 2014, pages e83418
YU, J.SUN, Z.LIU, W.XI, X.SONG, Y.XU, H.LV, Q.BAO, Q.MENGHE, B.SUN, T.: "Multilocus sequence typing of Streptococcus thermophilus from naturally fermented dairy foods in China and Mongolia", BMC MICROBIOL, vol. 15, 2015, pages 236, XP021231042, DOI: 10.1186/s12866-015-0551-0
Attorney, Agent or Firm:
NEDERLANDSCH OCTROOIBUREAU (NL)
Download PDF:
Claims:
Claims

1. A peptide or peptidomimetic able to induce bacteriocin production in a microbial organism with a length of at least 6 residues comprising, consisting essentially of, or consisting of the sequence motif Xaai-Trp-Xaa2-Xaa3-Xaa4-Xaa5 (SEQ ID NO: 1 ), wherein the sequence motif is located at the C-terminus of said peptide or peptidomimetic or located so that it is followed by 1 , 2, 3, 4 or 5 additional C-terminal residues, and wherein:

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His), Cys or Ser;

- Xaa2, Xaa3 and Xaa4 represent any residue; and

- Xaas represents Gly, lie or Val.

2. A peptide or peptidomimetic according to claim 1 , wherein the sequence motif is located at the C-terminus of said peptide or peptidomimetic. 3. A peptide or peptidomimetic according to claim 1 or 2, wherein at least one or at least two or all of the following conditions are fulfilled:

- the sequence motif is preceded immediately by a Pro residue;

- Xaa1 represents an aromatic residue (Phe, Tyr, Trp, His); and

- Xaa5 represents Gly.

4. A peptide or peptidomimetic according to any one of claims 1 to 3, wherein the sequence motif is Phe-T rp-Leu-Val-Leu-Gly (SEQ ID NO: 2).

5. A peptide or peptidomimetic according to claim 1 , wherein the peptide or peptidomimetic comprises, consists essentially of, or consists of one of the following sequences:

- Ala-Phe-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 3)

- Thr-Trp-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 4)

- Pro-T yr-T rp-Leu-G ly-Leu-G ly (SEQ ID NO: 5)

- Pro-Trp-Trp-Val-Ser-Val-Gly (SEQ ID NO: 6)

- Pro-Phe-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 7)

- Pro-Tyr-Trp-Leu-Leu-lle-Gly (SEQ ID NO: 8)

- Pro-Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 9)

- Pro-Phe-Trp-Val-Val-Ala-Gly (SEQ ID NO: 10)

- Pro-Phe-Trp-Leu-Ser-Val-Gly (SEQ ID NO: 1 1 )

- Pro-Tyr-Trp-Leu-Asp-Met-Gly (SEQ ID NO: 12)

- Pro-Tyr-Trp-Val-Thr-Met-Gly (SEQ ID NO: 13)

- Pro-Tyr-Trp-Val-Val-Leu-Gly (SEQ ID NO: 14)

- Pro-Ser-Trp-Leu-Val-Val-Gly (SEQ ID NO: 15)

- Pro-His-Trp-lle-Thr-lle-Gly (SEQ ID NO: 16)

- Pro-His-Trp-Cys-Val-Leu-Gly (SEQ ID NO: 17) - Pro-Phe-Trp-Leu-Ala-Leu-Gly (SEQ ID NO: 18)

- Pro-Phe-Trp-Cys-Val-Leu-Gly (SEQ ID NO: 19)

- Phe-T rp-Val-Glu-Leu-Gly (SEQ ID NO: 20)

- Tyr-T rp-Ala-Thr-Thr-Gly-Leu (SEQ ID NO: 21 )

- T rp-T rp-Gly-Thr-Met-lle (SEQ ID NO: 22)

- Pro-Tyr-Trp-Leu-Cys-lle-lle (SEQ ID NO: 23)

- Thr-Cys-T rp-Val-Cys-lle-Val (SEQ ID NO: 24)

- Leu-Ala-Phe-T rp-Asp-Ser-Leu-Gly (SEQ ID NO: 749)

6. A peptide or peptidominnetic according to any one of claims 1 to 5, wherein the peptide or peptidomimetic has a maximum length of 30 amino acids, preferably has a maximum length of 20 amino acids, more preferably has a maximum length of 10 amino acids.

7. A peptide or peptidomimetic according to any one of claims 1 to 6, wherein the peptide or peptidomimetic is able to induce bacteriocin production in a microbial organism without concomitantly inducing competence.

8. A polypeptide able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising a peptide or peptidomimetic as defined in any one of claims 1-7, wherein the peptide or peptidomimetic can be released from the polypeptide by natural, chemical or biological peptide hydrolysis.

9. A culture medium comprising a peptide or peptidomimetic according to any one of claims 1 to 7 and/or a polypeptide according to claim 8.

10. A culture medium according to claim 9, further comprising a signaling molecule and/or a quenching molecule and/or an antimicrobial peptide and/or a bacteriocin.

1 1. A composition for inducing bacteriocin production in a microbial organism, comprising a peptide or peptidomimetic according to any one of claims 1 to 7 and/or polypeptide according to claim 8 and a solvent.

12. A composition according to claim 1 1 , further comprising a signaling molecule and/or a quenching molecule and/or an antimicrobial peptide and/or a bacteriocin.

13. A microbial organism able to produce and/or secrete a peptide or peptidomimetic according to any one of claims 1 to 7 and/or a polypeptide according to claim 8.

14. A method for inducing bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising administering a peptide or peptidomimetic according to any one of claims 1 to 7 and/or a polypeptide according to claim 8 to the microbial organism, and/or culturing the microbial organism in a culture medium according to any one of claims 9 or 10, and/or administering a composition according to any one of claims 1 1 or 12 to the microbial organism.

15. A method for inducing bacteriocin production in a first microbial organism, preferably without concomitantly inducing competence, comprising administering to the first microbial organism a second microbial organism according to claim 13 and/or co-culturing the first microbial organism with the second microbial organism according to claim 13.

16. A method according to claim 14 or 15, wherein the bacteriocin-producing microbial organism belongs to the microbiota.

17. A method according to any one of claims 14 to 16, wherein the bacteriocin-producing microbial organism also produces a desired product.

18. A method according to any one of claims 14 to 17, wherein the bacteriocin-producing microbial organism is a Gram-positive bacterium, for example a lactic acid bacterium, such as a Streptococcus species, such as Streptococcus salivarius.

19. A method according to any one of claims 14 to 18, wherein the bacteriocin production in the microbial organism neutralizes a second, undesired microbial organism.

20. A method according to any one of claims 14 to 19, wherein the undesired microbial organism is a pathogenic microbial organism or a contaminant.

21. A method for identifying, selecting and/or optimizing peptide ligands of peptide-responsive transcriptional regulators, comprising the following:

- generating a library of randomized genes, wherein said genes are operably linked to an inducible promoter, inducible by an inducer molecule;

- transforming the library into a microbial organism which comprises a nucleic acid encoding a selectable marker conferring resistance to a selection agent, operably linked to a promoter which is controlled by the peptide-responsive transcriptional regulator; and

- selecting or enriching positive clones by growing the microbial organism in the presence of the inducer molecule and the selection agent.

22. A method according to claim 21 , wherein the nucleotide sequences of the randomized genes comprise both fixed-sequence codons and degenerate codons.

Description:
Peptides for inducing bacteriocin synthesis and methods to identify and/or select and/or optimize the same

Field

Aspects herein relate generally to peptides and peptide-responsive regulators. More particularly, some aspects herein relate to peptides, culture media, compositions, methods and uses for inducing bacteriocin production in a microbial organism. Some other aspects herein relate to a method for identifying, selecting and/or optimizing peptide ligands of peptide-responsive regulators.

Background

Populations of microbial organisms are involved in maintaining the health and metabolic functions of multicellular organisms, for example as the microbiota associated with the gut and skin of humans, or the roots of the plants. In addition, populations of microbial organisms are used for various industrial processes. Accordingly, tuning populations of microbial organisms, for example to reduce or eliminate or neutralize undesired microbial organisms, can be useful for maintaining robustness and consistency of industrial processes and maintaining the health of tissues that comprise microbial organisms. For example, WO 2015/024855 describes systems, methods and microbial cells for the controlled growth of microorganisms.

Extensive and widespread use of antimicrobial drugs to reduce or eliminate or neutralize undesired microbial organisms has led to the emergence of resistant strains of microbial organisms. These microbial organisms are no longer susceptible to the currently available antimicrobial drugs. Bacteriocins are proteinaceous or peptidic toxins produced by microbial organisms, typically to inhibit the growth of similar or closely related strain(s). Bacteriocins are able to overcome at least some of the drawbacks associated with antimicrobials, as they are still active against antimicrobial- resistant microbial organisms.

It is well known that regulation of bacteriocin production is tightly coupled to regulation of competence. Competence has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to a physiological state enabling bacteria to bind and take up high-molecular-weight exogenous DNA (transformation). Concomitant induction of competence upon inducing bacteriocin production may be disadvantageous, as it places a fitness burden on the microbial population, and increases the likelihood of unwanted genetic changes occurring in the microbial organisms in the population. For example, competence may increase the spread of antibiotic resistance genes and/or bacteriocin immunity genes.

Accordingly, to expand the currently available arsenal of compounds that can be used to tune populations of microbial organisms, there is a need for compounds which can activate bacteriocin production in microbial cells, preferably without concomitantly inducing competence to avoid the disadvantages thereof as indicated above. Summary

In a first aspect, there is provided a peptide or peptidomimetic with a length of at least 6 residues comprising, consisting essentially of, or consisting of the sequence motif Xaai-Trp-Xaa2- Xaa3-Xaa4-Xaa5 (SEQ ID NO: 1 ), wherein:

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His), Cys or Ser;

- Xaaå, Xaa3 and Xaa4 represent any residue; and

- Xaas represents Gly, lie or Val.

In a preferred embodiment, there is provided a peptide or peptidomimetic able to induce bacteriocin production in a microbial organism with a length of at least 6 residues comprising, consisting essentially of, or consisting of the sequence motif Xaai-Trp-Xaa2-Xaa3-Xaa4-Xaas (SEQ ID NO: 1 ), wherein the sequence motif is located at the C-terminus of said peptide or peptidomimetic or located so that it is followed by 1 , 2, 3, 4 or 5 additional C-terminal residues, and wherein:

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His), Cys or Ser;

- Xaaå, Xaa3 and Xaa4 represent any residue; and

- Xaas represents Gly, lie or Val.

Preferably, the sequence motif is located at the C-terminus of said peptide or peptidomimetic. Preferably, the peptide or peptidomimetic fulfills at least one or at least two or all of the following conditions:

- the sequence motif is preceded immediately by a Pro residue

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His)

- Xaas represents Gly

The sequence motif is preferably Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2). Preferably, the peptide or peptidomimetic comprises, consists essentialy of, or consists of any of the following sequences:

- Ala-Phe-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 3)

- Thr-Trp-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 4)

- Pro-T yr-T rp-Leu-G ly-Leu-G ly (SEQ ID NO: 5)

- Pro-Trp-Trp-Val-Ser-Val-Gly (SEQ ID NO: 6)

- Pro-Phe-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 7)

- Pro-Tyr-Trp-Leu-Leu-lle-Gly (SEQ ID NO: 8)

- Pro-Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 9)

- Pro-Phe-Trp-Val-Val-Ala-Gly (SEQ ID NO: 10)

- Pro-Phe-Trp-Leu-Ser-Val-Gly (SEQ ID NO: 1 1 )

- Pro-Tyr-Trp-Leu-Asp-Met-Gly (SEQ ID NO: 12)

- Pro-Tyr-Trp-Val-Thr-Met-Gly (SEQ ID NO: 13)

- Pro-Tyr-Trp-Val-Val-Leu-Gly (SEQ ID NO: 14)

- Pro-Ser-Trp-Leu-Val-Val-Gly (SEQ ID NO: 15)

- Pro-His-Trp-lle-Thr-lle-Gly (SEQ ID NO: 16)

- Pro-His-Trp-Cys-Val-Leu-Gly (SEQ ID NO: 17)

- Pro-Phe-Trp-Leu-Ala-Leu-Gly (SEQ ID NO: 18) - Pro-Phe-Trp-Cys-Val-Leu-Gly (SEQ ID NO: 19)

- Phe-T rp-Val-Glu-Leu-Gly (SEQ ID NO: 20)

- Tyr-T rp-Ala-Thr-Thr-Gly-Leu (SEQ ID NO: 21

- T rp-T rp-Gly-Thr-Met-lle (SEQ ID NO: 22)

- Pro-Tyr-Trp-Leu-Cys-lle-lle (SEQ ID NO: 23)

- Thr-Cys-T rp-Val-Cys-lle-Val (SEQ ID NO: 24)

- Leu-Ala-Phe-T rp-Asp-Ser-Leu-Gly (SEQ ID NO: 749)

Preferably, the peptide or peptidominnetic is able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence.

In a preferred embodiment, the peptide or peptidomimetic has a maximum length of 30 amino acids. In another preferred embodiment, the peptide or peptidomimetic has a maximum length of 20 amino acids. In another preferred embodiment, the peptide or peptidomimetic has a maximum length of 10 amino acids.

In a second aspect, there is provided a polypeptide able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising a peptide or peptidomimetic as described in the first aspect, wherein the peptide or peptidomimetic can be released from the polypeptide by natural, chemical or biological peptide hydrolysis.

In a third aspect, there is provided a culture medium comprising a peptide or peptidomimetic as described in the first aspect. Also provided is a culture medium comprising a peptide or peptidomimetic according to to the first aspect and/or a polypeptide according to the second aspect. The culture medium may further comprise a signaling molecule and/or a quenching molecule and/or an antimicrobial peptide and/or a bacteriocin.

In a fourth aspect, there is provided a composition for inducing bacteriocin production in a microbial organism, comprising a peptide or peptidomimetic as described in the first aspect and a solvent. Also provided is a composition for inducing bacteriocin production in a microbial organism, comprising a peptide or peptidomimetic according to the first aspect and/or polypeptide according to the second aspect. The composition may further comprise a signaling molecule and/or a quenching molecule and/or an antimicrobial peptide and/or a bacteriocin.

In a fifth aspect, there is provided a microbial organism able to produce and/or secrete a peptide or peptidomimetic according to the first aspect and/or a polypeptide according to the second aspect.

In a sixth aspect, there is provided a method for inducing bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising administering a peptide or peptidomimetic according to the first aspect to the microbial organism, and/or culturing the microbial organism in a culture medium according to the third aspect, and/or administering a composition according to the fourth aspect to the microbial organism. The bacteriocin production in the microbial organism can neutralize a second, undesired microbial organism. According to some embodiments, the microbial organism is a Gram-positive bacterium, for example a lactic acid bacterium, such as a Streptococcus species, such as Streptococcus salivarius. Also provided is a method for inducing bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising administering a peptide or peptidomimetic according to the first apect and/or a polypeptide according to the second aspect to the microbial organism, and/or culturing the microbial organism in a culture medium according to the third aspect, and/or administering a composition according to the fourth aspect to the microbial organism. Also provided is a method for inducing bacteriocin production in a first microbial organism, preferably without concomitantly inducing competence, comprising administering to the first microbial organism a second microbial organism according to the fifth aspect and/or co-culturing the first microbial organism with the second microbial organism according to the fifth aspect. Preferably, the bacteriocin-producing microbial organism belongs to the microbiota. Preferably the bacteriocin- producing microbial organism also produces a desired product. Preferably, the bacteriocin- producing microbial organism is a Gram-positive bacterium, for example a lactic acid bacterium, such as a Streptococcus species, such as Streptococcus salivarius. Preferably the undesired microbial organism is a pathogenic microbial organism or a contaminant.

In a seventh aspect, there is provided a method for identifying, selecting, and/or optimizing peptide ligands of peptide-responsive transcriptional regulators, comprising the following steps:

- generating a library of randomized genes, wherein said genes are operably linked to an inducible promoter, inducible by an inducer molecule;

- transforming the library into a microbial organism which comprises a nucleic acid encoding a selectable marker conferring resistance to a selection agent, operably linked to a promoter which is controlled by the peptide-responsive transcriptional regulator; and

- selecting or enriching positive clones by growing the microbial organism in the presence of the inducer molecule and the selection agent.

Preferably, the nucleotide sequences of the randomized genes comprise both fixed-sequence codons and degenerate codons.

Description The present inventors have surprisingly developed a method, by which they have identified a group of peptide ligands inducing a unique peptide-responsive regulator of bacteriocin production. Particularly, as elaborated in the experimental part, the present inventors have surprisingly found that a peptide or peptidomimetic according to the invention can act as a ligand of the peptide- responsive regulators ScuR and/or SarF, thereby inducing the production of a group of class II type bacteriocins without necessarily inducing competence. Accordingly, the aspects and embodiments of the present invention as described herein solve at least some of the problems and needs as discussed herein. Peptide

In a first aspect, there is provided a peptide or peptidomimetic with a length of at least 6 residues comprising, consisting essentially of, or consisting of the sequence motif Xaai-Trp-Xaa2- Xaa3-Xaa4-Xaa5 (SEQ ID NO: 1 ), wherein:

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His), Cys or Ser;

- Xaaå, Xaa3 and Xaa4 represent any residue; and

- Xaas represents Gly, lie or Val. The sequence motif Xaai-Trp-Xaa2-Xaa3-Xaa4-Xaas may also be denoted as a sequence pattern or, simply, a sequence. A“sequence motif” or“sequence pattern” has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to an amino-acid (or nucleotide) sequence that recurs, with a certain degree of variation, on several sites of a molecule or several different molecules and has, or is conjectured to have or is assumed to be linked to, a biological significance or exhibit a biological activity as described herein. A biological significance or biological activity of the peptide of the invention is preferably to be able to induce bacteriocin production in a microbial organism, more preferably without concomitantly inducing competence.

In some embodiments, a peptide or peptidomimetic according to the invention may have a length of at least 6, at least 7, at least 8, at least 9, at least 10, at least 1 1 , at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19 or at least 20 residues, or at least 25 residues, or at least 30 residues, or at least 35 residues, or at least 40 residues, or at least 45 residues, or at least 50 residues. Accordingly, in other embodiments, a peptide or peptidomimetic according to the invention may have a minimal length of 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19 or 20 residues and a maximal length of 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20,

21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46,

47, 48, 49 or 50 residues.

As used herein, a“peptide” should be understood to encompass any peptide, regardless of whether it is generated through recombinant protein synthesis, purified from a native producer microbial organism or generated by means of chemical peptide synthesis. In some embodiments, the peptide comprises, consists essentially of, or consists of a non-naturally occurring amino acid sequence, a naturally occurring amino acid sequence, or a combination of these. Accordingly, there is also provided a vector comprising a nucleic acid encoding a peptide according to the invention. The term“vector” as used herein has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to a nucleic acid molecule, such as a plasmid, bacteriophage or animal virus, capable of introducing a heterologous nucleic acid sequence into a host cell. Further provided is a recombinant host cell comprising a vector according to the invention. The recombinant host cell can be a microbial host cell. Exemplary microbial host cells that can be used in this context are described later herein. A“synthetic peptide” has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to a peptide which is generated by means of chemical peptide synthesis. Accordingly, in some embodiments, a peptide may be a synthetic peptide. A synthetic peptide according to the invention may be prepared or synthesized using conventional methods that are well known in the art. For instance, peptides can be synthesized by commonly used solid-phase synthesis methods such as those that involve a tert-butyloxycarbonyl protecting group (t-BOC) or fluorenylmethyloxycarbonyl protecting group (FMOC) for protection of alpha- amino groups. In such methods, amino acids are added sequentially to a growing amino acid chain. Such methods are, for instance, described in Merrifield (1963), J. Am. Chem. Soc. 85(14):2149- 2154; and Atherton & Sheppard, Solid Phase Peptide Synthesis: A practical Approach (IRL Press, Oxford, UK, 1999), both of which are incorporated herein by reference. A peptide may further be modified by natural processes, such as post-translational processing, or by chemical modification techniques. Such modifications may be inserted in the peptide at any location, including in the backbone, amino acid side-chains and at the N- or C-terminus. Multiple types of modifications may occur in a single peptide, or a peptide may comprise several modifications of a single type. Illustrative but non-limiting examples of modifications are alkylation, acetylation, amidation, acylation, phosphorylation, methylation, demethylation, ADP-ribosylation, disulfide bond formation, ubiquitination, gamma-carboxylation, glycosylation, hydroxylation, iodination, oxidation, pegylation, succinylation and sulfation.

As used herein, a “peptidomimetic” is understood to encompass all compounds whose essential elements (pharmacophore) mimic a natural peptide and which retain the ability to interact with the biological target and produce the same biological effect. In some embodiments, the peptidomimetic comprises, consists essentially of, or consists of a non-naturally occurring amino acid sequence. In an embodiment, the peptidomimetic does not occur in nature and is considered to be man-made. They typically arise either from modification of an existing peptide, or by designing similar systems that mimic peptides, such as peptoids and b-peptides. Structures and synthesis of peptidomimetics are for instance described in William D. Lubell (ed.), Peptidomimetics I and II, Topics in Heterocyclic Chemistry (Book 48), Springer 1st ed. 2017, XVI, 310 p, which is incorporated herein by reference. Modification of an existing peptide may be the result of natural processes, such as post-translational processing, or chemical modification techniques. In general, a peptidomimetic typically refers to a compound containing non-peptidic structural elements. Typical but non-limiting examples of non-peptidic structural elements are modifications of one or more existing amino acids, conformational restraints, cyclization of the polypeptide, isosteric replacement or other modifications. In some embodiments, a peptidomimetic may contain one or more or all substitutions of an amino acid by the corresponding D-amino acid. As used herein,“corresponding D-amino acid” denotes the D-amino acid counterpart of an L-amino acid. In some embodiments, a peptidomimetic may also contain non-natural amino acids. As used herein, "non-natural amino acid" has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to non-genetically encoded amino acids, irrespective of whether they appear in nature or not. Non-natural amino acids that can be present in a peptidomimetic as described herein include: b-amino acids; p-acyl-L-phenylalanine; N-acetyl lysine; O-4-allyl-L-tyrosine; 2-aminoadipic acid; 3- aminoadipic acid; beta-alanine; 4-tert-butyl hydrogen 2-azidosuccinate; beta-aminopropionic acid;

2-aminobutyric acid; 4-aminobutyric acid; 2,4-diamino butyric acid; 6-aminocaproic acid; 2- aminoheptanoic acid; 2-aminoisobutyric acid; 3-aminoisobutyric acid; 2- aminopimelic acid; p- aminophenylalanine; 2,3-diaminobutyric acid; 2,3-diamino propionic acid; 2,2'-diaminopinnelic acid; p-amino-L-phenylalanine; p-azido-L- phenylalanine; D-allyl glycine; p-benzoyl-L-phenylalanine; 3- benzothienyl alanine p-bromophenylalanine; t-butylalanine; t-butylglycine; 4-chlorophenylalanine; cyclohexylalanine; cysteic acid; D-citrulline; thio-L-citrulline; desmosine; epsilon-amino hexanoic acid; N-ethylglycine; N-ethylasparagine; 2-fluorophenylalanine; 3-fluorophenylalanine; 4- fluorophenylalanine; homoarginine; homocysteine; homoserine; hydroxy lysine; alio-hydroxy lysine;

3-(3-methyl-4-nitrobenzyl)-L-histidine methyl ester; isodesmosine; allo-isoleucine; isopropyl-L- phenylalanine; 3- methyl-phenylalanine; N-methylglycine; N-methylisoleucine; 6-N-methyllysine; O- methyl-L-tyrosine; N-methylvaline; methionin sulfoxide; 2-napthylalanine; L-3-(2-naphthyl)alanine; isoserine; 3-phenylserine; norvaline; norleucine; 5,5,5-trifluoro-DL-leucine; ornithine; 3-chloro- tyrosine; N5-carbamoylornithine; penicillamine; phenylglycine; piperidinic acid; pyridylalanine; 1 ,2,3,4-tetrahydro-isoquinoline-3-carboxylic acid; beta-2-thienylalanine; y-carboxy-DL-glutamic acid; 4-fluoro-DL-glutamic acid; D-thyroxine; allo-threonine; 5-hydroxy-tryptophan; 5-methoxy- tryptophan; 5-fluoro-tryptophan; 3-fluoro-valine. In some embodiments, a natural amino acid of a peptide or peptidomimetic according to the invention is substituted by a corresponding non-natural amino acid. As used herein, a "corresponding non-natural amino acid" refers to a non-natural amino acid that is a derivative of the reference natural amino acid. For instance, a natural amino acid can be substituted by the corresponding b-amino acid, which have their amino group bonded to the b- carbon rather than the ocarbon. According to some embodiments, a peptide or peptidomimetic of the invention can further be provided with a targeting moiety. It is known that peptidomimetics are able to circumvent some of the disadvantages associated with natural peptides: e.g. stability against proteolysis (duration of activity) and poor bioavailability. Certain other properties, such as receptor selectivity or potency, often can be substantially improved.

As used herein, a“peptide or peptidomimetic” is understood to refer to a chain of a limited number of amino acids, generally between 2 and 50.

In a preferred embodiment, the sequence motif as described herein is located at the C- terminus of the peptide or peptidomimetic. In some other embodiments, the sequence motif as described herein may be located so that it is followed by 1 , 2, 3, 4 or 5 additional C-terminal residues.

In some embodiments, a peptide or peptidomimetic as described herein does not comprise, consist essentially of, or consist of the amino acid sequence TNVTKSWWVLAGCNQWASNCNCGNVKGLT (SEQ ID NO: 751 ) as disclosed in US 7,662,592.

In some embodiments, a peptide or peptidomimetic as described herein does not comprise, consist essentially of, or consist of the sequence VKGLT. In some embodiments, a peptide or peptidomimetic as described herein does not comprise a VKGLT sequence (SEQ ID NO: 752) at its C-terminus or at its C-terminus followed by 1 , 2, 3, 4 or 5 additional C-terminal residues.

In some embodiments, a peptide or peptidomimetic according to the invention may be comprised in a polypeptide. In certain embodiments, the polypeptide can be processed by natural, chemical or biological peptide hydrolysis, including proteolysis mediated by recombinant or natural (endo- and exo-) proteases, in order to generate a peptide as described herein. Accordingly, in some embodiments there is provided a polypeptide able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising a peptide sequence as described herein, wherein the peptide or peptidomimetic can be released from the polypeptide by natural, chemical or biological peptide hydrolysis. Such polypeptide can be referred to as a“pro-polypeptide”.

In some embodiments a“pro-polypeptide”, i.e. a polypeptide able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising a peptide sequence as described herein may be a precursor peptide or peptidomimetic. Accordingly, in some embodiments there is provided a precursor peptide or peptidomimetic able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising a peptide or peptidomimetic as described herein, wherein the peptide or peptidomimetic can be released from the protein by natural, chemical or biological peptide hydrolysis. In some embodiments, a precursor peptide or peptidomimetic may comprise, consist essentially of, or consist of any of the sequences of SEQ ID NO: 3-24 and 749 followed by a tail of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more additional amino acids, for example a triple leucine tail. In some embodiments, a precursor peptide or peptidomimetic may comprise, consist essentially of, or consist of the sequence LAFWDSLGLLL (SEQ ID NO: 750).

In some embodiments, a polypeptide (“pro-polypeptide’) or a precursor peptide or peptidomimetic as described herein may have a length of at least 7, at least 8, at least 9, at least 10, at least 11 , at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21 , at least 22, at least 23, at least 24, at least 25, at least 26, at least 27, at least 28, at least 29, at least 30, at least 31 , at least 32, at least 33, at least 34, at least 35, at least 36, at least 37, at least 38, at least 39, at least 40, at least 41 , at least 42, at least 43, at least 44, at least 45, at least 46, at least 47, at least 48, at least 49, or at least 50 residues. Accordingly, in other embodiments, a polypeptide (“pro-polypeptide’) or a precursor peptide or peptidomimetic as described herein may have a minimal length of 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15,

16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49 or 50 residues and a maximal length of 6, 7, 8, 9, 10, 11 , 12, 13, 14,

15, 16, 17, 18, 19, 20, 21 , 22, 23, 24, 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, 40,

41 , 42, 43, 44, 45, 46, 47, 48, 49 or 50 residues. In some embodiments, a polypeptide (“propolypeptide’) may have a length of at least 50, at least 75, at least 100, at least 125, at least 150, at least 175, at least 200, at least 250, at least 300, at least 350, at least 400, at least 450, at least 500, at least 600, at least 700, at least 800, at least 900, or at least 1000 residues. In some embodiments, processing occurs in or by a microbial cell. In other embodiments, processing occurs in an animal or human host. In certain embodiments, a peptide or peptidomimetic according to the invention may be comprised in a polypeptide which is a“pro-polypeptide”, further containing one or more other peptides with a specific activity separated by cleavage sites. As used herein,“cleavage site” has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to a polypeptide sequence that mediates the cleavage of a polypeptide (for example by hydrolysis of a peptide bond) to separate a single polypeptide into two or more discrete polypeptides. In some embodiments, a cleavage site comprises, consists essentially of, or consists of a consensus polypeptide sequence for cleavage by a“cleavage enzyme,” such as a peptidase. In some embodiments, the cleavage enzyme is a wild-type, a variant, or a synthetic cleavage enzyme, for example a wild-type, variant, or synthetic endopeptidase. In some embodiments, the cleavage sites are for a cleavage enzyme selected from the group consisting of: Arg-C proteinase, Asp-N endopeptidase, BNPS-Skatole, Caspase 1 , Caspase 2, Caspase 3, Caspase 4, Caspase 5, Caspase 6, Caspase 7, Caspase 8, Caspase 9, Caspase 10, Chymotrypsin-high specificity, Clostripain (Clostridiopeptidase B), CNBr, Enterokinase, Factor Xa, Formic acid, Glutamyl endopeptidase, GranzymeB, Hydroxylamine, lodosobenzoic acid, LysC, Neutrophil elastase, NTCB (2-nitro-5-thiocyanobenzoic acid), Pepsin (pH 1.3), Pepsin (pH>2), Proline-endopeptidase, Proteinase K, Staphylococcal peptidase I, Thermolysin, Thrombin, or Trypsin. The one or more other peptides with a specific activity can be signal molecules and/or quenching molecules and/or antimicrobial peptides and/or bacteriocins as described herein. Upon processing, such pro-polypeptide releases the peptide or peptidomimetic according to the invention as well as the one or more other peptides with a specific activity. A possible advantage of a peptide or peptidomimetic according to the invention when it is comprised in a precursor peptide or peptidomimetic or in a polypeptide and can be released by natural, chemical or biological peptide hydrolysis, is that the released peptides are able to to induce bacteriocins while precursor peptide or peptidomimetic or the full protein is not. Therefore, this constitutes an inducible or switchable system. In some embodiments, the inducible/switchable system may comprise, consist essentially of, or consist of a protease released in the environment (culture medium, microbiota, ...). In some embodiments, the inducible/switchable system may comprise, consist essentially of, or consist of a sensor protein directly or indirectly coupled to the activation of a transcriptional regulator. Activation of the transcriptional regulator would result in the net production/secretion of a protease that will cleave the precursor and release the active part of the inducibles peptide or peptidomimetic. The sensor may be capable to perceive stimuli from physico-chemical (temperature, pH, ...) changes in the culture condition or from the addition of biological or chemical compounds (preferentially sugar, peptides or peptidomimetics). Inducible promoters as described herein, such as xylose-, arabinose- , nickel-, nisin-, IPTG-, and pheromone-inducible promoters, may also be used. In some specific cases, the active peptide or peptidomimetic released might be the stimulus by itself, creating a positive feedback loop that promotes a rapid and sharp accumulation of the peptide or peptidomimetic in the environment (culture medium, microbiota,...). In a preferred embodiment, a peptide or peptidomimetic as described herein fulfills at least one or at least two or all of the following conditions:

- the sequence motif is preceded immediately by a Pro residue;

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His);

- Xaas represents Gly.

In some embodiments, if all of the above-defined conditions are fulfilled, a peptide or peptidomimetic according to the invention may thus comprise, consist essentially of, or consist of the sequence motif Xaai-Trp-Xaa2-Xaa3-Xaa4-Gly (SEQ ID NO: 25), wherein:

- Xaai represents an aromatic residue (Phe, Tyr, Trp, His); and

- Xaaå, Xaa3 and Xaa4 represent any residue.

In some embodiments, Xaa å , Xaa3 and Xaa 4 may be hydrophobic residues.

Sequence motifs may be represented by a consensus sequence: a single sequence consisting of the most commonly encountered residues at each site. Hence, a peptide or peptidomimetic according to an embodiment of the invention may comprise, consist essentially of, or consist of a sequence motif having the consensus sequence Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2). Thus, in some embodiments, a peptide or peptidomimetic according to the invention may comprise, consist essentially of, or consist of the sequence motif Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2). In some embodiments, the sequence motif Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2) may contain amino acid substitutions at 1 , 2, 3, 4 or up to 5 positions. Amino acid substitutions can be conservative amino acid substitutions, as described herein. Examples of suitable amino acid substitutions in this context include the substitution of Phe for Tyr, Trp or His, and/or the substitution of Gly for Val or lie. In some embodiments, the sequence motif Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2) may contain amino acid substitutions at 1 , 2, 3, 4 or up to 5 positions, wherein Trp is not substituted. In some embodiments, the sequence motif Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2) contains amino acid substitutions at 1 , 2, 3, 4 or up to 5 positions, wherein Trp is not substituted and Gly is substituted by Val or lie. In some embodiments, the sequence motif Phe-Trp-Leu-Val- Leu-Gly (SEQ ID NO: 2) may contain amino acid substitutions at 1 , 2, 3 or up to 4 positions, wherein Trp and Gly are not substituted. In some embodiments, the sequence motif Phe-Trp-Leu-Val-Leu- Gly (SEQ ID NO: 2) may contain amino acid substitutions at 1 , 2, 3 or up to 4 positions, wherein Trp and Gly are not substituted and Phe is substituted by Tyr, Trp or His. In some embodiments, the sequence motif Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 2) may contain amino acid substitutions at 1 , 2, or up to 3 positions, wherein Trp, Gly and Phe are not substituted.

It will be appreciated that a consensus sequence can also be represented by a sequence logo. A sequence logo is a graphical representation of the consensus sequence, in which the size of a symbol is related to the frequency that a given nucleotide or amino acid occurs at a certain position. The more conserved the residue, the larger the symbol for that residue is drawn; the less conserved the residue, the smaller the symbol is drawn. Sequence logos can for example be generated using WebLogo, available at on the world wide web at weblogo.berkelv.edu. In some embodiments, the sequence motif Xaai-Trp-Xaa2-Xaa3-Xaa4-Xaas as described herein, can be represented by a sequence logo as depicted in FIG 2C.

According to a preferred embodiment, a peptide or peptidomimetic as described herein comprises, consists essentially of, or consists of one of the following sequences:

- Ala-Phe-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 3)

- Thr-T rp-T rp-Leu-l le-Leu-Gly (SEQ ID NO: 4)

- Pro-T yr-T rp-Leu-G ly-Leu-G ly (SEQ ID NO: 5)

- Pro-Trp-Trp-Val-Ser-Val-Gly (SEQ ID NO: 6)

- Pro-Phe-Trp-Leu-lle-Leu-Gly (SEQ ID NO: 7)

- Pro-Tyr-Trp-Leu-Leu-lle-Gly (SEQ ID NO: 8)

- Pro-Phe-Trp-Leu-Val-Leu-Gly (SEQ ID NO: 9)

- Pro-Phe-T rp-Val-Val-Ala-Gly (SEQ ID NO: 10)

- Pro-Phe-Trp-Leu-Ser-Val-Gly (SEQ ID NO: 1 1 )

- Pro-Tyr-Trp-Leu-Asp-Met-Gly (SEQ ID NO: 12)

- Pro-Tyr-Trp-Val-Thr-Met-Gly (SEQ ID NO: 13)

- Pro-Tyr-Trp-Val-Val-Leu-Gly (SEQ ID NO: 14)

- Pro-Ser-Trp-Leu-Val-Val-Gly (SEQ ID NO: 15)

- Pro-His-Trp-lle-Thr-lle-Gly (SEQ ID NO: 16)

- Pro-His-Trp-Cys-Val-Leu-Gly (SEQ ID NO: 17)

- Pro-Phe-Trp-Leu-Ala-Leu-Gly (SEQ ID NO: 18)

- Pro-Phe-Trp-Cys-Val-Leu-Gly (SEQ ID NO: 19)

- Phe-T rp-Val-Glu-Leu-Gly (SEQ ID NO: 20)

- Tyr-T rp-Ala-Thr-Thr-Gly-Leu (SEQ ID NO: 21

- T rp-T rp-Gly-Thr-Met-lle (SEQ ID NO: 22)

- Pro-Tyr-Trp-Leu-Cys-lle-lle (SEQ ID NO: 23)

- Thr-Cys-T rp-Val-Cys-lle-Val (SEQ ID NO: 24)

- Leu-Ala-Phe-T rp-Asp-Ser-Leu-Gly (SEQ ID NO: 749)

In some embodiments, there is also provided a peptide or peptidomimetic which comprises, consists essentially of, or consists of a sequence in which 1 , 2, 3, 4 or up to 5 residues are deleted, added or substituted compared to any of the sequences of SEQ ID NO’s: 3-24 and 749. Amino acid substitutions can be conservative amino acid substitutions, as described herein.

In a preferred embodiment, a peptide or peptidomimetic according to the invention is able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence.

Natural DNA transformation, or just “transformation”, refers to a lateral gene transfer mechanism during which bacteria take up naked DNA from their environment and stably integrate it in their genome. The physiological state during which bacteria become able to take up DNA is named competence. Although natural transformation drives genome plasticity and adaptability, competence induction may also be associated with several disadvantages. For example, competence induction is likely to cause deleterious effects in the chromosome of the recipient bacteria and negatively impact cell growth by imposing an important energy burden on the recipient cells (Fontaine et al. Infection, Genetics and Evolution 2015, 33:343-360). Competence is also associated with a suppression of basal metabolism, which may have consequences for the microbe's resilience to fluctuations in the environment, as competence is costly in terms of use of energy and protein translation (Zaccaria et al. Plos One 2016, 1 1 (5):e0153571 ). Also, competence induction may contribute to the unwanted spread of antibiotic resistance or bacteriocin immunity genes. The competence window is thus generally tightly regulated in response to species-specific environmental conditions or signaling oligopeptides called competence pheromones and limited to a proportion of the cell population.

In some embodiments, a peptide or peptidomimetic according to the invention is able to induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, by inducing a peptide-responsive regulator that regulates genes involved in the synthesis of bacteriocins. Within the context of this disclosure,“inducing bacteriocin production” may be assessed by a skilled person at the nucleic acid level and/or at the amino acid level using common known techniques. As soon as a bacteriocin is detectable using common known techniques, the peptide or peptidomimetic according to embodiments of the invention will be said to exhibit a biological activity. A bacteriocin may be detectable using common known techniques after 1 , 2, 3, 5, 10, 20, 30, 45, 60 or 90 minutes or also after 2, 3, 4, 5, 6, 8, 10, 12, 16, 20 or 24 hours, including intervals between any two of the listed values. In a preferred embodiment, the peptide-responsive regulator that regulates genes involved in the synthesis of bacteriocins does not regulate genes involved with competence, such as the central regulator of competence ComX (also denoted alternative sigma factor X, SigX or o x ), ComK, SigH (also denoted alternative sigma factor H or o H ) and TfoX (also denoted Sxy). As used herein, regulation includes both direct regulation as well as indirect regulation. Preferred peptide-responsive regulators regulate genes involved in the synthesis of bacteriocins but do not regulate genes involved with competence, such as ScuR and SarF.

A peptide or peptidomimetic as described herein is a ligand of a peptide-responsive regulator, preferably a peptide-responsive regulator that regulates genes involved in the synthesis of bacteriocins. Examples of suitable peptide-responsive regulators in this context are RRNPP type peptide-responsive regulators, such as ScuR and SarF.

Exemplary bacteriocins in this context are bacteriocins as described herein. Preferred bacteriocins in this context comprise, consist essentially of, or consist of class II bacteriocins, for example salivaricins. Culture medium

In a further aspect, there is provided a culture medium comprising a peptide or peptidomimetic as described herein, and/or a polypeptide as described herein. A culture medium can be either a liquid culture medium or a solid culture medium, such as an agar-based solid culture medium.

A preferred culture medium as described herein induces bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, when the microbial organism is cultured in the culture medium. Exemplary bacteriocins in this context are bacteriocins as described herein. Preferred bacteriocins in this context comprise, consist essentially of, or consist of class II bacteriocins, for example salivaricins.

Typically, a culture medium as described herein comprises a peptide or peptidomimetic as described herein in a concentration ranging from 1 nM (0.001 mM) to 10 mM. Thus, a suitable concentration may be at least 0.001 pM, at least 0.01 pM, at least 0.1 pM, at least 1 pM or at least 10 pM. Accordingly, in other embodiments, a suitable concentration may lie between a minimum of 0.001 pM, 0.01 pM, 0.1 pM, 1 pM or 10 pM and a maximum of 0.001 pM, 0.01 pM, 0.1 pM, 1 pM or 10 pM.

In some embodiments, a suitable concentration is a concentration which induces bacteriocin production in a microbial organism that is cultured in the culture medium. A skilled person knows how to determine such suitable concentrations, for example based on an assay as used in the experimental part.

In some embodiments, the culture medium further comprises a signal molecule and/or a quenching molecule and/or an antimicrobial peptide and/or a bacteriocin as described herein.

Composition

In a further aspect, there is provided a composition comprising a peptide or peptidomimetic as described herein, and/or a polypeptide as described herein, and a solvent.

Suitable solvents include any solvent or mixture of solvents in which a peptide or peptidomimetic or polypeptide as described herein can be dissolved at a suitable concentration. The number and types of ionic charges in the peptide determine its solubility in aqueous solutions. In general, the more charged residues the peptide possesses, the more soluble it is in aqueous solutions. In addition, peptides generally have more charges at pH 6-8 than at pH 2-6. It is for this reason that peptides are better dissolved at near neutral pH. Among the many exceptions to the rule are peptide sequences that are very hydrophobic and those that tend to aggregate. While the hydrophobicity of the sequence is the primary cause of aggregation, peptides can also aggregate or "gel" through extensive hydrogen bonding network. Non-limiting examples of solvents that can be used in the context of the invention are water, ethanol, ammoniumhydroxide, dimethylsulfoxide (DMSO), acetic acid, acetonitrile and dimethylformamide (DMF). Dissolution can be enhanced by sonication. A peptide or peptidomimetic or polypeptide according to the invention exhibits a number of activities that can be advantageously used in a wide range of applications, including therapeutic or agricultural applications, and applications in probiotics, cosmetology, cleaning of surfaces (including surfaces of chemically fragile medical devices), biotechnology, biofermentation processes, and food preservation. Provided therefore are compositions comprising a peptide or peptidomimetic or polypeptide as described herein or an acceptable salt thereof, and an acceptable carrier, diluent and/or excipient. Each of the acceptable salt, carrier, diluent and/or excipient can be a salt, carrier, diluent and/or excipient which is suitable for the intended use or application, for example pharmaceutical or agricultural.

A peptide or peptidomimetic or polypeptide according to the invention exhibits a number of activities that can be advantageously used in agricultural applications. Provided therefore are agricultural compositions comprising a peptide or peptidomimetic or polypeptide as described herein or an agriculturally acceptable salt thereof, and an agriculturally acceptable carrier, diluent and/or excipient.

Pharmaceutically and/or agriculturally acceptable salts include, but are not limited to, acid addition salts and base addition salts. As used herein, “pharmaceutically acceptable salt” of a peptide refers to a salt that retains the desired function of the peptide, and is suitable for administration to humans or animals. As used herein,“agriculturally acceptable salt” of a peptide refers to a salt that retains the desired function or activity of the peptide at least to some extent, and is suitable for use in the environment including animals, plants, water, air and/or soil. Within the context of the application,“at least to some extent” means that at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of the initial desired function or activity is retained. Methods for the preparation of salts of peptides are known in the art and generally involve mixing of the peptide with a pharmaceutically and/or agriculturally acceptable acid or base, for instance, by reacting the free acid or free base forms of the product with one or more equivalents of the appropriate acid or base in a solvent or medium in which the salt is insoluble, or in a solvent such as water, which is then removed by vacuum or by freeze-drying, or by exchanging the cations of an existing salt for another cation on a suitable ion exchange resin. Non-limiting examples of pharmaceutically and/or agriculturally acceptable acids and bases are organic and inorganic acids such as formic acid, acetic acid, propionic acid, lactic acid, glycolic acid, oxalic acid, pyruvic acid, succinic acid, maleic acid, malonic acid, trifluoroacetic acid, cinnamic acid, sulfuric acid, hydrochloric acid, hydrobromic acid, nitric acid, perchloric acid, phosphoric acid, and thiocyanic acid, which form ammonium salts with free amino groups of polypeptides, and bases that form carboxylate salts with free carboxylic groups of polypeptides, such as ethylamine, methylamine, dimethylamine, triethylamine, isopropylamine, diisopropylamine, and other mono-, di-and trialkylamines, and arylamines.

Preferred compositions, pharmaceutical compositions and agricultural compositions as described herein induce bacteriocin production in a microbial organism, preferably without concomitantly inducing competence. Thus, compositions, pharmaceutical compositions and agricultural compositions as described herein include compositions, pharmaceutical compositions and agricultural compositions for inducing bacteriocin production in a microbial organism, preferably without concomitantly inducing competence. Preferred bacteriocins in this context comprise, consist essentially of, or consist of class II bacteriocins, for example salivaricins.

Typically, compositions, pharmaceutical compositions and agricultural compositions as described herein comprise a peptide or peptidomimetic or polypeptide as earlier described herein at a concentration ranging from 0.001 to 10 mM. Thus, a suitable concentration may be at least 0.001 mM, at least 0.01 pM, at least 0.1 pM, at least 1 pM or at least 10 pM. Accordingly, in other embodiments, a suitable concentration may lie between a minimum of 0.001 pM, 0.01 pM, 0.1 pM, 1 pM or 10 pM and a maximum of 0.001 pM, 0.01 pM, 0.1 pM, 1 pM or 10 pM.

In some embodiments, a suitable concentration is a concentration which induces bacteriocin production in a microbial organism when the composition is administered to the microbial organism. A skilled person knows how to determine such suitable concentrations, for example based on an assay as used in the experimental part.

In some embodiments, the composition further comprises a signal molecule and/or a quenching molecule and/or an antimicrobial peptide and/or a bacteriocin.

Within the context of culture media and compositions and methods according to embodiments of the invention, a“signal molecule” as used herein has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to a secreted or released molecule that is capable of modulating, inducing, or inhibiting an activity or process in the cell that produced it, or in a different cell (a subject cell can be a microbial cell or a non-microbial cell, for example a cell of a multicellular organism such as an animal or plant).

In some embodiments, a signal molecule may be selected from the group consisting of: quorum sensing molecules, signal transduction receptor ligands, growth factors, hormones, and cytokines. A signal molecule as described herein can be wild-type, mutant, or synthetic. Examples of suitable signal molecules include quorum sensing peptides (also called pheromones). In some embodiments, a signal molecule comprises, consists essentially of, or consists of a signaling peptides, quorum sensing molecules (for example, quorum sensing peptides), signal transduction receptor ligands, growth factors, hormones, or cytokine. In some embodiments, a signal molecule comprises, consists essentially of, or consists of a combination of two or more of signaling peptides, quorum sensing molecules (for example, quorum sensing peptides), signal transduction receptor ligands, growth factors, hormones, and cytokines, which can include combinations of two or more of the same type of molecule (for example a combination of two signaling peptides or a combination of two receptor ligands), as well as combinations of two or more different kinds of molecules (e.g., a combination of a cytokine and a hormone). In some embodiments, the signal molecule stimulates, inhibits, increases, or decreases the production of bacteriocins and/or the growth rate of a subpopulation of a microbiota. In some embodiments, the signal molecule comprises, consists essentially of, or consists of a quorum sensing peptide, or a variant thereof as described herein.

Examples of quorum sensing peptides suitable for culture media and compositions according to embodiments of the invention include, but are not limited to, quorum sensing peptides such as the peptides shown in Table 1 below, including variants of these peptides, and combinations of two or more of any of these peptides.

In some embodiments, the quorum sensing peptides are naturally-occuring. In some embodiments, the quorum sensing peptide comprises, consists essentially of, or consists of a variant of a naturally-occurring quorum sensing peptide. In some embodiments, the quorum sensing peptide comprises, consists essentially of, or consists of a synthetic peptide. Information on quorum sensing peptides, including example sequences, can be found on the quorumpeps database, accessible on the world wide web at quorumpeps.uqent.be, which is hereby incorporated by reference in its entirety.

Table 1

Within the context of culture media and compositions and methods according to embodiments of the invention, a“quenching molecule” as used herein has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to (peptidic) quenching molecules that prevent the signal molecules to reach their cognate receptors by enzymatic activity such as proteolysis, addition of inactivating chemical groups or by competition with the signal molecule. Within the context of culture media and compositions and methods according to embodiments of the invention, an“antimicrobial peptide” as used herein has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to a class of peptides that confer innate immune activity to kill or arrest the growth of microbial organisms. Classically, antimicrobial peptides have been described as peptides produced by the innate immune systems of invertebrates and vertebrates. Thus, while bacteriocins have classically been referred to a class of microbial gene products that target microbial organisms, antimicrobial peptides have classically been referred to as a class of invertebrate and vertebrate gene products that target microbial organisms.

Examples of antimicrobial peptides suitable for peptides or peptidomimetics, culture media, compositions, methods and uses according to the invention are known in the art, and can be found, for example, at The Antimicrobial Peptide Database accessible on the world wide web at aps.unmc.edu/AP/main.php, which is incorporated herein by reference in its entirety. Over 1000 antimicrobial peptides and variants thereof have been identified and cataloged. The Antimicrobial Peptide Database is described in Wang et al. (2016), Nucleic Acids Res. 44:D1087-D1093, which is incorporated herein by reference in its entirety.

Over 1000 antimicrobial peptides and variants thereof have been identified and cataloged. The Antimicrobial Peptide Database is described in Wang et al. (2016), Nucleic Acids Res. 44(Database issue): D1087-D1093, which is incorporated herein by reference in its entirerty. Examples of antimicrobial peptides include bacteriocins, antibacterial, antiviral, anti-HIV, antifungal, antiparasitic and anticancer peptides, such as Dermaseptin-B2, Abaecin, Ct-AMPI, Andropin, Aurein 1.1 , Lactofericin B, and Heliomicin. Methods, culture media, and compositions of some embodiments comprise naturally occuring antimicrobial peptides, or a nucleic acid encoding the same, and/or nonnaturally occurring antimicrobial peptides, or a nucleic acid encoding the same. Methods, culture media, and compositions of some embodiments include antimicrobial peptides that comprise a mutation or variation in a naturally-occuring antimicrobial peptide, or a nucleic acid encoding the same. Methods, culture media, and compositions of some embodiments comprise antimicrobial peptides comprising, consisting essentially of, or consisting of non-naturally occuring peptide sequences, or nucleic acids encoding the same.

It is further contemplated that methods, culture media, and compositions of some embodiments herein can be in conjunction with naturally occurring antimicrobial peptides, variants of naturally occurring antimicrobial peptides, and/or synthetic antimicrobial peptides. As such, antimicrobial peptides of methods, culture media, and compositions of some embodiments can comprise, consist essentially of, or consist of naturally occurring antimicrobial peptides, variants of naturally occurring antimicrobial peptides, and/or synthetic antimicrobial peptides. In some embodiments, a variant antimicrobial peptide has at least 70%, 71 %, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to a reference antimicrobial peptide (for example Dermaseptin-B2, Abaecin, Ct-AMPI, Andropin, Aurein 1.1 , Lactoferricin B, or Heliomicin).

Microbial organism

In a further aspect, there is provided a microbial organism able to produce and/or secrete a peptide or peptidomimetic and/or a precursor peptide or peptidomimetic and/or a protein as described herein. A variety of bacterial species and strains can be used in accordance with embodiments herein, as explained in the section“General descriptions”. In some embodiments, the microbial organism is a Gram-positive bacterium, preferably a lactic acid bacterium. In some embodiments, the microbial organism is a Streptococcus sp., preferably Streptococcus salivarius. In some embodiments, the microbial organism is“generally recognized as safe” (GRAS). In some embodiments, the microbial organism is a commensal bacterium from the microbiota. Method and use

In a further aspect, there is provided a method for inducing bacteriocin production in a microbial organism, preferably without concomitantly inducing competence, comprising administering a peptide or peptidomimetic or polypeptide as described herein to the microbial organism, and/or culturing the microbial organism in a culture medium as described herein, and/or administering a composition as described herein to the microbial organism. In some embodiments of a method according to the invention, administering a peptide or peptidomimetic or polypeptide as described herein to the microbial organism and administering a composition as described herein to the microbial organism may optionally be followed by a culturing step. In some embodiments, a peptide or peptidomimetic or polypeptide or a composition as described herein can be administered to a microbial organism in a host, such as a patient having an infection or an imbalance in the microbiome.

Also provided is a use of a peptide or peptidomimetic or polypeptide as described herein, a culture medium as described herein, or a composition as described herein for inducing bacteriocin production in a microbial organism. The use can be without concomitantly inducing competence.

In a further aspect, there is provided a method for inducing bacteriocin production in a first microbial organism, preferably without concomitantly inducing competence, comprising administering to the first microbial organism a second microbial organism able to produce and/or secrete a peptide or peptidomimetic and/or a polypeptide as described herein and/or co-culturing the first microbial organism with the second microbial organism able to produce and/or secrete a peptide or peptidomimetic and/or a polypeptide as described herein.

Also provided is a use of a microbial organism able to produce and/or secrete a peptide or peptidomimetic and/or a polypeptide as described herein fo inducing bacteriocin production in a microbial organism. The use can be without concomitantly inducing competence.

In some embodiments of a method and/or a use as described herein, the bacteriocin production in the microbial organism neutralizes a second, undesired microbial organism.

As used herein,“neutralizes” and variations to this root term has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It includes any form of inhibition or arrest of microbial growth and/or division (bacteriostatic effect), as well as any cytotoxic or bactericidal effect (killing). Neutralization can be fully or partially. For example, the whole population or only a part of the targeted population may be growth-inhibited or killed. More particularly, partial neutralization may mean that at least 5%, at least 10%, at least 15%, at least

20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least

55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least

90%, at least 95%, or at least 99% of the microbial organisms is growth-inhibited or killed. A skilled person knows how to assess growth inhibition and killing, for example based on assays that are used in the experimental part.“Undesired” microbial organism as used herein refers to any microbial organism that is targeted for neutralization. For example, an undesired microbial organism may be a pathogenic microbial organism or a contaminant.

In some embodiments of a method and/or a use as described herein, the microbial organism that produces bacteriocin is a bacterium. In some embodiments, the microbial organism that produces bacteriocin is a Gram-negative bacterium. According to some embodiments, the microbial organism that produces bacteriocin is a Gram-positive bacterium, for example a lactic acid bacterium, such as a Streptococcus species, such as Streptococcus salivarius.

In the context of a method and/or a use according to the invention, the undesired microbial organism may be a pathogenic microbial organism, preferably a pathogenic microbial organism that is susceptible to neutralization by a bacteriocin as described herein. In some embodiments, the undesired microbial organism is a Gram-positive bacterium. In still some embodiments, the undesired microbial organism is a pathogenic Gram-positive bacteria. The pathogenic Grampositive bacteria can be selected from the group consisting of the following genera: Staphylococcus, Enterococcus, Streptococcus, Listeria, Bacillus, Brochothrix, Clostridium, Mycobacterium, Propionibacterium, or Corynebacterium. Accordingly, Gram-positive bacterial species may be Staphylococcus aureus, Enterococcus faecium, Streptococcus agalactiae, Streptococcus pneumoniae, Streptococcus pyogenes, Listeria monocytogenes, Bacillus cereus, Brochothrix thermosphacta, Staphylococcus epidermidis, Clostridium difficile, Clostridium perfringens, Mycobacterium tuberculosis, Propionibacterium acnes, and Corynebacterium diphteriae.

In some embodiments, a method and/or a use as described herein may be a method and/or a use wherein the microbial organism belongs to the microbiota. In other words, the microbial organism that produces bacteriocin is a microbial organism that belongs to the microbiota. In this context, a microbiota can be the microbiota of human, for example the microbiota of the skin and/or the gut and/or the mouth and/or the vagina, the microbiota of an animal, for example the microbiota of the skin and/or the gut and/or the mouth and/or the vagina, and/or the microbiota of a plant, for example the microbiota of the roots.

In other embodiments, a method and/or a use as described herein may be a method and/or a use wherein the microbial organism produces a desired product. In other words, the microbial organism that produces bacteriocin is a microbial organism that also produces a desired product. In this context, the microbial organism can exist in a number in commercially useful environments such as industrial cultures, fermenters, pharmaceutical, biological, and cosmetic manufacturing and in products, such as foods (for human and/or animals), drug products, and cosmetic products. Hence, in some embodiments, the undesired microbial organism may be a contaminant in commercially useful environments such as industrial cultures, fermenters, pharmaceutical, biological, and cosmetic manufacturing and in products, such as foods (for human and/or animals), drug products, and cosmetic products.

Exemplary bacteriocins in this context are bacteriocins as described herein. In the context of a method and/or a use according to the invention, the bacteriocin may be a class II bacteriocin, for example salivaricin. Method

In a further aspect, there is provided a method for identifying, selecting and/or optimizing peptide ligands of peptide-responsive transcriptional regulators, comprising the following:

- generating a library of randomized genes, wherein said genes are operably linked to an inducible promoter, inducible by an inducer molecule;

- transforming the library into a microbial organism which encodes a selectable marker conferring resistance to a selection agent, operably linked to a promoter which is controlled by the peptide-responsive transcriptional regulator; and

- selecting or enriching positive clones by growing the microbial organism in the presence of the inducer molecule and the selection agent.

In a preferred embodiment, there is provided a method for identifying, selecting and/or optimizing peptide ligands of peptide-responsive transcriptional regulators, comprising the following steps:

- generating a library of randomized genes, wherein said genes are operably linked to an inducible promoter, inducible by an inducer molecule;

- transforming the library into a microbial organism which encodes a selectable marker conferring resistance to a selection agent, operably linked to a promoter which is controlled by the peptide-responsive transcriptional regulator; and

- selecting or enriching positive clones by growing the microbial organism in the presence of the inducer molecule and the selection agent.

In a preferred embodiment, the peptide-responsive transcriptional regulator belongs to the RRNPP family. RRNPP proteins are named after the different sensors described: Rgg, Rap, NprR, PlcR, and PrgX. They are characterized at the structural level by tetratricopeptide repeat (TPR) domains, which are involved in the regulator/peptide interaction.

In a preferred embodiment, the peptide responsive regulator is a regulator of bacteriocin synthesis. The peptide responsive regulator of bacteriocin synthesis may be ScuR or SarF.

In a preferred embodiments, a method as described herein may be a method wherein the nucleotide sequences of the randomized genes comprise both fixed-sequence codons and degenerate codons. As used herein, degenerate codons include mixtures of nucleotides at one, two or three positions and are used for introducing random diversity in the genes during oligonucleotide synthesis. For example, the complete set of standard amino acids can be encoded using NNK or NNS codons, where N = A or T or G or C, K = G or T and S = C or G.

In some embodiments, the randomized genes are optionally further operably linked to a selectable marker. Accordingly, the transforming step of a method as described herein optionally involves a selection step for successful transformants by selecting or enriching transformants in the presence of the selectable marker. In some embodiments, the selectable marker encoded by the microbial organism is different from the selectable marker operably linked to the randomized genes. Exemplary selectable markers useful in some embodiments herein are antibiotic-resistance conferring genes such as cat (chloramphenicol acetyl transferase), erm (erythromycin ribosome methylation) and spec (spectinomycine resistance gene). Other suitable selectable markers may be bacteriocin immunity genes.

Inducible promoters are promoters which drive transcription only in the presence of a suitable inducer molecule. Inducible promoters useful in some embodiments herein are xylose-, arabinose- , nickel-, nisin-, IPTG-, and pheromone-inducible promoters. More exemplary promoters suitable for embodiments herein are given in Tables 2 and 3 below.

Table 2: Exemplary metal-sensitive promoters

Table 3: Exemplary Cell Signaling-Responsive Promoters

In some embodiments, the randomized genes may also be operably linked to a sequence which is homologous to a sequence of the microbial organism’s genome to allow recombination at a desired locus in the microbial organism’s genome. In some embodiments, the homologous sequence has a length of 50-2000 nucleotides, for example 100-1000 nucleotides, 200-800 nucleotides, or 500 nucleotides. In some embodiments of a method as described herein, the microbial organism that encodes a selectable marker is a bacterium. In some embodiments, the microbial organism that encodes a selectable marker is a Gram-negative bacterium. According to some embodiments, the microbial organism that encodes a selectable marker is a Gram-positive bacterium, for example a lactic acid bacterium, such as a Streptococcus species, such as Streptococcus salivarius.

Within the context of peptides or peptidomimetics, polypeptides, culture media, compositions, microbial organisms, and methods and uses according to embodiments of the invention, a “bacteriocin” as used herein has its customary and ordinary meaning as understood by one of skill in the art in view of this disclosure. It refers to proteinaceous toxins produced by bacteria to inhibit the growth of similar or closely related bacterial strain(s). A bacteriocin can neutralize at least one cell other than the individual host cell in which the polypeptide is made, including cells clonally related to the host cell and other microbial cells. Neutralization can be fully or partially as described herein. As used herein, bacteriocin also encompasses a cell-free or chemically synthesized version of such a polypeptide. A synthetic variant of a bacteriocin may be derived from the bacteriocin secreted by a host cell in any way as long as the synthetic variant still exhibits at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% of the activity of the corresponding bacteriocin secreted by a host cell.

Detailed descriptions of bacteriocins, including methods and compositions for using bacteriocins to control the growth of microbial cells can be found, for example, in U.S. Patent No. 9,333,227, which is hereby incorporated by reference.“Bacteriocin” is not limited by the origin of the polypeptide, and by way of example is contemplated to encompass any bacteriocin, such as naturally-occurring bacteriocins, synthetic bacteriocins, and variants and combinations thereof. A cell that expresses a particular "immunity modulator" is immune to the neutralizing effects of a particular bacteriocin or group of bacteriocins. As such, bacteriocins can neutralize a cell producing the bacteriocin and/or other microbial cells, so long as these cells are“susceptible”, i.e. do not produce an appropriate immunity modulator. As such, a bacteriocin can exert cytotoxic or growth-inhibiting effects on a plurality of other microbial organisms. In an embodiment, a bacteriocin is produced by the translational machinery (e.g. a ribosome, etc.) of a microbial cell. In another embodiment, a bacteriocin is chemically synthesized. Some bacteriocins can be derived from a polypeptide precursor. The polypeptide precursor can undergo cleavage (for example processing by a protease) to yield the polypeptide of the bacteriocin itself. As such, in some embodiments, a bacteriocin is produced from a precursor polypeptide. In some embodiments, a bacteriocin comprises, consists essentially of, or consists of a polypeptide that has undergone post - translational modifications, for example cleavage, or the addition of one or more functional groups.

Neutralizing activity of bacteriocins can include inhibition or arrest of microbial growth and/or division, or cytotoxicity. Some bacteriocins have cytotoxic activity (e.g. "bacteriocide" effects), and thus can kill microbial organisms, for example bacteria, yeast, algae, synthetic micoorganisms, and the like. Some bacteriocins can inhibit the growth and/or division of microbial organisms (e.g. "bacteriostatic" effects), for example bacteria, yeast, algae, synthetic microorganisms, and the like, for example by arresting the cell cycle.

A number of bacteriocins have been identified and characterized (see Table 4 and 5). Without being limited by any particular theory, exemplary bacteriocins can be classified as "class I" bacteriocins, which typically undergo post -translational modification, and "class II" bacteriocins, which are typically unmodified. Additionally, exemplary bacteriocins in each class can be categorized into various subgroups, as summarized in Cotter, P.D. et al. "Bacteriocins- a viable alternative to antibiotics" Nature Reviews Microbiology 11(2): 95-105, hereby incorporated by reference. Without being limited by any particular theory, bacteriocins can effect neutralization of a target microbial cell in a variety of ways. For example, a bacteriocin can permeabilize a cell wall, thus depolarizing the cell wall and interfering with respiration.

Table 4: Classification of Exemplary Bacteriocins

A number of bacteriocins can be used in accordance with embodiments herein. Exemplary bacteriocins are shown in Table 5. In some embodiments, at least one bacteriocin comprising, consisting essentially of, or consisting of a polypeptide sequence of Table 5 is provided. As shown in Table 5, some bacteriocins function as pairs of molecules. As such, it will be understood that unless explicity stated otherwise, when a functional "bacteriocin" or "providing a bacteriocin," or the like is discussed herein, functional bacteriocin pairs are included along with bacteriocins that function individually. With reference to Table 5, "organisms of origin" listed in parentheses indicate alternative names and/or strain information for organisms known to produce the indicated bacteriocin.

Embodiments herein also include peptides and proteins with identity to bacteriocins described in Table 5. The term "identity" is meant to include nucleic acid or protein sequence homology or three-dimensional homology. Several techniques exist to determine nucleic acid or polypeptide sequence homology and/or three-dimensional homology to polypeptides. These methods are routinely employed to discover the extent of identity that one sequence, domain, or model has to a target sequence, domain, or model. A vast range of functional bacteriocins can incorporate features of bacteriocins disclosed herein, thus providing for a vast degree of identity to the bacteriocins in Table 5. In some embodiments, a bacteriocin has at least 50% identity, for example, at least 50%, 51 %, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61 %, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71 %, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to any one of the polypeptides of Table 5.

While the bacteriocins in Table 5 are naturally-occurring, the skilled artisan will appreciate that variants of the bacteriocins of Table 5, naturally-occurring bacteriocins other than the bacteriocins of Table 5 or variants thereof, or synthetic bacteriocins can be used according to some embodiments herein. In some embodiments, such variants have enhanced or decreased levels of cytotoxic or growth inhibition activity on the same or a different microorganism or species of microorganism relative to the wild type protein. Several motifs have been recognized as characteristic of bacteriocins. For example, the motif YGXGV (SEQ ID NO: 236), wherein X is any amino acid residue, is an N-terminal consensus sequence characteristic of class lla bacteriocins. Accordingly, in some embodiments, a synthetic bacteriocin comprises an N-terminal sequence with at least 50% identity to SEQ ID NO: 236, for example at least 50%, 51 %, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61 %, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71 %, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81 %, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91 %, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 236. In some embodiments, a synthetic bacteriocin comprises a N-terminal sequence comprising SEQ ID NO: 236. Additionally, some class lib bacteriocins comprise a GXXXG motif (SEQ ID NO: 237) (X means any amino acid). Without being limited by any particular theory, it is believed that the GXXXG (SEQ ID NO: 237) motif can mediate association between helical proteins in the cell membrane, for example to facilitate bacteriocin-mediated neutralization through cell membrane interactions. As such, in some embodiments, the bacteriocin comprises a motif that facilitates interactions with the cell membrane. In some embodiments, the bacteriocin comprises a GXXXG (SEQ ID NO: 237) motif. Optionally, the bacteriocin comprising a GXXXG (SEQ ID NO: 237) motif can comprise a helical structure. In addition to structures described herein, "bacteriocin" as used herein also encompasses structures that have substantially the same effect on microbial cells as any of the bacteriocins explicitly provided herein.

It has been shown that fusion polypeptides comprising, consisting essentially of, or consisting of two or more bacteriocins or portions thereof can have neutralizing activity against a broader range of microbial organisms than either individual bacteriocin. For example, it has been shown that a hybrid bacteriocin, Ent35-MccV

(GKY Y GN G V SCNKKGC S VD W GRAIGIIGNN S AANL AT GG AAG WKS GGG AS GRD IAMAIGTLSGQFVAGGIGAAAGGVAGGAIYDYASTHKPNPAMSPSGLGGTIKQKP EGIPSE AWNYAAGRLCNWSPNNLSDVCL, SEQ ID NO: 238) displays antimicrobial activity against pathogenic Gram-positive and Gram-negative bacteria (Acuna et al. (2012), FEBS Open Bio, 2: 12-19). It is noted that that Ent35-MccV fusion bacteriocin comprises, from N -terminus to C-terminus, an N-terminal glycine, Enterocin CRL35, a linker comprising three glycines, and a C-terminal Microcin V. It is contemplated herein that bacteriocins can comprise fusions of two or more polypeptides having bacteriocin activity. In some embodiments, a fusion polypeptide of two or more bacteriocins is provided. In some embodiments, the two or more bacteriocins comprise, consist essentially of, or consist of polypeptides from Table 5, or modifications thereof. In some embodiments, the fusion polypeptide comprising of two or more bacteriocins has a broader spectrum of activity than either individual bacteriocin, for example having neutralizing activity against more microbial organisms, neutralizing activity under a broader range of environmental conditions, and/or a higher efficiency of neutralization activity. In some embodiments, a fusion of two or more bacteriocins is provided, for example two, three, four, five, six, seven, eight, nine, or ten bacteriocins. In some embodiments, two or more bacteriocin polypeptides are fused to each other via a covalent bond, for example a peptide linkage. In some embodiments, a linker is positioned between the two bacteriocin polypeptides. In some embodiments, the linker comprises, consists essentially of, or consists of one or more glycines, for example 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 glycines. In some embodiments, the linker is cleaved within the cell to produce the individual bacteriocins included in the fusion protein. In some embodiments, a bacteriocin as provided herein is modified to provide a desired spectrum of activity relative to the unmodified bacteriocin. For example, the modified bacteriocin may have enhanced or decreased activity against the same organisms as the unmodified bacteriocin. Alternatively, the modified bacteriocin may have enhanced activity against an organism against which the unmodified bacteriocin has less activity or no activity. Table 5: Exemplary Bacteriocins

For example, in some embodiments, an anti-fungal activity (such as anti- yeast activity) is desired. A number of bacteriocins with anti- fungal activity have been identified. For example, bacteriocins from Bacillus have been shown to have neutralizing activity against yeast strains (Adetunji and Olaoye (2013) Malaysian Journal of Microbiology 9: 130-13, hereby incorporated by reference), an Enterococcus faecalis peptide (WLPPAGLLGRCGRWFRPWLLWLQ SGAQY KWLGNLFGLGPK, SEQ ID NO: 727) has been shown to have neutralizing activity against Candida species {see Shekh and Roy (2012) BMC Microbiology 12: 132, hereby incorporated by reference in its entirety), and bacteriocins from Pseudomonas have been shown to have neutralizing activity against fungi such as Curvularia lunata, Fusarium species, Helminthosporium species, and Biopolaris species (Shalani and Srivastava (2008) The Internet Journal of Microbiology. Volume 5 Number 2, hereby incorporated by reference). By way of example, botrycidin AJ1316 (Zuber, P et al. (1993) Peptide Antibiotics. In Bacillus subtilis and Other Gram-Positive Bacteria: Biochemistry, Physiology, and Molecular Genetics ed Sonenshein et al., pp. 897-916, American Society for Microbiology, hereby incorporated by reference) and alirin Bl (Shenin et al. (1995) Antibiot Khimioter 50: 3-7, hereby incorporated by reference) from B. subtilis have been shown to have antifungal activities. As such, in some embodiments, for example embodiments in which neutralization of a fungal microbial organism is desired, a bacteriocin comprises at least one of botrycidin AJ 1316 or alirin B 1.

For example, in some embodiments, bacteriocin activity in a culture of cyanobacteria is desirable. In some embodiments, bacteriocins are provided to neutralize cyanobacteria. In some embodiments, bacteriocins are provided to neutralize invading microbial organisms typically found in a cyanobacteria culture environment. Clusters of conserved bacteriocin polypetides have been identified in a wide variety of cyanobacteria species. For example, at least 145 putative bacteriocin gene clusters have been identified in at least 43 cyanobacteria species, as reported in Wang et al. (201 1 ), Genome Mining Demonstrates the Widespread Occurrence of Gene Clusters Encoding Bacteriocins in Cyanobacteria. PLoS ONE 6(7): e22384, hereby incorporated by reference in its entirety. Exemplary cyanobacteria bacteriocins are shown in Table 5 as SEQ ID NO's 655, 657, 659, 661 , 663, 665, 667, 669, 671 , 673, 675, 677, 679, 681 , 683, and 685.

General descriptions

Unless stated otherwise, all technical and scientific terms used herein have the same meaning as customarily and ordinarily understood by a person of ordinary skill in the art to which this invention belongs, and read in view of this disclosure.

As used herein, the term "promoter" or "transcription regulatory sequence" refers to a nucleic acid fragment that functions to control the transcription of one or more coding sequences, and is located upstream with respect to the direction of transcription of the transcription initiation site of the coding sequence, and is structurally identified by the presence of a binding site for DNA- dependent RNA polymerase, transcription initiation sites and any other DNA sequences, including, but not limited to transcription factor binding sites, repressor and activator protein binding sites, and any other sequences of nucleotides known to one of skill in the art to act directly or indirectly to regulate the amount of transcription from the promoter. A "constitutive" promoter is a promoter that is active in most tissues under most physiological and developmental conditions. An "inducible" promoter is a promoter that is physiologically or developmentally regulated, e.g. by the application of a chemical inducer.

As used herein, a“regulator” or“transcriptional regulator” is a protein that controls the rate of transcription of genetic information from DNA to messenger RNA, by binding to a specific DNA sequence.

The term "selectable marker" is a term familiar to one of ordinary skill in the art and is used herein to describe any genetic entity which, when expressed, can be used to select for a cell or cells containing the selectable marker. Selectable markers may be dominant or recessive or bidirectional.

As used herein, the term "operably linked" refers to a linkage of polynucleotide elements in a functional relationship. A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. For instance, a transcription regulatory sequence is operably linked to a coding sequence if it affects the transcription of the coding sequence. Operably linked means that the DNA sequences being linked are typically contiguous and, where necessary to join two protein encoding regions, contiguous and in reading frame.

The terms "protein" or "polypeptide" are used interchangeably and refer to molecules consisting of a chain of amino acids, without reference to a specific mode of action, size, 3- dimensional structure or origin.

The term "gene" means a DNA fragment comprising a region (transcribed region), which is transcribed into an RNA molecule (e.g. an mRNA) in a cell, operably linked to suitable regulatory regions (e.g. a promoter). A gene will usually comprise several operably linked fragments, such as a promoter, a 5' leader sequence, a coding region and a 3'-nontranslated sequence (3'-end) e.g. comprising a polyadenylation- and/or transcription termination site.

"Expression of a gene" refers to the process wherein a DNA region which is operably linked to appropriate regulatory regions, particularly a promoter, is transcribed into an RNA, which is biologically active, i.e. which is capable of being translated into a biologically active protein or peptide.

In amino acid sequences as described herein, amino acids or“residues” are denoted by three-letter symbols. These three-letter symbols as well as the corresponding one-letter symbols are well known to the person skilled in the art and have the following meaning: A (Ala) is alanine, C (Cys) is cysteine, D (Asp) is aspartic acid, E (Glu) is glutamic acid, F (Phe) is phenylalanine, G (Gly) is glycine, H (His) is histidine, I (lie) is isoleucine, K (Lys) is lysine, L (Leu) is leucine, M (Met) is methionine, N (Asn) is asparagine, P (Pro) is proline, Q (Gin) is glutamine, R (Arg) is arginine, S (Ser) is serine, T (Thr) is threonine, V (Val) is valine, W (Trp) is tryptophan, Y (Tyr) is tyrosine. A residue may be any proteinogenic amino acid, but also any non-proteinogenic amino acid such as D-amino acids and modified amino acids formed by post-translational modifications, and also any non-natural amino acid, as described herein.

Sequence identity

The terms“homology”, “sequence identity” and the like are used interchangeably herein. Sequence identity is described herein as a relationship between two or more amino acid (polypeptide or protein) sequences or two or more nucleic acid (polynucleotide) sequences, as determined by comparing the sequences. In a preferred embodiment, sequence identity is calculated based on the full length of two given SEQ ID NO’s or on a part thereof. Part thereof preferably means at least 50%, 60%, 70%, 80%, 90%, or 100% of both SEQ ID NO’s. In the art, "identity" also refers to the degree of sequence relatedness between amino acid or nucleic acid sequences, as the case may be, as determined by the match between strings of such sequences. "Similarity" between two amino acid sequences is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide. "Identity" and "similarity" can be readily calculated by known methods, including but not limited to those described in Bioinformatics and the Cell: Modern Computational Approaches in Genomics, Proteomics and transcriptomics, Xia X., Springer International Publishing, New York, 2018; and Bioinformatics: Sequence and Genome Analysis, Mount D., Cold Spring Harbor Laboratory Press, New York, 2004.

“Sequence identity” and“sequence similarity” can be determined by alignment of two peptide or two nucleotide sequences using global or local alignment algorithms, depending on the length of the two sequences. Sequences of similar lengths are preferably aligned using a global alignment algorithms (e.g. Needleman-Wunsch) which aligns the sequences optimally over the entire length, while sequences of substantially different lengths are preferably aligned using a local alignment algorithm (e.g. Smith-Waterman). Sequences may then be referred to as "substantially identical” or “essentially similar” when they (when optimally aligned by for example the program EMBOSS needle or EMBOSS water using default parameters) share at least a certain minimal percentage of sequence identity (as described below).

A global alignment is suitably used to determine sequence identity when the two sequences have similar lengths. When sequences have a substantially different overall length, local alignments, such as those using the Smith-Waterman algorithm, are preferred. EMBOSS needle uses the Needleman-Wunsch global alignment algorithm to align two sequences over their entire length (full length), maximizing the number of matches and minimizing the number of gaps. EMBOSS water uses the Smith-Waterman local alignment algorithm. Generally, the EMBOSS needle and EMBOSS water default parameters are used, with a gap open penalty = 10 (nucleotide sequences) / 10 (proteins) and gap extension penalty = 0.5 (nucleotide sequences) / 0.5 (proteins). For nucleotide sequences the default scoring matrix used is DNAfull and for proteins the default scoring matrix is Blosum62 (Henikoff & Henikoff, 1992, PNAS 89, 915-919).

Alternatively percentage similarity or identity may be determined by searching against public databases, using algorithms such as FASTA, BLAST, etc. Thus, the nucleic acid and protein sequences of some embodiments of the present invention can further be used as a “query sequence” to perform a search against public databases to, for example, identify other family members or related sequences. Such searches can be performed using the BLASTn and BLASTx programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to oxidoreductase nucleic acid molecules of the invention. BLAST protein searches can be performed with the BLASTx program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17): 3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., BLASTx and BLASTn) can be used. See the homepage of the National Center for Biotechnology Information accessible on the world wide web at www.ncbi.nlm.nih.gov/.

Optionally, in determining the degree of amino acid similarity, the skilled person may also take into account so-called conservative amino acid substitutions.

As used herein, “conservative” amino acid substitutions refer to the interchangeability of residues having similar side chains. Examples of classes of amino acid residues for conservative substitutions are given in the Tables below.

Alternative conservative amino acid residue substitution classes :

Alternative physical and functional classifications of amino acid residues:

Microbial organism

As used herein, "microbial organism", "microorganism",“microbial cell” or“microbial host” and variations of these root terms (such as pluralizations and the like) have their customary and ordinary meanings as understood by one of skill in the art in view of this disclosure, including any naturally-occurring species or synthetic or fully synthetic prokaryotic or eukaryotic unicellular organism. Thus, this expression can refer to cells of any of the three domains Bacteria, Archaea and Eukarya. Exemplary microorganisms that can be used in accordance with embodiments herein include, but are not limited to, bacteria, yeast, filamentous fungi, and algae, for example photosynthetic microalgae. Furthermore, fully synthetic microorganism genomes can be synthesized and transplanted into single microbial cells, to produce synthetic microorganisms capable of continuous self-replication (see Gibson et al. (2010), "Creation of a Bacterial Cell Controlled by a Chemically Synthesized Genome," Science 329: 52-56, which is incorporated herein by reference). As such, in some embodiments, the microorganism is fully synthetic. A desired combination of genetic elements, including elements that regulate gene expression, and elements encoding gene products (for example immunity modulators, poison, antidote, and industrially useful molecules also called product of interest) can be assembled on a desired chassis into a partially or fully synthetic microorganism. Description of genetically engineered microbial organisms for industrial applications can also be found in Wright, et al. (2013) "Building-in biosafety for synthetic biology" Microbiology 159: 1221-1235, incorporated herein by reference.

A variety of bacterial species and strains can be used in accordance with embodiments herein, and genetically modified variants, or synthetic bacteria based on a "chassis" of a known species can be provided. Exemplary bacteria with industrially applicable characteristics, which can be used in accordance with embodiments herein include, but are not limited to, Bacillus species (for example Bacillus coagulans, Bacillus subtilis, and Bacillus licheniformis), Paenibacillus species, Streptomyces species, Micrococcus species, Corynebacterium species, Acetobacter species, Cyanobacteria species, Salmonella species, Rhodococcus species, Pseudomonas species, Lactobacillus species, Enterococcus species, Alcaligenes species, Klebsiella species, Paenibacillus species, Arthrobacter species, Corynebacterium species, Brevibacterium species, Thermus aquaticus, Pseudomonas stutzeri, Clostridium thermocellus, and Escherichia coli. A variety of yeast species and strains can be used in accordance with embodiments herein, and genetically modified variants, or synthetic yeast based on a "chassis" of a known species can be provided. Exemplary yeast with industrially applicable characteristics, which can be used in accordance with embodiments herein include, but are not limited to Saccharomyces species (for example, Saccharomyces cerevisiae, Saccharomyces bayanus, Saccharomyces boulardii), Candida species (for example, Candida utilis, Candida krusei), Schizosaccharomyces species (for example Schizosaccharomyces pombe, Schizosaccharomyces japonicus ), Pichia or Hansenula species (for example, Pichia pastoris or Hansenula polymorpha) species, and Brettanomyces species (for example, Brettanomyces claussenii).

A variety of algae species and strains can be used in accordance with embodiments herein, and genetically modified variants, or synthetic algae based on a "chassis" of a known species can be created. In some embodiments, the algae comprises, consists essentially of, or consists of photosynthetic microalgae. Exemplary algae species that can be useful for biofuels, and can be used in accordance with embodiments herein, include Botryococcus braunii, Chlorella species, Dunaliella tertiolecta, Gracilaria species, Pleurochrysis carterae, and Sargassum species. Additionally, many algaes can be useful for food products, fertilizer products, waste neutralization, environmental remediation, and carbohydrate manufacturing (for example, biofuels).

A variety of filamentous fungal species and strains can be used in accordance with embodiments herein, and genetically modified variants, or synthetic filamentous fungi based on a "chassis" of a known species can be provided. Exemplary filamentous fungi with industrially applicable characteristics, which can be used in accordance with embodiments herein include, but are not limited to an Acremonium, Agaricus, Alternaria, Aspergillus, Aureobasidium, Botryospaeria, Ceriporiopsis, Chaetomidium, Chrysosporium, Claviceps, Cochliobolus, Coprinopsis, Coptotermes, Corynascus, Cryphonectria, Cryptococcus, Diplodia, Exidia, Filibasidium, Fusarium, Gibberella, Holomastigotoides, Humicola, Irpex, Lentinula, Leptospaeria, Magnaporthe, Melanocarpus, Meripilus, Mucor, Myceliophthora, Neocaffimastix, Neurospora, Paecilomyces, Peniciffium, Phanerochaete, Piromyces, Poitrasia, Pseudoplectania, Pseudotrichonympha, Rhizomucor, Schizophyllum , Scytalidium, Talaromyces, Thermoascus, Thielavia, Tolypocladium, Trichoderma, Trichophaea, Verticillium, Volvariella, or Xylaria.

Species include Acremonium cellulolyticus, Aspergillus aculeatus, Aspergillus awamori, Aspergillus foetidus, Aspergillus fumigatus, Aspergillus japonicus, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae, Chrysosporium inops, Chrysosporium keratinophilum, Chrysosporium lucknowense, Chrysosporium merdarium, Chrysosporium pannicola, Chrysosporium queenslandicum, Chrysosporium tropicum, Chrysosporium zonatum, Fusarium bactridioides, Fusarium cerealis, Fusarium crookwellense, Fusarium culmorum, Fusarium graminearum, Fusarium graminum, Fusarium heterosporum, Fusarium negundi, Fusarium oxysporum, Fusarium reticulatum, Fusarium roseum, Fusarium sambucinum, Fusarium sarcochroum, Fusarium sporotrichioides, Fusarium sulphureum, Fusarium torulosum, Fusarium trichothecioides, Fusarium venenaturn, Humicola grisea, Humicola insolens, Humicola lanuginosa, Irpex lacteus, Mucor miehei, Myceliophthora thermophila, Neurospora crassa, Penicillium funiculosum, Penicillium purpurogenum, Phanerochaete chrysosporium, Thielavia achromatica, Thielavia albomyces, Thielavia albopilosa, Thielavia australeinsis, Thielavia fimeti, Thielavia microspora, Thielavia ovispora, Thielavia peruviana, Thielavia setosa, Thielavia spededonium, Thielavia subthermophila, Thielavia terrestris, Trichoderma harzianum, Trichoderma koningii, Trichoderma longibrachiatum, Trichoderma reesei, or Trichoderma viride.

In this document and in its claims, the verb "to comprise" and its conjugations is used in its non-limiting sense to mean that items following the word are included, but items not specifically mentioned are not excluded. In addition, the verb“to consist” may be replaced by“to consist essentially of” meaning that a peptide or peptidomimetic, a polypeptide, a culture medium, a microbial organism or a composition as described herein may comprise additional component(s) than the ones specifically identified, said additional component(s) not altering the unique characteristic of the invention. In addition, the verb“to consist” may be replaced by“to consist essentially of meaning that a method as described herein may comprise additional step(s) than the ones specifically identified, said additional step(s) not altering the unique characteristic of the invention.

Reference to an element by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element is present, unless the context clearly requires that there be one and only one of the elements. The indefinite article "a" or "an" thus usually means "at least one".

As used herein, with "at least" a particular value means that particular value or more. For example, "at least 2" is understood to be the same as "2 or more" i.e., 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , 12, 13, 14, 15, ..., etc.

The word“about” or“approximately” when used in association with a numerical value (e.g. about 10) preferably means that the value may be the given value (of 10) more or less 0.1 % of the value. As used herein, the term "and/or" indicates that one or more of the stated cases may occur, alone or in combination with at least one of the stated cases, up to with all of the stated cases.

Each embodiment as identified herein may be combined together unless otherwise indicated.

All patent applications, patents, and printed publications cited herein are incorporated herein by reference in the entireties, except for any definitions, subject matter disclaimers or disavowals, and except to the extent that the incorporated material is inconsistent with the express disclosure herein, in which case the language in this disclosure controls.

One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. Indeed, the present invention is in no way limited to the methods and materials described.

The present invention is further described by the following examples which should not be construed as limiting the scope of the invention.

Description of the figures

FIG 1. Competence-predation dependencies of ComR paralogs in S. salivarius

(A) Scheme of genomic organization and transcriptional dependencies (dashed arrows) between competence activation ( comX) and bacteriocins production ( blpK , slvX,...) in S. salivarius. Promoters are depicted with broken arrows. Regulators and the ComS pheromone are stained according to their encoding genes. The ComS precursor is produced (curled arrow) as an intracellular precursor (square) before secretion, maturation and import as an active pheromone (ellipses). The newly described two-Rgg system is shaded and the T arrow pinpoints the inhibitory role of SarF on ScuR.

(B, C, D and E) Maximum luciferase activity/ODeoo ratio (RLU/OD; logarithmic scale) of various competence or bacteriocin production-involved promoters fused to a iuxAB reporter system in WT or overexpressing backgrounds. (B) Promoter activation of genes upon sComS addition (full bars) vs mock condition (striped bars) in WT (light grey bars) or scuR overexpression mutant (sct/R ++ ; dark grey bars). (C) Activity of sptA, comS and slvX promoters in WT strain, and scuR or sarF ( sarF ++ ) overexpression mutants. (D) Activity of sptA and comS promoters in WT and scuR ++ mutant deleted or not for comR gene. (E) Activity of sptA and comS promoters in overexpression of scuR or sarF in a scuR locus ( AscuR-sarF ) deletion background. Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

(F) Maximum luciferase activity/ODeoo ratio (RLU/OD) of comS, blpK and slvX promoters fused to a IuxAB reporter system in WT (open bars) or AscuR-sarF (grey bars) strain activated with sComS. Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates. FIG 2. ScuR/SarF activating peptide identification

(A) Cartoon portraying the rational strategy for the peptide randomization-based screen. A library of randomized small genes under inducible promoter controls (P xy/ ) is transformed in a reporter strain in which the chloramphenicol resistance gene (cat) is translationally fused to sptA promoter. In absence of xylose or upon xylose induction of irrelevant peptides (square, hexagon and star), sptA promoter is maintained OFF and does not initiate cat transcription, causing cell sensitivity (Cm s ) on chloramphenicol-supplemented media. The xylose-driven intracellular production of a cognate peptide (ellipse) promotes chloramphenicol resistance (Cm R ), through P sp tA activation by ScuR/SarF (dashed arrow).

(B) Activity of sptA promoter in WT strains and various mutants expressing intracellularly activating peptides (cartoon) in medium supplemented with xylose (0.1 % or 1 %; grey bars) vs mock conditions (open bars). The BM 1 clone is an irrelevant peptide (negative control). Magnitude is expressed in percentage compared to the WT P sptA -luxAB reporter strain (Relative maximal luciferase activity). Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

(C) Weighted consensus sequence for the suite of 22 activating peptides identified in the randomization-based screen. Randomized residues are crowned with a horizontal black bar while the non-variable amino acids are grey-coloured. The Bits represent the relative frequency of residues. Information content is plotted as a function of residues position and reckoned from the N- terminus (1 to 12) or the C-terminus (-1 to -12). The sequence logo image was generated using the WebLogo application (accessible on the world wide web at webloqo.berkelev.edu/loqo.cgi).

(D) Promoter activity of the sptA gene in response to the BI7 encoded peptide in various rgg mutant backgrounds. Media were supplemented with 0.1 % xylose (open bars) or water (grey bars). Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

FIG 3. The ScuR/SarF system responds to exogenous peptides

(A) Cartoon depicting the ScuR/SarF-mediated activation of P sp tA upon addition of exogenous synthetic peptides.

(B) Fold increase in maximal P sp tA activity upon addition of representative synthetic peptide

(0.01 or 1 mM) vs mock conditions. Peptide sequences are correlated to peptide name and compared to the consensus motive (SEQ ID NO: 728) (open box). sBH6: SEQ ID NO: 729; sBI7: SEQ ID NO: 730; sBMO: SEQ ID NO: 731 ; sBJ1 : SEQ ID NO: 732; sBK1 : SEQ ID NO: 733; sBK3: SEQ ID NO: 734; sBK4: SEQ ID NO: 735; sBK8: SEQ ID NO: 736; sB02: SEQ ID NO: 737. The crucial W and G residues are highlighted with grey boxes.

(C) Maximal activity of P sp tA exposed to sBI7 WT and mutant peptides (1 nM).

(D) Dose response dot plot of sptA, slvX and comS promoter activity upon sBI7 induction at various concentrations (nM). Promoters were tested in WT strain and, AscuR or AsarF deletants.

(B, C and D) Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates. (E) Maximal activity of PsptA (RLU/OD; logarithmic scale) exposed to WT and mutant sBI7 peptides (1 mM). Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

(F) Activity of sptA, comS and slvX promoters (absolute maximal luciferase activity; ALU) in AscuR-sarF challenged with sBI7 synthetic peptide (grey bars) vs mock conditions (open bars).

Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

(G) Maximum luciferase activity/OD600 ratio (RLU/OD; logarithmic scale) of comS, slvX and sptA promoters fused to a luxAB reporter system in WT (open bars) or D comR (grey bars) strain activated with the sBI7 synthetic peptide. Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

FIG 4. Singularities in promoter recognition of Rgg paralogs

(A) Mobility shift assays of comX, comS, slvX and sptA promoter probes conducted with purified Rgg paralogs and decreasing concentrations of their cognate peptide (grey triangles; 2:2 dilutions from 20mM). Probes are 30bp (or 40bp for P sp w), were Cy3-conjugated and used at 40ng. Protein concentration remains constant (grey boxes; 4mM). Open triangles showcase ternary complexes (peptide-Rgg-DNA).

(B) Nucleotide alignment of promoters of comX (SEQ ID NO: 738), comS (SEQ ID NO: 739), slvX (SEQ ID NO: 740) and sptA (SEQ ID NO: 741 ). The palindromic stretches (inverted arrows) and the sigma-bound DNA sequence (-10 boxes) are shaded in grey. Boxed nucleotides highlight the potential mismatches in the hairpin structure of P ¥m x or P sp tA that were substituted to restore a genuine dyad symmetry sequence (see FIG 4C and 4D). A and T represent the position and nature of single nucleotide insertion in the sptA promoter (see FIG 4C).

(C and D) Fold increase in maximal activity of WT and mutated promoters of sptA (C) or comX (D) exposed to sBI7 or sComS (1 mM). Nucleotides substitutions and insertions are disclosed in FIG 4B. Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

(E) Mobility shift assays of mutated comX promoter probes conducted with a unique concentration of ScuR or SarF (grey boxes; 4mM) and decreasing concentrations of sBI7 peptide

(grey triangles; 2:2 dilutions from 20mM). Open triangles showcase ternary complexes (peptide- Rgg-DNA).

(F) Mobility shift assays of comS, slvX and sptA promoter probes conducted with purified ScuR and decreasing concentrations of the non-cognate sComS peptide (grey triangles; 2:2 dilutions from 20mM). Probes are 30bp (or 40bp for P sp w), were Cy3-conjugated and used at 40ng. Protein concentration remains constant (grey boxes; 4mM). Open triangle showcases binary complexes (ScuR-DNA).

(G) Mobility shift assays of slvX and sptA promoter probes conducted with sBI7 peptide and decreasing concentrations of purified ScuR (grey triangles; 2:2 dilutions from 8mM). Probes are 30bp (P s/v x) or 40bp (P SP M), were Cy3-conjugated and used at 40ng. Peptide concentration remains constant (grey boxes; 1 mM). Open triangles showcase ternary complexes (sBI7-ScuR-DNA).

(H) Nucleotide alignment of bacteriocin-related gene promoters. Psivx: SEQ ID NO: 740; Pooi7 6 : SEQ ID NO: 742; Poi58 4 : SEQ ID NO: 743; Pbi pK : SEQ ID NO: 744; Psiw: SEQ ID NO: 745; PbipG: SEQ ID NO: 746; Psivw: SEQ ID NO: 747; SEQ ID NO: 748. The palindromic stretches (inverted arrows) and the sigma-bound DNA sequence (-10 boxes) are shaded in grey. The characteristic T-rich region is grey font.

FIG 5. Activated ScuR drives bacteriocin secretion

(A and B) Bacteriocin inhibition assay of S. salivarius WT and mutant derivatives. Indicator strains ( L . lactis) were embedded in the top soft agar layer, while sBI7 was supplemented into the bottom agar layer as required. Producer strains were spotted on top of the two agar layers. (A) Killing properties of scuR or sarF overexpression mutants compared WT. (B) Effect of sBI7 addition (1 mM) on WT strain and scuR/sarF various mutants. sct/R ++ and bacteriocin null mutant ( Aslv5 ) were used as positive and negative control, respectively.

(C) Bacteriocin inhibition assay of S. salivarius WT and scuR/sarF mutant derivatives. Indicator strains (L lactis) were embedded in the top soft agar layer, while sBI7 was supplemented or not into the bottom agar layer as stated. Producer strains were spotted on top of the two agar layers. sct/R ++ and bacteriocin null mutant ( Aslv5 ) were used as positive and negative control, respectively.

FIG 6. Rgg members requisition predation control in S. salivarius

(A) Conservation of ScuR locus, and BlpRH system across S. salivarius. Functional BlpRH pair, ScuR, SptA, SptB, and SarF were sought for homologs in various S. salivarius strains. The phylogenetic tree (100 bootstrap replicates) was adapted from (Yu et al., 2015). An empty box means that no functional ortholog was found in the species genome. Scale bar: 0.01 substitution per site.

(B) Figurative illustration of RRNPPs vs two-component systems (TCSs) drift toward competence and predation regulation in paradigmatic streptococci (S. pneumoniae, S. mutans and S. salivarius).

FIG 7. Amino acid requirements for the sBI7-mediated effect

Maximal activity of salivaricin promoters (RLU/OD; logarithmic scale) exposed to sBI7 (WT) (SEQ ID NO: 36) and mutant peptides (1 mM). Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

FIG 8. sBI7 has no effect on the comX promoter activation

Maximal activity of P ¥m x exposed to the sComS or sBI7 peptide (1 pM) (SEQ ID NO: 36) in WT strain and AscuR and/or D sarF mutants. Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates. FIG 9. No activation of comX or late competence genes under ComX control by sBI7

Maximal activity of P ¥m x and ComX-dependent promoters exposed to the sComS (light grey) or sBI7 peptide (SEQ ID NO: 36) (dark grey) (1 mM) in comparison to basal activity (open box). Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

FIG 10. C-terminal tolerance of synthetic peptides

(A) Maximal activity of P spM exposed to LAFWDSLG (SEQ ID NO: 749), LAFWDSLGLLL (SEQ ID NO: 750) or the peptide sBI7 (SEQ ID NO: 36) (1 mM) in comparison to basal activity. Experimental values represent the averages (with standard error of the mean, SEM) of at least three independent biological replicates.

(B) Mobility shift assays of the sptA promoter probe conducted with purified ScuR and decreasing concentrations of the peptides LAFWDSLG (SEQ ID NO: 749) or LAFWDSLGLLL (SEQ ID NO: 750) (grey triangles; 2:2 dilutions from 20pM). The probe is 40bp, Cy3-conjugated and used at 40ng. Protein concentration remains constant (grey boxes; 4pM). Open triangle showcases single or binary complexes (ScuR-DNA).

Examples Experimental procedures

Bacterial strains, plasmids, oligonucleotides, and growth conditions

Bacterial strains, plasmids and oligonucleotides used in the Examples are listed and described in the tables below. Streptococcus salivarius HSISS4 and derivatives were grown at 37°C without shaking in M17 (Difco Laboratories, Detroit, Ml) or in CDM (Fontaine et al. Mol Microbiol 2013, 87: 1 1 13-1 132) supplemented with 1 % (w/v) glucose (M17G, CDMG, respectively). Escherichia coli TOP10 (Invitrogen) were cultivated with shaking at 37°C in LB. Electrotransformation of E. coli was performed as previously described (Mignolet et al. Elife 2016, 5:e18647). Lactococcus lactis was grown in M17 broth with 1 % glucose at 30°C without shaking. We added 1.5% (w/v) agar into M17 and LB plates, and bacteriocin inhibition tests were assayed on M17 plates containing 0.2% agar. We added D-xylose (0.1 or 1 %; w/v), ampicillin (250 pg/ml), spectinomycin (200 pg/ml), chloramphenicol (5 pg/ml; except if otherwise stated) or erythromycin (10 pg/ml), 5-FOA (1 mg/ml) (Melford Laboratories), as required. Synthetic peptides and sComS (purity of 95%; 1 pM, except if otherwise stated) were supplied by Peptide2.0 Inc. (Chantilly, VA, USA) and resuspended in DMSO. Solid plates inoculated with streptococci cells were incubated anaerobically (BBL GasPak systems, Becton Dickinson, Franklin lakes, NJ) at 37°C. Table 1. List of bacterial strains used in the Examples

Characteristics Reference/source

HSISS4 Wild-type gastro-intestinal tract isolate (Van den Bogert et al., 2014)

JM1004 HSISS4 AcomA::cat (Mignolet et al., 2018)

JM1013 HSISS4 Aslv5 (Mignolet et al., 2018)

JM1019 HSISS4 tRNA Thr ::Pco m s-luxAB-cat (Mignolet et al., 2018)

JM1020 HSISS4 tRNA Thr ::Pco m x-luxAB-cat (Mignolet et al., 2018)

JM1021 HSISS4 tRNA Thr ::P bipK -luxAB-cat (Mignolet et al., 2018)

JM1022 HSISS4 tRNA Thr :: P 176 -1 uxA B- cat (Mignolet et al., 2018)

JM1023 HSISS4 tRNA Thr : :P 58 4 -luxAB-cat (Mignolet et al., 2018)

JM1024 HSISS4 tRNA Thr ::Psivv-luxAB-cat (Mignolet et al., 2018)

JM1025 HSISS4 tRNA Thr ::P bip e-luxAB-cat (Mignolet et al., 2018)

JM1026 HSISS4 tRNA Thr ::Psivw-luxAB-cat (Mignolet et al., 2018)

JM1027 HSISS4 tRNA Thr ::Psivx-luxAB-cat (Mignolet et al., 2018)

JM1028 HSISS4 tRNA Thr ::Psiv Y -luxAB-cat (Mignolet et al., 2018)

JM1100 HSISS4 tRNA Thr ::Ps PtA -luxAB-cat This work

JM1101 HSISS4 tRNA Ser ::P3 2 -scuR-spec (scuFF + ) This work

JM1102 HSISS4 tRNA Ser ::P 32 -sarF-ST-spec ( sarF-ST ++ ) This work

JM1103 JM1019 tRNA Ser ::P3 2 -scuR-spec This work

JM1104 JM1020 tRNA Ser ::P3 2 -scuR-spec This work

JM1105 J M 1021 tRNA Ser ::P3 2 -scuR-spec This work

JM1106 JM1022 tRNA Ser ::P3 2 -scuR-spec This work

JM1107 JM1023 tRNA Ser ::P3 2 -scuR-spec This work

JM1108 JM1024 tRNA Ser ::P3 2 -scuR-spec This work

JM1109 JM1025 tRNA Ser ::P3 2 -scuR-spec This work

JM1110 JM1026 tRNA Ser ::P3 2 -scuR-spec This work

JM1111 JM1027 tRNA Ser ::P3 2 -scuR-spec This work

JM1112 JM1028 tRNA Ser ::P3 2 -scuR-spec This work

JM1113 JM1100 tRNA Ser ::P3 2 -scuR-spec This work

JM1114 JM1019 tRNA Ser ::P3 2 -sarF-spec This work

JM1115 JM1027 tRNA Ser ::P3 2 -sarF-spec This work

JM1116 JM1100 tRNA Ser ::P3 2 -sarF-spec This work

JM1117 JM1113 AcomRxery This work

JM1118 HSISS4 AscuR-sarF::ery This work

JM1119 JM1113 AscuR-sarF::ery This work

JM1120 JM1116 AscuR-sarF::ery This work

JM1121 HSISS4 tRNA Thr ::Pspt A -cat-spec This work

JM1122 HSISS4 tRNA Thr ::P SptA -cat-lox72 This work

JM1123 JM1122 tRNA Ser ::Pxyii-comR-spec This work

JM1124 JM1013 tRNA Thr ::P SptA -cat-lox72 This work

JM1125 JM1122 tRNA Ser ::P Xyi2 -BH6-spec This work

JM1126 JM1122 tRNA Ser ::Pxyii-BI1-spec This work

JM1127 JM1122 tRNA Ser :: P xy n- BI2- spec This work

JM1128 JM1122 tRNA Ser ::Pxyn-BI5-spec This work

JM1129 JM1122 tRNA Ser ::Pxyii-BI6-spec This work

JM1130 JM1122 tRNA Ser ::Pxyii-BI7-spec This work

JM1131 JM1122 tRNA Ser ::Pxyii-BI9-spec This work

JM1132 JM1122 tRNA Ser ::Pxyii-BI10-spec This work

JM1133 JM1122 tRNA Ser ::Pxyii-BI11-spec This work

JM1134 JM1122 tRNA Ser ::Pxyii-BI12-spec This work

JM1135 JM1122 tRNA Ser :: P xyi2 - BJ 1-spec This work

JM1136 JM1122 tRNA Ser ::Pxyii-BK1-spec This work

JM1137 JM1122 tRNA Ser ::Pxyii-BK2-spec This work

JM1138 JM1122 tRNA Ser ::Pxyii-BK3-spec This work

JM1139 JM1122 tRNA Ser ::Pxyii-BK4-spec This work

JM1140 JM1122 tRNA Ser ::Pxyii-BK5-spec This work

JM1141 JM1122 tRNA Ser ::Pxyii-BK6-spec This work

JM1142 JM1122 tRNA Ser ::Pxyii-BK7-spec This work

JM1143 JM1122 tRNA Ser ::Pxyii-BK8-spec This work

JM1144 JM1122 tRNA Ser ::Pxyii-BK9-spec This work

JM1145 JM1124 tRNA Ser ::P Xyii -BL 1-spec This work JM1146 JM1124 tRNA Ser ::Pxyii-BL2-spec This work

JM1147 JM1124 tRNA Ser ::Pxyii-BL3-spec This work

JM1148 JM1124 tRNA Ser ::Pxyi 2 -BM1-spec This work

JM1149 JM1124 tRNA Ser ::P Xyii -BN 1-spec This work

JM1150 JM1124 tRNA Ser ::Pxyii-BN2-spec This work

JM1151 JM1124 tRNA Ser ::Pxyii-BN3-spec This work

JM1152 JM1124 tRNA Ser ::Pxyii-BN4-spec This work

JM1153 JM1122 tRNA Ser ::Pxyi 2 -B01-spec This work

JM1154 JM1122 tRNA Ser ::Pxyi 2 -B02-spec This work

JM1155 JM1122 tRNA Ser ::Pxyii-BP1-spec This work

JM1156 JM1100 tRNA Ser ::Pxyii-BI5-spec This work

JM1157 JM1100 tRNA Ser ::Pxyii-BI7-spec This work

JM1158 JM1100 tRNA Ser ::Pxyii-BI10-spec This work

JM1159 JM1100 tRNA Ser :: P xyi - BJ 1-spec This work

JM1160 JM1100 tRNA Ser ::Pxyii-BK1-spec This work

JM1161 JM1100 tRNA Ser ::Pxyii-BK4-spec This work

JM1162 JM1100 tRNA Ser ::Pxyii-BK8-spec This work

JM1163 JM1100 tRNA Ser ::Pxyii-BK9-spec This work

JM1164 JM1100 tRNA Ser ::Pxyii-BL2-spec This work

JM1165 JM1100 tRNA Ser ::Pxyii-BL3-spec This work

JM1166 JM1100 tRNA Ser ::Pxyi 2 -BM1-spec This work

JM1167 JM1100 tRNA Ser ::Pxyii-BN2-spec This work

JM1168 JM1100 tRNA Ser ::Pxyii-BN3-spec This work

JM1169 JM1100 tRNA Ser ::Pxyi 2 -B02-spec This work

JM1170 JM1100 tRNA Ser ::Pxyii-BP1-spec This work

JM1171 JM1157 AscuRxery This work

JM1172 JM1157 AsarFxery This work

JM1173 JM1157 AscuR-sarF::ery This work

JM1174 JM1157 AcomRxery This work

JM1175 JM1100 AscuRxery This work

JM1176 JM1100 AsarF::ery This work

JM1177 JM1027 AscuRxery This work

JM1178 JM1027 AsarF::ery This work

JM1179 JM1019 AscuRxery This work

JM1180 JM1019 AsarFxery This work

JM1181 HSISS4 tRNA Thr ::P SptA CT ^ AC -luxAB-cat This work

JM1182 HSISS4 tRNA Thr ::Pspt A +1 -tuxAB-cat This work

JM1183 HSISS4 tRNA Thr ::P SptA +A -luxAB-cat This work

JM1184 HSISS4 tRNA Thr ::Pco m x G ^ A -luxAB-cat This work

JM1185 JM1101 AcomAxcat This work

JM1186 JM1101 AsptAxcat This work

JM1187 HSISS4 AscuR This work

JM1188 HSISS4 AsarF This work

JM1189 HSISS4 AsptAxcat This work

JM1190 JM1019 AscuR-sarFxery This work

JM1191 J M 1021 AscuR-sarFxery This work

JM1192 JM1027 AscuR-sarFxery This work

JM1193 JM1100 AscuR-sarFxery This work

Lactococcus lactis subsp. lactis

IL1403 Laboratory stra

Table 2. List of plasmids used in the Examples

Characteristics Reference/source pGhostcre Thermosensitive replication origin vector in S. (Fontaine et al., 2010) salivarius, encoding the Cre recombinase; ery R

pGIUD0855e/y pUC18 derivative containing the erm gene (Fontaine et al., 2010) pS E U DO - P usp45 - erm-oroP containing vector (Overkamp et al., 2013) stgfp( Bs)

pGILFspec pG + host9 derivative containing the spectinomycin (Haustenne et al., 2015) resistance cassette P Spec -spec downstream of luxAB

pJUD specmutl pGILFspec derivative in which a Spel restriction (Mignolet et al., 2018) site was mutated

pJUD specmutl - Terminator associated -gfp* ORF cloned in (Mignolet et al., 2018) gfp + ter pJUD specmutl PNZ5319 pACYC184 derivative containing the cat gene (Lambert et al., 2007) under the control of the P32 constitutive promoter

from Lactococcus lactis

pJIMcaf pG + host9 containing the luxAB genes of (Mignolet et al., 2018)

Photorhabdus luminescens, a transcriptional

terminator with a cat cassette

pBAD -comR-ST pBAD hisA derivative encoding ComR fused to a (Mignolet et al., 2018)

C-terminal Streptagll

pBAD -scuR-ST pBAD hisA derivative encoding ScuR fused to a This work

C-terminal Streptagll

pBAD -sarF-ST pBAD hisA derivative encoding SarF fused to a C- This work

terminal Streptagll

Table 3. List of oligonucleotides used in the Examples

Names Sequences SEQ ID NO

rggD_Ncol SS 5’- 88

AAAAAACCATGGCAGAAGATATTAAAA

TCAAGA-3’

rggD_Munl 5’- 89

AAAAAACAATT G CTTT G ACT CGTT ACTT GTAT-3’

rggC_Ncol SS 5’- 90

AAAAAACC AT GGCTGAAGATATT AAAA TCGAGA-3’

rggC RI SS 5’- 91

AAAAAAG AATT CTTCTATATTT AAAT CT TTTT-3’

Uplox66 5’-TAAGGAAGATAAATCCCATAAGG-3’ 92

DNIox71 5’-TT CACGTT ACT AAAGG G AAT GTA-3’ 93

lox66-ery 5’- 94

T AAGG AAG AT AAAT CCCAT AAG GT ACC T AAT AATTT AT CT ACATT CC-3’

lox71 -ery 5’- 95

TT CACGTT ACT AAAGG G AAT GT AAAAT GATACACCAATCAGTGC-3’

F spec 5’-TAATAAGGCCGGCCAAT AAA-3’ 96

R spec 5’-ATAGGAT GAGAACT CCCATG-3’ 97

UPery-oroP 5’-AAGGTT GAT GTTACTGCT GATA-3’ 98

DNery-oroP 5’-TGCT G ACTT G CACCAT AT CAT A-3’ 99

UF tRNAser 5’-CAAGATTAACCAT GACCTT C-3’ 100

UR tRNAser 5’-AGTAATTAAAAAGAAGATGG-3’ 101

DF tRNAser 5’-T ACCT AAAAAGTGT CCCTT C-3’ 102

DR2_tRNAser 5’-TTGG AT AAG GT CTT G ACTT C-3’ 103

UF tRNAthr 5’-T GT CAAAGG ATTAGGAAAAC-3’ 104

UR tRNAthr 5’-TT G ATTT AT ACCT CT CAATTT -3’ 105

DF tRNAthr 5’-AAAT CAACCT CTTT GAACATA-3’ 106

DR tRNAthr 5’- 107

AAAAAAGAATT CATT CAT GAT GAGCGG GTTCGTGAGA-3’

F_pZX9 5’- 108

CCAT CTT CTTTTT AATT ACTT CT AG ATT AT AT AT GAT AT GAT C-3’

R_pZX9_ATG 5’-CAT ATTT ACCT CCTTT G ATTT A-3’ 109

F luxAB ATG 5’-AT GAAATTTGGAAACTTTTTGC-3’ 1 10

R cat tRNAthr 5’- 1 1 1

TATGTT CAAAG AG GTT G ATTT CACGTT ACTAAAGGGAATGTA-3’

UFcomRJIM- 5’- 1 12

SS1 -4 GCAGTACCACTCTATGCT AAATTT G CC

AACTTTGA-3’

URcomRJIM- 5’- 1 13

SS1 -4 CCTTATGGGATTTATCTTCCTTAGAGA

CACT CCTTT ATTT C-3’

DFcomRJIM- 5’- 1 14

SS1 -4 T ACATT CCCTTT AGT AACGTG AAAAAT

GGTGGTGACAT AAA-3’ DRcomRJIM- 5’- 115

SS 1 -4 T G ACGTG ATTT CACCAGT ACG ACGT G A

ACTAAAGA-3’

Up comR SS1- 5’-TTGCTTACAGTTGCTATGGT-3’ 116

4

Down comR S 5’-T CAT CACAAT G GT CACAT CT-3’ 117

S1-4

UFrggC JIM 5’- 118

AAAACTGC AAGT AG AGT CG CCG AATT A GAA-3’

URrggC JIM 5’- 119

CCTT ATGG G ATTT AT CTT CCTT AACAT A ATT CCTT AT G ATTT AG A-3’

DFrggC JIM 5’- 120

T ACATT CCCTTT AGT AACGT G AAG ACA TT GATGTCCTTTT GA-3’

DRrggD SS 5’- 121

TAGCTT CATT CAT GT CATGTGTCGT CA AAA-3’

Up rggC JIM 5’- AAAT ATCGT CATT G CCAGT A-3’ 122

Down rggD SS 5’-CT GAATAAGTT CAGCAGGTT-3’ 123

Down rggD SS 5’- 124

T CACTTT G ACTCGTT ACTT GT G ATTTT A AT AT CTT CT G ACAT-3’

rggD_S4_3 5’- 125

ATGT CAG AAG AT ATTAAAAT C ACAAGT AACGAGTCAAAGTGA-3’ rggD_S4_4 5’- 126

TAT CAGCAGTAACAT CAACCTT CAT GT CATGTGTCGTCAAAA-3’ rggD_S4_5 5’- 127

TATGATATGGTGCAAGTCAG CAT CAT G AAGT CTCCTGTCTAT-3’

rggD_S4_6 5’-GGCTAGTACAGTAGCTGTAT-3’ 128

UFrggC SS 5’- 129

CT GTTT AGCCCT AT CTTT G AGTTT AT CA GT-3’

URrggD JIM 5’- 130

CCTT ATGG G ATTT AT CTT CCTT ATGTAT TCCCCTT G AGTTTT-3’

DFrggD JIM 5’- 131

T ACATT CCCTTT AGT AACGTG AAG AAA AT AT AT CAG CAACAT -3’

DRrggC SS 5’- 132

CAGT G GTTT G ACGTT GTTTTT G AAT AC GGT-3’

Up rggC SS 5’-AAGGTAGCCTAAACAACTCA-3’ 133

Down rggC SS 5’-TTTATTGGTACCAAACGCCA-3’ 134 rggC_S4_2 5’- 135

CT ATTCT AT ATTT AAAT CTTTG ATTTT AA T AT CTT CAG ACAT-3’

rggC_S4_3 5’- 136

ATGTCT G AAG AT ATT AAAAT CAAAG ATT TAAATATAGAATAG-3’

rggC_S4_4 5’- 137

T AT CAGCAGT AACAT CAACCTT G ACGT T GTTTTT G AAT ACG GT-3’ rggC_S4_5 5’- 138

T ATG AT ATG GTG C AAG TC AG C AACT AG ACATT CCT G AAGACT-3’

rggC_S4_6 5’-TCCGCTAGTAGGATAGCTTT-3’ 139

UF PcomRJux 5’-TAATTGAGGAGGTCTATGAG-3’ 140

AB

UR comA 5’- 141

CCTT ATGGG ATTT AT CTT CCTT AAT AT G GAT ATTTT G AC AT G G - 3’ DF comA 5’- 142

T ACATT CCCTTT AGT AACGTG AAGCT A ATTT CAAT CCATT CCAG-3’

DR comA 5’- ACAGT ACT CTTT ATTTGGT G-3’ 143

F comR 5’- 144

CT AG AG G AGG AATTT AG AT G AACAT AA AAGACAGCATT G -3’

Down_PcomS_ 5’-GACAAAGTAGTCAAGACCGT-3’ 145

JIMSS1-4

UF_potA2 5’- AT ACT AT ACCTTT CAAT GTC-3’ 146

UR_potA2 5’- 147

CCTT ATGGG ATTT AT CTT CCTT AAT AAG GTTT GT CAT AT CTT G-3’

DF_potA2 5’- 148

T ACATT CCCTTT AGT AACGT G AAGG AA AACTTAATGTTTAACC-3’

DR_potA2 5’-ACT GAT CCCT GAAAGCATTG-3’ 149

Up_potA2 5’-AGAGTATACCTTAAATGACC-3’ 150

Down_potA2 5’-GATTTAAAG ATTTCGT GAAC-3’ 151

F_PpotA2_tRN 5’- 152

Athr AAATT G AG AGGT AT AAAT CAAT CATTTT

GGAAGCAAAATAC-3’

R_PpotA2_luxA 5’- 153

B ATG G C AAAAAG TTT CC AAATTT CAT ATCTTG

ATTT CT CCAATTT G-3’

F rggD 5’- 154

CT AG AG G AGG AATTT AG AT GT CAG AA GATATTAAAATC-3’

R rggD 5’- 155

TTTATTGGCCGGCCTTATTATCACTTT GACT CGTTACTTG-3’

F rggC 5’- 156

CTAGAGG AGGAATTTAGAT GTCT GAAG ATATTAAAATC-3’

R StrepTag 5’- 157

TTTATTGGCCGGCCTTATTACTATTTCT CGAACTGCGG-3’

F P32- 5’- 158 gfp+ter_spec CCAT CTT CTTTTTAATTACTGTCCT CGG

GATATGATAAG -3’

R P32 5’-CAT CT AAATT CCTCCT CT AG-3’ 159

F cat ATG 5’- AT G AACTTT AAT AAAATT G ATT-3’ 160

R_cat_(spec) 5’- 161

TTTATTGGCCGGCCTTATTATAAAAGC CAGTCATTAGGC-3’

R_PpotA2_cat_ 5’- 162

ATG AAT C AATTTT ATT AAAG TT CAT ATCTTG

ATTT CT CCAATTT G-3’

Pxyl seq 5’-TT GTTT AT CCTCCT CT AGT C-3’ 163 spec2 5’- AACT CCT G ATCCAAACAT GTA-3’ 164

Cy3_F_PpotA2 5’-Cy3- 165

EMSA T AACG AGTCAAAGT G AC AT AG AT GTCC

TTTT GATT CGTTA-3’

R_PpotA2_EMS 5’- 166

A T AACG AAT CAAAAG G ACAT CTATGT CA

CTTT GACT CGTTA-3’

Cy3_F_P01665 5’-Cy3- 167

EMSA CT CCAT AGTG ACATTT ATGT CACT ATTT

TT-3’

R P01665 EM 5’- 168

SA AAAAAT AGT G ACAT AAAT GT CACT AT G

GAG-3’

Cy3_F_PcomS_ 5’-Cy3- 169

EMSA AAT G G TG GT G AC AT AAAT GTCACTACT

TTT-3’ R_PcomS_EMS 5’- 170

A AAAAGT AGT G ACATTTATGTCACCACC ATT-3’

Cy3_F_PcomX_ 5’-Cy3- 171

EMSA TTTTATAGTGACATATATGTCGCTATTT

TA-3’

R PcomX EMS 5’- 172

A T AAAAT AGCG ACAT ATATGT CACT AT A AAA-3’

Cy3_F_PcomX_ 5’-Cy3- 173

EMSArev TTTT AT AGT G ACAT ATATGT CACT ATTT

TA-3’

R PcomX EMS 5’- 174

Arev T AAAAT AGT G ACAT ATATGT CACT AT AA

AA-3’

F_PcomX_mut 5’-CAT ATATGT CACTATTTT ATT -3’ 175

R_PcomX_mut 5’- AAT AAAAT AGT G ACAT ATATG-3’ 176

F_PpotA2_mut2 5’- ACAT AG ATGTCACTTT GATT CGT-3’ 177

R_PpotA2_mut 5’- ACG AAT CAAAGTG ACAT CTATGT-3’ 178

2

F_PpotA2_mut+ 5’-T GATT CGTTATTTTTTTT GTTT-3’ 179

1

R_PpotA2_mut 5’-AAACAAAAAAAATAACGAAT CA-3’ 180

+1

F_PpotA2_mut+ 5’-CATAG AT GT CACTTTT GATT C-3’ 181

A

R_PpotA2_mut 5’- GAATCAAAAGTGACATCTATG-3’ 182

+A

F_pept_xyl 5’- 183

TAAATCAAAGGAGGTAAATATGATCGC AATCCTANNNNNNNNNNNNNNNNNNN NNTGAT AAT AAGGCCGGCCAAT AAA-3’

Table 4. List of EMSA annealings, overlapping and cloning PCR subfraqments amplified in this study

PCR/annealing Primer 1 _ Primer 2

scuR amplification for pBAD-scuR-ST cloning rggD_Ncol SS rggD_Munl sarF amplification for pBAD-sarF-ST cloning rggC_Ncol SS rggC RI SS

P 32-cat cassette amplification Uplox66 DNIox71

Erm cassette amplification lox66-ery lox71-ery spec cassette amplification for tRNA Ser locus F spec R spec

Random peptide gene and spec for tRNA Ser locus F_pept_xyl R spec erm-oroP cassette amplification UPery-oroP DNery-oroP

P xyii amplification F_pZX9 R_pZX9_ATG

P Xyi 2 amplification F_pZX9 R_pZX9_ATG luxAB-cat amplification F luxAB ATG R_cat_tRNAthr

P 3 2 amplification F_P32-gfp+ter_spec R P32

spec cassette amplification for tRNA Thr locus F spec R_cat_tRNAthr Upstream homologous region of tRNA Ser locus UF tRNAser UR tRNAser Downstream homologous region of tRNA Ser locus DF tRNAser DR2_tRNAser Upstream homologous region of tRNA Thr locus UF tRNAthr UR tRNAthr Downstream homologous region of tRNA Thr locus DF tRNAthr DR tRNAthr scuR amplification for P32-SCUR fusion at tRNA Ser F rggD R rggD locus

sarF-ST amplification for P 32-sarF-ST fusion at F_rggC R StrepTag tRNA Ser locus

Promoter of sptA for luxAB fusion F_PpotA2_tRNAthr R_PpotA2_luxAB_AT

G

Promoter of sptA CT ^ AC for luxAB fusion F_PpotA2_mut2 R_PpotA2_mut2 Promoter of sptA +1 for luxAB fusion F_PpotA2_mut+1 R_PpotA2_mut+1 Promoter of sptA +A for luxAB fusion F_PpotA2_m ut+A R_PpotA2_m ut+A Promoter of comX G ^ A for luxAB fusion F_PcomX_mut R_PcomX_mut Promoter of sptA for cat fusion (screen) F_PpotA2_tRNAthr R_PpotA2_cat_AT G cat cassette amplification for P sp w fusion (screen) F cat ATG R_cat_(spec) Diagnostic PCR for random peptide sequencing Pxyl seq spec2

PCR for random peptide backcross UF tRNAser DR2 tRNAser PCR for tRNA Ser ::Pxyii-comR-spec amplification UF tRNAser DR2_tRNAser

Upstream homologous region of scuR gene UFrggC JIM URrggC JIM

Downstream homologous region of scuR gene DFrggC JIM DRrggD SS

Upstream homologous region of scuR gene (in UFrggC JIM rggD_S4_2

frame deletion)

Downstream homologous region 1 of scuR gene rggD_S4_3 rggD_S4_4

(in-frame deletion)

Downstream homologous region2 of scuR gene rggD_S4_5 rggD_S4_6

(in-frame deletion)

Diagnostic PCR for scuR deletion Up rggC JIM Down rggD SS

Upstream homologous region of sarF gene UFrggC SS URrggD JIM

Downstream homologous region of sarF gene DFrggD JIM DRrggC SS

Upstream homologous region of sarF gene (in UFrggC SS rggC_S4_2

frame deletion)

Downstream homologous region 1 of sarF gene rggC_S4_3 rggC_S4_4

(in-frame deletion)

Downstream homologous region2 of sarF gene rggC_S4_5 rggC_S4_6

(in-frame deletion)

Diagnostic PCR for sarF deletion Up rggC SS Down_rggC SS

Diagnostic PCR for scuR-sarF deletion Up rggC JIM Down_rggC SS

Upstream homologous region of comR gene UFcomRJIM-SS1-4 URcomRJIM-SS1-4 Downstream homologous region of comR gene DFcomRJIM-SS1-4 DRcomRJIM-SS1-4 Diagnostic PCR for comR deletion Up_comR SS1-4 Down_com R_SS 1 -4 Upstream homologous region of comA gene UF PcomRJuxAB UR comA

Downstream homologous region of comA gene DF comA DR comA

Diagnostic PCR for comA deletion F comR Down_PcomS_JIMS

S1 -4

Upstream homologous region of sptA gene UF_potA2 UR_potA2

Downstream homologous region of sptA gene DF_potA2 DR_potA2

Diagnostic PCR for sptA deletion Up_potA2 Down_potA2

Promoter of sptA annealing for EMSA Cy 3_F_P pot A2_E MSA R_PpotA2_EMSA Promoter of slvX annealing for EMSA Cy3_F_P01665 E MSA R P01665 EMSA Promoter of comS annealing for EMSA Cy 3_F_P co m S_E M S A R PcomS EMSA Promoter of comX annealing for EMSA Cy3_F_PcomX_EMSA R PcomX EMSA Promoter of comX G ^ A annealing for EMSA Cy3 F PcomX E M S Arev R PcomX EMSArev

Randomized peptide screen

To generate the two DNA libraries encoding randomized sequence of small peptides, we performed overlapping PCRs to graft fragments encompassing the follow features : (1 ) a 5’ recombination arm (for the ectopic tRNA ser locus); (2) the xylR gene that codes for the xylose responsive regulator; (3) either P xy n (library I) or P xy /2 (library II) translationally-fused to a 12 codon long gene, for which the last 7 are randomized; (4) the spec gene; (5) a 3’ recombination arm (for the ectopic tRNA ser locus). To obtain the randomized DNA stretch, we used a 78 nucleotide long primer degenerated on 21 contiguous positions. Next, we transformed these two libraries in strains containing the sptA promoter translationally-fused to the cat gene (chloramphenicol resistance) in which the associated spec gene was excised by the previously described cre-lox method. The initial background of these strains were either a comR-overexpressing (P xy n-comR) or a salivaricin- deprived ( Aslv5 ) strain. We plated transformed cells on solid medium supplemented with xylose (either 0.1 or 1 %), chloramphenicol (2 mg/ml) and spectinomycin (200 mg/ml) and grew overnight. We restreaked single colonies on fresh chloramphenicol and spectinomycin solid medium supplemented or not with xylose. We finally collected clones that displayed an increased in growth on xylose vs non xylose medium (except for the clone BM1 that we used as a negative control). Mobility shift assays (EMSA)

All double-stranded DNA fragments (30 or 40bp) were obtained from annealing of single- stranded Cy3-labelled (at 5’ end) and unlabeled oligonucleotides. Primers used are listed in Table 3. Typically, a gel shift reaction (20 pi) was performed in a binding buffer (20 mM Tris-HCI pH 8.0, 150 mM NaCI, 1 mM EDTA, 1 mM DTT, 10% glycerol, 1 mg ml-1 BSA) and contained 150 ng labelled probe and 4 mM StrepTag proteins. When necessary, 8 mM of ComS peptides (unless otherwise stated) are added. The reaction is incubated at 37°C for 10 min prior to loading of the samples on a native TBE 5% gel. The gel is next subjected to 80 V for approximately 1 h in TBE buffer. DNA complexes were detected by fluorescence on the Ettan DIGE Imager with bandpass excitation filters (nm): 540/25 (Cy3) or 635/30 (Cy5) and bandpass emission filters: 595/25 (Cy3) or 680/30 (Cy5) (GE Healthcare, Waukesha, Wl).

Bacteriocin detection assay

The spot-on lawn (multilayer) detection method was performed as followed: 10 pi of overnight cultures of producer strains were diluted in fresh M17G medium and grown to reach mid-log phase (OD6OO = ~ 0.5). In parallel, we casted plates with a bottom feeding layer (M17G 1.5% agar) supplemented with a synthetic peptide where required. Next, we mixed 100 mI of an overnight culture of Lactococcus lactis IL1403 (indicator strain) in pre-warmed soft M17G medium (0.3% agar) and casted it as a top layer. Finally, we incubated mid-log phase cultures for 30 minutes with the corresponding synthetic peptides and spotted 3mI of the producer strains on the top layer. Plates were incubated overnight before analysis of the inhibition zones surrounding the producer colonies.

Competence induction, transformation rate and engineering of mutants

To induce competence, overnight CDMG precultures were diluted at a final Oϋboo of 0.05 in 300 mI (10 ml concerning the randomized peptide screen) of fresh CDMG and incubated 75 min at

37°C. Then, we added the pheromone sComS as well as DNA (overlapping PCRs or plasmids) and let the cells recover for 3 h at 37°C before plating on M17G agar supplemented with antibiotics where required. Null-mutants were constructed by exchanging (double homologous recombination) the coding sequences (CDS) of target genes (sequence between start and stop codons) for either chloramphenicol or erythromycin resistance cassette. If stated, mutants were cleaned for the lox site-flanked resistance cassette, as previously described (Fontaine et al. Mol Microbiol 2010; 87: 1 1 13-1 132). In case of deletion of multiple CDSs, the region between the start codon of the first CDS and the stop codon of the last CDS was deleted. Integration of the antibiotic resistance cassette at the right location was subsequently checked by PCR. The promoters of spfA genes was fused to the luxAB reporter genes and inserted with a chloramphenicol resistance cassette at the permissive tRNA threonine locus ( HSISS4_r00061 ) by double homologous recombination. In case of AscuR and AsarF in-frame deletion, we used the two-step selection/counter-selection strategy previously described (Mignolet et al. Cell Rep 2018, 22:1627-1638). We transformed the wild-type strain with an overlapping PCR product composed of 4 fragments: (I) the upstream region of scuR or sarF genes, (II) the downstream region of scuR or sarF genes, (III) a cassette that includes the erythromycin resistance gene ( erm ) and a gene encoding the orotate transporter oroP, and finally (IV) the downstream region of scuR or sarF genes. We selected a first event of double recombination on medium supplemented with erythromycin. Next, we selected an intramolecular recombination between region (I) and (IV) that excises the erm-oroP cassette by growing cells on M17G supplemented with the toxic 5-fluoro-orotic acid (5-FOA) compound. In absence of oroP, 5- FOA is not able to cross the membrane and penetrate the cytoplasm where it is deleteriously incorporated in the nucleotide metabolic pathway (Overkamp et al., 2013). At final, we engineered an in-frame deletion mutant of scuR or sarF in which the first seven codons were fused to the last six codons without any cassette scar (see below for detailed cloning method).

ComR, ScuR and SarF purification

The PCR-amplified scuR-StrepTag and gene sarF-StrepTag were cloned into the pBAD- comR-ST vector (see supplemental information). The ComR-StrepTag, ScuR-StrepTag and SarF- StrepTag recombinant proteins were overproduced in E. coli and purified as previously described (Fontaine et al., 2013) in standard native conditions on Strep-Tactin agarose beads (IBA).

Measurements of growth and luciferase activity

Overnight precultures were diluted at a final Oϋboo of 0.05. A volume of 300 pi of culture samples was incubated in the wells of a sterile covered white microplate with a transparent bottom (Greiner, Alphen a/d Rijn, The Netherlands) for 75 min at 37°C and then supplemented with synthetic peptides (1 mM, except if otherwise stated) or DMSO, and xylose where required. Growth (ODeoo) and luciferase (Lux) activity (expressed in relative light units) was monitored at 10 min intervals during 24 h in a multi-wells plate reader (Hidex Sense, Hidex, Turku, Finland) as previously described (Fontaine et al., 2013).

Deep sequencing (RNAseq) and data processing

S. salivarius WT, AscuR, AsarF, scuR ++ or AsarF-ST ++ strains were pre-cultured overnight in CDMG at 37°C. They were resuspended in 50 ml of fresh pre-warmed CDMG to a final Oϋboo of 0.05 and grown for approximately 2 h 30 min (Oϋboo = 0.3) at 37°C. Cells were harvested by centrifugation (10 min; 4,050 * g), the supernatant were discarded and the cell pellets were frozen with liquid nitrogen. Finally, RNA was extracted using the RiboPure bacteria kit (Ambion-Life Technologies) and the protocol provided by the manufacturer, with protocol changes to cell lysis and RNA precipitation. For lysis, cells were resuspended in RNAwiz buffer (Ambion-Life Technologies) supplemented with Zirconia beads and shaked for 40 sec (4 times) in a fastPrep homogenizer device (MP biomedicals). For RNA precipitation, a 1.25-ethanol volume (instead of 0.5) was added to partially purified RNAs. Total RNA was checked for quality on a RNA Nano chip (Agilent technologies) and concentration was measured using Ribogreen assay (Life technologies). rRNA depletion was performed on 2 pg total RNA with the Ribo-Zero rRNA removal kit for Grampositive bacteria (lllumina) according to manufacturer’s instructions. Total stranded mRNA libraries were prepped with the NEBNext Ultra Directional RNA Library Prep kit for lllumina (New England Biolabs). Library PCR was executed for 15 cycles. Quality of the libraries was evaluated with the use of a High sensitivity DNA chip (Agilent technologies) and concentrations were determined through qPCR according to lllumina protocol. Libraries were sequenced on a NextSeq 500 high- throughput run with 76 bp single reads. 2.3 pM of the library was loaded on the flowcell with a Phix spike-in of 5%. Sequenced mRNAs generated several million reads that were mapped on the WT S. salivarius chromosome and processed with both bowties V0.12.9 (http://bowtie- bio.sourceforge.net/bowtie2) and samtools VO.1.18 (http://samtools.sourceforge.net/) algorithms to yield BAM files containing the read coordinates. We imported these files into SeqMonk V0.23.0 (www.bioinformatics.babraham.ac.uk/projects/) to assess the total number of reads for each coding sequence (CDS). The dataset was exported into an excel file for further analyses. First, the dataset was standardized to CDS-mapped reads per million overall reads. Then, we estimated a ratio of CDS-mapped reads in mutants vs WT. All RNAseq data was deposited in the GEO database under accession number GSE120640.

Plasmid and Linear DNA fragment constructions

All DNA fragments were amplified by PCR using the Phusion high fidelity polymerase (www.thermoscientificbio.com/) following a protocol as recommended by the manufacturer. Overlapping PCR products were transferred in competence-induced HSISS4 derivatives (Mignolet at al. Genome Announc 2016, 4:e01637-01615). cat, erm, spec, erm-oroP, P xy n, P xyi 2, and luxAB- cat cassettes were amplified from pNZ5319, pGIUD0855ery, pJUD specm ut 1 -gfp + te , pSEUDO- P usp 45- sf gfp(Bs), pZX9, pZX10 and pJIMcaf, respectively. comX and sptA mutated promoter were amplified from the WT com and sptA luxAB-fused promoter strain, respectively. The sarF-ST allele was amplified from pBAD-sarF-ST. The full P xy n-comR-spec at tRNA Ser locus was amplified in one block from the strain tRNA Ser ::P xy n-comR-spec (Mignolet et al. Cell Rep 2018, 22: 1627-1638). All the constructed plasmids were sequence-verified. The sci/R-coding sequence was PCR amplified using the rggD_Ncol SS

and rggD_Munl primers. This scuR fragment was digested with Nco\IMun\ and cloned into L/col/EcoRI-digested pBAD-comR-ST (Mignolet et al. Cell Rep 2018, 22:1627-1638). pBAD-sarF-ST. The sarF-coding sequence was PCR amplified using the rggC_Ncol SS and rggC_RI SS primers. This sarF fragment was digested with Nco\l EcoR\ and cloned into L/col/EcoRI-digested pBAD-comR-ST (Mignolet et al. Cell Rep 2018, 22:1627-1638).

Example 1 : Requlon interweaving in ComR paraloqs

The bacteriocin short-circuitry imposed by ComR in the S. salivarius species is startling and suggests a positive selection for species-specific strategies that participate in niche adaptation. Interestingly, the S. salivarius HSISS4 genome encodes five RRNPP transcriptional factors, including ComR. Besides it, the two regulators ScuR (HSISS4_01 166; stands for salivaricins- competence uncoupling regulator) and SarF (HSISS4_01 169; ScuR-associated Rgg factor) share a high level of similarity with ComR (64 and 63%, respectively). The residues involved in HTH sequestration and homodimerization of ComR are well-conserved in both ScuR and SarF, suggesting that they could display a similar mode of activation. Moreover, the paralogous ScuR and SarF proteins are highly identical (similarity of 91 %). Strikingly, residue divergences are nearly all concentrated in only 3 amino-acids stretches, one of which overlaps the a-helices 13 and 14 that form a part of the peptide recognition pocket. This indicates that the two proteins are likely to homodimerize and could accommodate specific peptides. On the chromosome, the scuR and sarF genes are located in a unique locus and separated by two genes that code for two predicted subunits of an ABC transporter, SptA and SptB (for ScuR-promoted transporter A and B, respectively) (FIG 1A). In contrast to characterized rgglcomR loci, no small coding sequence was distinguishable upstream or downstream of scuR and sarF genes, arguing for a different genomic coding topology of the communication system.

Due to the huge conservation between ComR, ScuR and SarF, especially in the DNA binding domain, we questioned whether the two uncharacterized paralogs are capable to control competence and predation as well. Hence, we extracted mRNA of wild-type (WT) and engineered in-frame deletion mutants ( AscuR and AsarF) and carried out a deep sequencing (RNAseq). With no hint about the genuine activating pheromones, we included in our high-throughput transcriptomic analyses overexpression mutants ( scuR ++ and sarF-ST ++ ), in order to exacerbate the regulation phenotype. Indeed, a strong overproduction of ComR was reported to be sufficient for activation of its target promoters, even in absence of ComS (from endogenous production or synthetic peptide addition). Both deletion mutants did not dramatically affect the transcriptome compared to the WT strain, meaning that the function of ScuR and SarF is barely noticeable during standard growth conditions. However, the SarF loss slightly increased scuR expression, while the differential sptA and sptB mRNA level almost reached the arbitrary 5-fold induction cut-off, suggesting that SarF could be a repressor/antagonist of the ScuR-SptAB system. In contrast, the strong overexpression of scuR (28-fold) elicited a tremendous activation of the sptA-sptB operon (about 2000-fold). Furthermore, a second cluster of genes, all located inside salivaricin loci, was robustly influenced, even if with a lower magnitude of activation (ranging from 35- to 140-fold). Surprisingly, comX mRNA levels remained approximately stable in all mutants.

Altogether, these results imply that the ComR, ScuR and SarF paralogs might shape overlapping but dedicated regulatory networks.

Example 2: ScuR is an alternative self-sufficient pathway that controls salivaricin production but maintains the competence off

In order to validate our transcript profile analyses, we performed promoter-probe assays, as previously described for ComR. We first expanded our collection of luciferase reporter strains (composed of comS, comX and bacteriocin gene promoters) to include and monitor the sptA promoter (P sp w) and, next transformed all of them with the scuR overexpression cassette. Finally, we measured promoter activity in presence or absence of sComS (synthetic peptide) during cell growth (FIG 1 B). In agreement with our RNA-seq data, sptA and bacteriocin promoters were all markedly up in the sct/R ++ strain, irrespective of the addition of sComS, and with no significant synergy. In addition, we observed no activation of the P ¥m x due to ScuR accumulation, disabling ScuR as a trigger of competence state. Nonetheless, the P ¥m s showed a 35-fold change in activity, suggesting that ScuR might modulate the ComR cell signaling. To complement our understanding of this bipartite system, we assessed the activity of P sp w, P ¥m s and P s/V x in a sarF-ST ++ strain, and noticed that scuR and sarF overexpression governs P sp tA activation amplitude in a similar range, while P com s and P s/V x are irresponsive to SarF (FIG 1 C).

Considering that the effects of transcriptional regulators could be indirect, we were prompted to inactivate one Rgg by gene deletion in our reporter strains and test the residual activity of the others. We discovered that ScuR still controls both P sp tA and P ¥m s in comR (FIG 1 D) or sarF deleted strains (FIG 1 E), while SarF is sufficient to activate P sp tA even in absence of scuR ( AscuR-sarF/sarF - ST ++ mutant) (FIG 1 E). Finally, the sComS-mediated regulation of ComR is not crippled in absence of both ScuR and SarF (FIG 1 F), suggesting that each regulator can stand alone to fulfill its function and work in parallel.

Taken together, our results suggest that the 3 transcriptional factors have only partly redundant functions, with regulatory network specificities, presumably to ensure a broader diversity of cellular response to environment stresses. ScuR and SarF, but not ComR, control the sptAB operon, while ScuR alone has a ComR-independent extra regulatory role on bacteriocin production. Even though ScuR raises ComS production, this regulator does not act on comX promoter and is likely to disconnect the competence-predation coupling compelled by ComR.

Example 3: Randomization-based screen for pheromone identification

Typically, the major challenge to address the transduction mechanism of cell-cell communication sensor is the identification of the ligand(s) or the perceived signal(s). As inspection of the genomic neighborhood did not reveal any small encoded peptide in the vicinity of scuR-sarF locus, we decided to conduct an empirical screen to unearth peptides able to activate the ScuR- SarF system (FIG 2A). Thence, we constructed on one side a strain harboring a translational fusion of the ScuR/SarF-specific P sp tA to a gene conferring resistance to chloramphenicol (cat). On the other side, we amplified a DNA fragment that allows recombination at a permissive locus (tRNA Ser ) and encompasses, under xylose control, a 12 codons-long nucleotide sequence, the last 7 of which are randomized (see Experimental Procedures). We finally transformed this PCR product in the above-mentioned reporter mutant and selected clones on plates supplemented with chloramphenicol and xylose (0.1 or 1 %). Note that, in order to increase the transformation rate or decrease the cytotoxicity due to concomitant bacteriocin production, we worked in comR overexpression (P xy n-comR) or salivaricin deprived ( Aslv5 ) background, respectively. In total, over 9 runs of screen, we collected a hundred of clones that we streaked again on selective medium with and without xylose. We sent for sequencing clones that showed a clear improvement of growth due to xylose on chloramphenicol (Table 5). As a negative control, we included in our further analyses a clone (BM1 ) with a xylose-independent growth. Out of the 30 positive clones, 22 harbored a non- redundant peptide/nucleotide sequence. Table 5

In order to discard clones with secondary mutations for which the survival phenotype was not related to the peptide nature, we amplified for each clone the full locus that encodes the small peptide and backcrossed it into WT or ( Aslv5 ) backgrounds. We then confirmed on solid media that chloramphenicol resistance qualitatively increased upon xylose addition. We used the same PCR products to transform a strain bearing the P sptA -luxAB report fusion and quantitatively estimate the influence of peptide production. Again, we noticed that xylose addition potentiated the promoter activity with values ranging from 5 to 100 fold, while it has no effect on BM1 (negative control) and WT strains (FIG 2B).

We aligned the 22 unique peptide sequences to elicit common chemical properties (FIG 2C).

Strikingly, a tryptophan residue is highly conserved at position -5 from the C-terminus. On the top of this, the adjoined position (-6) is mainly occupied with an aromatic residue. Finally, the position - 1 is preferentially a glycine. Positions -2, -3 and -4 are more erratic, even if we observed a tendency for hydrophobic amino acids. Given that it encodes a peptide (MIAILPFWJJLG) that neatly mimics the consensus sequence (MIAILPFWLVLG), we decided to hereafter focus on clone BI7, and we disclosed that ScuR is specifically responsible for the xylose-driven phenotype. Indeed, neither comR nor sarF deletion has a dramatic effect, while ScuR loss annihilates both xylose induction and basal leaky expression (FIG 2D). Example 4: Exogenous pheromones activate the ScuR-SarF tandem

We next checked whether we could, akin to ComS toward ComR, activate the system with synthetic peptides (FIG 3A). We therefore selected a representative panel of peptides from our screen and ordered the synthesis of the last 8 amino acids and tested P sp tA activation (FIG 3B). Whatever their degree of kinship toward the consensus motif, the peptides were capable of inducing light production when supplemented to the medium. However, a weaker activation was displayed by the peptides that diverge the most from the paradigm such as sBK3, which does not harbor a C- terminus glycine, or sBK4, for which the tryptophan and glycine are shifted of one position (exacerbated effect at the non-saturating concentration of 0.01 mM). The huge variability in sequence and the similar amplitude of activation for all other peptides emphasizes that (1 ) the residues between the conserved tryptophan and glycine and the proline (position -7) are not essential for ScuR or SarF transactivation, while the position -6 tolerates substitutions as far as the peptide nature is aromatic. Moreover, rational mutations of the ultra-conserved tryptophan evidenced the strict requirement of the indole moiety, considering that neither alanine (sBI7 w ^ A ) nor phenylalanine (sBI7 w ^ F ) variants sustains luciferase transcription at low peptide concentration (0.001 mM, FIG 3C). A similar strategy for the C-terminal glycine showed that its substitution by an alanine (sBI7 G ^ A ) decreases the P sp tA response although to a lesser extent compare to the tryptophan (FIG 3C). It is noteworthy that high concentration of sBI7 w ^ F and sBI7 G ^ A (but not sBI7 w ^ A ) can bypass the requirement of the tryptophan and glycine and activate P ca t at a similar range than the WT sBI7 peptide (FIG 3E), suggesting that the mutations do not totally abrogate the ScuR/SarF activation but rather modulate the kinetics of interaction.

Example 5: Selective recognition of targeted promoters

To refine our view of the 2 Rgg-systems, P sp tA, P ¥ms and P s/V x were challenged with increasing amount of sBI7 at low concentration in WT, AscuR or AsarF strains (FIG 3D). In line with our overexpression data, P ¥m s and P s/V xwere totally insensitive to SarF (no activity in AscuR), while both ScuR and SarF can turn on P sp tA independently of each other. Furthermore, in WT backgrounds, we observed that all promoters were responsive to less than 1 nM of peptide, with the highest amplitude for P sp tA and the lowest for P ¥m s· Whereas activity of all promoters in AsarF were slightly up compared to the WT, supporting the notion that SarF might have a mild inhibitory effect on ScuR function. Expectedly, the induction provoked by sBI7 addition is utterly erased in a AscuR- SarF double mutant (FIG 3F), however sBI7 is surprisingly able to induce the SarF-mediated P sp tA response (FIG 3D). This discrepancy in regard to the genome-encoded peptide results (FIG 2D) might be due to inherent differences imposed by the screening method compared to the exogenous supplementation of a synthetic peptide (variable intracellular concentration, different peptide length, lower activation rate of SarF,...). But it underlines that the peptide-binding pocket of both Rgg could accommodate a unique pheromone.

Next, we carried out in vitro mobility shift assays to assess the direct interaction between proteins and promoter probes in absence or presence of decreasing concentration of synthetic peptides. We included a P ¥m x probe as a negative control. Anew, we corroborated our promoter activity data at the magnitude and protein-peptide/DNA specificity level (FIG 4A). The ScuR regulator displayed the weakest affinity for P ¥m s, and the strongest one toward P SP M, starting from 0.08nM of sBI7 peptide and complexing the full amount of probe at maximal concentrations (presence of a doublet presumably due to a second higher order of oligomerization) (FIG 4A). We observed similar results with unique concentration of sBI7 and decreasing concentration of ScuR on P sp tA and P s/V x probe (FIG 4G). Moreover, in comparison to the sComS-bound ComR, the ScuR » sBI7 affinity for P ¥m s and P s/V x appears weaker with a less stable complex (probe smear) (FIG 4A). Remarkably, the ScuR and SarF binding progressively and linearly increased with the amount of peptide, contrasting with ComR that showed a smaller interval between sub-activating and saturating concentrations of sComS. This suggests ScuR and SarF have a different dynamic of binding compared to ComR that might reflect the congruence between reactivity and physiological function. Finally, the ComR » sComS pair can unexpectedly bind the P sp tA probe, indicating that the sptA promoter topology, and presumably the distance between the palindrome center and the -10 box, might be crucial to dictate the specific ScuR/SarF-driven transactivation (FIG 4A).

The topology of ScuR, SarF or ComR responsive promoters appears extremely similar (FIG 4B and 4H). The architecture of every promoter includes a conserved nucleotide stretch of dyad symmetry and a T-rich spacer that separates it from the sigma-bound -10 box (exact same length in all promoters, P sp tA apart). However, the core palindromic region of the ComR-specific P ¥m x includes a mismatch, while the equivalent stretch in the ScuR- and SarF-specific P sp tA is more extended, comprises 2 mismatches and is closer to the -10 box from one nucleotide (FIG 4B). Considering the high degree of similitude between promoters at the primary sequence level (FIG 4B and 4H), we were prompted to address the nucleotide determinants that impose the protein- DNA selectivity. We therefore mutated comX and sptA promoters to sensitize them toward ScuR and ComR, respectively. In P ¥m x, we substituted a guanosine for an adenosine (P ¥m x G ^) to reconstitute the palindromic region observed in P ¥m s and P s/V x (FIG 4B). Concerning P sp w, we performed three kinds of mutation (FIG 4B). We reconstituted the symmetric region with 2 substitutions (P sp tA CT ^ AC ), we inserted a nucleoside in the T-rich stretch to redefine the same distance between palindrome center and -10 box (P sPM +i ), and finally, with the mere insertion of an adenosine in the palindrome (P sp tA +A ), we redesigned both space and palindrome. Non- ambiguously, all these mutations impinged on selectivity at different extent, driving the engineered promoters sensitive to both pheromones (FIG 4C and 4D). In agreement with this, ScuR, but not SarF, is able to shift the P COm x G ^ A probe (FIG 4E). As competence entry hinders cell fitness whereas the mutation in comX promoter bolstered the ComRS-mediated activation (FIG 4D), we suspect that evolution maintains a selective pressure to ensure an appropriate expression in time and scale of comX compatible with the bacterium life cycle.

Example 6: Pheromone-induced ScuR promotes bacteriocin production

The lower reactivity of salivaricin promoters toward ScuR (vs ComR) pheromone incited us to investigate the phenotypical output at the bacteriocin production level. Thence, we performed a standard bacteriocin test on soft overlay and showed that the sct/R ++ (but not sarF-ST ++ ) overexpression mutant is able to produce a comA-dependent inhibitory halo in bacteriocin tests (FIG 5A). Even if the overexpression of scuR is more potent, sBI7 induced a small halo of inhibition around the WT strain for concentration ranging from 1 nM to 1 mM (FIG 5B). This effect is eradicated in single AscuR or double AscuR-sarF mutants, or in a strain deprived of bacteriocin genes ( Aslv5 ), demonstrating that the toxicity is due to bacteriocins and mediated by ScuR (FIG 5B). Example 7: Rqq members requisition predation control in S. salivarius

ScuR tends to compensate for the loss/lack of a functional BlpRH system, as they usually do not co-exist in the same strain (FIG 6A). Oddly, the direct control of competence and bacteriocin network drifts from a full TCS control in Streptococcus pneumoniae (ComCDE and BlpRHC) to a full Rgg regulation in S. salivarius (ComRS and ScuR), going through an hybrid mechanism in mutans, bovis and pyogenes streptococci groups (ComRS and BlpRHC) (FIG 6B). Why opposing extracellular sensing vs intracellular sensing inside streptococci is a vast question. As a genuine member of the gastro-intestinal tract, S. salivarius is under a grueling selective pressure, competing for resources and territories in a constantly changing environment. The most tantalizing hypothesis is that a cytoplasmic receptor could be better protected from communication interferences. Indeed, quenching molecules extruded by competitors face the chemical selectivity of the semi-permeable cell membrane to penetrate the cell. A second hypothesis is that internal cues, such as starvation/lushness, might impinge on the nutritional oligopermease system (Opp) to globally modulate small peptide inward fluxes and make the cell transiently communication-less or superreactive to pheromones in particular stressful situations.

All our phenotypical and molecular data coincide to conclude that ScuR is strictly devoted for predation in contrast to competence. Although highly similar to P ¥m s and P s/v x at primary sequence level, comX promoter cannot be occupied (FIG 4A) nor activated (FIG 1 B and 4D) by ScuR. Inconsistently, this regulator turns on P ¥m s and should somehow modulate the ComRS activity through a flicking impetus in the positive feedback loop. An obvious reason for this discrepancy might be that the ScuR-driven expression of comS (FIG 1 B and 3D) is weaker compared to our previous observations for ComR. Therefore it is not sufficient to reach the activation threshold, but it might ensure a basal production of ComA to extrude bacteriocins. Alternatively, we suspect that promoters are not responsive to ComR and ScuR pheromone with the same timeframe. In this case, ScuR might promote ComS production in a time interval during which the comX promoter is silenced or unreactive.

Example 8: induction of bacteriocin promoters, no induction of competence genes

Confirming the importance of the ultra-conserved tryptophane residue (see FIG 3C), we observed similar trends for all bacteriocin gene promoters (FIG 7), with a total inefficacy of the peptide sBI7 w ^ A , and a low to high activation disability for peptides sBI7 w ^ F and sBI7 G ^ A .

It was also confirmed that ScuR and SarF have no effect on the sComS-mediated competence entry (FIG 8). Further confirming this finding, sBI7 cannot activate comX and late competence genes under direct ComX control (FIG 9), and is ineffective to support natural transformation (Table 6). Table 6: Competence development (transformation frequency 3 ) in S. salivarius HSISS4 derivatives

acalculated as the ratio of transformants (chloramphenicol-resistant CFU) to the total CFU count per 0.1 ug of linear DNA. Transformation frequencies are expressed as the arithemtic mean of three independent experiments. Geometric means ± standard deviations are provided. ND: not detected (<1.0 E-08), NA: not applicable.

Example 9: C-terminal residues and bacteriocin induction activity

To evaluate whether the synthetic pheromones could tolerate amino acid addition at the C- terminus, we tested the close-to-consensus petide LAFWDSLG (SEQ ID NO: 749) as well as the peptide LAFWDSLGLLL (SEQ ID NO: 750) that harbors a triple leucine extension at the C-terminus. Strikingly, both peptides were able to induce the sptA promoter in vivo. However, the longer peptide was much more active at 1 mM concentration than the shorter one, with an induction magnitude comparable to the peptide sBI7 (FIG 10A). By contrast, mobility shift assay performed with the very same peptides showed that ScuR binding to P sp tA is more intense with LAFWDSLG (SEQ ID NO: 749), while it requires extreme concentration of the peptide LAFWDSLGLLL (SEQ ID NO: 750) to get activated (FIG 10B). This suggests that LAFWDSLGLLL (SEQ ID NO: 750) can barely bind ScuR by itself but is processed in vivo, by one or more peptidases, to liberate an active peptide, presumably LAFWDSLG (SEQ ID NO: 749). It is thus clear that the peptides can tolerate amino acids at the C-terminus and still be active in vivo.

References

Chopin, A., Chopin, M.C., Moillo-Batt, A., and Langella, P. (1984). Two plasmid-determined restriction and modification systems in Streptococcus lactis. Plasmid 11, 260-263.

Fontaine, L., Dandoy, D., Boutry, C., Delplace, B., de Frahan, M.H., Fremaux, C., Horvath, P., Boyaval, P., and Hols, P. (2010). Development of a versatile procedure based on natural transformation for marker-free targeted genetic modification in Streptococcus thermophilus. Appl Environ Microbiol 76, 7870-7877.

Fontaine, L., Goffin, P., Dubout, H., Delplace, B., Baulard, A., Lecat-Guillet, N., Chambellon, E., Gardan, R., and Hols, P. (2013). Mechanism of competence activation by the ComRS signalling system in streptococci. Mol Microbiol 87, 1 1 13-1 132.

Haustenne, L., Bastin, G., Hols, P., and Fontaine, L. (2015). Modeling of the ComRS Signaling Pathway Reveals the Limiting Factors Controlling Competence in Streptococcus thermophilus. Front Microbiol 6, 1413. Lambert, J.M., Bongers, R.S., and Kleerebezem, M. (2007). Cre-lox-based system for multiple gene deletions and selectable-marker removal in Lactobacillus plantarum. Appl Environ Microbiol 73, 1 126-1 135.

Mignolet, J., Fontaine, L., Kleerebezem, M., and Hols, P. (2016a). Complete Genome Sequence of Streptococcus salivarius HSISS4, a Human Commensal Bacterium Highly Prevalent in the Digestive Tract. Genome Announc 4, e01637-01615

Mignolet, J., Fontaine, L., Sass, A., Nannan, C., Mahillon, J., Coenye, T., and Hols, P. (2018). Circuitry Rewiring Directly Couples Competence to Predation in the Gut Dweller Streptococcus salivarius. Cell Rep 22, 1627-1638.

Mignolet, J., Holden, S., Berge, M., Panis, G., Eroglu, E., Theraulaz, L., Manley, S., and

Viollier, P.H. (2016b). Functional dichotomy and distinct nanoscale assemblies of a cell cycle- controlled bipolar zinc-finger regulator. Elife 5, e18647.

Overkamp, W., Beilharz, K., Detert Oude Weme, R., Solopova, A., Karsens, H., Kovacs, A., Kok, J., Kuipers, O.P., and Veening, J.W. (2013). Benchmarking various green fluorescent protein variants in Bacillus subtilis, Streptococcus pneumoniae, and Lactococcus lactis for live cell imaging . Appl Environ Microbiol 79, 6481-6490.

Van den Bogert, B., Boekhorst, J., Herrmann, R., Smid, E.J., Zoetendal, E.G., and Kleerebezem, M. (2014). Comparative genomics analysis of Streptococcus isolates from the human small intestine reveals their adaptation to a highly dynamic ecosystem. PLoS One 8, e83418.

Yu, J., Sun, Z„ Liu, W„ Xi, X., Song, Y„ Xu, H., Lv, Q., Bao, Q., Menghe, B„ and Sun, T.

(2015). Multilocus sequence typing of Streptococcus thermophilus from naturally fermented dairy foods in China and Mongolia. BMC Microbiol 15, 236.