E. Abranches, A. M. Guedes, M. Moravec, H. Maamar, P. Svoboda et al., Stochastic NANOG fluctuations allow mouse embryonic stem cells to explore pluripotency, Development, vol.141, pp.2770-2779, 2014.

H. F. Blum, Time's Arrow and Evolution, 1968.

J. P. Capp, Stochastic gene expression, disruption of tissue averaging effects and cancer as a disease of development, Bioessays, vol.27, pp.1277-1285, 2005.

J. P. Capp, Tissue disruption increases stochastic gene expression thus producing tumors: cancer initiation without driver mutation, Int. J. Cancer, vol.140, pp.2408-2413, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01600555

I. Chambers, J. Silva, D. Colby, J. Nichols, B. Nijmeijer et al., Nanog safeguards pluripotency and mediates germline development, Nature, vol.450, pp.1230-1234, 2007.

H. H. Chang, M. Hemberg, M. Barahona, D. E. Ingber, and S. Huang, , 2008.

, Transcriptome-wide noise controls lineage choice in mammalian progenitor cells, Nature, vol.453, pp.544-547

K. R. Duffy, C. J. Wellard, J. F. Markham, J. H. Zhou, R. Holmberg et al., Activation-induced B cell fates are selected by intracellular stochastic competition, Science, vol.335, pp.338-341, 2012.

S. Efroni, R. Duttagupta, J. Cheng, H. Dehghani, D. J. Hoeppner et al., Global transcription in pluripotent embryonic stem cells, Cell Stem Cell, vol.2, pp.437-447, 2008.

S. Efroni, S. Melcer, M. Nissim-rafinia, and E. Meshorer, Stem cells do play with dice: a statistical physics view of transcription, Cell Cycle, vol.8, pp.43-48, 2009.

M. B. Elowitz, A. J. Levine, E. D. Siggia, and P. S. Swain, Stochastic gene expression in a single cell, Science, vol.297, pp.1183-1186, 2002.

F. Emmert-streib, M. Dehmer, and B. Haibe-kains, Gene regulatory networks and their applications: understanding biological and medical problems in terms of networks, Front. Cell Dev. Biol, vol.2, p.38, 2014.

M. Fang, H. Xie, S. K. Dougan, H. Ploegh, and A. Van-oudenaarden, Stochastic cytokine expression induces mixed T helper cell States, PLoS Biol, vol.11, 2013.

E. Farge, Mechanical induction of Twist in the Drosophila foregut/ stomodeal primordium, Curr Biol, vol.13, pp.576-577, 2003.
URL : https://hal.archives-ouvertes.fr/inserm-02440678

K. Featherstone, C. V. Harper, A. Mcnamara, S. Semprini, D. G. Spiller et al., Pulsatile patterns of pituitary hormone gene expression change during development, J. Cell Sci, vol.124, pp.3484-3491, 2011.

K. Featherstone, K. Hey, H. Momiji, A. V. Mcnamara, A. L. Patist et al., Spatially coordinated dynamic gene transcription in living pituitary tissue, vol.5, p.8494, 2016.

C. Furusawa and K. Kaneko, A dynamical-systems view of stem cell biology, Science, vol.338, pp.215-217, 2012.

N. P. Gao, O. Gandrillon, A. Páldi, U. Herbach, and R. Gunawan, Universality of cell differentiation trajectories revealed by a reconstruction of transcriptional uncertainty landscapes from single-cell transcriptomic data, 2020.

A. Giladi, F. Paul, Y. Herzog, Y. Lubling, A. Weiner et al., , 2018.

, Single-cell characterization of haematopoietic progenitors and their trajectories in homeostasis and perturbed haematopoiesis, Nat. Cell Biol, vol.20, pp.836-846

J. Glisse, Simulation d'un Modèle Darwinien de Différenciation Cellulaire, 2009.

M. A. Goodell, H. Nguyen, and N. Shroyer, Somatic stem cell heterogeneity: diversity in the blood, skin and intestinal stem cell compartments, Nat. Rev. Mol. Cell Biol, vol.16, pp.299-309, 2015.

G. Guo, M. Huss, G. Q. Tong, C. Wang, L. Li-sun et al., Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst, Dev. Cell, vol.18, pp.675-685, 2010.

P. Guye, M. R. Ebrahimkhani, N. Kipniss, J. J. Velazquez, E. Schoenfeld et al., Genetically engineering self-organization of human pluripotent stem cells into a liver bud-like tissue using Gata6, Nat. Commun, vol.7, p.10243, 2016.

K. Hayashi, S. M. De-sousa-lopes, F. Tang, K. Lao, and M. A. Surani, Dynamic equilibrium and heterogeneity of mouse pluripotent stem cells with distinct functional and epigenetic states, Cell Stem Cell, vol.3, pp.391-401, 2008.

S. R. Hough, M. Thornton, E. Mason, J. C. Mar, C. A. Wells et al., Single-cell gene expression profiles define self-renewing, pluripotent, and lineage primed states of human pluripotent stem cells, Stem Cell Rep, vol.2, pp.881-895, 2014.

S. Huang, Systems biology of stem cells: three useful perspectives to help overcome the paradigm of linear pathways, Philos. Trans. R. Soc. Lond. B Biol. Sci, vol.366, pp.2247-2259, 2011.

S. Huang, I. Ernberg, and S. Kauffman, Cancer attractors: a systems view of tumors from a gene network dynamics and developmental perspective, Semin. Cell Dev. Biol, vol.20, pp.869-876, 2009.

D. A. Hume, Probability in transcriptional regulation and its implications for leukocyte differentiation and inducible gene expression, Blood, vol.96, pp.2323-2328, 2000.

N. Ji, T. C. Middelkoop, R. A. Mentink, M. C. Betist, S. Tonegawa et al., Feedback control of gene expression variability in the Caenorhabditis elegans Wnt pathway, Cell, vol.155, pp.869-880, 2013.

T. Kalmar, C. Lim, P. Hayward, S. Munoz-descalzo, J. Nichols et al., Regulated fluctuations in nanog expression mediate cell fate decisions in embryonic stem cells, PLoS Biol, vol.7, p.1000149, 2009.

K. Kaneko, Characterization of stem cells and cancer cells on the basis of gene expression profile stability, plasticity, and robustness: dynamical systems theory of gene expressions under cell-cell interaction explains mutational robustness of differentiated cells and suggests how cancer cells emerge, Bioessays, vol.33, pp.403-413, 2011.

S. Kauffman, Homeostasis and differentiation in random genetic control networks, Nature, vol.224, pp.177-178, 1969.

A. M. Klein, L. Mazutis, I. Akartuna, N. Tallapragada, A. Veres et al., Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells, Cell, vol.161, pp.1187-1201, 2015.

S. Knaan-shanzer, I. Van-der-velde-van-dijke, M. J. Van-de-watering, P. J. De-leeuw, D. Valerio et al., Phenotypic and functional reversal within the early human hematopoietic compartment, Stem Cells, vol.26, pp.3210-3217, 2008.

J. J. Kupiec, A probabilist theory for cell differentiation, embryonic mortality and DNA c-value paradox, Specul. Sci. Technol, vol.6, pp.471-478, 1983.

J. J. Kupiec, A chance-selection model for cell differentiation, Cell Death Differ, vol.3, pp.385-390, 1996.

J. J. Kupiec, A Darwinian theory for the origin of cellular differentiation, Mol. Gen. Genet, vol.255, pp.201-208, 1997.

J. J. Kupiec, The Origin of Individuals, 2009.

B. Laforge, Emergent properties in biological systems as a result of competition between internal and external dynamics, J. Biol. Phys. Chem, vol.9, pp.5-9, 2009.

B. Laforge, D. Guez, M. Martinez, and J. J. Kupiec, Modeling embryogenesis and cancer: an approach based on an equilibrium between the autostabilization of stochastic gene expression and the interdependence of cells for proliferation, Prog. Biophys. Mol. Biol, vol.89, pp.93-120, 2005.

B. J. Lesch, A. R. Gehrke, M. L. Bulyk, and C. I. Bargmann, Transcriptional regulation and stabilization of left-right neuronal identity in C. elegans, Genes Dev, vol.23, pp.345-358, 2009.

R. Levins and R. Lewontin, The Dialectic Biologist, 1985.

M. Li and J. C. Belmonte, Deconstructing the pluripotency gene regulatory network, Nat. Cell Biol, vol.20, pp.382-392, 2018.

B. D. Macarthur and I. R. Lemischka, Statistical mechanics of pluripotency, Cell, vol.154, pp.484-489, 2013.

B. D. Macarthur, A. Sevilla, M. Lenz, F. J. Muller, B. M. Schuldt et al., Nanog-dependent feedback loops regulate murine embryonic stem cell heterogeneity, Nat. Cell Biol, vol.14, pp.1139-1147, 2012.

R. Marsland and J. England, Limits of predictions in thermodynamic systems: a review, Rep. Prog. Phys, vol.81, p.16601, 2018.

E. Meshorer, D. Yellajoshula, E. George, P. J. Scambler, D. T. Brown et al., Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells, Dev. Cell, vol.10, pp.105-116, 2006.

V. Moignard, S. Woodhouse, L. Haghverdi, A. J. Lilly, Y. Tanaka et al., Decoding the regulatory network of early blood development from single-cell gene expression measurements, Nat. Biotechnol, vol.33, pp.269-276, 2015.

M. Mojtahedi, A. Skupin, J. Zhou, I. G. Castano, R. Y. Leong-quong et al., Cell fate decision as high-dimensional critical state transition, PLoS Biol, vol.14, p.2000640, 2016.

N. Moris, C. Pina, and A. M. Arias, Transition states and cell fate decisions in epigenetic landscapes, Nat. Rev. Genet, vol.17, pp.693-703, 2016.

A. Moussy, J. Cosette, R. Parmentier, C. Da-silva, G. Corre et al., Integrated time-lapse and single-cell transcription studies highlight the variable and dynamic nature of human hematopoietic cell fate commitment, PLoS Biol, vol.15, p.2001867, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01934491

M. Muers, Development: many roads lead to commitment, Nat. Rev. Genet, vol.13, p.224, 2012.

Y. Ohnishi, W. Huber, A. Tsumura, M. Kang, P. Xenopoulos et al., Cell-to-cell expression variability followed by signal reinforcement progressively segregates early mouse lineages, Nat. Cell Biol, vol.16, pp.27-37, 2014.

A. Olsson, M. Venkatasubramanian, V. K. Chaudhri, B. J. Aronow, N. Salomonis et al., Single-cell analysis of mixed-lineage states leading to a binary cell fate choice, Nature, vol.537, pp.698-702, 2016.

S. H. Orkin and K. Hochedlinger, Chromatin connections to pluripotency and cellular reprogramming, Cell, vol.145, pp.835-850, 2011.

A. Paldi, Stochastic gene expression during cell differentiation: order from disorder? Cell Mol, Life Sci, vol.60, pp.1775-1778, 2003.

A. Paldi, What makes the cell differentiate?, Prog. Biophys. Mol. Biol, vol.110, pp.41-43, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02881149

N. Pelaez, A. Gavalda-miralles, B. Wang, H. T. Navarro, H. Gudjonson et al., Dynamics and heterogeneity of a fate determinant during transition towards cell differentiation, vol.4, p.8924, 2015.

C. Pina, C. Fugazza, A. J. Tipping, J. Brown, S. Soneji et al., Inferring rules of lineage commitment in haematopoiesis, Nat. Cell Biol, vol.14, pp.287-294, 2012.

I. Prigogine, Structure, dissipation and life, Proceedings of the 1st International Conference on Physique Théorique et Biologie, 1967.

A. Radzisheuskaya, B. Chia-gle, R. L. Santos, T. W. Theunissen, L. F. Castro et al., A defined Oct4 level governs cell state transitions of pluripotency entry and differentiation into all embryonic lineages, Nat. Cell Biol, vol.15, pp.579-590, 2013.

A. Raj, S. A. Rifkin, E. Andersen, and A. Van-oudenaarden, Variability in gene expression underlies incomplete penetrance, Nature, vol.463, pp.913-918, 2010.

E. V. Ram and E. Meshorer, Transcriptional competence in pluripotency, Genes Dev, vol.23, pp.2793-2798, 2009.

A. Richard, L. Boullu, U. Herbach, A. Bonnafoux, V. Morin et al., Single-cell-based analysis highlights a surge in cell-to-cell molecular variability preceding irreversible commitment in a differentiation process, PLoS Biol, vol.14, p.1002585, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01934489

S. Semrau, J. E. Goldmann, M. Soumillon, T. S. Mikkelsen, R. Jaenisch et al., Dynamics of lineage commitment revealed by singlecell transcriptomics of differentiating embryonic stem cells, Nat. Commun, vol.8, p.1096, 2017.

C. S. Simon, A. K. Hadjantonakis, and C. Schroter, Making lineage decisions with biological noise: lessons from the early mouse embryo, Wiley Interdiscip. Rev. Dev. Biol, vol.7, p.319, 2018.

J. M. Slack, Conrad Hal Waddington: the last renaissance biologist?, Nat. Rev. Genet, vol.3, pp.889-895, 2002.

M. Spivakov and A. G. Fisher, Epigenetic signatures of stem-cell identity, Nat. Rev. Genet, vol.8, pp.263-271, 2007.

P. S. Stumpf, R. C. Smith, M. Lenz, A. Schuppert, F. J. Muller et al., Stem cell differentiation as a non-markov stochastic process, Cell Syst, vol.5, pp.268-282, 2017.

K. Sunadome, T. Suzuki, M. Usui, Y. Ashida, and E. Nishida, Antagonism between the master regulators of differentiation ensures the discreteness and robustness of cell fates, Mol. Cell, vol.54, pp.526-535, 2014.

A. V. Terskikh, T. Miyamoto, C. Chang, L. Diatchenko, and I. L. Weissman, Gene expression analysis of purified hematopoietic stem cells and committed progenitors, Blood, vol.102, pp.94-101, 2003.

P. W. Tetteh, H. F. Farin, and H. Clevers, Plasticity within stem cell hierarchies in mammalian epithelia, Trends Cell Biol, vol.25, pp.100-108, 2015.

M. Thery and M. Bornens, Cell shape and cell division, Curr. Opin. Cell Biol, vol.18, pp.648-657, 2006.

J. A. Thomson, J. Itskovitz-eldor, S. S. Shapiro, M. A. Waknitz, J. J. Swiergiel et al., Embryonic stem cell lines derived from human blastocysts, Science, vol.282, pp.1145-1147, 1998.

M. E. Torres-padilla and I. Chambers, Transcription factor heterogeneity in pluripotent stem cells: a stochastic advantage, Development, vol.141, pp.2173-2181, 2014.

S. Varahan, A. Walvekar, V. Sinha, S. Krishna, and S. Laxman, Metabolic constraints drive self-organization of specialized cell groups, vol.8, p.46735, 2019.

M. Villani, A. Barbieri, and R. Serra, A dynamical model of genetic networks for cell differentiation, PLoS One, vol.6, p.17703, 2011.

A. Wabik and P. H. Jones, Switching roles: the functional plasticity of adult tissue stem cells, Embo J, vol.34, pp.1164-1179, 2015.

M. F. Wernet, E. O. Mazzoni, A. Celik, D. M. Duncan, I. Duncan et al., Stochastic spineless expression creates the retinal mosaic for colour vision, Nature, vol.440, pp.174-180, 2006.

R. Yamamoto, Y. Morita, J. Ooehara, S. Hamanaka, M. Onodera et al., Clonal analysis unveils self-renewing lineage-restricted progenitors generated directly from hematopoietic stem cells, Cell, vol.154, pp.1112-1126, 2013.

R. A. Young, Control of the embryonic stem cell state, Cell, vol.144, pp.940-954, 2011.

D. Zipori, The nature of stem cells: state rather than entity, Nat. Rev. Genet, vol.5, pp.873-878, 2004.

D. Zipori, The stem state: plasticity is essential, whereas self-renewal and hierarchy are optional, Stem Cells, vol.23, pp.719-726, 2005.