R. M. Donlan, Biofilms: microbial life on surfaces, Emerging infectious diseases, vol.8, issue.9, pp.881-890, 2002.

H. Flemming and S. Wuertz, Bacteria and archaea on Earth and their abundance in biofilms, Nature Reviews Microbiology, vol.17, issue.4, pp.247-260, 2019.

R. Sender, S. Fuchs, and R. Milo, Revised estimates for the number of human and bacteria cells in the body, PLOS Biology, vol.14, issue.8, p.1002533, 2016.

L. M. Feazel, L. K. Baumgartner, K. L. Peterson, D. N. Frank, J. K. Harris et al., Opportunistic pathogens enriched in showerhead biofilms, Proc Natl Acad Sci, vol.106, issue.38, p.16393, 2009.

L. Neu, C. Bänziger, C. R. Proctor, Y. Zhang, W. Liu et al., Ugly ducklings-the dark side of plastic materials in contact with potable water, npj Biofilms and Microbiomes, vol.4, issue.1, p.7, 2018.

S. Yoshida, A bacterium that degrades and assimilates poly(ethylene terephthalate), Science, vol.351, issue.6278, p.1196, 2016.

L. , C. Simões, and M. Simões, Biofilms in drinking water: problems and solutions, RSC Adv, vol.3, issue.8, pp.2520-2533, 2013.

H. Flemming, J. Wingender, U. Szewzyk, P. Steinberg, S. A. Rice et al., Biofilms: an emergent form of bacterial life, Nature Reviews Microbiology, vol.14, p.563, 2016.

N. Høiby, A personal history of research on microbial biofilms and biofilm infections, Pathogens and Disease, vol.70, issue.3, pp.205-211, 2014.

H. Flemming and J. Wingender, The biofilm matrix, Nature Reviews Microbiology, vol.8, p.623, 2010.

G. A. O'toole and G. C. Wong, Sensational biofilms: surface sensing in bacteria, Current Opinion in Microbiology, vol.30, pp.139-146, 2016.

P. Stoodley, K. Sauer, D. G. Davies, and J. W. Costerton, Biofilms as complex differentiated communities, Annu. Rev. Microbiol, vol.56, issue.1, pp.187-209, 2002.

, National Institute of Health, Research on microbial biofilms: PA Number: PA-03-047, 2002.

W. M. Dunne, Bacterial adhesion: seen any good biofilms lately?, Clinical microbiology reviews, vol.15, issue.2, pp.155-166, 2002.

G. B. Pier, Human monoclonal antibodies to Pseudomonas aeruginosa alginate that protect against Infection by both mucoid and nonmucoid Strains, J. Immunol, vol.173, issue.9, p.5671, 2004.

S. Ramírez-estrada, B. Borgatta, and J. Rello, Pseudomonas aeruginosa ventilator-associated pneumonia management, Infection and drug resistance, vol.9, pp.7-18, 2016.

N. Safdar, C. Dezfulian, H. Collard, and S. Saint, Clinical and economic consequences of ventilatorassociated pneumonia: a systematic review, Critical Care Medicine, vol.33, issue.10, pp.2184-2193, 2005.

D. A. Rosen, T. M. Hooton, W. E. Stamm, P. A. Humphrey, and S. J. Hultgren, Detection of intracellular bacterial communities in human urinary tract infection, PLoS medicine, vol.4, issue.12, pp.329-329, 2007.

, Department of Health and Human Services, Centers for Disease Control and Prevention, issue.06, 2019.

M. J. Soto-giron, Biofilms on hospital shower hoses: characterization and implications for nosocomial infections, Applied and environmental microbiology, vol.82, issue.9, pp.2872-2883, 2016.

H. Whiley, J. Hinds, J. Xi, and R. Bentham, Real-time continuous surveillance of temperature and flow events presents a novel monitoring approach for hospital and healthcare water distribution systems, International Journal of Environmental Research and Public Health, vol.16, issue.8, 2019.

Y. Zhang and W. Liu, The application of molecular tools to study the drinking water microbiomecurrent understanding and future needs, Critical Reviews in Environmental Science and Technology, vol.49, issue.13, pp.1188-1235, 2019.

A. A. Latorre, Biofilm in milking equipment on a dairy farm as a potential source of bulk tank milk contamination with Listeria monocytogenes, Journal of Dairy Science, vol.93, issue.6, pp.2792-2802, 2010.

S. Galié, C. García-gutiérrez, E. M. Miguélez, C. J. Villar, and F. Lombó, Biofilms in the food industry: health aspects and control methods, Frontiers in microbiology, vol.9, pp.898-898, 2018.

J. Whitehead and B. Lake, Recent trends in unpasteurized fluid milk outbreaks, legalization, and consumption in the United States, PLoS currents, vol.10, 2018.

R. C. Santos-mendonça, A. M. Morelli, J. A. Pereira, M. M. De-carvalho, and N. L. De-souza, Prediction of Escherichia coli O157:H7 adhesion and potential to form biofilm under experimental conditions, Food Control, vol.23, issue.2, pp.389-396, 2012.

J. Denny, M. Bhat, and K. Eckmann, Outbreak of Escherichia coli O157:H7 associated with raw milk consumption in the Pacific Northwest, Foodborne Pathogens and Disease, vol.5, issue.3, pp.321-328, 2008.

, Alerte sanitaire. « La Fromagerie Alpine » : informations complémentaires sur les fromages au lait cru concernés par le rappel. Site du Ministère de l'Agriculture et d'Alimentation, 2019.

N. Marouani-gadri, O. Firmesse, D. Chassaing, D. Sandris-nielsen, N. Arneborg et al., Potential of Escherichia coli O157:H7 to persist and form viable but non-culturable cells on a foodcontact surface subjected to cycles of soiling and chemical treatment, International Journal of Food Microbiology, vol.144, issue.1, pp.96-103, 2010.

L. L. Nesse, Potentially pathogenic Escherichia coli can form a biofilm under conditions relevant to the food production chain, Appl. Environ. Microbiol, vol.80, issue.7, p.2042, 2014.

S. Singh, S. K. Singh, I. Chowdhury, and R. Singh, Understanding the mechanism of bacterial biofilms resistance to antimicrobial agents, The open microbiology journal, vol.11, pp.53-62, 2017.

W. Hengzhuang, H. Wu, O. Ciofu, Z. Song, and N. Høiby, In vivo pharmacokinetics/pharmacodynamics of colistin and imipenem in Pseudomonas aeruginosa biofilm infection, Antimicrobial agents and chemotherapy, vol.56, issue.5, pp.2683-2690, 2012.

D. Campoccia, L. Montanaro, and C. R. Arciola, A review of the biomaterials technologies for infectionresistant surfaces, Biomaterials, vol.34, issue.34, pp.8533-8554, 2013.

F. Achouri, ZnO nanorods with high photocatalytic and antibacterial activity under solar light irradiation, Materials, vol.11, issue.11, p.2158, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01910693

H. A. Foster, I. B. Ditta, S. Varghese, and A. Steele, Photocatalytic disinfection using titanium dioxide: spectrum and mechanism of antimicrobial activity, Applied Microbiology and Biotechnology, vol.90, issue.6, pp.1847-1868, 2011.

H. Wu, C. Moser, H. Wang, N. Høiby, and Z. Song, Strategies for combating bacterial biofilm infections, International journal of oral science, vol.7, issue.1, pp.1-7, 2014.

C. T. O'loughlin, L. C. Miller, A. Siryaporn, K. Drescher, M. F. Semmelhack et al., A quorum-sensing inhibitor blocks Pseudomonas aeruginosa virulence and biofilm formation, Proc Natl Acad Sci, vol.110, issue.44, p.17981, 2013.

T. Bjarnsholt, Garlic blocks quorum sensing and promotes rapid clearing of pulmonary Pseudomonas aeruginosa infections, Microbiology, vol.151, issue.12, pp.3873-3880, 2005.

Z. Song, Panax ginseng has anti-infective activity against opportunistic pathogen Pseudomonas aeruginosa by inhibiting quorum sensing, a bacterial communication process critical for establishing infection, Phytomedicine : international journal of phytotherapy and phytopharmacology, vol.17, issue.13, pp.1040-1046, 2010.

K. Lovetri and S. Madhyastha, Antimicrobial and antibiofilm activity of quorum sensing peptides and peptide analogues against oral biofilm bacteria, Antimicrobial Peptides: Methods and Protocols, pp.383-392, 2010.

U. Römling, M. Y. Galperin, and M. Gomelsky, Cyclic di-GMP: the first 25 years of a universal bacterial second messenger, Microbiology and molecular biology reviews : MMBR, vol.77, issue.1, pp.1-52, 2013.

R. M. Dedrick, Engineered bacteriophages for treatment of a patient with a disseminated drugresistant Mycobacterium abscessus, Nature Medicine, vol.25, issue.5, pp.730-733, 2019.

S. K. Hansen, P. B. Rainey, J. A. Haagensen, and S. Molin, Evolution of species interactions in a biofilm community, Nature, vol.445, p.533, 2007.

S. Elias and E. Banin, Multi-species biofilms: living with friendly neighbors, FEMS Microbiology Reviews, vol.36, issue.5, pp.990-1004, 2012.

C. Hill, The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic, Nature Reviews Gastroenterology &Amp

, Hepatology, vol.11, p.506, 2014.

M. E. Sanders, How do we know when something called 'probiotic' is really a probiotic? A guideline for consumers and health care professionals, Functional Food Reviews, vol.1, pp.1-12, 2009.

V. Hancock, M. Dahl, and P. Klemm, Probiotic Escherichia coli strain Nissle 1917 outcompetes intestinal pathogens during biofilm formation, Journal of Medical Microbiology, vol.59, issue.4, pp.392-399, 2010.

I. Y. Hwang, Engineered probiotic Escherichia coli can eliminate and prevent Pseudomonas aeruginosa gut infection in animal models, Nature Communications, vol.8, p.15028, 2017.

K. Fang, X. Jin, and S. H. Hong, Probiotic Escherichia coli inhibits biofilm formation of pathogenic E. coli via extracellular activity of DegP, Scientific Reports, vol.8, issue.1, p.4939, 2018.

Q. Chen, Probiotic E. coli Nissle 1917 biofilms on silicone substrates for bacterial interference against pathogen colonization, Acta Biomaterialia, vol.50, pp.353-360, 2017.

K. Lohith and K. A. Anu-appaiah, Antagonistic effect of Saccharomyces cerevisiae KTP and Issatchenkia occidentalis ApC on hyphal development and adhesion of Candida albicans, Medical Mycology, vol.56, issue.8, pp.1023-1032, 2018.

E. Metchnikoff, The Prolongation of Life. Optimistic Studies, p.96, 1908.

P. A. Mackowiak, Recycling metchnikoff: probiotics, the intestinal microbiome and the quest for long life, Frontiers in public health, vol.1, pp.52-52, 2013.

, Probiotics Market Size, Share & Trends Analysis Report By Application (Food & Beverages, Dietary Supplements, Animal Feed), By End-use, By Region, And Segment Forecast, 2018.

M. Bermudez-brito, J. Plaza-díaz, S. Muñoz-quezada, C. Gómez-llorente, and A. Gil, Probiotic mechanisms of action, Annals of Nutrition and Metabolism, vol.61, issue.2, pp.160-174, 2012.

M. Nair, M. A. Amalaradjou, and K. Venkitanarayanan, Chapter One -Antivirulence properties of probiotics in combating microbial pathogenesis, Advances in Applied Microbiology, vol.98, pp.1-29, 2017.

F. and B. Taheur, Anti-bacterial and anti-biofilm activity of probiotic bacteria against oral pathogens, Microbial Pathogenesis, vol.97, pp.213-220, 2016.

C. M. Chapman, G. R. Gibson, and I. Rowland, Effects of single-and multi-strain probiotics on biofilm formation and in vitro adhesion to bladder cells by urinary tract pathogens, Anaerobe, vol.27, pp.71-76, 2014.

T. Fiedler, C. Riani, D. Koczan, K. Standar, B. Kreikemeyer et al., Protective mechanisms of respiratory tract Streptococci against Streptococcus pyogenes biofilm formation and epithelial cell infection, Applied and environmental microbiology, vol.79, issue.4, pp.1265-1276, 2013.

E. G. Lopes, D. A. Moreira, P. Gullón, B. Gullón, A. Cardelle-cobas et al., Topical application of probiotics in skin: adhesion, antimicrobial and antibiofilm in vitro assays, Journal of Applied Microbiology, vol.122, issue.2, pp.450-461, 2017.

P. Varma, N. Nisha, K. R. Dinesh, A. V. Kumar, and R. Biswas, Anti-infective properties of Lactobacillus fermentum against Staphylococcus aureus and Pseudomonas aeruginosa, Journal of Molecular Microbiology and Biotechnology, vol.20, issue.3, pp.137-143, 2011.

E. Ciandrini, R. Campana, and W. Baffone, Live and heat-killed Lactobacillus spp. interfere with Streptococcus mutans and Streptococcus oralis during biofilm development on titanium surface, Archives of Oral Biology, vol.78, pp.48-57, 2017.

F. Schwendicke, F. Korte, C. E. Dörfer, S. Kneist, K. El-sayed et al., Inhibition of Streptococcus mutans growth and biofilm formation by probiotics in vitro, Caries Research, vol.51, issue.2, pp.87-95, 2017.

R. D. Rossoni, Inhibitory effect of probiotic Lactobacillus supernatants from the oral cavity on Streptococcus mutans biofilms, Microbial Pathogenesis, vol.123, pp.361-367, 2018.

H. Park, Autoinducer-2 associated inhibition by Lactobacillus sakei NR28 reduces virulence of enterohaemorrhagic Escherichia coli O157:H7, Food Control, vol.45, pp.62-69, 2014.

D. Bujnakova, E. Strakova, and V. Kmet, In vitro evaluation of the safety and probiotic properties of Lactobacilli isolated from chicken and calves, Anaerobe, vol.29, pp.118-127, 2014.

R. Ben-slama, B. Kouidhi, T. Zmantar, K. Chaieb, and A. Bakhrouf, Anti-listerial and anti-biofilm activities of potential probiotic Lactobacillus strains isolated from Tunisian traditional fermented food, Journal of Food Safety, vol.33, issue.1, pp.8-16, 2013.

H. Frickmann, C. Klenk, P. Warnke, S. Redanz, and A. Podbielski, Influence of probiotic culture supernatants on in vitro biofilm formation of staphylococci, European Journal of Microbiology and Immunology, vol.8, issue.4, pp.119-127, 2018.

J. Woo and J. Ahn, Probiotic-mediated competition, exclusion and displacement in biofilm formation by food-borne pathogens, Letters in Applied Microbiology, vol.56, issue.4, pp.307-313, 2013.

E. Turhan, Z. Erginkaya, M. Hatice, and E. Ozer, Inactivation effect of probiotic biofilms on growth of Listeria monocytogenes, Journal of the Faculty of Veterinary Medicine, vol.23, issue.4, pp.541-546, 2017.

N. C. Gómez, J. M. Ramiro, B. X. Quecan, B. D. De-melo, and . Franco, Use of potential probiotic lactic acid bacteria (LAB) biofilms for the control of Listeria monocytogenes, Salmonella Typhimurium, and Escherichia coli O157:H7 biofilms formation, Frontiers in Microbiology, vol.7, p.863, 2016.

P. Ommen, N. Zobek, and R. L. Meyer, Quantification of biofilm biomass by staining: Non-toxic safranin can replace the popular crystal violet, Journal of Microbiological Methods, vol.141, pp.87-89, 2017.

B. W. Trautner, Nanoscale surface modification favors benign biofilm formation and impedes adherence by pathogens, Nanomedicine : nanotechnology, biology, and medicine, vol.8, issue.3, pp.261-270, 2012.

M. E. Segers and S. Lebeer, Towards a better understanding of Lactobacillus rhamnosus GG -host interactions, Microbial Cell Factories, vol.13, issue.1, p.7, 2014.

A. Tarrah, Probiotic potential and biofilm inhibitory activity of Lactobacillus casei group strains isolated from infant feces, Journal of Functional Foods, vol.54, pp.489-497, 2019.

M. Kankainen, Comparative genomic analysis of Lactobacillus rhamnosus GG reveals pili containing a human-mucus binding protein, Proceedings of the National Academy of Sciences of the United States of America, vol.106, issue.40, pp.17193-17198, 2009.

R. M. Sullan, Single-cell force spectroscopy of pili-mediated adhesion, Nanoscale, vol.6, issue.2, pp.1134-1143, 2014.

S. Lebeer, T. L. Verhoeven, M. Perea, J. Vélez, S. C. Vanderleyden et al., Impact of environmental and genetic factors on biofilm formation by the probiotic strain Lactobacillus rhamnosus GG, Applied and Environmental Microbiology, vol.73, issue.21, pp.6768-6775, 2007.

R. Lu, N. Madayiputhiya, N. P. Morin, J. Nataro, and A. Fasano, Isolation, identification, and characterization of small bioactive peptides from Lactobacillus GG conditional media that exert both anti-Gram-negative and Gram-positive bactericidal activity, Journal of Pediatric Gastroenterology and Nutrition, vol.49, issue.1, pp.23-30, 2009.

M. I. Petrova, Lectin-Like molecules of Lactobacillus rhamnosus GG inhibit pathogenic Escherichia coli and Salmonella biofilm formation, PloS one, vol.11, issue.8, pp.161337-0161337, 2016.

C. N. Allonsius, Inhibition of Candida albicans morphogenesis by chitinase from Lactobacillus rhamnosus GG, Scientific Reports, vol.9, issue.1, p.2900, 2019.

S. Doron, D. R. Snydman, and S. L. Gorbach, Lactobacillus GG: bacteriology and clinical applications, Gastroenterol Clin North Am, vol.34, issue.3, pp.483-498, 2005.

M. Kalliomäki, S. Salminen, T. Poussa, and E. Isolauri, Probiotics during the first 7 years of life: A cumulative risk reduction of eczema in a randomized, placebo-controlled trial, Journal of Allergy and Clinical Immunology, vol.119, issue.4, pp.1019-1021, 2007.

R. and B. Canani, Extensively hydrolyzed casein formula containing Lactobacillus rhamnosus GG reduces the occurrence of other allergic manifestations in children with cow's milk allergy: 3-year randomized controlled trial, Journal of Allergy and Clinical Immunology, vol.139, issue.6, 1906.

G. Harata, Probiotics modulate gut microbiota and health status in Japanese cedar pollinosis patients during the pollen season, European Journal of Nutrition, vol.56, issue.7, pp.2245-2253, 2017.

L. Näse, Effect of Long-Term Consumption of a Probiotic Bacterium, Lactobacillus rhamnosus GG, in Milk on Dental Caries and Caries Risk in Children, Caries Research, vol.35, issue.6, pp.412-420, 2001.

A. Toiviainen, Impact of orally administered lozenges with Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis BB-12 on the number of salivary mutans streptococci, amount of plaque, gingival inflammation and the oral microbiome in healthy adults, Clinical oral investigations, vol.19, issue.1, pp.77-83, 2015.

E. Bruzzese, Effect of Lactobacillus GG supplementation on pulmonary exacerbations in patients with cystic fibrosis: A pilot study, Clinical Nutrition, vol.26, issue.3, pp.322-328, 2007.

L. E. Morrow, M. H. Kollef, and T. B. Casale, Probiotic prophylaxis of ventilator-associated pneumonia: a blinded, randomized, controlled trial, American journal of respiratory and critical care medicine, vol.182, issue.8, pp.1058-1064, 2010.

S. W. Seow, Lactobacillus rhamnosus GG induces tumor regression in mice bearing orthotopic bladder tumors, Cancer Science, vol.101, issue.3, pp.751-758, 2010.

M. Kandasamy, B. Bay, Y. Lee, and R. Mahendran, Lactobacilli secreting a tumor antigen and IL15 activates neutrophils and dendritic cells and generates cytotoxic T lymphocytes against cancer cells, Cellular Immunology, vol.271, issue.1, pp.89-96, 2011.

K. Laitinen, T. Poussa, and E. Isolauri, Probiotics and dietary counselling contribute to glucose regulation during and after pregnancy: a randomised controlled trial, British Journal of Nutrition, vol.101, issue.11, pp.1679-1687, 2008.

I. Spacova, Expression of fluorescent proteins in Lactobacillus rhamnosus to study host-microbe and microbe-microbe interactions, Microbial Biotechnology, vol.11, issue.2, pp.317-331, 2018.

Q. Jiang, V. Kainulainen, I. Stamatova, R. Korpela, and J. H. Meurman, Lactobacillus rhamnosus GG in experimental oral biofilms exposed to different carbohydrate sources, Caries Research, vol.52, issue.3, pp.220-229, 2018.

S. C. De-keersmaecker, T. L. Verhoeven, J. Desair, K. Marchal, J. Vanderleyden et al., Strong antimicrobial activity of Lactobacillus rhamnosus GG against Salmonella typhimurium is due to accumulation of lactic acid, FEMS Microbiology Letters, vol.259, issue.1, pp.89-96, 2006.

L. Makras, Kinetic analysis of the antibacterial activity of probiotic lactobacilli towards Salmonella enterica serovar Typhimurium reveals a role for lactic acid and other inhibitory compounds, Research in Microbiology, vol.157, issue.3, pp.241-247, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00175483

G. Schaule, T. Griebe, and H. Flemming, Steps in biofilm sampling and characterization in biofouling cases, Biofilms: Investigative Methods and Applications, vol.268, 2000.

C. Hannig, M. Hannig, O. Rehmer, G. Braun, E. Hellwig et al., Fluorescence microscopic visualization and quantification of initial bacterial colonization on enamel in situ, Archives of Oral Biology, vol.52, issue.11, pp.1048-1056, 2007.

A. Delille, F. Quilès, and F. Humbert, In situ monitoring of the nascent Pseudomonas fluorescens biofilm response to variations in the dissolved organic carbon level in low-nutrient water by Attenuated Total Reflectance-Fourier Transform Infrared Spectroscopy, Applied and Environmental Microbiology, vol.73, issue.18, pp.5782-5788, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01849021

L. Drago, S. Agrappi, M. Bortolin, M. Toscano, C. L. Romanò et al., How to study biofilms after microbial colonization of materials used in orthopaedic implants, International journal of molecular sciences, vol.17, issue.3, pp.293-293, 2016.

A. Fahs, F. Quilès, D. Jamal, F. Humbert, and G. Francius, In situ analysis of bacterial extracellular polymeric substances from a Pseudomonas fluorescens biofilm by combined vibrational and single molecule force spectroscopies, J. Phys. Chem. B, vol.118, issue.24, pp.6702-6713, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01503745

M. M. Baum, Characterization of structures in biofilms formed by a Pseudomonas fluorescens isolated from soil, BMC microbiology, vol.9, pp.103-103, 2009.

C. Wilson, Quantitative and qualitative assessment methods for biofilm growth: a mini-review, Research & reviews. Journal of engineering and technology, vol.6, issue.4, 2017.

M. V. Berridge, P. M. Herst, and A. S. Tan, Tetrazolium dyes as tools in cell biology: New insights into their cellular reduction, Biotechnology Annual Review, vol.11, pp.127-152, 2005.

S. Ullrich, B. Karrasch, H. Hoppe, K. Jeskulke, and M. Mehrens, Toxic effects on bacterial metabolism of the redox dye 5-cyano-2,3-ditolyl tetrazolium chloride, Appl. Environ. Microbiol, vol.62, issue.12, p.4587, 1996.

S. N. Rampersad, Multiple applications of Alamar Blue as an indicator of metabolic function and cellular health in cell viability bioassays, Sensors, vol.12, issue.9, pp.12347-12360, 2012.

J. R. Lawrence, D. R. Korber, and T. R. Neu, Analytical Imaging and Microscopy Techniques, pp.40-68, 2007.

S. Lebeer, FISH analysis of Lactobacillus biofilms in the gastrointestinal tract of different hosts, Letters in Applied Microbiology, vol.52, issue.3, pp.220-226, 2011.

K. S. Brandenburg, Development of Pseudomonas aeruginosa Biofilms in Partial-Thickness Burn Wounds Using a Sprague-Dawley Rat Model, Journal of burn care & research : official publication of the American Burn Association, vol.40, issue.1, pp.44-57, 2019.

S. Srey, I. K. Jahid, and S. Ha, Biofilm formation in food industries: A food safety concern, Food Control, vol.31, issue.2, pp.572-585, 2013.

G. Francius, Bacterial surface appendages strongly impact nanomechanical and electrokinetic properties of Escherichia coli cells subjected to osmotic stress, PloS one, vol.6, issue.5, pp.20066-20066, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01393507

J. Burgain, Impacts of pH-mediated EPS structure on probiotic bacterial pili-whey proteins interactions, Colloids and Surfaces B: Biointerfaces, vol.134, pp.332-338, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01248665

J. Guerin, Adhesion of Lactobacillus rhamnosus GG surface biomolecules to milk proteins, Food Hydrocolloids, vol.82, pp.296-303, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01898949

S. Lebeer, Functional analysis of Lactobacillus rhamnosus GG pili in relation to adhesion and immunomodulatory interactions with intestinal epithelial cells, Applied and environmental microbiology, vol.78, issue.1, pp.185-193, 2012.

R. R. Burgess, Chapter 32 Elution of proteins from gels, Methods in Enzymology, vol.463, pp.565-572, 2009.

T. Mahmood and P. Yang, Western blot: technique, theory, and trouble shooting, N Am J Med Sci, vol.4, issue.9, pp.429-434, 2012.

I. J. Claes, Genetic and biochemical characterization of the cell wall hydrolase activity of the major secreted protein of Lactobacillus rhamnosus GG, PLOS ONE, vol.7, issue.2, p.31588, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01189702

R. Diez, M. Herbstreith, C. Osorio, and O. Alzate, Chapter 42-D fluorescence difference gel electrophoresis (DIGE) in neuroproteomics, Neuroproteomics, 2010.

J. Koponen, Effect of acid stress on protein expression and phosphorylation in Lactobacillus rhamnosus GG, Journal of Proteomics, vol.75, issue.4, pp.1357-1374, 2012.

K. Koskenniemi, Proteome analysis of Lactobacillus rhamnosus GG using 2-D DIGE and mass spectrometry shows differential protein production in laboratory and industrial-type growth media, J. Proteome Res, vol.8, issue.11, pp.4993-5007, 2009.

N. Ali, R. De-cássia-pontello-rampazzo, A. Dias-tavares-costa, and M. A. Krieger, Current nucleic acid extraction methods and their implications to point-of-care diagnostics, BioMed Research International, vol.2017, 2017.

G. Schochetman, C. Ou, and W. K. Jones, Polymerase chain reaction, The Journal of Infectious Diseases, vol.158, issue.6, pp.1154-1157, 1988.

L. Garibyan and N. Avashia, Research techniques made simple: polymerase chain reaction (PCR), J Invest Dermatol, vol.133, issue.3, pp.1-4, 2013.

S. A. Barghouthi, A universal method for the identification of bacteria based on general PCR primers, Indian J Microbiol, vol.51, issue.4, pp.430-444, 2011.

A. Endo, J. Aakko, and S. Salminen, Evaluation of strain-specific primers for identification of Lactobacillus rhamnosus GG, FEMS Microbiology Letters, vol.337, issue.2, pp.120-125, 2012.

P. Treven, V. Mrak, B. Matija?i?, S. Horvat, and I. Rogelj, Administration of probiotics Lactobacillus rhamnosus GG and Lactobacillus gasseri K7 during pregnancy and lactation changes mouse mesenteric lymph nodes and mammary gland microbiota, Journal of Dairy Science, vol.98, issue.4, pp.2114-2128, 2015.

T. Ahlroos and S. Tynkkynen, Quantitative strain-specific detection of Lactobacillus rhamnosus GG in human faecal samples by real-time PCR, Journal of Applied Microbiology, vol.106, issue.2, pp.506-514, 2009.

S. S. Nielsen, Phenol-sulfuric acid method for total carbohydrates, Food Analysis Laboratory Manual, Food Science Texts Series, pp.47-53, 2010.

P. M. Bales, E. M. Renke, S. L. May, Y. Shen, and D. C. Nelson, Purification and characterization of biofilm-associated EPS exopolysaccharides from ESKAPE organisms and other pathogens, PLoS One, vol.8, issue.6, pp.67950-67950, 2013.

S. Jachlewski, W. D. Jachlewski, U. Linne, C. Bräsen, J. Wingender et al., Isolation of extracellular polymeric substances from biofilms of the thermoacidophilic Archaeon Sulfolobus acidocaldarius, Front Bioeng Biotechnol, vol.3, pp.123-123, 2015.

C. N. Allonsius, Interplay between Lactobacillus rhamnosus GG and Candida and the involvement of exopolysaccharides, Microb Biotechnol, vol.10, issue.6, pp.1753-1763, 2017.

P. Stoodley, 5 -Biofilms, Biomaterials, and device-related infections, Handbook of Polymer Applications in Medicine and Medical, pp.77-101, 2013.

C. Hannig, M. Follo, E. Hellwig, and A. Al-ahmad, Visualization of adherent micro-organisms using different techniques, Journal of Medical Microbiology, vol.59, issue.1, pp.1-7, 2010.

M. A. Auty, Direct in situ viability assessment of bacteria in probiotic dairy products using viability staining in conjunction with confocal scanning laser microscopy, Appl Environ Microbiol, vol.67, issue.1, pp.420-425, 2001.

F. Ou, C. Mcgoverin, S. Swift, and F. Vanholsbeeck, Absolute bacterial cell enumeration using flow cytometry, Journal of Applied Microbiology, vol.123, issue.2, pp.464-477, 2017.

M. Erriu, G. Genta, E. Tuveri, G. Orrù, G. Barbato et al., Microtiter spectrophotometric biofilm production assay analyzed with metrological methods and uncertainty evaluation, Measurement, vol.45, issue.5, pp.1083-1088, 2012.

O. Zagorodko, Surface Plasmon Resonance (SPR) for the Evaluation of Shear-Force-Dependent Bacterial Adhesion, Biosensors (Basel), vol.5, issue.2, pp.276-287, 2015.

J. Guo, Microbial attachment and adsorption-desorption kinetic of tightly bound extracellular polymeric substances on model organic surfaces, Chemical Engineering Journal, vol.279, pp.516-521, 2015.

A. Pranzetti, Model organic surfaces to probe marine bacterial adhesion kinetics by surface plasmon resonance, Advanced Functional Materials, vol.22, issue.17, pp.3672-3681, 2012.

P. Larkin, Infrared and Raman Spectroscopy: Principles and Spectral Interpretation, 2011.

W. E. Huang, R. I. Griffiths, I. P. Thompson, M. J. Bailey, and A. S. Whiteley, Raman Microscopic Analysis of Single Microbial Cells, Anal. Chem, vol.76, issue.15, pp.4452-4458, 2004.

M. Hlaing, B. Wood, D. Mcnaughton, D. Ying, and M. A. Augustin, Raman spectroscopic analysis of Lactobacillus rhamnosus GG in response to dehydration reveals DNA conformation changes, Journal of Biophotonics, vol.10, issue.4, pp.589-597, 2017.

V. Emmanuel, B. Odile, and R. Céline, FTIR spectroscopy of woods: A new approach to study the weathering of the carving face of a sculpture, Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, vol.136, pp.1255-1259, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01304928

M. J. Baker, Using Fourier transform IR spectroscopy to analyze biological materials, Nat Protoc, vol.9, issue.8, pp.1771-1791, 2014.

P. Zarnowiec, L. Lechowicz, G. Czerwonka, and W. Kaca, Fourier Transform Infrared Spectroscopy (FTIR) as a Tool for the Identification and Differentiation of Pathogenic Bacteria, Current Medicinal Chemistry, vol.22, issue.14, pp.1710-1718, 2015.

H. J. Busscher, H. C. Van-der, and M. , Microbial adhesion in flow displacement systems, Clin Microbiol Rev, vol.19, issue.1, pp.127-141, 2006.

P. R. Rutter and B. Vincent, Attachment mechanisms in the surface growth of microorganisms, Physiological models in microbiology, pp.87-107

N. Mohamed, T. R. Rainier, and J. M. Ross, Novel experimental study of receptor-mediated bacterial adhesion under the influence of fluid shear, Biotechnology and Bioengineering, vol.68, issue.6, pp.628-636, 2000.

W. E. Thomas, E. Trintchina, M. Forero, V. Vogel, and E. V. Sokurenko, Bacterial adhesion to target cells enhanced by shear force, Cell, vol.109, issue.7, pp.913-923, 2002.

P. Qi, W. Wang, and Z. Qi, Effect of shear stress on biofilm morphological characteristics and the secretion of extracellular polymeric substances, 2008 2nd International Conference on Bioinformatics and Biomedical Engineering, pp.3438-3441, 2008.

J. Hou, D. H. Veeregowda, B. Van-de-belt-gritter, H. J. Busscher, H. C. Van-der et al., Extracellular polymeric matrix production and relaxation under fluid shear and mechanical pressure in Staphylococcus aureus Biofilms, Applied and Environmental Microbiology, vol.84, issue.1, 2018.

M. Lemos, F. Mergulhão, L. Melo, and M. Simões, The effect of shear stress on the formation and removal of Bacillus cereus biofilms, Food and Bioproducts Processing, vol.93, pp.242-248, 2015.

K. P. Norris, Infra-red spectroscopy and its application to microbiology, J Hyg (Lond), vol.57, issue.3, pp.326-345, 1959.

R. H. Bordner, P. W. Kabler, B. A. Kenner, J. W. Riddle, S. W. Rockwood et al., Bacterial identification by infrared spectrophotometry, J Bacteriol, vol.72, issue.5, pp.593-603, 1956.

D. E. Nivens, J. Q. Chambers, T. R. Anderson, A. Tunlid, J. Smit et al., Monitoring microbiol adhesion and biofilm formation by attenuated total reflection/Fourier transform infrared spectroscopy, Journal of Microbiological Methods, vol.17, issue.3, pp.199-213, 1993.

D. C. White, A. A. Arrage, D. E. Nivens, R. J. Palmer, J. F. Rice et al., Biofilm ecology: Online methods bring new insights into mic and microbial biofouling, Biofouling, vol.10, issue.1-3, pp.3-16, 1996.

D. E. Nivens, D. E. Ohman, J. Williams, and M. J. Franklin, Role of alginate and its O acetylation in formation of Pseudomonas aeruginosa microcolonies and biofilms, J Bacteriol, vol.183, issue.3, pp.1047-1057, 2001.

J. Schmitt, D. Nivens, D. C. White, and H. Flemming, Changes of biofilm properties in response to sorbed substances -an FTIR-ATR study, Water Science and Technology, vol.32, issue.8, pp.149-155, 1995.

P. A. Suci, G. G. Geesey, and B. J. Tyler, Integration of Raman microscopy, differential interference contrast microscopy, and attenuated total reflection Fourier transform infrared spectroscopy to investigate chlorhexidine spatial and temporal distribution in Candida albicans biofilms, Journal of Microbiological Methods, vol.46, issue.3, pp.193-208, 2001.

P. A. Suci, J. D. Vrany, and M. W. Mittelman, Investigation of interactions between antimicrobial agents and bacterial biofilms using attenuated total reflection Fourier transform infrared spectroscopy, Biomaterials, vol.19, issue.4, pp.327-339, 1998.

R. M. Donlan, Model system for growing and quantifying Streptococcus pneumoniae biofilms in situ and in real time, Applied and Environmental Microbiology, vol.70, issue.8, pp.4980-4988, 2004.

M. Boualam, F. Quilès, L. Mathieu, and J. Block, Monitoring the effect of organic matter on biofilm growth in low nutritive waters by ATR/FT-IR Spectroscopy, Biofouling, vol.18, issue.1, pp.73-81, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01810352

S. J. Parikh and J. Chorover, FTIR spectroscopic study of biogenic Mn-Oxide formation by Pseudomonas putida GB-1, Geomicrobiology Journal, vol.22, issue.5, pp.207-218, 2005.

J. J. Ojeda, M. E. Romero-gonzalez, H. M. Pouran, and S. A. Banwart, In situ monitoring of the biofilm formation of Pseudomonas putida on hematite using flow-cell ATR-FTIR spectroscopy to investigate the formation of inner-sphere bonds between the bacteria and the mineral, Mineralogical Magazine, vol.72, issue.1, pp.101-106, 2008.

F. Quilès, F. Humbert, and A. Delille, Analysis of changes in attenuated total reflection FTIR fingerprints of Pseudomonas fluorescens from planktonic state to nascent biofilm state, Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, vol.75, issue.2, pp.610-616, 2010.

J. Huang, E. J. Elzinga, Y. Brechbuehl, A. Voegelin, and R. Kretzschmar, Impacts of Shewanella putrefaciens strain CN-32 cells and extracellular polymeric substances on the sorption of As(V) and As(III) on Fe(III)-(Hydr)oxides, Environ. Sci. Technol, vol.45, issue.7, pp.2804-2810, 2011.

G. S. Lorite, C. M. Rodrigues, A. A. Souza, C. Kranz, B. Mizaikoff et al., The role of conditioning film formation and surface chemical changes on Xylella fastidiosa adhesion and biofilm evolution, Journal of Colloid and Interface Science, vol.359, issue.1, pp.289-295, 2011.

G. S. Lorite, A. A. Souza, D. Neubauer, B. Mizaikoff, C. Kranz et al., On the role of extracellular polymeric substances during early stages of Xylella fastidiosa biofilm formation, Colloids and Surfaces B: Biointerfaces, vol.102, pp.519-525, 2013.

L. Giotta, Reversible binding of metal ions onto bacterial layers revealed by protonation-induced ATR-FTIR difference spectroscopy, Langmuir, vol.27, issue.7, pp.3762-3773, 2011.

H. Wang, S. Ding, G. Wang, X. Xu, and G. Zhou, In situ characterization and analysis of Salmonella biofilm formation under meat processing environments using a combined microscopic and spectroscopic approach, International Journal of Food Microbiology, vol.167, issue.3, pp.293-302, 2013.

S. J. Parikh, F. N. Mukome, and X. Zhang, ATR-FTIR spectroscopic evidence for biomolecular phosphorus and carboxyl groups facilitating bacterial adhesion to iron oxides, Colloids and Surfaces B: Biointerfaces, vol.119, pp.38-46, 2014.

F. Quilès, S. Saadi, G. Francius, J. Bacharouche, and F. Humbert, In situ and real time investigation of the evolution of a Pseudomonas fluorescens nascent biofilm in the presence of an antimicrobial peptide, Biochimica et Biophysica Acta (BBA) -Biomembranes, vol.1858, issue.1, pp.75-84, 2016.

S. Kumari, N. Mangwani, and S. Das, Interaction of Pb(II) and biofilm associated extracellular polymeric substances of a marine bacterium Pseudomonas pseudoalcaligenes NP103, Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, vol.173, pp.655-665, 2017.

O. Freudenthal, F. Quilès, and G. Francius, Discrepancies between cyclic and linear antimicrobial peptide actions on the spectrochemical and nanomechanical fingerprints of a young biofilm, ACS Omega, vol.2, issue.9, pp.5861-5872, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02202492

L. Zhou, Surface interaction between metazeunerite and an indigenous microorganism Kocuria rosea: Implications for bioremediation of As-U tailings, Chemical Engineering Journal, vol.359, pp.393-401, 2019.

A. Fanesi, A. Zegeye, C. Mustin, and A. Cébron, Soil particles and phenanthrene interact in defining the metabolic profile of Pseudomonas putida G7: a vibrational spectroscopy approach, Frontiers in Microbiology, vol.9, p.2999, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02317659

M. Pousti and J. Greener, Altered biofilm formation at plasma bonded surfaces in microchannels studied by attenuated total reflection infrared spectroscopy, Surface Science, vol.676, pp.56-60, 2018.

R. Davis and L. J. Mauer, Fourier tansform infrared (FT-IR) spectroscopy: A rapid tool for detection and analysis of foodborne pathogenic bacteria, Current Research, 2010.

K. Kamila, In vivo atomic force microscopy-infrared spectroscopy of bacteria, Journal of The Royal Society Interface, vol.15, issue.140, p.20180115, 2018.

S. Moussaoui, C. Carteret, D. Brie, and A. Mohammad-djafari, Bayesian analysis of spectral mixture data using Markov Chain Monte Carlo Methods, Chemometrics and Intelligent Laboratory Systems, vol.81, issue.2, pp.137-148, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00022304

N. Dobigeon, S. Moussaoui, J. Tourneret, and C. Carteret, Bayesian separation of spectral sources under non-negativity and full additivity constraints, Signal Processing, vol.89, issue.12, pp.2657-2669, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00548756

A. Hartstein, J. R. Kirtley, and J. C. Tsang, Enhancement of the Infrared Absorption from Molecular Monolayers with Thin Metal Overlayers, Phys. Rev. Lett, vol.45, issue.3, pp.201-204, 1980.

M. Osawa and M. Ikeda, Surface-enhanced infrared absorption of p-nitrobenzoic acid deposited on silver island films: contributions of electromagnetic and chemical mechanisms, J. Phys. Chem, vol.95, issue.24, pp.9914-9919, 1991.

G. T. Merklin and P. R. Griffiths, Influence of chemical interactions on the surface-enhanced infrared absorption spectrometry of nitrophenols on copper and silver films, Langmuir, vol.13, issue.23, pp.6159-6163, 1997.

J. P. Busalmen, A. Esteve-nuñez, A. Berná, and J. M. Feliu, ATR-SEIRAs characterization of surface redox processes in G. sulfurreducens, Bioelectrochemistry, vol.78, issue.1, pp.25-29, 2010.

Y. Liu, A. Berná, V. Climent, and J. M. Feliu, Real-time monitoring of electrochemically active biofilm developing behavior on bioanode by using EQCM and ATR/FTIR, Sensors and Actuators B: Chemical, vol.209, pp.781-789, 2015.

P. Toziou, P. Barmpalexis, P. Boukouvala, S. Verghese, and I. Nikolakakis, Quantification of live Lactobacillus acidophilus in mixed populations of live and killed by application of attenuated reflection Fourier transform infrared spectroscopy combined with chemometrics, Journal of Pharmaceutical and Biomedical Analysis, vol.154, pp.16-22, 2018.

R. Tareb, M. Bernardeau, C. Amiel, and J. P. Vernoux, Usefulness of FTIR spectroscopy to distinguish rough and smooth variants of Lactobacillus farciminis CNCM-I-3699, FEMS Microbiology Letters, vol.364, issue.2, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02374480

G. Deepika, R. J. Green, R. A. Frazier, and D. Charalampopoulos, Effect of growth time on the surface and adhesion properties of Lactobacillus rhamnosus GG, Journal of Applied Microbiology, vol.107, issue.4, pp.1230-1240, 2009.

G. Posselt, S. Backert, and S. Wessler, The functional interplay of Helicobacter pylori factors with gastric epithelial cells induces a multi-step process in pathogenesis, Cell Communication and Signaling, vol.11, issue.1, p.77, 2013.

J. M. Koczan, B. R. Lenneman, M. J. Mcgrath, and G. W. Sundin, Cell surface attachment structures contribute to biofilm formation and xylem colonization by Erwinia amylovora, Appl Environ Microbiol, vol.77, issue.19, pp.7031-7039, 2011.

Y. Hong and D. G. Brown, Electrostatic behavior of the charge-regulated bacterial cell surface, Langmuir, vol.24, issue.9, pp.5003-5009, 2008.

G. Capitani, O. Eidam, R. Glockshuber, and M. G. Grütter, Structural and functional insights into the assembly of type 1 pili from Escherichia coli, Microbes and Infection, vol.8, issue.8, pp.2284-2290, 2006.

J. Ryu, H. Kim, J. F. Frank, and L. R. Beuchat, Attachment and biofilm formation on stainless steel by Escherichia coli O157:H7 as affected by curli production, Letters in Applied Microbiology, vol.39, issue.4, pp.359-362, 2004.

C. I. Pereni, Q. Zhao, Y. Liu, and E. Abel, Surface free energy effect on bacterial retention, Colloids and Surfaces B: Biointerfaces, vol.48, issue.2, pp.143-147, 2006.

N. P. Boks, W. Norde, H. C. Van-der-mei, and H. J. Busscher, Forces involved in bacterial adhesion to hydrophilic and hydrophobic surfaces, Microbiology, vol.154, issue.10, pp.3122-3133, 2008.

D. Cunliffe, C. A. Smart, C. Alexander, and E. N. Vulfson, Bacterial adhesion at synthetic surfaces, Appl Environ Microbiol, vol.65, issue.11, pp.4995-5002, 1999.

B. Li and B. E. Logan, Bacterial adhesion to glass and metal-oxide surfaces, Colloids and Surfaces B: Biointerfaces, vol.36, issue.2, pp.81-90, 2004.

X. Dou, D. Zhang, C. Feng, and L. Jiang, Bioinspired Hierarchical Surface Structures with Tunable Wettability for Regulating Bacteria Adhesion, ACS Nano, vol.9, issue.11, pp.10664-10672, 2015.

Y. Yuan, M. P. Hays, P. R. Hardwidge, and J. Kim, Surface characteristics influencing bacterial adhesion to polymeric substrates, RSC Adv, vol.7, issue.23, pp.14254-14261, 2017.

X. Zhang, L. Wang, and E. Levänen, Superhydrophobic surfaces for the reduction of bacterial adhesion, RSC Adv, vol.3, issue.30, pp.12003-12020, 2013.

W. Barthlott and C. Neinhuis, Purity of the sacred lotus, or escape from contamination in biological surfaces, Planta, vol.202, issue.1, pp.1-8, 1997.

C. Gómez-suárez, H. J. Busscher, H. C. Van-der, and M. , Analysis of Bacterial Detachment from Substratum Surfaces by the Passage of Air-Liquid Interfaces, Appl. Environ. Microbiol, vol.67, issue.6, p.2531, 2001.

W. G. Pitt, M. O. Mcbride, A. J. Barton, and R. D. Sagers, Air-water interface displaces adsorbed bacteria, Biomaterials, vol.14, issue.8, pp.605-608, 1993.

J. K. Oh, The influence of surface chemistry on the kinetics and thermodynamics of bacterial adhesion, Scientific Reports, vol.8, issue.1, p.17247, 2018.

C. Valotteau, Nanoscale antiadhesion properties of sophorolipid-coated surfaces against pathogenic bacteria, Nanoscale Horiz, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02093308

J. Verran, A. Packer, P. J. Kelly, and K. A. Whitehead, Use of the Atomic Force Microscope to Determine the Strength of Bacterial Attachment to Grooved Surface Features, Journal of Adhesion Science and Technology, vol.24, pp.2271-2285, 2010.

S. J. Parikh and J. Chorover, ATR-FTIR spectroscopy reveals bond formation during bacterial adhesion to iron oxide, Langmuir, vol.22, issue.20, pp.8492-8500, 2006.

M. J. Mcwhirter, A. J. Mcquillan, and P. J. Bremer, Influence of ionic strength and pH on the first 60 min of Pseudomonas aeruginosa attachment to ZnSe and to TiO2 monitored by ATR-IR spectroscopy, Colloids and Surfaces B: Biointerfaces, vol.26, issue.4, pp.365-372, 2002.

S. E. Glassford, B. Byrne, and S. G. Kazarian, Recent applications of ATR FTIR spectroscopy and imaging to proteins, Biochimica et Biophysica Acta (BBA) -Proteins and Proteomics, vol.1834, issue.12, pp.2849-2858, 2013.

J. Schartner, Chemical functionalization of germanium with dextran brushes for immobilization of proteins revealed by Attenuated Total Reflection Fourier Transform Infrared Difference Spectroscopy, Anal. Chem, vol.87, issue.14, pp.7467-7475, 2015.

C. S. Riccardi, D. W. Hess, and B. Mizaikoff, Surface-modified ZnSe waveguides for label-free infrared attenuated total reflection detection of DNA hybridization, Analyst, vol.136, issue.23, pp.4906-4911, 2011.

I. C. Stefan, D. Mandler, and D. A. Scherson, In Situ FTIR-ATR studies of functionalized self-assembled bilayer interactions with metal ions in aqueous solutions, Langmuir, vol.18, issue.18, pp.6976-6980, 2002.

B. A. Jucker, H. Harms, and A. J. Zehnder, Adhesion of the positively charged bacterium Stenotrophomonas (Xanthomonas) maltophilia 70401 to glass and Teflon, J Bacteriol, vol.178, issue.18, pp.5472-5479, 1996.

B. Gottenbos, H. C. Van-der-mei, and H. J. Busscher, Initial adhesion and surface growth of Pseudomonas aeruginosa on negatively and positively charged poly(methacrylates), Journal of Materials Science: Materials in Medicine, vol.10, pp.853-855, 1999.

C. Chen, T. Petterson, J. Illergård, M. Ek, and L. Wågberg, Influence of cellulose charge on bacteria adhesion and viability to PVAm/CNF/PVAm-modified cellulose model surfaces, Biomacromolecules, vol.20, issue.5, pp.2075-2083, 2019.

A. Terada, A. Yuasa, T. Kushimoto, S. Tsuneda, A. Katakai et al., Bacterial adhesion to and viability on positively charged polymer surfaces, Microbiology, vol.152, issue.12, pp.3575-3583, 2006.

B. Gottenbos, D. W. Grijpma, H. C. Van-der-mei, J. Feijen, and H. J. Busscher, Antimicrobial effects of positively charged surfaces on adhering Gram-positive and Gram-negative bacteria, Journal of Antimicrobial Chemotherapy, vol.48, issue.1, pp.7-13, 2001.

J. L. Clement and P. S. Jarrett, Antibacterial silver, Met Based Drugs, vol.1, issue.5-6, pp.467-482, 1994.

M. G. Schmidt, Copper surfaces are associated with significantly lower concentrations of bacteria on selected surfaces within a pediatric intensive care unit, American Journal of Infection Control, vol.44, issue.2, pp.203-209, 2016.

K. A. Whitehead, J. Colligon, and J. Verran, Retention of microbial cells in substratum surface features of micrometer and sub-micrometer dimensions, Colloids and Surfaces B: Biointerfaces, vol.41, issue.2, pp.129-138, 2005.

J. Verran, A. Packer, P. Kelly, and K. A. Whitehead, The retention of bacteria on hygienic surfaces presenting scratches of microbial dimensions, Letters in Applied Microbiology, vol.50, issue.3, pp.258-263, 2010.

D. Perera-costa, J. M. Bruque, M. L. González-martín, A. C. Gómez-garcía, and V. Vadillo-rodríguez, Studying the influence of surface topography on bacterial adhesion using spatially organized microtopographic surface patterns, Langmuir, vol.30, issue.16, pp.4633-4641, 2014.

M. Lorenzetti, The influence of surface modification on bacterial adhesion to titanium-based substrates, ACS Appl. Mater. Interfaces, vol.7, issue.3, pp.1644-1651, 2015.

G. Feng, Y. Cheng, S. Wang, D. A. Borca-tasciuc, R. W. Worobo et al., Bacterial attachment and biofilm formation on surfaces are reduced by small-diameter nanoscale pores: how small is small enough?, Npj Biofilms And Microbiomes, vol.1, p.15022, 2015.

A. Bhattacharjee, M. Khan, M. Kleiman, and A. I. Hochbaum, Effects of growth surface topography on bacterial signaling in coculture biofilms, ACS Appl. Mater. Interfaces, vol.9, issue.22, pp.18531-18539, 2017.

B. Derjaguin and L. Landau, Theory of the stability of strongly charged lyophobic sols and of the adhesion of strongly charged particles in solutions of electrolytes, Acta physicochimica URSS, vol.14, pp.633-662, 1941.

J. H. Adair, E. Suvaci, and J. Sindel, Surface and colloid chemistry, Encyclopedia of Materials: Science and, pp.1-10, 2001.

S. Perni, E. C. Preedy, and P. Prokopovich, Success and failure of colloidal approaches in adhesion of microorganisms to surfaces, Advances in Colloid and Interface Science, vol.206, pp.265-274, 2014.

M. J. Salas-jara, E. A. Sanhueza, A. Retamal-díaz, C. González, H. Urrutia et al., Probiotic Lactobacillus fermentum UCO-979C biofilm formation on AGS and Caco-2 cells and Helicobacter pylori inhibition, Biofouling, vol.32, issue.10, pp.1245-1257, 2016.

N. Aoudia, Biofilms of Lactobacillus plantarum and Lactobacillus fermentum: Effect on stress responses, antagonistic effects on pathogen growth and immunomodulatory properties, Food Microbiology, vol.53, pp.51-59, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01432554

M. J. Mcshane and Y. M. Lvov, Electrostatic self-assembly: layer-by-layer, Dekker Encyclopedia of Nanoscience and Nanotechnology, Third Edition, 2014.

, Layer Films for Biomedical Applications, 2015.

D. Gundogdu, V. Bütün, and I. Erel-göktepe, Preparation of layer-by-layer films with remarkably different pH-stability and release properties using dual responsive block copolymer micelles, Macromolecular Chemistry and Physics, vol.219, issue.15, p.1800128, 2018.

R. W. Hutkins and N. L. Nannen, pH Homeostasis in lactic acid bacteria, Journal of Dairy Science, vol.76, issue.8, pp.2354-2365, 1993.

S. A. Hussain, B. Dey, D. Bhattacharjee, and N. Mehta, Unique supramolecular assembly through Langmuir -Blodgett (LB) technique, Heliyon, vol.4, issue.12, p.1038, 2018.

D. Y. Takamoto, Stable Ordering in Langmuir-Blodgett Films, Science, vol.293, issue.5533, p.1292, 2001.

O. , Nanoscale investigation of the interaction of colistin with model phospholipid membranes by Langmuir technique, and combined infrared and force spectroscopies, Biochimica et Biophysica Acta (BBA) -Biomembranes, vol.1858, issue.11, pp.2592-2602, 2016.

N. Kumar, L. Wang, I. Siretanu, M. Duits, and F. Mugele, Salt dependent stability of stearic acid Langmuir-Blodgett films exposed to aqueous electrolytes, Langmuir, vol.29, issue.17, pp.5150-5159, 2013.

S. Feng, K. Gong, and J. Chew, Molecular interactions between a lipid and an antineoplastic drug Paclitaxel (Taxol) within the lipid monolayer at the air/water interface, Langmuir, vol.18, issue.10, pp.4061-4070, 2002.

J. B. Brzoska, I. B. Azouz, and F. Rondelez, Silanization of solid substrates: a step toward reproducibility, Langmuir, vol.10, issue.11, pp.4367-4373, 1994.

J. Böhmler, A. Ponche, K. Anselme, and L. Ploux, Self-assembled molecular platforms for bacteria/material biointerface studies: importance to control functional group accessibility, ACS Appl. Mater. Interfaces, vol.5, issue.21, pp.10478-10488, 2013.

A. R. Yadav, R. Sriram, J. A. Carter, and B. L. Miller, Comparative study of solution-phase and vaporphase deposition of aminosilanes on silicon dioxide surfaces, Mater Sci Eng C Mater Biol Appl, vol.35, pp.283-290, 2014.

J. C. Love, L. A. Estroff, J. K. Kriebel, R. G. Nuzzo, and G. M. Whitesides, Self-assembled monolayers of thiolates on metals as a form of nanotechnology, Chem. Rev, vol.105, issue.4, pp.1103-1170, 2005.

M. Kind and C. Wöll, Organic surfaces exposed by self-assembled organothiol monolayers: Preparation, characterization, and application, Progress in Surface Science, vol.84, issue.7-8, pp.230-278, 2009.

C. D. Bain, E. B. Troughton, Y. T. Tao, J. Evall, G. M. Whitesides et al., Formation of monolayer films by the spontaneous assembly of organic thiols from solution onto gold, J. Am. Chem. Soc, vol.111, issue.1, pp.321-335, 1989.

C. Vericat, M. E. Vela, G. Benitez, P. Carro, and R. C. Salvarezza, Self-assembled monolayers of thiols and dithiols on gold: new challenges for a well-known system, Chem. Soc. Rev, vol.39, issue.5, pp.1805-1834, 2010.

L. G. Dubois and R. G. Nuzzo, Synthesis, structure, and properties of model organic surfaces, Annual Review of Physical Chemistry, vol.43, issue.1, pp.437-463, 1992.

A. Ulman, Formation and structure of self-assembled monolayers, Chem. Rev, vol.96, issue.4, pp.1533-1554, 1996.

G. M. Whitesides and P. E. Laibinis, Wet chemical approaches to the characterization of organic surfaces: self-assembled monolayers, wetting, and the physical-organic chemistry of the solid-liquid interface, Langmuir, vol.6, issue.1, pp.87-96, 1990.

D. K. Schwartz, Mechanisms and kinetics of self-assembled monolayer formation, Annu. Rev. Phys. Chem, vol.52, issue.1, pp.107-137, 2001.

G. Yang and G. Liu, New insights for self-assembled monolayers of organothiols on Au(111) revealed by scanning tunneling microscopy, J. Phys. Chem. B, vol.107, issue.34, pp.8746-8759, 2003.

G. E. Poirier, Characterization of organosulfur molecular monolayers on Au(111) using scanning tunneling microscopy, Chem. Rev, vol.97, issue.4, pp.1117-1128, 1997.

L. J. Cristina, G. Ruano, R. Salvarezza, and J. Ferrón, Thermal stability of self-assembled monolayers of n-Hexanethiol on Au(111)-(1 × 1) and Au(001)-(1 × 1), J. Phys. Chem. C, vol.121, issue.50, pp.27894-27904, 2017.

H. Wang, S. Chen, L. Li, and S. Jiang, Improved method for the preparation of carboxylic acid and amine terminated self-assembled monolayers of alkanethiolates, Langmuir, vol.21, issue.7, pp.2633-2636, 2005.

A. R. Noble-luginbuhl and R. G. Nuzzo, Assembly and characterization of SAMs formed by the adsorption of alkanethiols on zinc selenide substrates, Langmuir, vol.17, issue.13, pp.3937-3944, 2001.

T. M. Mcintire, O. Ryder, and B. J. Finlayson-pitts, Secondary ozonide formation from the ozone oxidation of unsaturated self-assembled monolayers on zinc selenide Attenuated Total Reflectance crystals, J. Phys. Chem. C, vol.113, issue.25, pp.11060-11065, 2009.

K. M. Wiencek and M. Fletcher, Effects of substratum wettability and molecular topography on the initial adhesion of bacteria to chemically defined substrata, Biofouling, vol.11, issue.4, pp.293-311, 1997.

B. Stuart, Infrared spectroscopy : fundamentals and applications, 2004.

F. Humbert and F. Quilès, In-situ study of early stages of biofilm formation under different environmental stresses by ATR-FTIR spectroscopy, Science against microbial pathogens: communicating current research and technological advances, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01935510

E. K. Plyler, Infrared spectra of methanol, ethanol, and n-propanol, Journal of Research of the National Bureau of Standards, vol.48, issue.4, p.2314, 1952.

J. P. Maity, Identification and discrimination of bacteria using Fourier transform infrared spectroscopy, Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, vol.116, pp.478-484, 2013.

Y. Yuan and T. R. Lee, Contact angle and wetting properties, Surface Science Techniques, pp.3-34, 2013.

, KRÜSS Technical Note: So you want to measure surface energy? Models for surface free energy calculation

F. M. Fowkes, Additivity of intermolecular forces at intefaces. I. Determination of the contribution to surface and interfacial tensions of dispersion forces in vatious liquids, J. Phys. Chem, vol.67, issue.12, pp.2538-2541, 1963.

W. Chu, J. W. Mayer, and M. Nicolet, Chapter 1 -Introduction, pp.1-20, 1978.

M. Mayer, Improved physics in SIMNRA 7, Nuclear Instruments and Methods in Physics Research Section B: Beam Interactions with Materials and Atoms, vol.332, pp.176-180, 2014.

A. T. Demissie, G. Haugstad, and C. D. Frisbie, Quantitative surface coverage measurements of selfassembled monolayers by Nuclear Reaction Analysis of Carbon-12, J. Phys. Chem. Lett, vol.7, issue.17, pp.3477-3481, 2016.

J. Mayer, L. A. Giannuzzi, T. Kamino, and J. Michael, TEM sample preparation and FIB-induced damage, MRS Bulletin, vol.32, issue.5, pp.400-407, 2007.

P. Tripathi, Towards a nanoscale view of lactic acid bacteria, Micron, vol.43, issue.12, pp.1323-1330, 2012.

J. B. Kaper, J. P. Nataro, and H. L. Mobley, Pathogenic Escherichia coli, Nature Reviews Microbiology, vol.2, issue.2, pp.123-140, 2004.

L. A. Pratt and R. Kolter, Genetic analysis of Escherichia coli biofilm formation: roles of flagella, motility, chemotaxis and type I pili, Molecular Microbiology, vol.30, issue.2, pp.285-293, 1998.

J. C. De-man, M. Rogosa, and S. M. Elisabeth, A medium for the cultivation of Lactobacilli, Journal of Applied Bacteriology, vol.23, issue.1, pp.130-135, 1960.

F. Quilès and F. Humbert, On the production of glycogen by Pseudomonas fluorescens during biofilm development: an in situ study by attenuated total reflection-infrared with chemometrics, Biofouling, vol.30, issue.6, pp.709-718, 2014.

, ThermoFisher Scientific. Fluorescence SpectraViewer

B. M. Corcoran, C. Stanton, G. F. Fitzgerald, and R. P. Ross, Survival of probiotic Lactobacilli in acidic environments is enhanced in the presence of metabolizable sugars, Appl. Environ. Microbiol, vol.71, issue.6, p.3060, 2005.

G. Cassanas, M. Morssli, E. Fabrègue, and L. Bardet, Vibrational spectra of lactic acid and lactates, Journal of Raman Spectroscopy, vol.22, issue.7, pp.409-413, 1991.

P. J. Bremer and G. G. Geesey, An evaluation of biofilm development utilizing non-destructive attenuated total reflectance Fourier transform infrared spectroscopy, Biofouling, vol.3, issue.2, pp.89-100, 1991.

A. R. Berry, C. M. Franco, W. Zhang, and A. P. Middelberg, Growth and lactic acid production in batch culture of Lactobacillus rhamnosus in a defined medium, Biotechnology Letters, vol.21, issue.2, pp.163-167, 1999.

V. Bottazzi, An introduction to rod-shaped lactic-acid bacteria, Biochimie, vol.70, issue.3, pp.303-315, 1988.

T. F. Cálix-lara, T. Duong, and T. M. Taylor, Addition of a surfactant to tryptic soy broth allows growth of a Lactic Acid Bacteria food antimicrobial, Escherichia coli O157:H7, and Salmonella enterica, Letters in Applied Microbiology, vol.54, issue.5, pp.392-397, 2012.

H. Siegumfeldt, K. Björn-rechinger, and M. Jakobsen, Dynamic changes of intracellular pH in individual lactic acid bacterium cells in response to a rapid drop in extracellular pH, Applied and Environmental Microbiology, vol.66, issue.6, pp.2330-2335, 2000.

S. Lebeer, Identification of a gene cluster for the biosynthesis of a long, galactose-rich exopolysaccharide in Lactobacillus rhamnosus GG and functional analysis of the priming glycosyltransferase, Appl Environ Microbiol, vol.75, issue.11, pp.3554-3563, 2009.

K. D. Young, The Selective value of bacterial shape, Microbiology and Molecular Biology Reviews, vol.70, issue.3, pp.660-703, 2006.

C. J. Ingham, M. Beerthuyzen, J. Van-hylckama, and . Vlieg, Population heterogeneity of Lactobacillus plantarum WCFS1 microcolonies in response to and recovery from acid stress, Applied and environmental microbiology, vol.74, issue.24, pp.7750-7758, 2008.

D. C. Yang, K. M. Blair, and N. R. Salama, Staying in shape: the impact of cell shape on bacterial survival in diverse environments, Microbiol. Mol. Biol. Rev, vol.80, issue.1, p.187, 2016.

E. Cefalì, Morphologic variations in bacteria under stress conditions: Near-field optical studies, Scanning, vol.24, issue.6, pp.274-283, 2002.

A. Chien, N. S. Hill, and P. A. Levin, Cell size control in bacteria, Current Biology, vol.22, issue.9, pp.340-349, 2012.

A. Typas, M. Banzhaf, C. A. Gross, and W. Vollmer, From the regulation of peptidoglycan synthesis to bacterial growth and morphology, Nature Reviews Microbiology, vol.10, p.123, 2011.

A. J. Egan, R. M. Cleverley, K. Peters, R. J. Lewis, and W. Vollmer, Regulation of bacterial cell wall growth, The FEBS Journal, vol.284, issue.6, pp.851-867, 2016.

L. Liu, G. Lindauer, W. B. Alexander, and P. H. Holloway, Surface preparation of ZnSe by chemical methods, Journal of Vacuum Science & Technology B: Microelectronics and Nanometer Structures Processing, vol.13, issue.6, pp.2238-2244, 1995.

A. Kita, M. Ozawa, and C. D. Gutleben, XPS analysis of chemically etched II-VI semiconductor surfaces, Applied Surface Science, vol.100, pp.652-655, 1996.

M. W. Cho, Surface treatment of znse substrate and homoepitaxy of znse, Journal of Electronic Materials, vol.26, issue.5, pp.423-428, 1997.

T. F. Mcgee and H. J. Cornelissen, X-ray photoelectron spectroscopy of etched ZnSe, Applied Surface Science, vol.35, issue.3, pp.371-379, 1989.

J. L. Meléndez and C. R. Helms, Wet chemical cleaning and surface analysis of ZnSe, Journal of The Electrochemical Society, vol.141, issue.7, pp.1973-1976, 1994.

, The crystran handbook of infra-red and ultra-violet optical materials, 2016.

H. Gong, H. Huang, L. Ding, M. Wang, and K. Liu, Characterization and optical properties of ZnSe prepared by hydrothermal method, Journal of Crystal Growth, vol.288, issue.1, pp.96-99, 2006.

S. Joshi, P. Pellacani, T. A. Van-beek, H. Zuilhof, and M. W. Nielen, Surface characterization and antifouling properties of nanostructured gold chips for imaging surface plasmon resonance biosensing, Sensors and Actuators B: Chemical, vol.209, pp.505-514, 2015.

W. Shen, Blocking agent optimization for nonspecific binding on phage based magnetoelastic biosensors, Journal of The Electrochemical Society, vol.159, issue.10, pp.818-823, 2012.

Z. Hussain, M. A. Salim, M. A. Khan, and E. E. Khawaja, X-ray photoelectron and auger spectroscopy study of copper-sodium-germanate glasses, Journal of Non-Crystalline Solids, vol.110, issue.1, pp.44-52, 1989.

R. G. Greenler, Infrared study of adsorbed molecules on metal surfaces by reflection techniques, J. Chem. Phys, vol.44, issue.1, pp.310-315, 1966.

S. Brown, J. P. Santa-maria, and S. Walker, Wall teichoic acids of gram-positive bacteria, Annu Rev Microbiol, vol.67, pp.313-336, 2013.

V. Lacour, K. Moumanis, W. M. Hassen, C. Elie-caille, T. Leblois et al., Formation kinetics of mixed self-assembled monolayers of alkanethiols on GaAs(100), Langmuir, vol.35, issue.13, pp.4415-4427, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01914370

M. R. Kosuri, R. Cone, Q. Li, S. M. Han, B. C. Bunker et al., Adsorption kinetics of 1-alkanethiols on hydrogenated Ge, vol.20, pp.835-840, 2004.

S. S. Cheng, D. A. Scherson, and C. N. Sukenik, In situ observation of monolayer self-assembly by FTIR/ATR, J. Am. Chem. Soc, vol.114, issue.13, pp.5436-5437, 1992.

N. Goutev and M. Futamata, Attenuated Total Reflection Surface-Enhanced Infrared Absorption Spectroscopy of carboxyl terminated self-assembled monolayers on gold, Appl Spectrosc, vol.57, issue.5, pp.506-513, 2003.

M. D. Porter, T. B. Bright, D. L. Allara, and C. E. Chidsey, Spontaneously organized molecular assemblies. 4. Structural characterization of n-alkyl thiol monolayers on gold by optical ellipsometry, infrared spectroscopy, and electrochemistry, J. Am. Chem. Soc, vol.109, issue.12, pp.3559-3568, 1987.

R. Arnold, W. Azzam, A. Terfort, and C. Wöll, Preparation, modification, and crystallinity of aliphatic and aromatic carboxylic acid terminated self-assembled monolayers, Langmuir, vol.18, issue.10, pp.3980-3992, 2002.

M. I. Béthencourt, L. Srisombat, P. Chinwangso, and T. R. Lee, SAMs on gold derived from the direct adsorption of alkanethioacetates are inferior to those derived from the direct adsorption of alkanethiols, Langmuir, vol.25, issue.3, pp.1265-1271, 2009.

M. R. Anderson, M. N. Evaniak, and M. Zhang, Influence of solvent on the interfacial structure of selfassembled alkanethiol monolayers, Langmuir, vol.12, issue.10, pp.2327-2331, 1996.

S. Hillebrandt, T. Glaser, and A. Pucci, Investigation of self-assembled monolayer formation using infrared-reflection-absorption-spectroscopy, presented at the Proc.SPIE, vol.9137, 2014.

T. Baum, S. Ye, and K. Uosaki, Formation of self-assembled monolayers of alkanethiols on GaAs surface with in situ surface activation by ammonium hydroxide, Langmuir, vol.15, issue.25, pp.8577-8579, 1999.

A. Urakawa, T. Buergi, and A. Baiker, Modulation Excitation PM-IRRAS: a new possibility for simultaneous monitoring of surface and gas species and surface properties, Chimia, vol.60, issue.4, pp.231-233, 2006.

J. Zhao, M. H. Na, E. H. Lee, H. C. Chang, J. A. Gardella et al., Surface chemistry of II-VI semiconductor ZnSe studied by time of flight secondary ion mass spectrometry and x-ray photoelectron spectroscopy, Journal of Vacuum Science & Technology B: Microelectronics and Nanometer Structures Processing, vol.16, issue.6, pp.3048-3054, 1998.

S. Skoglund, J. Hedberg, E. Yunda, A. Godymchuk, E. Blomberg et al., Difficulties and flaws in performing accurate determinations of zeta potentials of metal nanoparticles in complex solutions-Four case studies, PLOS ONE, vol.12, issue.7, p.181735, 2017.

M. Shenasa, S. Sainkar, and D. Lichtman, XPS study of some selected selenium compounds, Journal of Electron Spectroscopy and Related Phenomena, vol.40, issue.4, pp.329-337, 1986.

M. Hövel, B. Gompf, and M. Dressel, Dielectric properties of ultrathin metal films around the percolation threshold, Phys. Rev. B, vol.81, issue.3, p.35402, 2010.

B. Gompf, J. Beister, T. Brandt, J. Pflaum, and M. Dressel, Nanometer-thick Au-films as antireflection coating for infrared light, Opt. Lett, vol.32, issue.11, pp.1578-1580, 2007.

A. Klimovskaya, A. Sarikov, Y. Pedchenko, A. Voroshchenko, O. Lytvyn et al., Study of the formation processes of gold droplet arrays on Si substrates by high temperature anneals, Nanoscale Res Lett, vol.6, issue.1, pp.151-151, 2011.

E. N. Aybeke, Y. Lacroute, C. Elie-caille, A. Bouhelier, E. Bourillot et al., Homogeneous large-scale crystalline nanoparticle-covered substrate with high SERS performance, Nanotechnology, vol.26, issue.24, p.245302, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02868345

K. Ataka, S. T. Stripp, and J. Heberle, Surface-enhanced infrared absorption spectroscopy (SEIRAS) to probe monolayers of membrane proteins, Biochimica et Biophysica Acta (BBA) -Biomembranes, vol.1828, issue.10, pp.2283-2293, 2013.

S. Lortal, M. Rousseau, P. Boyaval, and J. Van-heijenoort, Cell wall and autolytic system of Lactobacillus helveticus ATCC 12046, Microbiology, vol.137, issue.3, pp.549-559, 1991.
URL : https://hal.archives-ouvertes.fr/hal-02715426

M. Bieri, T. Bürgi-;-atr-ir, P. , and Q. Study, Adsorption kinetics, orientation, and self-assembling of N-Acetyl-l-cysteine on gold, J. Phys. Chem. B, vol.109, issue.47, pp.22476-22485, 2005.

P. Tripathi, Adhesion and nanomechanics of pili from the probiotic Lactobacillus rhamnosus GG, ACS Nano, vol.7, issue.4, pp.3685-3697, 2013.

Q. Wang and T. J. Webster, Nanostructured selenium for preventing biofilm formation on polycarbonate medical devices, Journal of Biomedical Materials Research Part A, vol.100, issue.12, pp.3205-3210, 2012.

S. Kesel, A. Mader, P. H. Seeberger, O. Lieleg, and M. Opitz, Carbohydrate coating reduces adhesion of biofilm-forming Bacillus subtilis to gold surfaces, Appl. Environ. Microbiol, vol.80, issue.19, p.5911, 2014.

M. Jarosz, Novel bioelectrodes based on polysaccharide modified gold surfaces and electrochemically active Lactobacillus rhamnosus GG biofilms, Electrochimica Acta, vol.296, pp.999-1008, 2019.

J. Maillard, Bacterial target sites for biocide action, Journal of Applied Microbiology, vol.92, issue.s1, pp.16-27, 2002.

P. Maris, Scientific and Technical Review of the, Office International des Epizooties (Paris), vol.14, issue.1, pp.47-55, 1995.

C. P. Gerba, Quaternary ammonium biocides: efficacy in application, Appl. Environ. Microbiol, vol.81, issue.2, p.464, 2015.

K. P. Fears, S. E. Creager, and R. A. Latour, Determination of the surface pK of carboxylic-and amineterminated alkanethiols using surface plasmon resonance spectroscopy, Langmuir, vol.24, issue.3, pp.837-843, 2008.

T. H. Degefa, P. Schön, D. Bongard, and L. Walder, Elucidation of the electron transfer mechanism of marker ions at SAMs with charged head groups, Journal of Electroanalytical Chemistry, vol.574, issue.1, pp.49-62, 2004.

J. Riet, AFM topography and friction studies of hydrogen-bonded bilayers of functionalized alkanethiols, Soft Matter, vol.6, issue.15, pp.3450-3454, 2010.

D. Enders, T. Nagao, A. Pucci, T. Nakayama, and M. Aono, Surface-enhanced ATR-IR spectroscopy with interface-grown plasmonic gold-island films near the percolation threshold, Phys. Chem. Chem. Phys, vol.13, issue.11, pp.4935-4941, 2011.

F. Quilès, F. Humbert, and A. Delille, Analysis of changes in attenuated total reflection FTIR fingerprints of Pseudomonas fluorescens from planktonic state to nascent biofilm state, Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, vol.75, issue.2, pp.610-616, 2010.

S. Kang, P. J. Bremer, K. Kim, and A. J. Mcquillan, Monitoring metal ion binding in single-layer Pseudomonas aeruginosa biofilms using ATR?IR spectroscopy, Langmuir, vol.22, issue.1, pp.286-291, 2006.

M. C. Terraf, M. S. Tomás, L. Rault, Y. L. Loir, S. Even et al., Biofilms of vaginal Lactobacillus reuteri CRL 1324 and Lactobacillus rhamnosus CRL 1332: kinetics of formation and matrix characterization, Archives of Microbiology, vol.198, issue.7, pp.689-700, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01454610

H. Czerski, Small is beautiful: Surface tension and viscosity, Storm in a Teacup: The Physics of Everyday Life, pp.83-85, 2016.

M. V. Zubkov, B. M. Fuchs, H. Eilers, P. H. Burkill, and R. Amann, Determination of total protein content of bacterial cells by SYPRO staining and flow cytometry, Appl. Environ. Microbiol, vol.65, issue.7, p.3251, 1999.
URL : https://hal.archives-ouvertes.fr/hal-01651710

J. Riet, Molecular friction as a tool to identify functionalized alkanethiols, Langmuir, vol.26, issue.9, pp.6357-6366, 2010.

M. M. Khan, L. K. Ista, G. P. Lopez, and A. J. Schuler, Experimental and theoretical examination of surface energy and adhesion of nitrifying and heterotrophic bacteria using self-assembled monolayers, Environ. Sci. Technol, vol.45, issue.3, pp.1055-1060, 2011.

Y. Arima and H. Iwata, Effects of surface functional groups on protein adsorption and subsequent cell adhesion using self-assembled monolayers, J. Mater. Chem, vol.17, issue.38, pp.4079-4087, 2007.

B. Berron and G. K. Jennings, Loosely packed hydroxyl-terminated SAMs on gold, Langmuir, vol.22, issue.17, pp.7235-7240, 2006.

D. R. Absolom, F. V. Lamberti, Z. Policova, W. Zingg, C. J. Van-oss et al., Surface thermodynamics of bacterial adhesion, Appl. Environ. Microbiol, vol.46, issue.1, p.90, 1983.

M. R. Alexander and P. Williams, Water contact angle is not a good predictor of biological responses to materials, Biointerphases, vol.12, issue.2, pp.2-201, 2017.

R. Kügler, O. Bouloussa, and F. Rondelez, Evidence of a charge-density threshold for optimum efficiency of biocidal cationic surfaces, Microbiology, vol.151, issue.5, pp.1341-1348, 2005.

A. M. Bieser and J. C. Tiller, Mechanistic considerations on contact-active antimicrobial surfaces with controlled functional group densities, Macromolecular Bioscience, vol.11, issue.4, pp.526-534, 2011.

A. Beaussart, Single-cell force spectroscopy of probiotic bacteria, Biophys J, vol.104, issue.9, pp.1886-1892, 2013.

M. V. Riquelme, H. Zhao, V. Srinivasaraghavan, A. Pruden, P. Vikesland et al., Optimizing blocking of nonspecific bacterial attachment to impedimetric biosensors, Sensing and Bio-Sensing Research, vol.8, pp.47-54, 2016.

R. Konradi, B. Pidhatika, A. Mühlebach, and M. Textor, Poly-2-methyl-2-oxazoline: a peptide-like polymer for protein-repellent surfaces, Langmuir, vol.24, issue.3, pp.613-616, 2008.

M. Heuberger, T. Drobek, and N. D. Spencer, Interaction forces and morphology of a protein-resistant Poly(ethylene glycol) layer, Biophysical Journal, vol.88, issue.1, pp.495-504, 2005.

I. Connell, W. Agace, P. Klemm, M. Schembri, S. M?rild et al., Type 1 fimbrial expression enhances Escherichia coli virulence for the urinary tract, Proc Natl Acad Sci U S A, vol.93, issue.18, pp.9827-9832, 1996.

T. K. Wood, Insights on Escherichia coli biofilm formation and inhibition from whole-transcriptome profiling, Environ Microbiol, vol.11, issue.1, pp.1-15, 2009.

A. F. Barrios, R. Zuo, Y. Hashimoto, L. Yang, W. E. Bentley et al., Autoinducer 2 controls biofilm formation in Escherichia coli through a novel motility quorum-sensing regulator (MqsR, B3022), J Bacteriol, vol.188, issue.1, pp.305-316, 2006.

M. Guo, S. Gamby, Y. Zheng, and H. O. Sintim, Small molecule inhibitors of AI-2 signaling in bacteria: state-of-the-art and future perspectives for anti-quorum sensing agents, Int J Mol Sci, vol.14, issue.9, pp.17694-17728, 2013.

K. B. Xavier and B. L. Bassler, Regulation of uptake and processing of the quorum-sensing autoinducer AI-2 in Escherichia coli, J Bacteriol, vol.187, issue.1, pp.238-248, 2005.

G. Aidelberg, B. D. Towbin, D. Rothschild, E. Dekel, A. Bren et al., Hierarchy of non-glucose sugars in Escherichia coli, BMC Syst Biol, vol.8, pp.133-133, 2014.

C. Holm and L. Jespersen, A flow-cytometric gram-staining technique for milk-associated bacteria, Appl Environ Microbiol, vol.69, issue.5, pp.2857-2863, 2003.

D. J. Mason, S. Shanmuganathan, F. C. Mortimer, and V. A. Gant, A fluorescent Gram stain for flow cytometry and epifluorescence microscopy, Appl Environ Microbiol, vol.64, issue.7, pp.2681-2685, 1998.

A. H. Anvarian, Y. Cao, S. Srikumar, S. Fanning, and K. Jordan, Flow cytometric and 16S sequencing methodologies for monitoring the physiological status of the microbiome in powdered infant formula production, Front Microbiol, vol.7, pp.968-968, 2016.

J. D. Durtschi, Increased sensitivity of bacterial detection in cerebrospinal fluid by fluorescent staining on low-fluorescence membrane filters, Journal of Medical Microbiology, vol.54, issue.9, pp.843-850, 2005.

A. I. Lopez, A. Kumar, M. R. Planas, Y. Li, T. V. Nguyen et al., Biofunctionalization of silicone polymers using poly(amidoamine) dendrimers and a mannose derivative for prolonged interference against pathogen colonization, Biomaterials, vol.32, issue.19, pp.4336-4346, 2011.