C. Kelly and O. Rahn, The Growth Rate of Individual Bacterial Cells, J Bacteriol, vol.23, pp.147-153, 1932.

P. Maloney and B. Rotman, Distribution of suboptimally induced ??-d-galactosidase in Escherichia coli, Journal of Molecular Biology, vol.73, issue.1, pp.77-91, 1973.
DOI : 10.1016/0022-2836(73)90160-5

J. Spudich, D. Koshland, and . Jr, Non-genetic individuality: chance in the single cell, Nature, vol.2, issue.5568, pp.467-471, 1976.
DOI : 10.1038/262467a0

P. Schaeffer, J. Millet, and J. Aubert, Catabolic repression of bacterial sporulation., Proceedings of the National Academy of Sciences, vol.54, issue.3, pp.704-711, 1965.
DOI : 10.1073/pnas.54.3.704

B. Haijema, J. Hahn, J. Haynes, and D. Dubnau, A ComGA-dependent checkpoint limits growth during the escape from competence, Molecular Microbiology, vol.177, issue.1, pp.52-64, 2001.
DOI : 10.1046/j.1365-2958.2001.02363.x

B. Brehm-stecher and E. Johnson, Single-Cell Microbiology: Tools, Technologies, and Applications, Microbiology and Molecular Biology Reviews, vol.68, issue.3, pp.538-559, 2004.
DOI : 10.1128/MMBR.68.3.538-559.2004

M. Lidstrom and M. Konopka, The role of physiological heterogeneity in microbial population behavior, Nature Chemical Biology, vol.25, issue.10, pp.705-712, 2010.
DOI : 10.1038/nchembio.436

A. Loewer and G. Lahav, We are all individuals: causes and consequences of non-genetic heterogeneity in mammalian cells, Current Opinion in Genetics & Development, vol.21, issue.6, pp.753-758, 2011.
DOI : 10.1016/j.gde.2011.09.010

A. Levchenko and I. Nemenman, Cellular noise and information transmission, Current Opinion in Biotechnology, vol.28, pp.156-164, 2014.
DOI : 10.1016/j.copbio.2014.05.002

J. Capp, Noise-Driven Heterogeneity in the Rate of Genetic-Variant Generation as a Basis for Evolvability, Genetics, vol.185, issue.2, pp.395-404, 2010.
DOI : 10.1534/genetics.110.118190

A. Sanchez, S. Choubey, and J. Kondev, Regulation of Noise in Gene Expression, Annual Review of Biophysics, vol.42, issue.1, pp.469-491, 2013.
DOI : 10.1146/annurev-biophys-083012-130401

H. Mcadams and A. Arkin, Stochastic mechanisms in gene expression, Proceedings of the National Academy of Sciences, vol.94, issue.3, pp.814-819, 1997.
DOI : 10.1073/pnas.94.3.814

J. Skommer, S. Raychaudhuri, and D. Wlodkowic, Timing is everything: stochastic origins of cell-to-cell variability in cancer cell death, Front Biosci, vol.16, pp.307-314, 2011.

J. Wu and E. Tzanakakis, Deconstructing stem cell population heterogeneity: Single-cell analysis and modeling approaches, Biotechnology Advances, vol.31, issue.7, pp.1047-1062, 2013.
DOI : 10.1016/j.biotechadv.2013.09.001

S. Huang, Non-genetic heterogeneity of cells in development: more than just noise, Development, vol.136, issue.23, pp.3853-3862, 2009.
DOI : 10.1242/dev.035139

A. Colman-lerner, A. Gordon, E. Serra, T. Chin, O. Resnekov et al., Regulated cell-to-cell variation in a cell-fate decision system, Nature, vol.13, issue.7059, pp.699-706, 2005.
DOI : 10.1016/S0092-8674(01)00596-7

W. Wright and J. Shay, Historical claims and current interpretations of replicative aging, Nature Biotechnology, vol.20, issue.7, pp.682-688, 2002.
DOI : 10.1038/nbt0702-682

K. Steinkraus, M. Kaeberlein, and B. Kennedy, Replicative Aging in Yeast: The Means to the End, Annual Review of Cell and Developmental Biology, vol.24, issue.1, pp.29-54, 2008.
DOI : 10.1146/annurev.cellbio.23.090506.123509

V. Longo and P. Fabrizio, Chronological Aging in Saccharomyces cerevisiae, Subcell Biochem, vol.57, pp.101-121, 2012.
DOI : 10.1007/978-94-007-2561-4_5

Y. Pan and . Mitochondria, Mitochondria, reactive oxygen species, and chronological aging: A message from yeast, Experimental Gerontology, vol.46, issue.11, pp.847-852, 2011.
DOI : 10.1016/j.exger.2011.08.007

V. Longo, G. Shadel, M. Kaeberlein, and B. Kennedy, Replicative and Chronological Aging in Saccharomyces cerevisiae, Cell Metabolism, vol.16, issue.1, pp.18-31, 2012.
DOI : 10.1016/j.cmet.2012.06.002

A. Roux, P. Chartrand, G. Ferbeyre, and L. Rokeach, Fission Yeast and Other Yeasts as Emergent Models to Unravel Cellular Aging in Eukaryotes, The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, vol.65, issue.1, pp.1-8, 2010.
DOI : 10.1093/gerona/glp152

L. Vachova, M. Cap, and Z. Palkova, Yeast Colonies: A Model for Studies of Aging, Environmental Adaptation, and Longevity, Oxidative Medicine and Cellular Longevity, vol.180, issue.15, pp.601836-601846, 2012.
DOI : 10.1007/s00248-006-9063-7

M. Barker and R. Walmsley, Replicative ageing in the fission yeast Schizosaccharomyces pombe, Yeast, vol.15, issue.14, pp.1511-1518, 1999.
DOI : 10.1002/(SICI)1097-0061(199910)15:14<1511::AID-YEA482>3.3.CO;2-P

R. Amann and B. Fuchs, Single-cell identification in microbial communities by improved fluorescence in situ hybridization techniques, Nature Reviews Microbiology, vol.56, issue.5, pp.339-348, 2008.
DOI : 10.1016/0022-2836(81)90508-8

J. Boedicker, M. Vincent, and R. Ismagilov, Microfluidic confinement of single cells of bacteria in small volumes initiates high-density behavior of quorum sensing and growth and reveals its variability

L. Boitard, D. Cottinet, C. Kleinschmitt, N. Bremond, J. Baudry et al., Monitoring single-cell bioenergetics via the coarsening of emulsion droplets, Proceedings of the National Academy of Sciences, vol.109, issue.19, pp.7181-7186, 2012.
DOI : 10.1073/pnas.1200894109

URL : https://hal.archives-ouvertes.fr/hal-00760719

K. Czechowska, D. Johnson, and J. Van-der-meer, Use of flow cytometric methods for single-cell analysis in environmental microbiology, Current Opinion in Microbiology, vol.11, issue.3, pp.205-212, 2008.
DOI : 10.1016/j.mib.2008.04.006

A. Dichosa, A. Daughton, K. Reitenga, M. Fitzsimons, and C. Han, Capturing and cultivating single bacterial cells in gel microdroplets to obtain near-complete genomes, Nature Protocols, vol.28, issue.3, pp.608-621, 2014.
DOI : 10.1016/j.watres.2007.07.009

L. Garay, K. Boundy-mills, and J. German, Accumulation of High-Value Lipids in Single-Cell Microorganisms: A Mechanistic Approach and Future Perspectives, Journal of Agricultural and Food Chemistry, vol.62, issue.13, 2014.
DOI : 10.1021/jf4042134

A. Garz, M. Sandmann, M. Rading, S. Ramm, R. Menzel et al., Cell-to-Cell Diversity in a Synchronized Chlamydomonas Culture As Revealed by Single-Cell Analyses, Biophysical Journal, vol.103, issue.5, pp.1078-1086, 2012.
DOI : 10.1016/j.bpj.2012.07.026

A. Grunberger, W. Wiechert, and D. Kohlheyer, Single-cell microfluidics: opportunity for bioprocess development, Current Opinion in Biotechnology, vol.29, pp.15-23, 2014.
DOI : 10.1016/j.copbio.2014.02.008

U. Hofer, Environmental microbiology: Exploring diversity with single-cell genomics, Nature Reviews Microbiology, vol.110, issue.9, pp.598-608, 2013.
DOI : 10.1038/nrmicro3095

K. Matsumura, T. Yagi, A. Hattori, M. Soloviev, and K. Yasuda, Using single cell cultivation system for on-chip monitoring of the interdivision timer in Chlamydomonas reinhardtii cell cycle, Journal of Nanobiotechnology, vol.8, issue.1, pp.23-33, 2010.
DOI : 10.1186/1477-3155-8-23

E. Shapiro, T. Biezuner, and S. Linnarsson, Single-cell sequencing-based technologies will revolutionize whole-organism science, Nature Reviews Genetics, vol.4, issue.9, pp.618-630, 2013.
DOI : 10.1186/gb-2013-14-4-r31

R. Stepanauskas, Single cell genomics: an individual look at microbes. Current Opinion in Microbiology, pp.613-620, 2012.

H. Yin and D. Marshall, Microfluidics for single cell analysis, Current Opinion in Biotechnology, vol.23, issue.1, pp.110-119, 2012.
DOI : 10.1016/j.copbio.2011.11.002

Y. Eun, A. Utada, M. Copeland, S. Takeuchi, and D. Weibel, Encapsulating Bacteria in Agarose Microparticles Using Microfluidics for High-Throughput Cell Analysis and Isolation, ACS Chemical Biology, vol.6, issue.3, pp.260-266, 2011.
DOI : 10.1021/cb100336p

C. Hoeck, Algae: an introduction to phycology Cell-to-Cell Heterogeneity in Chlamydomonas, 1995.

A. Richmond, Handbook of microalgal culture: biotechnology and applied phycology, 2008.
DOI : 10.1002/9781118567166

R. Leon, A. Galvan, and E. Fernandez, Transgenic Microalgae as Green Cell Factories, 2007.
DOI : 10.1007/978-0-387-75532-8

J. Gimpel, E. Specht, D. Georgianna, and S. Mayfield, Advances in microalgae engineering and synthetic biology applications for biofuel production, Current Opinion in Chemical Biology, vol.17, issue.3, pp.489-495, 2013.
DOI : 10.1016/j.cbpa.2013.03.038

C. Jones and S. Mayfield, Algae biofuels: versatility for the future of bioenergy, Current Opinion in Biotechnology, vol.23, issue.3, pp.346-351, 2012.
DOI : 10.1016/j.copbio.2011.10.013

M. Esquivel, H. Amaro, T. Pinto, P. Fevereiro, and F. Malcata, Efficient H2 production via Chlamydomonas reinhardtii, Trends in Biotechnology, vol.29, issue.12, pp.595-600, 2011.
DOI : 10.1016/j.tibtech.2011.06.008

B. Ghysels and F. Franck, Hydrogen photo-evolution upon S deprivation stepwise: an illustration of microalgal photosynthetic and metabolic flexibility and a step stone for future biotechnological methods of renewable H2 production, Photosynthesis Research, vol.214, issue.Suppl, pp.145-154, 2010.
DOI : 10.1007/s11120-010-9582-4

J. Rupprecht, From systems biology to fuel???Chlamydomonas reinhardtii as a model for a systems biology approach to improve biohydrogen production, Journal of Biotechnology, vol.142, issue.1, pp.10-20, 2009.
DOI : 10.1016/j.jbiotec.2009.02.008

S. Scott, M. Davey, J. Dennis, I. Horst, C. Howe et al., Biodiesel from algae: challenges and prospects, Current Opinion in Biotechnology, vol.21, issue.3, pp.277-286, 2010.
DOI : 10.1016/j.copbio.2010.03.005

L. Brennan and P. Owende, Biofuels from microalgae???A review of technologies for production, processing, and extractions of biofuels and co-products, Renewable and Sustainable Energy Reviews, vol.14, issue.2, pp.557-577, 2010.
DOI : 10.1016/j.rser.2009.10.009

S. Bae, C. Kim, J. Choi, J. Yang, and T. Seo, An integrated microfluidic device for the high-throughput screening of microalgal cell culture conditions that induce high growth rate and lipid content, Analytical and Bioanalytical Chemistry, vol.11, issue.1, pp.9365-9374, 2013.
DOI : 10.1007/s00216-013-7389-9

W. Yu, W. Ansari, N. Schoepp, M. Hannon, S. Mayfield et al., Modifications of the metabolic pathways of lipid and triacylglycerol production in microalgae, Microbial Cell Factories, vol.10, issue.1, pp.91-22047615, 2011.
DOI : 10.1186/1475-2859-10-91

I. Blaby, C. Blaby-haas, N. Tourasse, E. Hom, D. Lopez et al., The Chlamydomonas genome project: a decade on, Trends in Plant Science, vol.19, issue.10, 2014.
DOI : 10.1016/j.tplants.2014.05.008

T. Proschold, E. Harris, and A. Coleman, Portrait of a Species: Chlamydomonas reinhardtii, Genetics, vol.170, issue.4, pp.1601-1610, 2005.
DOI : 10.1534/genetics.105.044503

J. Rochaix, , a model system for studying the assembly and dynamics of photosynthetic complexes, FEBS Letters, vol.3, issue.1, pp.34-38, 2002.
DOI : 10.1016/S0014-5793(02)03181-2

J. Rochaix, The three genomes of Chlamydomonas, Photosynth Res, vol.73, pp.285-293, 2002.
DOI : 10.1007/1-4020-3324-9_92

M. Hippler, K. Redding, and J. Rochaix, Chlamydomonas genetics, a tool for the study of bioenergetic pathways, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1367, issue.1-3, pp.1-62, 1998.
DOI : 10.1016/S0005-2728(98)00136-4

J. Rochaix, as the Photosynthetic Yeast, Annual Review of Genetics, vol.29, issue.1, pp.209-230, 1995.
DOI : 10.1146/annurev.ge.29.120195.001233

E. Harris, The Chlamydomonas Sourcebook, Canada, 2009.

S. Merchant, S. Prochnik, O. Vallon, E. Harris, S. Karpowicz et al., The Chlamydomonas Genome Reveals the Evolution of Key Animal and Plant Functions, Science, vol.318, issue.5848, pp.245-250, 2007.
DOI : 10.1126/science.1143609

URL : https://hal.archives-ouvertes.fr/hal-00198837

A. Grossman, E. Harris, C. Hauser, P. Lefebvre, D. Martinez et al., Chlamydomonas reinhardtii at the Crossroads of Genomics, Eukaryotic Cell, vol.2, issue.6, pp.1137-1150, 2003.
DOI : 10.1128/EC.2.6.1137-1150.2003

A. Grossman, Chlamydomonas reinhardtii and photosynthesis: genetics to genomics, Current Opinion in Plant Biology, vol.3, issue.2, pp.132-137, 2000.
DOI : 10.1016/S1369-5266(99)00053-9

S. Eberhard, G. Finazzi, and F. Wollman, The Dynamics of Photosynthesis, Annual Review of Genetics, vol.42, issue.1, pp.463-515, 2008.
DOI : 10.1146/annurev.genet.42.110807.091452

F. Wollman, L. Minai, and R. Nechushtai, The biogenesis and assembly of photosynthetic proteins in thylakoid membranes1This article is dedicated to the memory of our colleague and friend, Alma Gal. Her presence played a major role in our decision to prepare this review article.1, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1411, issue.1, pp.21-85, 1999.
DOI : 10.1016/S0005-2728(99)00043-2

C. Silflow and P. Lefebvre, Assembly and Motility of Eukaryotic Cilia and Flagella. Lessons from Chlamydomonas reinhardtii, PLANT PHYSIOLOGY, vol.127, issue.4, pp.1500-1507, 2001.
DOI : 10.1104/pp.010807

W. Snell, J. Pan, and Q. Wang, Cilia and Flagella Revealed, Cell, vol.117, issue.6, pp.693-697, 2004.
DOI : 10.1016/j.cell.2004.05.019

L. Lee, Mechanisms of mammalian ciliary motility: Insights from primary ciliary dyskinesia genetics, Gene, vol.473, issue.2
DOI : 10.1016/j.gene.2010.11.006

J. Pan, Q. Wang, and W. Snell, Cilium-generated signaling and cilia-related disorders, Laboratory Investigation, vol.28, issue.4, pp.452-463, 2005.
DOI : 10.1038/labinvest.3700253

S. Rosales-mendoza, L. Paz-maldonado, and R. Soria-guerra, Chlamydomonas reinhardtii as a viable platform for the production of recombinant proteins: current status and perspectives, Plant Cell Reports, vol.12, issue.3, pp.479-494, 2012.
DOI : 10.1007/s00299-011-1186-8

S. Mayfield, A. Manuell, S. Chen, J. Wu, M. Tran et al., Chlamydomonas reinhardtii chloroplasts as protein factories, Current Opinion in Biotechnology, vol.18, issue.2, pp.126-133, 2007.
DOI : 10.1016/j.copbio.2007.02.001

E. Specht and S. Mayfield, Algae-based oral recombinant vaccines, Frontiers in Microbiology, vol.5, p.24596570, 2014.
DOI : 10.3389/fmicb.2014.00060

P. Spolaore, C. Joannis-cassan, E. Duran, and A. Isambert, Commercial applications of microalgae, Journal of Bioscience and Bioengineering, vol.101, issue.2, pp.87-96, 2006.
DOI : 10.1263/jbb.101.87

URL : https://hal.archives-ouvertes.fr/hal-00133263

K. Skjanes, C. Rebours, and P. Lindblad, Potential for green microalgae to produce hydrogen, pharmaceuticals and other high value products in a combined process, Critical Reviews in Biotechnology, vol.60, issue.2, pp.172-215, 2013.
DOI : 10.1080/00071660310001616200

N. Velmurugan, M. Sung, S. Yim, M. Park, J. Yang et al., Evaluation of intracellular lipid bodies in Chlamydomonas reinhardtii strains by flow cytometry, Bioresource Technology, vol.138, pp.30-37, 2013.
DOI : 10.1016/j.biortech.2013.03.078

D. Lee, C. Bae, J. Han, and J. Park, In Situ Analysis of Heterogeneity in the Lipid Content of Single Green Microalgae in Alginate Hydrogel Microcapsules, Analytical Chemistry, vol.85, issue.18, pp.8749-8756, 2013.
DOI : 10.1021/ac401836j

J. Pan, A. Stephenson, E. Kazamia, W. Huck, J. Dennis et al., Quantitative tracking of the growth of individual algal cells in microdroplet compartments, Integrative Biology, vol.33, issue.10, pp.1043-1051, 2011.
DOI : 10.1039/c1ib00033k

A. Dewan, J. Kim, R. Mclean, S. Vanapalli, and M. Karim, Growth kinetics of microalgae in microfluidic static droplet arrays, Biotechnology and Bioengineering, vol.117, issue.4, pp.2987-2996, 2012.
DOI : 10.1002/bit.24568

L. Baraban, F. Bertholle, M. Salverda, N. Bremond, P. Panizza et al., Millifluidic droplet analyser for microbiology, Lab on a Chip, vol.29, issue.6, pp.4057-4062, 2011.
DOI : 10.1039/c1lc20545e

URL : https://hal.archives-ouvertes.fr/hal-00647595

E. Harris, The Chlamydomonas Sourcebook. A Comprehensive Guide to Biology and Laboratory Use; Press A, editor, p.17756009, 1989.

B. Fischer, M. Wiesendanger, and R. Eggen, Growth Condition-Dependent Sensitivity, Photodamage and Stress Response of Chlamydomonas reinhardtii Exposed to High Light Conditions, Plant and Cell Physiology, vol.47, issue.8, pp.1135-1145, 2006.
DOI : 10.1093/pcp/pcj085

J. Spudich and R. Sager, Regulation of the Chlamydomonas cell cycle by light and dark, The Journal of Cell Biology, vol.85, issue.1, pp.136-145, 1980.
DOI : 10.1083/jcb.85.1.136

S. Lemaire, M. Hours, C. Gerard-hirne, A. Trouabal, O. Roche et al., Analysis of light/dark synchronization of cell-wall-less Chlamydomonas reinhardtii (Chlorophyta) cells by flow cytometry, European Journal of Phycology, vol.34, issue.3, pp.279-286, 1999.
DOI : 10.1017/S096702629900222X

D. Kürsten, J. Cao, A. Funfak, P. Müller, and J. Köhler, Cultivation of Chlorella vulgaris in microfluid segments and microtoxicological determination of their sensitivity against CuCl2 in the nanoliter range, Engineering in Life Sciences, vol.13, issue.6, pp.580-587, 2011.
DOI : 10.1002/elsc.201100023

W. Ratcliff, M. Herron, K. Howell, J. Pentz, F. Rosenzweig et al., Experimental evolution of an alternating uni- and multicellular life cycle in Chlamydomonas reinhardtii, Nature Communications, vol.88, pp.2742-2752, 2013.
DOI : 10.1046/j.1529-8817.1999.3510104.x

T. Strovas, L. Sauter, X. Guo, and M. Lidstrom, Cell-to-Cell Heterogeneity in Growth Rate and Gene Expression in Methylobacterium extorquens AM1, Journal of Bacteriology, vol.189, issue.19, pp.7127-7133, 2007.
DOI : 10.1128/JB.00746-07

K. Gerdes and E. Maisonneuve, Bacterial Persistence and Toxin-Antitoxin Loci, Annual Review of Microbiology, vol.66, issue.1, pp.103-123, 2012.
DOI : 10.1146/annurev-micro-092611-150159

E. Maisonneuve, M. Castro-camargo, and K. Gerdes, (p)ppGpp Controls Bacterial Persistence by Stochastic Induction of Toxin-Antitoxin Activity, Cell, vol.154, issue.5, pp.1140-1150, 2013.
DOI : 10.1016/j.cell.2013.07.048

P. Cell-to-cell-heterogeneity-in-chlamydomonas, . One, A. Doi-joers, N. Kaldalu, and T. Tenson, The frequency of persisters in Escherichia coli reflects the kinetics of awakening from dormancy, J Bacteriol, vol.192, pp.10-1371, 2010.

H. Moyed and K. Bertrand, hipA, a newly recognized gene of Escherichia coli K-12 that affects frequency of persistence after inhibition of murein synthesis, J Bacteriol, vol.155, pp.768-775, 1983.

B. Claudi, P. Sprote, A. Chirkova, N. Personnic, J. Zankl et al., Phenotypic Variation of Salmonella in Host Tissues Delays Eradication by Antimicrobial Chemotherapy, Cell, vol.158, issue.4, pp.722-733, 2014.
DOI : 10.1016/j.cell.2014.06.045

S. Helaine, J. Thompson, K. Watson, M. Liu, C. Boyle et al., Dynamics of intracellular bacterial replication at the single cell level, Proceedings of the National Academy of Sciences, vol.107, issue.8, pp.3746-3751, 2010.
DOI : 10.1073/pnas.1000041107

S. Helaine and D. Holden, Heterogeneity of intracellular replication of bacterial pathogens, Current Opinion in Microbiology, vol.16, issue.2, pp.184-191, 2013.
DOI : 10.1016/j.mib.2012.12.004

R. Demets, A. Tomson, E. Ran, C. Sigon, D. Stegwee et al., Synchronization of the Cell Division Cycle of Chlamydomonas eugametos, Microbiology, vol.131, issue.11, pp.2919-2924, 1985.
DOI : 10.1099/00221287-131-11-2919

E. Stewart, R. Madden, G. Paul, and F. Taddei, Aging and Death in an Organism That Reproduces by Morphologically Symmetric Division, PLoS Biology, vol.297, issue.2, pp.45-15685293, 2005.
DOI : 10.1371/journal.pbio.0030045.sv001

URL : https://hal.archives-ouvertes.fr/inserm-00080154

C. Rang, A. Peng, and L. Chao, Temporal Dynamics of Bacterial Aging and Rejuvenation, Current Biology, vol.21, issue.21, pp.1813-1816, 2011.
DOI : 10.1016/j.cub.2011.09.018

R. Flatscher, B. Frajman, P. Schonswetter, and O. Paun, Environmental Heterogeneity and Phenotypic Divergence: Can Heritable Epigenetic Variation Aid Speciation?, Genetics Research International, vol.72, issue.4, pp.698421-698431, 2012.
DOI : 10.1016/j.gde.2008.01.014

C. Richards, O. Bossdorf, and M. Pigliucci, What Role Does Heritable Epigenetic Variation Play in Phenotypic Evolution?, BioScience, vol.60, issue.3, pp.232-237, 2010.
DOI : 10.1525/bio.2010.60.3.9

D. Strenkert, S. Schmollinger, and M. Schroda, Protocol: methodology for chromatin immunoprecipitation (ChIP) in Chlamydomonas reinhardtii, Plant Methods, vol.7, issue.1, pp.35-45, 2011.
DOI : 10.1093/nar/11.5.1389

R. Ness, A. Morgan, N. Colegrave, and P. Keightley, Estimate of the Spontaneous Mutation Rate in Chlamydomonas reinhardtii, Genetics, vol.192, issue.4, pp.1447-1454, 2012.
DOI : 10.1534/genetics.112.145078

M. Perrineau, J. Gross, E. Zelzion, D. Price, O. Levitan et al., Using Natural Selection to Explore the Adaptive Potential of Chlamydomonas reinhardtii, PLoS ONE, vol.21, issue.3, p.24658261, 2014.
DOI : 10.1371/journal.pone.0092533.s009

W. Sung, M. Ackerman, S. Miller, T. Doak, and M. Lynch, Drift-barrier hypothesis and mutation-rate evolution, Proceedings of the National Academy of Sciences, vol.109, issue.45, pp.18488-18492, 2012.
DOI : 10.1073/pnas.1216223109

V. Cooper, The Origins of Specialization: Insights from Bacteria Held 25 Years in Captivity, PLoS Biology, vol.104, issue.1, p.24558348, 2014.
DOI : 10.1371/journal.pbio.1001790.g001

P. Wang, L. Robert, J. Pelletier, W. Dang, F. Taddei et al., Robust Growth of Escherichia coli, Current Biology, vol.20, issue.12, pp.1099-1103, 2010.
DOI : 10.1016/j.cub.2010.04.045

B. Wu, A novel method to study aging in bacteria that reproduce by morphologically symmetric fission, Journal of Theoretical Biology, vol.347, pp.1-6, 2013.
DOI : 10.1016/j.jtbi.2013.12.001

M. Kupiec, Biology of telomeres: lessons from budding yeast, FEMS Microbiology Reviews, vol.38, issue.2, pp.144-171, 2014.
DOI : 10.1111/1574-6976.12054

J. Campisi, Aging, Cellular Senescence, and Cancer, Annual Review of Physiology, vol.75, issue.1, pp.685-705, 2013.
DOI : 10.1146/annurev-physiol-030212-183653

A. Maklakov, Aging: Why Do Organisms Live Too Long?, Current Biology, vol.23, issue.22, pp.1003-1005, 2013.
DOI : 10.1016/j.cub.2013.10.002

T. Rando, The Ins and Outs of Aging and Longevity, Annual Review of Physiology, vol.75, issue.1, pp.617-619, 2013.
DOI : 10.1146/annurev-physiol-092712-103439

J. Vijg and Y. Suh, Genome Instability and Aging, Annual Review of Physiology, vol.75, issue.1, pp.645-668, 2013.
DOI : 10.1146/annurev-physiol-030212-183715

A. Aguilera and T. Garcia-muse, Causes of Genome Instability, Annual Review of Genetics, vol.47, issue.1, pp.1-32, 2013.
DOI : 10.1146/annurev-genet-111212-133232

S. Ghosh and Z. Zhou, Genetics of aging, progeria and lamin disorders, Current Opinion in Genetics & Development, vol.26, pp.41-46, 2014.
DOI : 10.1016/j.gde.2014.05.003

C. Harley, A. Futcher, and C. Greider, Telomeres shorten during ageing of human fibroblasts, Nature, vol.345, issue.6274, pp.458-460, 1990.
DOI : 10.1038/345458a0

B. Bernardes-de-jesus and M. Blasco, Telomerase at the intersection of cancer and aging, Trends in Genetics, vol.29, issue.9, pp.513-520, 2013.
DOI : 10.1016/j.tig.2013.06.007

H. Saeed and M. Iqtedar, Stem cell function and maintenance ??? ends that matter: Role of telomeres and telomerase, Journal of Biosciences, vol.17, issue.Pt 1, pp.641-649, 2013.
DOI : 10.1007/s12038-013-9346-3

P. Mason and N. Perdigones, Telomere biology and translational research, Translational Research, vol.162, issue.6, pp.333-342, 2013.
DOI : 10.1016/j.trsl.2013.08.009

V. Lundblad and J. Szostak, A mutant with a defect in telomere elongation leads to senescence in yeast, Cell, vol.57, issue.4, pp.633-643, 1989.
DOI : 10.1016/0092-8674(89)90132-3

J. Fulneckova, T. Sevcikova, J. Fajkus, A. Lukesova, M. Lukes et al., A Broad Phylogenetic Survey Unveils the Diversity and Evolution of Telomeres in Eukaryotes, Genome Biology and Evolution, vol.5, issue.3, pp.468-483, 2013.
DOI : 10.1093/gbe/evt019

T. Hails, O. Huttner, and A. Day, Isolation of a Chlamydomonas reinhardtii telomer by functional complementation in yeast, Current Genetics, vol.29, issue.5, pp.437-440, 1995.
DOI : 10.1007/BF00310812

M. Petracek and J. Berman, telomere repeats form unstable structures involving guanine-guanine base pairs, Nucleic Acids Research, vol.20, issue.1, pp.89-95, 1992.
DOI : 10.1093/nar/20.1.89

M. Petracek, L. Konkel, M. Kable, and J. Berman, A Chlamydomonas protein that binds singlestranded G-strand telomere DNA, EMBO J, vol.13, pp.3648-3658, 1994.

M. Petracek, P. Lefebvre, C. Silflow, and J. Berman, Chlamydomonas telomere sequences are A+T-rich but contain three consecutive G-C base pairs., Proceedings of the National Academy of Sciences, vol.87, issue.21, pp.8222-8226, 1990.
DOI : 10.1073/pnas.87.21.8222

O. Misumi, Y. Nishimura, and T. Kuroiwa, Effects of Chloroplast DNA Content on the Cell Proliferation and Aging in Chlamydomonas reinhardtii, Journal of Plant Research, vol.114, issue.2, pp.125-131, 2001.
DOI : 10.1007/PL00013975