D. I. Andersson, D. Hughes, and . Drug-resist, Update, vol.15, p.162, 2012.

A. V. Khan, Q. U. Ahmed, I. Shukla, and A. A. Khan, Asian Pac. J. Trop. Biomed, vol.2, p.189, 2012.

M. Simões, R. N. Bennett, and E. A. Rosa, Nat. Prod. Rep, vol.26, p.746, 2009.

S. Nabavi, A. Lorenzo, M. Izadi, E. Sobarzo-sánchez, M. Daglia et al., Nutrients, vol.7, p.7729, 2015.

G. Shrestha, J. Raphael, S. D. Leavitt, . St, and L. L. Clair, Pharm. Biol, vol.52, p.1262, 2014.

G. Shrestha, L. L. Clair, and . St, Phytochem. Rev, vol.12, p.229, 2013.

J. Boustie and M. Grube, Plant Genet. Resour. Charact. Util, vol.3, p.273, 2005.

V. Shukla, G. P. Joshi, and M. S. Rawat, Phytochem. Rev, vol.9, p.303, 2010.

M. Ba?korová, R. Jend?elovskyjend?elovsky´jend?elovsky´, M. Kello, M. Ba?kor, J. Mike? et al., Int. J. Publ. Assoc. Bibra, vol.26, p.462, 2012.

J. Yombi, L. Cyr;-belkhir, S. Jonckheere, D. Wilmes, O. Cornu et al., BMC Infect. Dis, vol.12, p.215, 2012.

N. J. Wood, H. F. Jenkinson, S. A. Davis, S. ;. Mann, D. J. O'sullivan et al., J. Mater. Sci. Mater. Med, 0201.

R. Huang, M. Li, M. Ye, K. Yang, X. Xu et al., Appl. Environ. Microbiol, vol.80, p.7212, 2014.

M. Kuboniwa, R. J. Lamont, and . Periodontology, , vol.52, 2000.

C. Y. Loo, D. A. Corliss, and N. Ganeshkumar, J. Bacteriol, vol.182, p.1374, 2000.

J. D. Rogers, E. M. Haase, A. E. Brown, C. W. Douglas, J. P. Gwynn et al., Read. Engl, vol.144, p.1223, 1998.

C. J. Cavallito, D. M. Fruehauf, and J. H. Bailey, J. Am. Chem. Soc, vol.70, p.3724, 1948.

S. Braukmüller and R. Brückner, Eur. J. Org. Chem, p.2110, 2006.

A. K. Perepogu, D. Raman, U. S. Murty, and V. Rao, J. Synth. Commun, p.686, 2010.

J. D. Guzman, A. Gupta, D. Evangelopoulos, C. Basavannacharya, L. C. Pabon et al., J. Antimicrob. Chemother, p.2101, 2010.

Y. Yang, X. Ye, X. Li, J. Zhen, B. Zhang et al., Planta Med, vol.73, p.602, 2007.

F. L. Sebastianes, N. Cabedo, N. E. Aouad, A. M. Valente, P. T. Lacava et al., Curr. Microbiol, vol.65, p.622, 2012.

S. Kitani, K. T. Miyamoto, S. Takamatsu, E. Herawati, H. Iguchi et al., Proc. Natl. Acad. Sci. U.S.A, vol.108, 2011.

T. Berkenbusch and R. Brückner, Tetrahedron, vol.54, p.11471, 1998.

R. A. Fernandes and A. K. Chowdhury, Eur. J. Org. Chem, 1106.

F. P. Kuhajda, E. S. Pizer, J. N. Li, N. S. Mani, G. L. Frehywot et al., Proc. Natl. Acad. Sci. U.S.A, p.3450, 2000.

J. Boustie, M. Galibert-anne, F. Devehat, M. Chollet-krugler, S. Tomasi et al., , vol.20, p.459, 2015.

S. Huneck, R. Z. Takeda, and . Naturforsch, , vol.4, p.7, 1992.

S. Huneck, K. Schreiber, G. Höfle, and G. Snatzke, J. Hattori Bot. Lab, vol.45, p.1, 1979.

O. Nicolle, A. Rouillon, H. Guyodo, Z. Tamanai-shacoori, F. Chandad et al., FEMS Immunol. Med. Microbiol, 2010.

R. M. Donlan and J. W. Costerton, Clin. Microbiol. Rev, vol.15, p.167, 2002.

D. W. Hecht, National Committee for Clinical Laboratory Standards Methods for antimicrobial Susceptibility Testing of Anaerobic Bacteria: Approved Standard, 2004.

T. Mosmann, J. Immunol. Methods, vol.5, p.5, 1983.

N. A. Ahmed, F. C. Petersen, and A. A. Scheie, AI-2/LuxS Is Involved in Increased Biofilm Formation by Streptococcus intermedius in the Presence of Antibiotics, Antimicrob Agents Chemother, vol.53, pp.4258-4263, 2009.

S. T. Aka and S. H. Haji, Sub-MIC of antibiotics induced biofilm formation of Pseudomonas aeruginosa in the presence of chlorhexidine, Braz J Microbiol, vol.46, pp.149-154, 2015.

D. I. Andersson and D. Hughes, Microbiological effects of sublethal levels of antibiotics, Nat Rev Microbiol, vol.12, pp.465-478, 2014.

P. M. Bartold and A. S. Narayanan, Molecular and cell biology of healthy and diseased periodontal tissues, Periodontol, vol.40, pp.29-49, 2000.

P. Batchelor, Is periodontal disease a public health problem?, Br Dent J, vol.217, pp.405-409, 2014.

J. Boustie and M. Grube, Lichens-a promising source of bioactive secondary metabolites, Plant Genet Resour Charact Util, vol.3, pp.273-287, 2005.

J. Bueno, Anti-Biofilm Drug Susceptibility Testing Methods: Looking for New Strategies against Resistance Mechanism, J Microb Biochem Technol, 2011.

C. A. Capestany, G. D. Tribble, K. Maeda, D. R. Demuth, and R. J. Lamont, Role of the Clp System in Stress Tolerance, Biofilm Formation, and Intracellular Invasion in Porphyromonas gingivalis, J Bacteriol, vol.190, pp.1436-1446, 2008.

C. J. Cavallito, D. M. Fruehauf, and J. H. Bailey, Lactone aliphatic acids as antibacterial agents, J Am Chem Soc, vol.70, pp.3724-3726, 1948.

W. Chen, K. Honma, A. Sharma, and H. K. Kuramitsu, A universal stress protein of Porphyromonas gingivalis is involved in stress responses and biofilm formation, Fems Microbiol Lett, vol.264, pp.15-21, 2006.

G. D. Christensen, W. A. Simpson, J. J. Younger, L. M. Baddour, F. F. Barrett et al., Adherence of coagulase-negative staphylococci to plastic tissue culture plates: a quantitative model for the adherence of staphylococci to medical devices, J Clin Microbiol, vol.22, pp.996-1006, 1985.

D. Dû, L. , E. Kolenbrander, and P. , Identification of Saliva-Regulated Genes of Streptococcus gordonii DL1 by Differential Display Using Random Arbitrarily Primed PCR, Infect Immun, vol.68, pp.4834-4837, 2000.

J. F. Gerster, S. R. Rohlfing, S. E. Pecore, R. M. Winandy, R. M. Stern et al., Synthesis, absolute configuration, and antibacterial activity of 6,7-dihydro-5,8dimethyl-9-fluoro-1-oxo-1H,5H-benzo[ij]quinolizine-2-carboxylic acid, J Med Chem, vol.30, pp.839-843, 1987.

K. Y. How, K. P. Song, and K. G. Chan, Porphyromonas gingivalis: An overview of periodontopathic pathogen below the gum line, Front Microbiol, 2016.

H. F. Jenkinson and D. R. Demuth, Structure, function and immunogenicity of streptococcal antigen I/II polypeptides, Mol Microbiol, vol.23, pp.183-190, 1997.

J. Kreth, J. Merritt, and F. Qi, Bacterial and Host Interactions of Oral Streptococci, Dna Cell Biol, vol.28, pp.397-403, 2009.

M. Kuboniwa and R. J. Lamont, Subgingival biofilm formation, Periodontol, vol.52, pp.38-52, 2000.

M. Kuboniwa and R. J. Lamont, Subgingival biofilm formation: Subgingival biofilm formation, Periodontol, vol.52, pp.38-52, 2000.

L. Bars, H. , L. Gall-david, S. Renoux, V. M. Bonnaure-mallet et al., Impact of a mutator phenotype on motility and cell adherence in Salmonella Heidelberg, Vet Microbiol, vol.159, pp.99-106, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00796481

C. Y. Loo, D. A. Corliss, and N. Ganeshkumar, Streptococcus gordonii biofilm formation: identification of genes that code for biofilm phenotypes, J Bacteriol, vol.182, pp.1374-1382, 2000.

R. D. Lunsford and J. London, Natural genetic transformation in Streptococcus gordonii: comX imparts spontaneous competence on strain wicky, J Bacteriol, vol.178, pp.5831-5835, 1996.

T. Mauro, A. Rouillon, and B. Felden, Insights into the regulation of small RNA expression: SarA represses the expression of two sRNAs in Staphylococcus aureus, Nucleic Acids Res gkw777, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01365876

R. Mcnab, S. K. Ford, A. El-sabaeny, B. Barbieri, G. S. Cook et al., LuxS-based signaling in Streptococcus gordonii: autoinducer 2 controls carbohydrate metabolism and biofilm formation with Porphyromonas gingivalis, J Bacteriol, vol.185, pp.274-284, 2003.

J. Mysak, S. Podzimek, P. Sommerova, Y. Lyuya-mi, J. Bartova et al., Porphyromonas gingivalis : Major Periodontopathic Pathogen Overview, J Immunol Res, vol.2014, pp.1-8, 2014.

R. Nakao, H. Senpuku, and H. Watanabe, Porphyromonas gingivalis galE Is Involved in Lipopolysaccharide O-Antigen Synthesis and Biofilm Formation, Infect Immun, vol.74, pp.6145-6153, 2006.

R. Nakao, H. Senpuku, and H. Watanabe, Porphyromonas gingivalis galE Is Involved in Lipopolysaccharide O-Antigen Synthesis and Biofilm Formation, Infect Immun, vol.74, pp.6145-6153, 2006.

O. Nicolle, A. Rouillon, H. Guyodo, Z. Tamanai-shacoori, F. Chandad et al., Development of SNAP-tag-mediated live cell labeling as an alternative to GFP in Porphyromonas gingivalis: New tool for labeling live Porphyromonas gingivalis, Fems Immunol Med Microbiol, 2010.

O. 'toole, G. Kaplan, H. B. Kolter, and R. , Biofilm Formation as Microbial Development, Annu Rev Microbiol, vol.54, pp.49-79, 2000.

J. H. Park, J. Lee, H. Um, C. Lee, and S. , A periodontitis-associated multispecies model of an oral biofilm, J Periodontal Implant Sci, vol.44, p.79, 2014.

A. Podbielski and B. Kreikemeyer, Cell density-dependent regulation: basic principles and effects on the virulence of Gram-positive cocci, Int J Infect Dis, vol.8, pp.81-95, 2004.

A. Podbielski and B. Kreikemeyer, Cell density-dependent regulation: basic principles and effects on the virulence of Gram-positive cocci, Int J Infect Dis, vol.8, pp.81-95, 2004.

R. Ramanan, B. Kim, D. Cho, H. Oh, and H. Kim, Algae-bacteria interactions: Evolution, ecology and emerging applications, Biotechnol Adv, vol.34, pp.14-29, 2015.

J. D. Rogers, E. M. Haase, A. E. Brown, C. Douglas, J. P. Gwynn et al., Identification and analysis of a gene (abpA) encoding a major amylase-binding protein in Streptococcus gordonii, Microbiology, vol.144, pp.1223-1233, 1998.

G. Shrestha, . St, and L. L. Clair, Lichens: a promising source of antibiotic and anticancer drugs, Phytochem Rev, vol.12, pp.229-244, 2013.

V. Shukla, G. P. Joshi, and M. Rawat, Lichens as a potential natural source of bioactive compounds: a review, Phytochem Rev, vol.9, pp.303-314, 2010.

S. K. Singhrao, A. Harding, S. Poole, L. Kesavalu, and S. Crean, Porphyromonas gingivalis: Periodontal infection and its putative links with Alzheimer's disease, Mediators Inflamm, vol.2015, pp.1-10, 2015.

H. Song, J. Lee, H. Um, C. Lee, S. Lee et al., Phototoxic effect of blue light on the planktonic and biofilm state of anaerobic periodontal pathogens, J Periodontal Implant Sci, vol.43, p.72, 2013.

, How to Overcome the Antibiotic Crisis, 2016.

A. Sweidan, M. Chollet-krugler, P. Van-de-weghe, A. Chokr, S. Tomasi et al., Design, synthesis and biological evaluation of potential antibacterial butyrolactones, Bioorg Med Chem, vol.24, pp.5823-5833, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01398051

L. R. Swem, D. L. Swem, O. 'loughlin, C. T. Gatmaitan, R. Zhao et al., A Quorum-Sensing Antagonist Targets Both Membrane-Bound and Cytoplasmic Receptors and Controls Bacterial Pathogenicity, Mol Cell, vol.35, pp.143-153, 2009.

Y. Wakiyama, K. Kumura, E. Umemura, K. Ueda, S. Masaki et al., Synthesis and structure-activity relationships of novel lincomycin derivatives. Part 1. Newly generated antibacterial activities against Gram-positive bacteria with erm gene by C-7 modification, J Antibiot (Tokyo), vol.69, pp.368-380, 2016.

M. Wilson, Susceptibility of oral bacterial biofilms to antimicrobial agents, J Med Microbiol, vol.44, pp.79-87, 1996.

J. Wu, X. Lin, and H. Xie, Porphyromonas gingivalis short fimbriae are regulated by a FimS/FimR two-component system, Fems Microbiol Lett, vol.271, pp.214-221, 2007.

. Yombi-j-cyr, L. Belkhir, S. Jonckheere, D. Wilmes, O. Cornu et al., Streptococcus gordonii septic arthritis : two cases and review of literature, Bmc Infect Dis, vol.12, p.215, 2012.

O. Özgenç, J. World, . Methodol, A. Borges, A. Abreu et al., REFERENCES, vol.6, issue.1, p.877, 2016.

G. Shrestha, . St, L. L. Clair, J. Boustie, M. Grube et al., Plant Genet. Resour. Charact. Util, vol.12, issue.1, p.5823, 2005.

K. Y. How, K. P. Song, and K. G. Chan, Front. Microbiol, vol.7, p.53, 2016.

P. P. Pita, J. A. Rodrigues, C. Ota-tsuzuki, T. F. Miato, E. G. Zenobio et al., Biomed Res. Int, issue.10, p.60, 2015.

F. Guilhelmelli, N. Vilela, P. Albuquerque, L. Derengowski, S. Da et al., Front. Microbiol, issue.4, p.353, 2013.

J. Van-heijenoort, . Nat, . Prod, A. Rep-;-typas, M. Banzhaf et al., 123. (14) Smith, Nat. Rev. Microbiol, vol.18, issue.5, p.689, 1997.

M. Matija?i?, V. Muni?-kos, K. Nuji?, S. ?u?i?, J. Padovan et al., 332. (21) Tennessen, J. Periodontal Implant Sci, vol.2012, issue.4, p.79, 2005.

D. J. Arndt-jovin, T. M. Jovin, S. Utaida, P. M. Dunman, D. Macapagal et al., 143. (26) Hoch, Biomed. Pharmacother. Biomedecine Pharmacother, vol.30, issue.24, p.936, 1989.

V. K. Gupta, S. Verma, S. Gupta, A. Singh, A. Pal et al., J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol, vol.2012, issue.12, p.5823, 2016.

S. Leejae, P. W. Taylor, S. P. Voravuthikunchai, H. Le-bars, ;. Gall-david et al., J. Med. Microbiol, vol.62, issue.36, p.99, 2013.

M. Kuboniwa and R. J. Lamont, Subgingival biofilm formation, Periodontol, vol.52, pp.38-52, 2000.

J. Cyr-yombi, L. Belkhir, S. Jonckheere, D. Wilmes, O. Cornu et al., Streptococcus gordonii septi aathhitis : tto ases aad eie of liteatuue, B IIfet. Dis, vol.12, p.215, 2012.

N. J. Wood, H. F. Jeekiso, ;. Daais, &. Maa, D. J. O'"ulliaa et al., Chloheeidie hexametaphosphate nanoparticles as a novel antimicrobial coating for dental implants, J. Mater. Sci. Mater. Med, vol.26, p.201, 2015.

R. Huang, M. Li, M. Ye, K. Yang, X. Xu et al., Effects of Nicotine on Streptococcus gordonii Growth, Biofilm Formation, and Cell Aggregation, vol.80, pp.7212-7218, 2014.

P. M. Bartold and A. S. Narayanan, Molecular and cell biology of healthy and diseased periodontal tissues, Periodontol, vol.40, pp.29-49, 2000.

S. K. Singhrao, A. Harding, S. Poole, L. Kesavalu, and S. Crean, Porphyromonas gingivalis Periodontal IIfeetio aad Its Putatie Liks ith Alzheiee's Disease, Mediatos IIfla

C. Y. Loo, D. A. Corliss, and N. Ganeshkumar, Streptococcus gordonii biofilm formation: identification of genes that code for biofilm phenotypes, J. Bacteriol, vol.182, pp.1374-1382, 2000.

J. D. Rogers, E. M. Haase, A. E. Brown, C. W. Douglas, J. P. Gwynn et al., Identification and analysis of a gene (abpA) encoding a major amylase-binding protein in Streptococcus gordonii, Microbiol. Read. Engl, vol.144, pp.1223-1233, 1998.

D. I. Andersson and D. Hughes, Evolution of antibiotic resistance at non-lethal drug concentrations, Drug Resist. Updat, vol.15, pp.162-172, 2012.

G. Shrestha, J. Raphael, S. D. Leavitt, L. L. St, and . Clair, In vitro evaluation of the antibacterial activity of extracts from 34 species of North American lichens, Pharm. Biol, vol.52, pp.1262-1266, 2014.

A. V. Khan, Q. U. Ahmed, I. Shukla, and A. A. Khan, Antibacterial activity of leaves extracts of Trifolium alexandrinum Linn. against pathogenic bacteria causing tropical diseases, Asian Pac, J. Trop. Biomed, vol.2, pp.60040-60049, 2012.

S. Nabavi, A. D. Lorenzo, M. Izadi, E. Sobarzo-sánchez, M. Daglia et al., Antibacterial Effects of Cinnamon: From Farm to Food, Cosmetic and Pharmaceutical Industries, Nutrients, vol.7, pp.7729-7748, 2015.

G. Shrestha, L. L. St, and . Clair, Lichens: a promising source of antibiotic and anticancer drugs, Phytochem. Rev, vol.12, pp.229-244, 2013.

J. Boustie and M. Grube, Lichens-a promising source of bioactive secondary metabolites, Plant Genet. Resour. Charact. Util, vol.3, pp.273-287, 2005.

V. Shukla, G. P. Joshi, and M. S. Rawat, Lichens as a potential natural source of bioactive compounds: a review, Phytochem. Rev, vol.9, pp.303-314, 2010.

N. M. Islam, I. Bhattacharyya, and D. M. Cohen, Common Oral Manifestations of Systemic Disease, Otolaryngol. Clin. North Am, vol.44, pp.161-182, 2011.

F. E. Dewhirst, T. Chen, J. Izard, B. J. Paster, A. C. Tanner et al., The Human Oral Microbiome, J. Bacteriol, vol.192, pp.5002-5017, 2010.

A. Nezar, Khat and Oral Microbiota, A microbial study with relevance to periodontitis and dental caries, 2005.

F. J. Laine and W. R. Smoker, Oral cavity: anatomy and pathology, Semin. Ultrasound. CT MR, vol.16, pp.527-545, 1995.

V. M. Zohrabian, C. S. Poon, and J. J. Abrahams, Embryology and Anatomy of the Jaw and Dentition, Ultrasound Ct Mri, vol.36, pp.397-406, 2015.

R. C-;-heid and J. B. Woelfel, Woelfel's deetal aaato, 2012.

R. M. Donlan and J. W. Costerton, Biofilms: Survival Mechanisms of Clinically Relevant Microorganisms, Clin. Microbiol. Rev, vol.15, pp.167-193, 2002.

R. Banthia, R. Chandki, and P. Banthia, Biofilms: A microbial home, J. Indian Soc. Periodontol, vol.15, p.111, 2011.

N. Rabin, Y. Zheng, C. Opoku-temeng, Y. Du, E. Bonsu et al., Biofilm formation mechanisms and targets for developing antibiofilm agents, Future Med. Chem, vol.7, pp.493-512, 2015.

R. Vasudevan, Biofilms: Microbial Cities of Scientific Significance, J. Microbiol. Exp, vol.1, 2014.

I. Cabarkapa, J. Levic, O. Djuragic, and B. , Microb. Pathog. Strat. Combat. Them Sci. Technol. Educ., Formatex research center, pp.42-51, 2013.

T. Mattila-"adholm and G. Wirtanen, Biofilm formation in the industry: A review, Food Rev. Int, vol.8, pp.573-603, 1992.

R. Van-houdt and C. W. Michiels, Biofilm formation and the food industry, a focus on the bacterial outer surface, J. Appl. Microbiol, vol.109, pp.1117-1131, 2010.

J. Klahre, Monitoring of biofouling in papermill process waters, Water Res, vol.34, pp.3657-3665, 2000.

N. Høiby, O. Ciofu, H. K. Johansen, Z. Song, C. Moser et al., The liial ipaat of ateial iofils, IIt. J. OOal

, Encyclopaedia Britannica, Human body, Encylopaedia Br, 2017.

S. Kackar, E. Suman, . Ms, and . Kotian, Bacterial and fungal biofilm formation on contact lenses and their susceptibility to lens care solutions, Indian J. Med. Microbiol, vol.35, p.80, 2017.

J. H. Park, J. Lee, H. Um, B. Chang, and S. Lee, A periodontitis-associated multispecies model of an oral biofilm, J. Periodontal Implant Sci, vol.44, p.79, 2014.

S. Ji, Y. S. Choi, and Y. Choi, Bacterial invasion and persistence: critical events in the pathogenesis of periodontitis?, J. Periodontal Res, vol.50, pp.570-585, 2015.

H. F. Jenkinson and R. J. Lamont, Oral microbial communities in sickness and in health, Trends Microbiol, vol.13, pp.589-595, 2005.

F. A. Scannapieco, Saliva-bacterium interactions in oral microbial ecology, Crit. Rev. Oral Biol. Med. Off. Publ. Am. Assoc. Oral Biol, vol.5, pp.203-248, 1994.

M. Costalonga and M. C. Herzberg, The oral microbiome and the immunobiology of periodontal disease and caries, Immunol. Lett, vol.162, pp.22-38, 2014.

M. Quirynen, M. De-soete, K. Dierickx, and D. Van-steenberghe, The intra-oral translocation of periodontopathogens jeopardises the outcome of periodontal therapy. A review of the literature, J. Clin. Periodontol, vol.28, pp.499-507, 2001.

L. Sbordone and C. Bortolaia, Oral microbial biofilms and plaque-related diseases: microbial communities and their role in the shift from oral health to disease, Clin. Oral Investig, vol.7, pp.181-188, 2003.

S. E. Whitmore and R. J. Lamont, The pathogenic persona of community-associated oral streptococci, Mol. Microbiol, vol.81, pp.305-314, 2011.

H. M. Lappin-scott and J. W. Consterton, The Press Syndicate of The University of Cambridge, Microbial biofilms, 1995.

M. C. Sánchez, A. Llama-palacios, V. Blanc, R. León, D. Herrera et al., Structure, viability and bacterial kinetics of an in vitro biofilm model using six bacteria from the subgingival microbiota: An in vitro subgingival biofilm model, J. Periodontal Res, vol.46, pp.252-260, 2011.

S. S. Socransky, A. D. Haffajee, M. A. Cugini, C. Smith, and R. L. Kent, Microbial complexes in subgingival plaque, J. Clin. Periodontol, vol.25, pp.134-144, 1998.

A. D. Haffajee, S. S. Socransky, M. R. Patel, and X. Song, Microbial complexes in supragingival plaque, Oral Microbiol. Immunol, vol.23, pp.196-205, 2008.

R. M. Donlan and J. W. Costerton, Biofilms: Survival Mechanisms of Clinically Relevant Microorganisms, Clin. Microbiol. Rev, vol.15, pp.167-193, 2002.

, Periodontal (gum) disease. Causes, symptoms and treatments, Anonymous, 2013.

G. D. Tribble and R. J. Lamont, Bacterial invasion of epithelial cells and spreading in periodontal tissue, Periodontol, vol.52, pp.68-83, 2000.

P. J. Thomson, C. S. Potten, and D. R. Appleton, In vitro labelling studies and the measurement of epithelial cell proliferative activity in the human oral cavity, Arch. Oral Biol, vol.46, pp.65-71, 2001.

M. E. Ryan, Nonsurgical Approaches for the Treatment of Periodontal Diseases, Dent. Clin. North Am, vol.49, pp.611-636, 2005.

M. G. Newman and H. H. Takei, Caaza's liial peeiodotolog, 2012.

J. Sahni, S. Talegaonkar, M. Tariq, Z. Ahmad, J. Ali et al., Treatment modalities and evaluation models for periodontitis, Int. J. Pharm. Investig, vol.2, p.106, 2012.

R. Nagpal, Y. Yamashiro, and Y. Izumi, The Two-Way Association of Periodontal Infection with Systemic Disorders: An Overview, Mediators Inflamm, pp.1-9, 2015.

A. Arigbede, . Bo, . Babatope, . Mk, and . Bamidele, Periodontitis and systemic diseases: A literature review, J. Indian Soc. Periodontol, vol.16, p.487, 2012.

C. B. Weibe and E. E. Putnins, The periodontal disease classification system of the American Academy of Periodontology-An update, J. Candian Dent. Assoc, vol.66, pp.594-597, 2000.

G. C. Armitage and M. P. Cullinan, Comparison of the clinical features of chronic and aggressive periodontitis, Periodontol, vol.53, pp.12-27, 2000.

, The Firmicutes, vol.2, 2009.

R. J. Lamont and H. F. Jenkinson, Oral microbiology at a glance

, Oral microbial ecology: current research and new perspectives, 2013.

L. D. Dû and P. E. Kolenbrander, Identification of Saliva-Regulated Genes of Streptococcus gordonii DL1 by Differential Display Using Random Arbitrarily Primed PCR, Infect. Immun, vol.68, pp.4834-4837, 2000.

H. F. Jenkinson and D. R. Demuth, Structure, function and immunogenicity of streptococcal antigen I/II polypeptides, Mol. Microbiol, vol.23, pp.183-190, 1997.

J. D. Rogers, E. M. Haase, A. E. Brown, C. W. Douglas, J. P. Gwynn et al., Identification and analysis of a gene (abpA) encoding a major amylase-binding protein in Streptococcus gordonii, Microbiology, vol.144, pp.1223-1233, 1998.

A. Podbielski and B. Kreikemeyer, Cell density-dependent regulation: basic principles and effects on the virulence of Gram-positive cocci, Int. J. Infect. Dis, vol.8, pp.81-95, 2004.

C. Y. Loo, D. A. Corliss, and N. Ganeshkumar, Streptococcus gordonii biofilm formation: identification of genes that code for biofilm phenotypes, J. Bacteriol, vol.182, pp.1374-1382, 2000.

R. D. Lunsford and J. London, Natural genetic transformation in Streptococcus gordonii: comX imparts spontaneous competence on strain wicky, J. Bacteriol, vol.178, pp.5831-5835, 1996.

R. Mcnab, S. K. Ford, A. El-sabaeny, B. Barbieri, G. S. Cook et al., LuxS-based signaling in Streptococcus gordonii: autoinducer 2 controls carbohydrate metabolism and biofilm formation with Porphyromonas gingivalis, J. Bacteriol, vol.185, pp.274-284, 2003.

N. R. Krieg, J. T. Staley, D. R. Brown, B. P. Hedlund, B. J. Paster et al., Beegee's Maaual® of "steati Baateiolog, 2010.

K. Y. How, K. P. Song, and K. G. Chan, Porphyromonas gingivalis: An overview of periodontopathic pathogen below the gum line, Front. Microbiol, vol.7, 2016.

J. Mysak, S. Podzimek, P. Sommerova, Y. Lyuya-mi, J. Bartova et al., Porphyromonas gingivalis : Major Periodontopathic Pathogen Overview, J. Immunol. Res, pp.1-8, 2014.

S. K. Singhrao, A. Harding, S. Poole, L. Kesavalu, and S. Crean, Porphyromonas gingivalis: Periodontal ifeetio aad its putatie liks ith Alzheiee's disease, Mediatos IIfla

M. Kuboniwa and R. J. Lamont, Subgingival biofilm formation: Subgingival biofilm formation, Periodontol, vol.52, pp.38-52, 2000.

W. Chen, K. Honma, A. Sharma, and H. K. Kuramitsu, A universal stress protein of Porphyromonas gingivalis is involved in stress responses and biofilm formation, Fems Microbiol. Lett, vol.264, pp.15-21, 2006.

R. Nakao, H. Senpuku, and H. Watanabe, Porphyromonas gingivalis galE Is Involved in Lipopolysaccharide O-Antigen Synthesis and Biofilm Formation, Infect. Immun, vol.74, pp.6145-6153, 2006.

C. A. Capestany, G. D. Tribble, K. Maeda, D. R. Demuth, and R. J. Lamont, Role of the Clp System in Stress Tolerance, Biofilm Formation, and Intracellular Invasion in Porphyromonas gingivalis, J. Bacteriol, vol.190, pp.1436-1446, 2008.

P. Ooua, W. ?aauz, M. Maak, and . Chosz-gajewska, Antimicrobial photodynamic therapy-A discovery originating from the pre-antibiotic era in a novel periodontal therapy, Photodiagnosis Photodyn. Ther, vol.12, pp.612-618, 2015.

K. Ingólfsdóttir, Usnic acid, Phytochemistry, vol.61, pp.383-390, 2002.

. Periodontal, Gum) Disease: Causes, Symptoms, and Treatments, 2013.

R. Teanpaisan, S. Senapong, and J. Puripattanavong, In vitro Antimicrobial and Antibiofilm Activity of Artocarpus Lakoocha (Moraceae) Extract against Some Oral Pathogens, Trop. J. Pharm. Res, vol.13, p.1149, 2014.

F. Ben-taheur, B. Kouidhi, K. Fdhila, H. Elabed, R. Ben et al., Anti-bacterial and anti-biofilm activity of probiotic bacteria against oral pathogens, Microb. Pathog, vol.97, pp.213-220, 2016.

J. Latimer, J. L. Munday, K. M. Buzza, S. Forbes, P. K. Sreenivasan et al., Antibacterial and anti-biofilm activity of mouthrinses containing cetylpyridinium chloride and sodium fluoride, Bmc Microbiol, vol.15, 2015.

C. Cabral-romero, J. J. Martinez-sanmiguel, D. Résendez-pérez, M. Del, S. Flores-goonzaléz et al., Antibacterial and Anti-Biofilm Activities of Ambroxol Against Oral Bacteria, Pharma Innov, vol.2, 2013.

K. Ouhara, Susceptibilities of periodontopathogenic and cariogenic bacteria to antibacterial peptides,-defensins and LL37, produced by human epithelial cells, J. Antimicrob. Chemother, vol.55, pp.888-896, 2005.

A. Kapoor, R. Malhotra, V. Grover, and D. Grover, Systemic antibiotic therapy in periodontics, Denta Res. J, vol.9, pp.505-515, 2012.

A. Prakasam, . Ss, R. Elavarasu, and . Natarajan, Antibiotics in the management of aggressive periodontitis, J. Pharm. Bioallied Sci, vol.4, p.252, 2012.

, Anonymous, Antimicrobial resistance, 2015.

A. J. Alanis, Resistance to Antibiotics: Are We in the Post-Antibiotic Era?, Arch. Med. Res, vol.36, pp.697-705, 2005.

P. R. Philip, R. Lee, and C. Lin, The Antibiotic Paradox: How the Misuse of Antibiotics Destroys Their Curative Powers (review), Perspect, Biol. Med, vol.46, pp.603-604, 2003.

C. H. Rammelkamp and T. Maxon, Resistance of Staphylococcus aureus to the Action of Penicillin, Exp. Biol. Med, vol.51, pp.386-389, 1942.

H. W. Jaffe, J. W. Biddle, S. R. Johnson, and P. J. Wiesner, Infections due to penicillinase-producing Neisseria gonorrhoeae in the United States: 1976-1980, J. Infect. Dis, vol.144, pp.191-197, 1981.

I. Lind, Epidemiology of antibiotic resistant Neisseria gonorrhoeae in industrialized and developing countries, Scand. J. Infect. Dis. Suppl, vol.69, pp.77-82, 1990.

J. D. Williams and F. Moosdeen, Antibiotic Resistance in Haemophilus influenzae: Epidemiology, Mechanisms, and Therapeutic Possibilities, vol.8, pp.555-561, 1986.

J. H. Jorgensen, Update on Mechanisms and Prevalence of Antimicrobial Resistance in Haemophilus influenzae, Clin. Infect. Dis, vol.14, pp.1119-1123, 1992.

F. D. Lowy, Staphylococcus aureus Infections, vol.339, pp.520-532, 1998.

S. Deresinski, Methicillin-Resistant Staphylococcus aureus: An Evolutionary, Epidemiologic, and Therapeutic Odyssey, Clin. Infect. Dis, vol.40, pp.562-573, 2005.
DOI : 10.1086/427701

URL : https://academic.oup.com/cid/article-pdf/40/4/562/1010555/40-4-562.pdf

F. D. Lowy, Antimicrobial resistance: the example of Staphylococcus aureus, J. Clin. Invest, vol.111, pp.1265-1273, 2003.
DOI : 10.1172/jci200318535

URL : http://www.jci.org/articles/view/18535/files/pdf

T. J. Foste, The "taphloous aueus supeeeug, J. Cli. IIest, pp.1693-1696

A. Pablos-méndez, M. C. Raviglione, A. Laszlo, N. Binkin, H. L. Rieder et al., Global Surveillance for Antituberculosis-Drug Resistance, vol.338, pp.1641-1649, 1994.

M. A. Espinal, A. Laszlo, L. Simonsen, F. Boulahbal, S. J. Kim et al., Global Trends in Resistance to Antituberculosis Drugs, vol.344, pp.1294-1303, 2001.
DOI : 10.1056/nejm200104263441706

G. W. Waterer and R. G. Wunderink, Increasing threat of Gram-negative bacteria, Crit. Care Med, vol.29, pp.75-81, 2001.
DOI : 10.1097/00003246-200104001-00004

M. E. Rupp and P. D. Fey, Extended spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae: considerations for diagnosis, prevention and drug treatment, Drugs, vol.63, pp.353-365, 2003.
DOI : 10.2165/00003495-200363040-00002

D. G. White, S. Zhao, R. Sudler, S. Ayers, S. Friedman et al., The Isolation of Antibiotic-Resistant Salmonella from Retail Ground Meats, N. Engl. J. Med, vol.345, pp.1147-1154, 2001.

K. E. Smith, J. M. Besser, C. W. Hedberg, F. T. Leano, J. B. Bender et al., Quinolone-Resistant Campylobacter jejuni Infections in Minnesota, N. Engl. J. Med, vol.340, pp.1525-1532, 1992.

H. C. Wegener, The Consequences for Food Safety of the Use of Fluoroquinolones in Food Animals, N. Engl. J. Med, vol.340, pp.1581-1582, 1999.

P. D. Fey, T. J. Safranek, M. E. Rupp, E. F. Dunne, E. Ribot et al., Ceftriaxone-Resistant Salmonella Infection Acquired by a Child from Cattle, N. Engl. J. Med, vol.342, pp.1242-1249, 2000.
DOI : 10.1056/nejm200004273421703

URL : http://digitalcommons.unl.edu/cgi/viewcontent.cgi?article=1043&context=zoonoticspub

S. Seltzer and I. B. Bender, The Use of Chloramphenicol (Chloromycetin) for Killing Some PenicillinStreptomycin Resistant Organisms Encountered in Infected Root Canals, J. Dent. Res, vol.29, pp.825-830, 1950.

H. A. Lyttle and G. H. Bowden, The Resistance and Adaptation of Selected Oral Bacteria to Mercury and Its Impact on Their Growth, J. Dent. Res, vol.72, pp.1325-1330, 1993.

M. C. Roberts, Antibiotic Resistance in Oral/Respiratory Bacteria, Crit. Rev. Oral Biol. Med, vol.9, pp.522-540, 1998.
DOI : 10.1177/10454411980090040801

URL : http://journals.sagepub.com/doi/pdf/10.1177/10454411980090040801

H. G. Saleem, C. A. Seers, A. N. Sabri, and E. C. Reynolds, Dental plaque bacteria with reduced susceptibility to chlorhexidine are multidrug resistant, Bmc Microbiol, vol.16, 2016.
DOI : 10.1186/s12866-016-0833-1

URL : https://bmcmicrobiol.biomedcentral.com/track/pdf/10.1186/s12866-016-0833-1?site=bmcmicrobiol.biomedcentral.com

M. Haenni and P. Moreillon, Fitness cost and impaired survival in penicillin-resistant Streptococcus gordonii isolates selected in the laboratory, Antimicrob. Agents Chemother, vol.52, pp.337-339, 2008.

A. Itzek, L. Zheng, Z. Chen, J. Merritt, and J. Kreth, Hydrogen peroxide-dependent DNA release and transfer of antibiotic resistance genes in Streptococcus gordonii, J. Bacteriol, vol.193, pp.6912-6922, 2011.

Y. Sanai, G. R. Persson, J. R. Starr, H. S. Luis, M. Bernardo et al., Presence and antibiotic resistance of Porphyromonas gingivalis, Prevotella intermedia, and Prevotella nigrescens in children, J. Clin. Periodontol, vol.29, pp.929-934, 2002.
DOI : 10.1034/j.1600-051x.2002.291008.x

URL : http://repositorio.ul.pt/bitstream/10451/34286/1/WU113_LUIh%20P_AI_2002.pdf

G. Bachrach, H. Altman, P. E. Kolenbrander, N. I. Chalmers, M. Gabai-gutner et al., Resistance of Porphyromonas gingivalis ATCC 33277 to direct killing by antimicrobial peptides is protease independent, Antimicrob. Agents Chemother, vol.52, pp.638-642, 2008.
DOI : 10.1128/aac.01271-07

URL : https://aac.asm.org/content/52/2/638.full.pdf

C. L. Ventole, The antibiotic resistance crisis, vol.40, pp.277-283, 2015.

G. O'toole, H. B. Kaplaa, and R. Koltee, Biofil Formation as Microbial Development, Annu. Rev. Microbiol, vol.54, pp.49-79, 2000.

D. I. Andersson and D. Hughes, Microbiological effects of sublethal levels of antibiotics, Nat. Rev. Microbiol, vol.12, pp.465-478, 2014.

V. Lorian, Some effects of subinhibitory concentrations on bacteria, 1974.

G. G. Zanel, D. Hoban, and G. Harding, Subinhibitory antimicrobial concentrations: A review of in vitro and in vivo data, Can, J. Infect. Dis, vol.3, pp.193-201, 1992.

T. M. Bakheet and A. J. Doig, Properties and identification of antibiotic drug targets, BMC Bioinformatics, vol.11, p.195, 2010.

K. Lewis, Platforms for antibiotic discovery, Nat. Rev. Drug Discov, vol.12, pp.371-387, 2013.

M. A. Kohanski, D. J. Dwyer, and J. J. Collins, How antibiotics kill bacteria: from targets to networks, Nat. Rev. Microbiol, vol.8, pp.423-435, 2010.

M. A. Azad, B. A. Finnin, A. Poudyal, K. Davis, J. Li et al., Polymyxin B Induces Apoptosis in Kidney Proximal Tubular Cells, vol.57, pp.4329-4335, 2013.

E. C. Böttger, B. Springer, T. Prammananan, Y. Kidan, and P. Sander, Structural basis for selectivity and toxicity of ribosomal antibiotics, EMBO Rep, vol.2, pp.318-323, 2001.

C. U. Chukwudi, rRNA Binding Sites and the Molecular Mechanism of Action of the Tetracyclines, Antimicrob. Agents Chemother, vol.60, pp.4433-4441, 2016.

H. Babich and D. ,

. Tipton, In vitro response of human gingival epithelioid S-G cells to minocycline, Toxicol. In Vitro, vol.16, pp.11-21, 2002.

Y. Pommier, Drugging Topoisomerases: Lessons and Challenges, Acs Chem. Biol, vol.8, pp.82-95, 2013.

D. I. Andersson and D. Hughes, Antibiotic resistance and its cost: is it possible to reverse resistance?, Nat. Rev. Microbiol, 2010.

B. Kouidhi, Y. M. Qurashi, and K. Chaieb, Drug resistance of bacterial dental biofilm and the potential use of natural compounds as alternative for prevention and treatment, Microb. Pathog, vol.80, pp.39-49, 2015.

D. Sriramulu and D. Sriramulu, Evolution and Impact of Bacterial Drug Resistance in the Context of Cystic Fibrosis Disease and Nosocomial Settings, Microbiol. Insights, p.29, 2013.

M. Jm and A. Ca, Mechanisms of Antibiotic Resistance, Virulence Mech. Bact. Pathog. Fifth Ed, pp.481-511, 2016.

J. Bueno, Anti-Biofilm Drug Susceptibility Testing Methods: Looking for New Strategies against Resistance Mechanism, J. Microb. Biochem. Technol, vol.3, 2011.

R. R. Tembhurne, N. S. Mali, S. U. Shinde, D. S. Bhise, and S. M. Satpute, , 2016.

V. Ahmadjian and M. E. Hale, The lichens, 1973.

A. Aptroot and F. Schumm, Fruticose Roccellaceae: an anatomical-microscopical atlas and guide with a worldwide keyand further notes on some crustose roccellaceae or similar lichens, 2011.

C. Delzeeee-vaa, J. Halu, and . Ga, Guide des lichens de France: lichens des sols, 2012.

M. E. Hale and M. Cole, Lichens of California, 1988.

I. M. Brodo, S. D. Sharnoff, and S. Sharnoff, Lichens of North America, 2001.

J. D. Fish, Fish, A studeet's guide to the seashoe, UUi Haa, Lodo ; Bosto

M. Millot, Étude phytohiiue de ttois lihees ustaa s du littoal eto : Ohholeehia paaella, Tephoela atta et Diploiia aaessees : eeheeehe d'ue aatiit photopoteetie, 2008.

P. D. Coley, Effects of plant growth rate and leaf lifetime on the amount and type of antiherbivore defense, Oecologia, vol.74, pp.531-536, 1988.

A. P. Podterob, Chemical composition of lichens and their medical applications, Pharm. Chem. J, vol.42, pp.582-588, 2008.

E. Stocker-wörgötter, Secondary chemistry of cultured mycobionts: formation of a complete chemosyndrome by the lichen fungus of Lobaria spathulata, The Lichenologist, vol.34, pp.351-359, 2002.

P. A. Cox, S. A. Banack, S. J. Murch, U. Rasmussen, G. Tien et al., Diverse taxa of cyanobacteria produce-N-methylamino-L-alanine, a neurotoxic amino acid, Proc. Natl. Acad. Sci, vol.102, pp.5074-5078, 2005.

X. Yang, Y. Shimizu, J. R. Steiner, and J. Clardy, Nostoclide I and II, extracellular metabolites from a symbiotic cyanobacterium, Nostoc sp., from the lichen Peltigera canina, Tetrahedron Lett, vol.34, issue.93, p.89005, 1993.

S. Huneck and I. Yoshimura, Identification of lichen substances, 1996.

B. Raakoi?, Lihee seeodaaa etaaolites: bioactive properties and pharmaceutical potential, 2015.

M. Grube, T. Cernava, J. Soh, S. Fuchs, I. Aschenbrenner et al., Exploring functional contexts of symbiotic sustain within lichenassociated bacteria by comparative omics, Isme J, vol.9, pp.412-424, 2015.

D. Parrot, N. Legrave, D. Delmail, M. Grube, M. Suzuki et al., Review-Lichen-associated bacteria as a hot spot of chemodiversity: focus on Uncialamycin, a promising compound for future medicinal applications, Planta Med, vol.82, pp.1143-4452, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01360065

M. R. González-tejero, J. Molero-mesa, M. Casares-porcel, and M. J. Martínez-lirola, New contributions to the ethnopharmacology of Spain, J. Ethnopharmacol, vol.45, pp.157-165, 1995.

F. E. Dayan and J. G. Romagni, Lichens as a potential source of pesticides, Pestic. Outlook, vol.12, pp.229-232, 2001.

T. Mitrovic, S. Stamenkovic, V. Cvetkovic, M. Nikolic, S. Tosic et al., Lichens as source of versatile bioactive compounds, Biol. Nyssana, vol.2, pp.1-6, 2011.

I. Oksanen, Ecological and biotechnological aspects of lichens, Appl. Microbiol. Biotechnol, vol.73, pp.723-734, 2006.

P. R. Burkholder, A. W. Evans, I. Mcveich, and H. K. Thornton, Antibiotic activity of lichens, Proc. Natl. Acad. Sci. U. S. A, vol.30, pp.250-255, 1944.

B. Segatore, P. Bellio, D. Setacci, F. Brisdelli, M. Piovano et al., In vitro interaction of usnic acid in combination with antimicrobial agents against methicillin-resistant Staphylococcus aureus clinical isolates determined FICI aad ?E odel ethods

N. T. Manojlovic, P. J. Vasiljevic, P. Z. Maskovic, M. Juskovic, and G. Bogdanovic-dusanovic, Chemical Composition, Antioxidant, and Antimicrobial Activities of Lichen Umbilicaria cylindrica (L.) Delise (Umbilicariaceae), pp.1-8, 2012.

N. K. Honda, F. R. Pavan, R. G. Coelho, S. R. De-andrade-leite, A. C. Micheletti et al., Antimycobacterial activity of lichen substances, vol.17, pp.328-332, 2010.

B. Gökals? and N. C. , esal, Lihee seeodaaa etaaolite eeeeei aaid as poteetial uou seesig inhibitor against Pseudomonas aeruginosa, World J. Microbiol. Biotechnol, vol.32, 2016.

T. Kokubun, W. Shiu, and S. Gibbons, Inhibitory activities of lichen-derived compounds against methicillin-and multidrug-resistant Staphylococcus aureus, Planta Med, vol.73, pp.176-179, 2007.

A. Sweidan, M. Chollet-krugler, P. Van-de-weghe, A. Chokr, S. Tomasi et al., Bousarghin, Design, synthesis and biological evaluation of potential antibacterial butyrolactones, Bioorg. Med. Chem, vol.24, pp.5823-5833, 2016.

K. S. Nishanth, R. S. Sreerag, I. Deepa, C. Mohandas, and B. Nambisan, Protocetraric acid: an excellent broad spectrum compound from the lichen Usnea albopunctata against medically important microbes, Nat. Prod. Res, vol.29, pp.574-577, 2015.

M. Baldry, A. Nielsen, M. S. Bojer, Y. Zhao, C. Friberg et al., Norlichexanthone reduces virulence, gene expression and biofilm formation in Staphylococcus aureus, Plos One, vol.11, p.168305, 2016.

M. Xu, S. Heidmarsson, E. S. Olafsdottir, R. Buonfiglio, T. Kogej et al., Secondary metabolites from cetrarioid lichens: Chemotaxonomy, biological activities and pharmaceutical potential, Phytomedicine, vol.23, pp.441-459, 2016.

I. Kurobane, L. C. Vining, and A. G. Mclnnes, Secalonic acids, US 4424373, 1984.

M. Lauterwein, M. Oethinger, K. Belsner, T. Peters, and R. Marre, In vitro activities of the lichen secondary metabolites vulpinic acid, (+)-usnic acid, and (-)-usnic acid against aerobic and anaerobic microorganisms, Antimicrob. Agents Chemother, vol.39, pp.2541-2543, 1995.

G. Shrestha, A. Thompson, R. Robison, L. L. St, and . Clair, Letharia vulpina , a vulpinic acid containing lichen, targets cell membrane and cell division processes in methicillin-resistant Staphylococcus aureus, Pharm. Biol, vol.54, pp.413-418, 2016.

M. Millot, A. Dieu, and S. Tomasi, Dibenzofurans and derivatives from lichens and ascomycetes, Nat Prod Rep, vol.33, pp.801-811, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01274836

C. Carpentier, E. F. Queiroz, L. Marcourt, J. Wolfender, J. Azelmat et al., Dibenzofurans and pseudodepsidones from the lichen Stereocaulon paschale collected in northern Quebec, J. Nat. Prod, vol.80, pp.210-214, 2017.

A. Karagöz, N. Dogruöz, Z. Zeybek, and A. Aslan, Antibacterial activity of some lichen extracts, J. Med. Plants Res, vol.3, pp.1034-1039, 2009.

S. S-c and J. Savita, Antibacterial Activity of the Himalayan Lichen Parmotrema nilgherrense Extracts, Br. Microbiol. Res. J, vol.1, pp.26-32, 2011.

N. Perry, Antimicrobial, antiviral and cytotoxic activity of New Zealand lichens, The Lichenologist, vol.31, pp.627-636, 1999.

A. Ö. Tüük, M. Y?laz, M. K?aaç, and H. Tüük, The AAtiioial AAtiit of EEttats of the Lihee Cetraria aculeata and Its Protolichesterinic Acid Constituent, Z. Für Naturforschung C, vol.58, 2003.

O. Özgenç, Methodology in improving antibiotic implementation policies, World J. Methodol, vol.6, p.143, 2016.

A. Borges, A. Abreu, C. Dias, M. Saavedra, F. Borges et al., New perspectives on the use of phytochemicals as an emergent strategy to control bacterial infections including biofilms, Molecules, vol.21, p.877, 2016.

P. Batchelor, Is periodontal disease a public health problem?, Br. Dent. J, vol.217, pp.405-409, 2014.

V. M. Thadhani, M. I. Choudhary, S. Khan, and V. Karunaratne, Antimicrobial and toxicological activities of some depsides and depsidones, J. Natl. Sci. Found. Sri Lanka, p.40, 2012.

, ANNEXE Original articles

A. Sweidan, M. Chollet-krugler, P. Van-de-weghe, A. Chokr, S. Tomasi et al., Bousarghin, Design, synthesis and biological evaluation of potential antibacterial butyrolactones, Bioorg. Med. Chem, vol.24, pp.5823-5833, 2016.

A. Sweidan, M. Chollet-krugler, A. Sauvager, P. Van-de-weghe, A. Chokr et al., Antibacterial activities of natural lichen compounds against Streptococcus gordonii and Porphyromonas gingivalis, Fitoterapia
URL : https://hal.archives-ouvertes.fr/hal-01579650

A. Sweidan, Z. Tamanai-shacoori, M. Chollet-krugler, N. Oliviero, A. Chokr et al., Antibiofilm activity of butyrotactone derivatives against oral bacteria, Applied Microbiology and Biotechnology

A. Sweidan, I. Smida, M. Chollet-krugler, A. Sauvager, J. Vallet et al., Lichen butyrolactone derivatives disrupted the cell wall of oral bacteria, Journal

A. Sweidan, M. Chollet-krugler, P. Van-de-weghe, A. Chokr, S. Tomasi et al., Antibacterial activity of some lichen compounds analogues on Streptococcus gordonii, p.6

, Faculty of Pharmacy, poster and oral communication, pp.20-21

A. Sweidan, M. Chollet-krugler, P. Van-de-weghe, A. Chokr, S. Tomasi et al., Treatment of Streptococcus gordonii infections by lichen butyrolactones. Young Research Fellow Meeting, pp.15-17, 2016.

A. Sweidan, M. Chollet-krugler, P. Van-de-weghe, A. Chokr, S. Tomasi et al., Antibacterial activity of butyrolactone analogues on Streptococcus gordonii. Conférence Internationale : Host-pathogen interactons : from bench to bedside, 2016.

A. Sweidan, Z. Tamanai-shacoori, M. Chollet-krugler, N. Oliviero, A. Chokr et al., Antibacterial and antibiofilm activities of a butyrolactone series against oral bacteria. 7ème Journée des doctorants, pp.18-19