L. Y. Chan, Refactoring bacteriophage T7, Molecular Systems Biology, vol.340, issue.1, p.18, 2005.
DOI : 10.1038/msb4100025

T. S. Gardner, Construction of a genetic toggle switch in Escherichia coli, Nature, vol.403, pp.339-342, 2000.

T. Danino, O. Mondragón?palomino, L. Tsimring, and J. Hasty, A synchronized quorum of genetic clocks, Nature, vol.44, issue.7279, pp.326-330, 2010.
DOI : 10.1038/nature08753

T. S. Moon, Genetic programs constructed from layered logic gates in single cells, Nature, vol.377, issue.7423, pp.249-253, 2012.
DOI : 10.1038/nature11516

P. E. Purnick and R. Weiss, The second wave of synthetic biology: from modules to systems, Nature Reviews Molecular Cell Biology, vol.307, issue.6, pp.410-422, 2009.
DOI : 10.1038/nrm2698

B. Wang, R. I. Kitney, N. Joly, and M. Buck, Engineering modular and orthogonal genetic logic gates for robust digital-like synthetic biology, Nature Communications, vol.26, p.508, 2011.
DOI : 10.1038/ncomms1516

A. E. Friedland, Synthetic Gene Networks That Count, Science, vol.324, issue.5931, pp.1199-1202, 2009.
DOI : 10.1126/science.1172005

M. B. Elowitz and S. Leibler, A synthetic oscillatory network of transcriptional regulators, Nature, vol.403, issue.6767, pp.335-338, 2000.
DOI : 10.1038/35002125

P. Siuti, J. Yazbek, and T. K. Lu, Synthetic circuits integrating logic and memory in living cells, Nature Biotechnology, vol.487, issue.5, pp.448-452, 2013.
DOI : 10.1073/pnas.022642299

L. You, Programmed population control by cell???cell communication and regulated killing, Nature, vol.428, issue.6985, pp.868-871, 2004.
DOI : 10.1038/nature02491

M. ?. Chen and R. Weiss, Artificial cell-cell communication in yeast Saccharomyces cerevisiae using signaling elements from Arabidopsis thaliana, Nature Biotechnology, vol.21, issue.12, pp.1551-1555, 2005.
DOI : 10.1038/nbt1162

A. Prindle, A sensing array of radically coupled genetic ???biopixels???, Nature, vol.271, issue.7379, pp.39-44, 2012.
DOI : 10.1038/nature10722

B. P. Kramer, An engineered epigenetic transgene switch in mammalian cells, Nature Biotechnology, vol.12, issue.7, pp.867-870, 2004.
DOI : 10.1038/nbt980

S. Ausländer, D. Ausländer, M. Müller, M. Wieland, and M. M. Fussenegger, Programmable single?cell mammalian biocomputers A synthetic mammalian gene circuit reveals antituberculosis compounds, Proceedings of the National Academy of Sciences 105, pp.9994-9998, 2008.

Z. Xie, Multi-Input RNAi-Based Logic Circuit for Identification of Specific Cancer Cells, Science, vol.333, issue.6047, pp.1307-1311, 2011.
DOI : 10.1126/science.1205527

C. Kemmer, Self-sufficient control of urate homeostasis in mice by a synthetic circuit, Nature Biotechnology, vol.267, issue.4, pp.355-360, 2010.
DOI : 10.1093/rheumatology/ken457

W. Bacchus, Synthetic two-way communication between mammalian cells, Nature Biotechnology, vol.13, issue.10, pp.991-996, 2012.
DOI : 10.1038/nm889

R. P. Ryan and J. M. Dow, Diffusible signals and interspecies communication in bacteria, Microbiology, vol.154, issue.7, pp.1845-1858, 2008.
DOI : 10.1099/mic.0.2008/017871-0

D. O. Peterson, K. K. Beifuss, and K. L. Morley, Context?dependent gene expression: cis? acting negative effects of specific procaryotic plasmid sequences on eucaryotic genes. 21. Human gene expression, 1999.

L. M. Nilsson, Mouse Genetics Suggests Cell-Context Dependency for Myc-Regulated Metabolic Enzymes during Tumorigenesis, PLoS Genetics, vol.180, issue.Pt 2, p.1002573, 2012.
DOI : 10.1371/journal.pgen.1002573.s010

C. ?. Chen, J. Krohn, S. Bhattacharya, and B. Davies, A Comparison of Exogenous Promoter Activity at the ROSA26 Locus Using a PhiC31 Integrase Mediated Cassette Exchange Approach in Mouse ES Cells, PLoS ONE, vol.122, issue.8, p.23376, 2011.
DOI : 10.1371/journal.pone.0023376.s002

J. Y. Qin, Systematic Comparison of Constitutive Promoters and the Doxycycline-Inducible Promoter, PLoS ONE, vol.420, issue.5, p.10611, 2010.
DOI : 10.1371/journal.pone.0010611.s001

C. Lois, Germline Transmission and Tissue-Specific Expression of Transgenes Delivered by Lentiviral Vectors, Science, vol.295, issue.5556, pp.868-872, 2002.
DOI : 10.1126/science.1067081

S. Knowling and K. Morris, Epigenetic Regulation of Gene Expression in Human Cells by Noncoding RNAs, V. in Progress in Molecular Biology and Translational Science, vol.102, pp.1-10, 2011.
DOI : 10.1016/B978-0-12-415795-8.00003-9

J. Eyquem, L. Poirot, R. Galetto, A. M. Scharenberg, and J. Smith, Characterization of three loci for homologous gene targeting and transgene expression, Biotechnology and Bioengineering, vol.117, issue.21, pp.2225-2235, 2013.
DOI : 10.1002/bit.24892

R. Schneider and R. Grosschedl, Dynamics and interplay of nuclear architecture, genome organization, and gene expression. genesdev.cshlp.org 29. Recillas?Targa, F. et al. Position?effect protection and enhancer blocking by the chicken beta ?globin insulator are separable activities, pp.6883-6888, 2002.

A. Lombardo, Site-specific integration and tailoring of cassette design for sustainable gene transfer, Nature Methods, vol.14, issue.10, pp.861-869, 2011.
DOI : 10.1038/ni883

N. Sharma, The Impact of cHS4 Insulators on DNA Transposon Vector Mobilization and Silencing in Retinal Pigment Epithelium Cells, PLoS ONE, vol.36, issue.10, p.48421, 2012.
DOI : 10.1371/journal.pone.0048421.s007

J. D. Jordan, E. M. Landau, and R. Iyengar, Signaling Networks, Cell, vol.103, issue.2, p.193, 2000.
DOI : 10.1016/S0092-8674(00)00112-4

L. Bögre, N. J. Lanning, and C. Carter?su, doi:10.1002/9780470015902 Recent advances in growth hormone signaling, Cell Signalling Mechanisms in Plants. els.net Rev Endocr Metab Disord, vol.7, pp.225-235, 2001.

E. R. Andersson, R. Sandberg, and U. Lendahl, Notch signaling: simplicity in design, versatility in function. dev.biologists.org 36. Bacchus, W. & Fussenegger, M. Engineering of synthetic intercellular communication systems, Metabolic Engineering, vol.16, pp.33-41, 2013.

S. Payne and L. You, Engineered Cell???Cell Communication and Its Applications, Synthetic biology: Let's start talking. Nat Meth 9, pp.1052-1052, 2012.
DOI : 10.1007/10_2013_249

M. Matsuda, M. Koga, E. Nishida, and M. Ebisuya, Synthetic Signal Propagation Through Direct Cell-Cell Interaction, Science Signaling, vol.5, issue.220, pp.31-31, 2012.
DOI : 10.1126/scisignal.2002764

W. Weber, EngineeredStreptomyces quorum-sensing components enable inducible siRNA-mediated translation control in mammalian cells and adjustable transcription control in mice, The Journal of Gene Medicine, vol.100, issue.4, pp.518-525, 2005.
DOI : 10.1002/jgm.682

W. ?. Wang, Z. ?. Chen, B. ?. Kang, and R. Li, Construction of an artificial intercellular communication network using the nitric oxide signaling elements in mammalian cells, Experimental Cell Research, vol.314, issue.4, pp.699-706, 2008.
DOI : 10.1016/j.yexcr.2007.11.023

W. Weber, M. Schuetz, N. Dénervaud, and M. Fussenegger, A synthetic metabolite-based mammalian inter-cell signaling system, Molecular BioSystems, vol.5, issue.7, p.757, 2009.
DOI : 10.1039/b902070p

W. Weber, Synthetic ecosystems based on airborne inter- and intrakingdom communication, Proceedings of the National Academy of Sciences, vol.104, issue.25, pp.10435-10440, 2007.
DOI : 10.1073/pnas.0701382104

L. Bleris, Synthetic incoherent feedforward circuits show adaptation to the amount of their genetic template, Molecular Systems Biology, vol.105, issue.1, p.519, 2011.
DOI : 10.1016/j.tig.2008.05.005

S. Mukherji, MicroRNAs can generate thresholds in target gene expression, Nature Genetics, vol.43, issue.9, pp.854-859, 2011.
DOI : 10.1126/science.1137999

M. Tigges, A tunable synthetic mammalian oscillator, Nature, vol.16, issue.7227, pp.309-312, 2009.
DOI : 10.1038/nature07616

V. Siciliano, Construction and Modelling of an Inducible Positive Feedback Loop Stably Integrated in a Mammalian Cell-Line, PLoS Computational Biology, vol.2, issue.3, p.1002074, 2011.
DOI : 10.1371/journal.pcbi.1002074.t001

E. Lanitis, Chimeric Antigen Receptor T Cells with Dissociated Signaling Domains Exhibit Focused Antitumor Activity with Reduced Potential for Toxicity In Vivo, Cancer Immunology Research, vol.1, issue.1, pp.43-53, 2013.
DOI : 10.1158/2326-6066.CIR-13-0008

D. Grushkin, The new drug circuit, Nature Medicine, vol.487, issue.10, pp.1452-1454, 2012.
DOI : 10.1016/j.jmb.2005.10.076

W. Ruder, T. Lu, and J. Collins, Synthetic Biology Moving into the Clinic, Science, vol.333, issue.6047, pp.1248-1252, 2011.
DOI : 10.1126/science.1206843

M. Folcher and M. Fussenegger, Synthetic biology advancing clinical applications, Current Opinion in Chemical Biology, vol.16, issue.3-4, pp.345-354, 2012.
DOI : 10.1016/j.cbpa.2012.06.008

W. Bacchus, D. Aubel, and M. Fussenegger, Biomedically relevant circuit-design strategies in mammalian synthetic biology, Molecular Systems Biology, vol.13, issue.1, 2013.
DOI : 10.1126/science.1226854

H. Ye, D. Aubel, and M. Fussenegger, Synthetic mammalian gene circuits for biomedical applications, Current Opinion in Chemical Biology, vol.17, issue.6, pp.910-917, 2013.
DOI : 10.1016/j.cbpa.2013.10.006

H. Ye, A Synthetic Optogenetic Transcription Device Enhances Blood-Glucose Homeostasis in Mice, Science, vol.332, issue.6037, pp.1565-1568, 2011.
DOI : 10.1126/science.1203535

E. A. Moehle, Targeted gene addition into a specified location in the human genome using designed zinc finger nucleases, Proceedings of the National Academy of Sciences, vol.104, issue.9, pp.3055-3060, 2007.
DOI : 10.1073/pnas.0611478104

D. F. Carlson, S. C. Fahrenkrug, and P. B. Hackett, Targeting DNA With Fingers and TALENs, Molecular Therapy - Nucleic Acids, vol.1, p.3, 2012.
DOI : 10.1038/mtna.2011.5

D. Reyon, FLASH assembly of TALENs for high-throughput genome editing, Nature Biotechnology, vol.649, issue.5, pp.460-465, 2012.
DOI : 10.1038/nbt1317

P. Perez?pinera, Gene targeting to the ROSA26 locus directed by engineered zinc finger nucleases, Nucleic Acids Research, vol.40, issue.8, pp.3741-3752, 2012.
DOI : 10.1093/nar/gkr1214

A. Hendel, Quantifying Genome-Editing Outcomes at Endogenous Loci with SMRT Sequencing, Cell Reports, vol.7, issue.1, pp.293-305, 2014.
DOI : 10.1016/j.celrep.2014.02.040

F. Zhu, DICE, an efficient system for iterative genomic editing in human pluripotent stem cells, Nucleic Acids Research, vol.42, issue.5, p.34, 2014.
DOI : 10.1093/nar/gkt1290

E. C. Olivares, Phage R4 integrase mediates site-specific integration in human cells, Gene, vol.278, issue.1-2, pp.167-176, 2001.
DOI : 10.1016/S0378-1119(01)00711-9

H. Zhou, Z. ?. Liu, Z. ?. Sun, Y. Huang, W. Yu et al., Generation of stable cell lines by site-specific integration of transgenes into engineered Chinese hamster ovary strains using an FLP-FRT system, Journal of Biotechnology, vol.147, issue.2, pp.122-129, 2010.
DOI : 10.1016/j.jbiotec.2010.03.020

F. Bushman, Efficient integration of transgenes into a defined locus in human embryonic stem cells, Nucleic Acids Res, vol.38, p.96, 2010.

H. Bai, Single-molecule analysis reveals the molecular bearing mechanism of DNA strand exchange by a serine recombinase, Proceedings of the National Academy of Sciences, vol.108, issue.18, pp.7419-7424, 2011.
DOI : 10.1073/pnas.1018436108

D. Hockemeyer, Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases, Nature Biotechnology, vol.2, issue.9, pp.851-857, 2009.
DOI : 10.1038/nbt.1562

M. Sadelain, E. P. Papapetrou, and F. Bushman, Safe harbours for the integration of new DNA in the human genome, Nature Reviews Cancer, vol.115, pp.51-58, 2012.
DOI : 10.1038/nrc3179

E. Weber, C. Engler, R. Gruetzner, S. Werner, and S. Marillonnet, A Modular Cloning System for Standardized Assembly of Multigene Constructs, PLoS ONE, vol.177, issue.2, p.16765, 2011.
DOI : 10.1371/journal.pone.0016765.s001

C. Engler, Golden Gate Shuffling: A One-Pot DNA Shuffling Method Based on Type IIs Restriction Enzymes, PLoS ONE, vol.23, issue.5, p.5553, 2009.
DOI : 10.1371/journal.pone.0005553.s004

J. P. Torella, Rapid construction of insulated genetic circuits via synthetic sequence-guided isothermal assembly, Nucleic Acids Research, vol.42, issue.1, pp.681-689, 2014.
DOI : 10.1093/nar/gkt860

P. Guye, Y. Li, L. Wroblewska, X. Duportet, and R. Weiss, Rapid, modular and reliable construction of complex mammalian gene circuits, Nucleic Acids Research, vol.41, issue.16, p.156, 2013.
DOI : 10.1093/nar/gkt605

URL : https://hal.archives-ouvertes.fr/hal-00926598

J. L. Schmid?burgk, Rapid hierarchical assembly of medium-size DNA cassettes, Nucleic Acids Research, vol.40, issue.12, p.92, 2012.
DOI : 10.1093/nar/gks236

H. Shiyuza, Cloning and stable maintenance of 300-kilobase-pair fragments of human DNA in Escherichia coli using an F-factor-based vector., Proceedings of the National Academy of Sciences, vol.89, issue.18, pp.8794-8797, 1992.
DOI : 10.1073/pnas.89.18.8794

C. Chevalier, I. Henry, and J. Nicolas, CpG content affects gene silencing in mice: evidence from novel transgenes, Genome Biology, vol.4, issue.9, p.53, 2003.
DOI : 10.1186/gb-2003-4-9-r53

Z. Chen, Improved Production and Purification of Minicircle DNA Vector Free of Plasmid Bacterial Sequences and Capable of Persistent Transgene Expression In Vivo

K. Anastassiadis, Dre recombinase, like Cre, is a highly efficient site-specific recombinase in E. coli, mammalian cells and mice, Disease Models & Mechanisms, vol.2, issue.9-10, p.156, 2013.
DOI : 10.1242/dmm.003087

Y. Chen, Genetic control of mammalian T-cell proliferation with synthetic RNA regulatory systems, Proceedings of the National Academy of Sciences, vol.107, issue.19, pp.8531-8536, 2010.
DOI : 10.1073/pnas.1001721107

J. L. Hartley, G. F. Temple, and M. A. Brasch, DNA Cloning Using In Vitro Site-Specific Recombination, Genome Research, vol.10, issue.11, pp.1788-1795, 2000.
DOI : 10.1101/gr.143000

S. A. Stewart, Lentivirus-delivered stable gene silencing by RNAi in primary cells, RNA, vol.9, issue.4, pp.493-501, 2003.
DOI : 10.1261/rna.2192803

N. Malchin, Optimization of coliphage HK022 Integrase activity in human cells, Gene, vol.437, issue.1-2, pp.9-13, 2009.
DOI : 10.1016/j.gene.2009.02.013

R. Krishnakumar, Simultaneous non?contiguous deletions using large synthetic DNA and site?specific recombinases doi:10.1093/nar/gku509 81: transgene activation and inactivation using lox and rox in zebrafish, Nucleic Acids Res. PLoS ONE, vol.8, pp.85218-85218, 2012.

D. Long, In??vivo site-specific integration of transgene in silkworm via PhiC31 integrase-mediated cassette exchange, Insect Biochemistry and Molecular Biology, vol.43, issue.11, pp.997-1008, 2013.
DOI : 10.1016/j.ibmb.2013.08.001

S. Turan, Expanding Flp?RMCE options: the potential of Recombinase Mediated Twin?Site Targeting (RMTT) Osterwalder, M. et al. Dual RMCE for efficient re?engineering of mouse mutant alleles, Gene Nat Meth, vol.7, pp.893-895, 2010.

P. Jain, Specialized Transduction Designed for Precise High-Throughput Unmarked Deletions in Mycobacterium tuberculosis, mBio, vol.5, issue.3, pp.1245-1259, 2014.
DOI : 10.1128/mBio.01245-14

Y. Wang, Targeted DNA recombination in vivo using an adenovirus carrying the cre recombinase gene., Proceedings of the National Academy of Sciences, vol.93, issue.9, pp.3932-3936, 1996.
DOI : 10.1073/pnas.93.9.3932

N. D. Grindley, K. L. Whiteson, and P. A. Rice, Mechanisms of Site-Specific Recombination, Annual Review of Biochemistry, vol.75, issue.1, pp.567-605, 2006.
DOI : 10.1146/annurev.biochem.73.011303.073908

G. Pan, K. L. Pan, and K. L. , Mechanism of cleavage and ligation by FLP recombinase: classification of mutations in FLP protein by in vitro complementation analysis., Molecular and Cellular Biology, vol.13, issue.6, p.3167, 1993.
DOI : 10.1128/MCB.13.6.3167

D. Esposito, D. Esposito, J. J. Scocca, and J. J. Scocca, The integrase family of tyrosine recombinases: evolution of a conserved active site domain, Nucleic Acids Research, vol.25, issue.18, pp.3605-3614, 1997.
DOI : 10.1093/nar/25.18.3605

J. P. Mumm, A. Landy, and J. Gelles, Viewing single ?? site-specific recombination events from start to finish, The EMBO Journal, vol.67, issue.19, pp.4586-4595, 2006.
DOI : 10.1038/sj.emboj.7601325

Z. Zhang, B. L. Zuwen-zhang, and B. L. , Cre recombinase-mediated inversion using lox66 and lox71: method to introduce conditional point mutations into the CREB-binding protein, Nucleic Acids Research, vol.30, issue.17, p.90, 2002.
DOI : 10.1093/nar/gnf089

T. Gaj, Structure-guided reprogramming of serine recombinase DNA sequence specificity, Proceedings of the National Academy of Sciences, vol.108, issue.2, pp.498-503, 2011.
DOI : 10.1073/pnas.1014214108

E. J. Hubbard, E. J. Hubbard, . Flp, . Frt, and S. Cre, Recombinase?mediated cassette exchange (RMCE) ? a rapidly?expanding toolbox for targeted genomic modifications, Thorpe, H. M. Diversity in the serine recombinases, pp.1-27, 2002.

Z. Xu, Accuracy and efficiency define Bxb1 integrase as the best of fifteen candidate serine recombinases for the integration of DNA into the human genome, BMC Biotechnology, vol.13, issue.1, 2013.
DOI : 10.1093/nar/gkm475

B. Thyagarajan, Site-Specific Genomic Integration in Mammalian Cells Mediated by Phage ??C31 Integrase, Molecular and Cellular Biology, vol.21, issue.12, pp.3926-3934, 2001.
DOI : 10.1128/MCB.21.12.3926-3934.2001

A. I. Kim, Mycobacteriophage Bxb1 integrates into the Mycobacterium smegmatis groEL1 gene, Molecular Microbiology, vol.179, issue.2, pp.463-473, 2003.
DOI : 10.1046/j.1365-2958.2003.03723.x

S. Singh, K. Rockenbach, R. M. Dedrick, A. P. Vandemark, and G. F. Hatfull, Cross-talk between Diverse Serine Integrases, Journal of Molecular Biology, vol.426, issue.2, pp.318-331, 2014.
DOI : 10.1016/j.jmb.2013.10.013

G. F. Hatfull, Complete Genome Sequences of 138 Mycobacteriophages, Journal of Virology, vol.86, issue.4, pp.2382-2384, 2012.
DOI : 10.1128/JVI.06870-11

A. Park, H. ?. Jeong, J. Lee, C. Lee, and . ?s, The inhibitory effect of phloretin on the formation of Escherichia coli O157:H7 biofilm in a microfluidic system, BioChip Journal, vol.82, issue.3, pp.299-305, 2012.
DOI : 10.1007/s13206-012-6313-2

D. Barreca, Biochemical and antimicrobial activity of phloretin and its glycosilated derivatives present in apple and kumquat, Food Chemistry, vol.160, pp.292-297, 2014.
DOI : 10.1016/j.foodchem.2014.03.118

W. Teran, Antibiotic-Dependent Induction of Pseudomonas putida DOT-T1E TtgABC Efflux Pump Is Mediated by the Drug Binding Repressor TtgR, Antimicrobial Agents and Chemotherapy, vol.47, issue.10, pp.3067-3072, 2003.
DOI : 10.1128/AAC.47.10.3067-3072.2003

K. Rutherford, P. Yuan, K. Perry, R. Sharp, and G. D. Van-duyne, Attachment site recognition and regulation of directionality by the serine integrases, Nucleic Acids Research, vol.41, issue.17, pp.8341-8356, 2013.
DOI : 10.1093/nar/gkt580

S. Singh, P. Ghosh, and G. Hatfull, Attachment Site Selection and Identity in Bxb1 Serine Integrase-Mediated Site-Specific Recombination, PLoS Genetics, vol.276, issue.5, p.1003490, 2013.
DOI : 10.1371/journal.pgen.1003490.s006

W. Terán, Effector-Repressor Interactions, Binding of a Single Effector Molecule to the Operator-bound TtgR Homodimer Mediates Derepression, Journal of Biological Chemistry, vol.281, issue.11, pp.7102-7109, 2006.
DOI : 10.1074/jbc.M511095200

M. Gitzinger, Controlling transgene expression in subcutaneous implants using a skin lotion containing the apple metabolite phloretin, Proceedings of the National Academy of Sciences, vol.106, issue.26, pp.10638-10643, 2009.
DOI : 10.1073/pnas.0901501106

K. T. Watts, P. C. Lee, and C. Schmidt?dannert, Exploring Recombinant Flavonoid Biosynthesis in Metabolically Engineered Escherichia coli, ChemBioChem, vol.10, issue.211, pp.500-507, 2004.
DOI : 10.1002/cbic.200300783

H. Jiang, Metabolic Engineering of the Phenylpropanoid Pathway in Saccharomyces cerevisiae, Applied and Environmental Microbiology, vol.71, issue.6, pp.2962-2969, 2005.
DOI : 10.1128/AEM.71.6.2962-2969.2005

I. Hwang, E. Kaneko, M. Ohnishi, Y. Horinouchi, and S. , Production of Plant-Specific Flavanones by Escherichia coli Containing an Artificial Gene Cluster, Applied and Environmental Microbiology, vol.69, issue.5, pp.2699-2706, 2003.
DOI : 10.1128/AEM.69.5.2699-2706.2003

T. Krell, Optimization of the Palindromic Order of the TtgR Operator Enhances Binding Cooperativity, Journal of Molecular Biology, vol.369, issue.5, pp.1188-1199, 2007.
DOI : 10.1016/j.jmb.2007.04.025

S. J. Kaczmarczyk, K. Sitaraman, H. A. Young, S. H. Hughes, and D. K. Chatterjee, Protein delivery using engineered virus-like particles, Proceedings of the National Academy of Sciences, vol.108, issue.41, pp.16998-17003, 2011.
DOI : 10.1073/pnas.1101874108

D. Farley, Factors that influence VSV-G pseudotyping and transduction efficiency of lentiviral vectors???in vitro andin vivo implications, The Journal of Gene Medicine, vol.74, issue.5, pp.345-356, 2007.
DOI : 10.1002/jgm.1022

B. Thyagarajan, Site-Specific Genomic Integration in Mammalian Cells Mediated by Phage ??C31 Integrase, Molecular and Cellular Biology, vol.21, issue.12, pp.3926-3934, 2001.
DOI : 10.1128/MCB.21.12.3926-3934.2001

E. C. Olivares, Phage R4 integrase mediates site-specific integration in human cells, Gene, vol.278, issue.1-2, pp.167-176, 2001.
DOI : 10.1016/S0378-1119(01)00711-9

S. Fukushige, S. Fukushige, B. Sauer, and B. Sauer, Genomic targeting with a positive-selection lox integration vector allows highly reproducible gene expression in mammalian cells., Proceedings of the National Academy of Sciences, vol.89, issue.17, pp.7905-7909, 1992.
DOI : 10.1073/pnas.89.17.7905

O. 'gorman, S. Fox, D. T. Wahl, and G. M. , Recombinase-mediated gene activation and site-specific integration in mammalian cells, Science, vol.251, issue.4999, pp.1351-1355, 1991.
DOI : 10.1126/science.1900642

R. 1. Weber, W. Fussenegger, and M. , Emerging biomedical applications of synthetic biology, Nature Reviews Genetics, vol.4, pp.21-35, 2012.
DOI : 10.1038/nrg3094

W. C. Ruder, T. Lu, and J. J. Collins, Synthetic Biology Moving into the Clinic, Science, vol.333, issue.6047, pp.1248-1252, 2011.
DOI : 10.1126/science.1206843

S. Ausla¨nderausla¨nder, D. Ausla¨nderausla¨nder, M. Muïler, M. Wieland, and M. Fussenegger, Programmable single-cell mammalian biocomputers, Nature, vol.487, pp.123-127, 2012.

T. Vierbuchen, A. Ostermeier, Z. P. Pang, Y. Kokubu, T. C. Su¨dhofsu¨dhof et al., Direct conversion of fibroblasts to functional neurons by defined factors, Nature, vol.48, issue.7284, pp.1035-1041, 2010.
DOI : 10.1038/nature08797

D. Sprinzak, A. Lakhanpal, L. Lebon, L. A. Santat, M. E. Fontes et al., Cis-interactions between Notch and Delta generate mutually exclusive signalling states, Nature, vol.118, issue.7294, pp.86-90, 2010.
DOI : 10.1038/nature08959

A. Kriz, K. Schmid, N. Baumgartner, U. Ziegler, I. Berger et al., A plasmid-based multigene expression system for mammalian cells, Nature Communications, vol.4, issue.8, p.120, 2010.
DOI : 10.1038/ncomms1120

J. L. Schmid-burgk, Z. Xie, S. Frank, V. Winter, S. Mitschka et al., Rapid hierarchical assembly of medium-size DNA cassettes, Nucleic Acids Research, vol.40, issue.12, p.92, 2012.
DOI : 10.1093/nar/gks236

A. Sarrion-perdigones, E. E. Falconi, S. I. Zandalinas, P. Jua´rezjua´rez, A. Ferna´ndezferna´ndez-del-carmen et al., GoldenBraid: An Iterative Cloning System for Standardized Assembly of Reusable Genetic Modules, PLoS ONE, vol.122, issue.7, p.21622, 2011.
DOI : 10.1371/journal.pone.0021622.g005

M. Li, Harnessing homologous recombination in vitro to generate recombinant DNA via SLIC, Nature Methods, vol.14, issue.3, pp.251-256, 2007.
DOI : 10.1038/nmeth1010

D. G. Gibson, G. A. Benders, K. C. Axelrod, J. Zaveri, M. A. Algire et al., One-step assembly in yeast of 25 overlapping DNA fragments to form a complete synthetic Mycoplasma genitalium genome, Proc. Natl Acad. Sci. USA, pp.20404-20409, 2008.
DOI : 10.1073/pnas.0811011106

K. Yahata, K. Maeshima, T. Sone, T. Ando, M. Okabe et al., cHS4 Insulator-mediated Alleviation of Promoter Interference during Cell-based Expression of Tandemly Associated Transgenes, Journal of Molecular Biology, vol.374, issue.3, pp.580-590, 2007.
DOI : 10.1016/j.jmb.2007.09.054

D. Gibson, L. Young, R. Chuang, and J. Venter, Enzymatic assembly of DNA molecules up to several hundred kilobases, Nature Methods, vol.102, issue.5, pp.12-16, 2009.
DOI : 10.1038/nmeth.1318

W. R. Brown, N. C. Lee, Z. Xu, and M. C. Smith, Serine recombinases as tools for genome engineering, Methods, vol.53, issue.4, pp.372-379, 2011.
DOI : 10.1016/j.ymeth.2010.12.031

R. Godiska, D. Mead, V. Dhodda, C. Wu, R. Hochstein et al., Linear plasmid vector for cloning of repetitive or unstable sequences in Escherichia coli, Nucleic Acids Research, vol.38, issue.6, p.88, 2010.
DOI : 10.1093/nar/gkp1181

Y. Sasaki, Evidence for high specificity and efficiency of multiple recombination signals in mixed DNA cloning by the Multisite Gateway system, Journal of Biotechnology, vol.107, issue.3, pp.233-243, 2004.
DOI : 10.1016/j.jbiotec.2003.10.001

K. Ishihara, M. Oshimura, and M. Nakao, CTCF-Dependent Chromatin Insulator Is Linked to Epigenetic Remodeling, Molecular Cell, vol.23, issue.5, pp.733-742, 2006.
DOI : 10.1016/j.molcel.2006.08.008

A. M. Bushey, E. R. Dorman, and V. G. Corces, Chromatin Insulators: Regulatory Mechanisms and Epigenetic Inheritance, Molecular Cell, vol.32, issue.1, pp.1-9, 2008.
DOI : 10.1016/j.molcel.2008.08.017

A. L. Slusarczyk, A. Lin, and R. Weiss, Foundations for the design and implementation of synthetic genetic circuits, Nature Reviews Genetics, vol.498, issue.6, pp.406-420, 2012.
DOI : 10.1101/gr.4431306

K. D. Litcofsky, R. B. Afeyan, R. J. Krom, A. S. Khalil, and J. J. Collins, Iterative plug-and-play methodology for constructing and modifying synthetic gene networks, Nature Methods, vol.9, issue.11, pp.1077-1080, 2012.
DOI : 10.1093/nar/25.6.1203

Z. I. Botev, J. F. Grotowski, and D. P. Kroese, Kernel density estimation via diffusion, The Annals of Statistics, vol.38, issue.5, pp.2916-2957, 2010.
DOI : 10.1214/10-AOS799

M. R. Shortreed, S. B. Chang, D. Hong, M. Phillips, B. Campion et al., A thermodynamic approach to designing structure-free combinatorial DNA word sets, Nucleic Acids Research, vol.33, issue.15, pp.4965-4977, 2005.
DOI : 10.1093/nar/gki812

M. Andronescu, Z. C. Zhang, and A. Condon, Secondary Structure Prediction of Interacting RNA Molecules, Journal of Molecular Biology, vol.345, issue.5, pp.987-1001, 2005.
DOI : 10.1016/j.jmb.2004.10.082

M. Andronescu, RNAsoft: a suite of RNA secondary structure prediction and design software tools, Nucleic Acids Research, vol.31, issue.13, pp.3416-3422, 2003.
DOI : 10.1093/nar/gkg612

D. Hockemeyer, F. Soldner, C. Beard, Q. Gao, M. Mitalipova et al., Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases, Nature Biotechnology, vol.2, issue.9, pp.851-857, 2009.
DOI : 10.1038/nbt.1562

Z. Xie, L. Wroblewska, L. Prochazka, R. Weiss, and Y. Benenson, Multi-Input RNAi-Based Logic Circuit for Identification of Specific Cancer Cells, Science, vol.333, issue.6047, pp.1307-1311, 2011.
DOI : 10.1126/science.1205527

J. Livet, T. A. Weissman, H. Kang, R. W. Draft, J. Lu et al., Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system, Nature, vol.20, issue.7166, pp.56-62, 2007.
DOI : 10.1038/nature06293

. Ingénierie-métabolique-chez-la-levure, Ingénierie combinatoire de biocatalyseurs et de structures protéonucléiques artificielles. Contact Biologie de Synthèse: denis.pompon@insa-toulouse.fr Site web: http://www.lisbp.fr/fr/la_recherche/axe_biocatalyse/ingenierie_moleculaire_et_metabolique

. Sysdiag-est-un-laboratoire-né-en, de l'alliance de deux partenaires aux ambitions complémentaires : le CNRS et Bio-Rad, dédié à la recherche, découverte et innovation dédié au diagnostic en santé humaine, 2007.

. Etude-du-vieillissement,-de-la-diversité-phénotypique and . Et-de-la-coopération-chez-les-bactéries, Contacts Biologie de Synthèse: ariel, CNRS ISSB

. Etude-du-contrôle-de-la-traduction-chez-les-bactéries, Contact Biologie de Synthèse: springer@ibpc.fr Site web

. Génomique-fonctionnelle-pour-la-découverte-de-nouveaux-biocatalyseurs, Contact Biologie de Synthèse: vberard@genoscope.cns.fr Site web: http://www.cns.fr/spip/Equipe-LCAB.html ? Metamorphosys, CNRS ISSB

. Post-doc-dans-l-'équipe-de-luciano-marraffini and . Contact, ? Jérôme Bonnet (University of California Stanford, USA) Post-doc dans l'équipe de Drew Endy Contac: bonnet@stanford.edu Site web: http://openwetware.org, USA) Post-doc dans l'équipe de Adam Arkin Contact: cambray.guillaume@gmail.com Site web

D. @bullet-xavier, INRIA, France et le Massachusetts Institute of Technology, USA) Doctorant chez Gregory Batt à l'INRIA et chez Ron Weiss au MIT. Contact: duportet@mit.edu Sites web: http://groups.csail.mit

A. Professor, Synthetic Biology Research Group Contact: jpeccoud@vbi.vt

S. A. Cellectis, est le pionnier mondial en ingénierie des génomes La société conçoit et commercialise des outils innovants, des nucléases telles que les méganucléases et les TALENs®, permettant d'intervenir de façon maîtrisée sur l'ADN, ainsi que d'autres produits résultant de cette technologie. Cellectis a pour but à exploiter tout le potentiel de cette technologie en médecine

@. Cotée-sur-le-marché-alternext-de and N. , Euronext à Paris depuis 2007 ? Personnel: 220 personnes dont 74 PhD ? 4 filiales: Cellectis bioresearch, Cellectis therapeutics, Cellectis plant sciences Cellectis stem cells, composée d'Ectycell & Cellartis ? Localisation, Goteborg (Suède), Saint-Paul (USA) et Cambridge (USA) ? Site

L. Société-eukarÿs and S. Développé, technologie C3P3 qui permet l'expression ou l'inhibition virtuelle de tous gènes, dans toutes espèces eucaryotes et dans tous systèmes biologiques. Le premier marché visé est celui de la génomique fonctionnelle. Le second marché est celui des services de recherche

@. Créée-en-par, R. Pandjaitan, @. Ceo:-r, @. Pandjaitan, and . Localisation, France) ? Site web: www.eviagenics.com ? Contact: contact@eviagenics.com Page 11 ? Global Bioenergies (Evry) Global Bioenergies développe un procédé unique de production biologique d'isobutène à, 2004.

@. Créée-en-par, M. Delcourt, P. Marlière, @. M. Ceo, . Delcourt et al., VP Chemical Engineering: R. Bockrath ? Revenus: 1,8M? ? Cotée sur le marché Alternext de NYSE-Euronext à Paris depuis 2011 ? Personnel: 40 personnes dont 10 Phd ? Localisation, Iowa (USA) ? Site web: www.global-bioenergies.com ? Contact: info@global-bioenergies.com ? METabolic EXplorer, 2008.

. Amyris, spécialiste des carburants et des produits chimiques issus de matières premières renouvelables, et Michelin, leader de l'innovation dans l'industrie pneumatique, collaborent en vue de développer et de commercialiser l'isoprène renouvelable No Compromise®, l'isoprène étant un composé

. Aux-termes-de, Michelin et Amyris contribueront ensemble au financement et aux ressources techniques permettant le développement de la technologie d'Amyris afin de produire de l'isoprène à partir de matières premières renouvelables

L. Été-créée-en, initiative de Joël de Rosnay, dans le but de promouvoir le conseil stratégique en matière de nouvelles technologies, l'approche systémique appliquée à la prospective et à l'éducation, notamment dans les secteurs de l'Internet et des biotechnologies, 1992.

P. Http, Information and Interactions, Systems and Synthetic Biology

!. Http, Cours de Première Année «Biologie synthétique: une introduction», Mines ParisTech (Paris) https://sgs.mines-paristech.fr/prod/sgs/ensmp/catalog/course/detail.php?code=S3212&lang=FR&year=1A ? Equipe iGEM Bordeaux (Université Bordeaux Segalen, Bordeaux) ? Participations: 2012, 2013 ? Contact: igem-bordeaux2-2012@googlegroups, igem-competition ? Equipe iGEM Grenoble (INP, Grenoble) ? Participations: 2011, 2013 ? Contact: igem.grenoble2012@gmail.com ? Equipe iGEM INSA Lyon, pp.2010-2012, 2011.

@. Participations, ? Contact: contact@igem-paris.org ? Site web, 2007.

D. Bikard, S. Julié-galau, G. Cambray, and D. Mazel, The synthetic integron: an in vivo genetic shuffling device, Nucleic Acids Research, vol.38, issue.15, 2009.
DOI : 10.1093/nar/gkq511

F. Delaplace, H. Klaudel, and A. Cartier-michaud, Discrete Causal Model View of Biological Networks CMSB, 2010.

P. Carbonell, A. Planson, D. Fichera, and J. Faulon, A retrosynthetic biology approach to metabolic pathway design for therapeutic production, BMC Systems Biology, vol.5, issue.1, pp.122-132, 2011.
DOI : 10.1016/j.ymben.2011.01.006

C. Delebecque, A. Lindner, P. Silver, and F. Aldaye, Organization of intracellular reactions with rationally designed RNA assemblies. Science, 2011.

A. Jaramillo and J. Faulon, Editorial: Synthetic Biology - applying new paradigms at the interface of fundamental research and innovation, Biotechnology Journal, vol.6, issue.7, pp.766-773, 2011.
DOI : 10.1002/biot.201100254

A. Laisne, M. Ewald, T. Ando, E. Lesniewska, and D. Pompon, Self-Assembly Properties and Dynamics of Synthetic Proteo???Nucleic Building Blocks in Solution and on Surfaces, Bioconjugate Chemistry, vol.22, issue.9, pp.1824-1858, 2011.
DOI : 10.1021/bc2002264

A. Randall, P. Guye, S. Gupta, X. Duportet, and R. Weiss, Design and Connection of Robust Genetic Circuits, Methods Enzymol, vol.497, pp.159-86, 2011.
DOI : 10.1016/B978-0-12-385075-1.00007-X

A. Basso-blandin, F. Delaplace, G. Batt, B. Besson, P. Ciron et al., Behavior-based Language for Open System Dedicated to Synthetic Biology Genetic network analyzer: a tool for the qualitative modeling and simulation of bacterial regulatory networks, Methods Mol Biol, vol.804, pp.439-62, 2012.

C. Delebecque, P. Silver, and A. Lindner, Designing and using RNA scaffolds to assemble proteins in vivo, Nature Protocols, vol.4, issue.10, pp.1797-807, 2012.
DOI : 10.1186/gb-2002-3-7-research0034

A. Planson, P. Carbonell, I. Grigoras, and J. Faulon, A retrosynthetic biology approach to therapeutics: from conception to delivery, Current Opinion in Biotechnology, vol.23, issue.6, pp.948-56, 2012.
DOI : 10.1016/j.copbio.2012.03.009

G. Rodrigo and A. Jaramillo, AutoBioCAD: Full Biodesign Automation of Genetic Circuits, ACS Synthetic Biology, vol.2, issue.5, 2012.
DOI : 10.1021/sb300084h

G. Rodrigo, T. Landrain, and A. Jaramillo, De novo automated design of small RNA circuits for engineering synthetic riboregulation in living cells, Proceedings of the National Academy of Sciences, vol.109, issue.38, pp.15271-15277, 2012.
DOI : 10.1073/pnas.1203831109

V. Singh, D. Chaudhary, and I. Mani, Gene network analysis of Aeromonas hydrophila for novel drug target discovery, Systems and Synthetic Biology, vol.53, issue.3, pp.23-30, 2012.
DOI : 10.1007/s11693-012-9093-z

J. Uhlendorf, A. Miermont, T. Delaveau, G. Charvin, F. Fages et al., Long-term model predictive control of gene expression at the population and single-cell levels, Proceedings of the National Academy of Sciences, vol.109, issue.35, 2012.
DOI : 10.1073/pnas.1206810109

M. Val, O. Skovgaard, M. Ducos-galand, M. Bland, and D. Mazel, Genome Engineering in Vibrio cholerae: A Feasible Approach to Address Biological Issues, PLoS Genetics, vol.163, issue.1, 2012.
DOI : 10.1371/journal.pgen.1002472.s005

URL : https://hal.archives-ouvertes.fr/inserm-01285625

A. Beli?, D. Pompon, K. Monostory, D. Kelly, S. Kelly et al., An algorithm for rapid computational construction of metabolic networks: A cholesterol biosynthesis example, Computers in Biology and Medicine, vol.43, issue.5, pp.471-80, 2013.
DOI : 10.1016/j.compbiomed.2013.02.017

P. Carbonell, A. Planson, and J. Faulon, Retrosynthetic Design of Heterologous Pathways, Methods Mol Biol, vol.985, pp.149-73, 2013.
DOI : 10.1007/978-1-62703-299-5_9

URL : https://hal.archives-ouvertes.fr/hal-01204312