D. C. Adler-wailes, A. T. Alberobello, X. Ma, L. Hugendubler, E. A. Stern et al., Analysis of variants and mutations in the human winged helix FOXA3 gene and associations with metabolic traits, Int. J. Obes, vol.39, pp.888-892, 2005.

Y. Ang, S. Tsai, D. Lee, J. Monk, J. Su et al., Wdr5 Mediates Self-Renewal and Reprogramming via the Embryonic Stem Cell Core Transcriptional Network, Cell, vol.145, pp.183-197, 2011.
DOI : 10.1016/j.cell.2011.03.003

URL : https://doi.org/10.1016/j.cell.2011.03.003

E. Van-anken, E. P. Romijn, C. Maggioni, A. Mezghrani, R. Sitia et al., Sequential Waves of Functionally Related Proteins Are Expressed When B Cells Prepare for Antibody Secretion, Immunity, vol.18, pp.243-253, 2003.

A. M. Arensdorf, D. Dezwaan-mccabe, R. J. Kaufman, and D. T. Rutkowski, , 2013.

, Temporal clustering of gene expression links the metabolic transcription factor HNF4? to the ER stress-dependent gene regulatory network

C. Atkins, Q. Liu, E. Minthorn, S. Zhang, D. J. Figueroa et al., Characterization of a Novel PERK Kinase Inhibitor with Antitumor and Antiangiogenic Activity, Cancer Res, vol.73, 1993.

C. E. Badr, J. W. Hewett, X. O. Breakefield, and B. A. Tannous, A Highly Sensitive Assay for Monitoring the Secretory Pathway and ER Stress, PLoS ONE, vol.2, p.571, 2007.

A. Banito, S. T. Rashid, J. C. Acosta, S. Li, C. F. Pereira et al., Senescence impairs successful reprogramming to pluripotent stem cells, Genes Dev, vol.23, pp.2134-2139, 2009.

S. Benosman, P. Ravanan, R. G. Correa, Y. Hou, M. Yu et al., Interleukin-1 Receptor-Associated Kinase-2 (IRAK2) Is a Critical Mediator of Endoplasmic Reticulum (ER) Stress Signaling, PLOS ONE, vol.8, p.64256, 2013.

C. F. Bento, M. Renna, G. Ghislat, C. Puri, A. Ashkenazi et al., Mammalian Autophagy: How Does It Work?, Annu. Rev. Biochem, vol.85, 2016.

S. Bernales, K. L. Mcdonald, and P. Walter, Autophagy Counterbalances Endoplasmic Reticulum Expansion during the Unfolded Protein Response, PLOS Biol, vol.4, p.423, 2006.

H. M. Blau, C. Chiu, and C. Webster, Cytoplasmic activation of human nuclear genes in stable heterocaryons, Cell, vol.32, pp.1171-1180, 1983.

C. Bock, E. Kiskinis, G. Verstappen, H. Gu, G. Boulting et al., Reference Maps of Human ES and iPS Cell Variation Enable High-Throughput Characterization of Pluripotent Cell Lines, Cell, vol.144, pp.439-452, 2011.

G. Boden, X. Duan, C. Homko, E. J. Molina, W. Song et al., Increase in Endoplasmic Reticulum Stress-Related Proteins and Genes in Adipose Tissue of Obese, Insulin-Resistant Individuals. Diabetes, vol.57, pp.2438-2444, 2008.

L. Bouman, A. Schlierf, A. K. Lutz, J. Shan, A. Deinlein et al., Parkin is transcriptionally regulated by ATF4: evidence for an interconnection between mitochondrial stress and ER stress, Cell Death Differ, vol.18, pp.769-782, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00595932

L. A. Boyer, T. I. Lee, M. F. Cole, S. E. Johnstone, S. S. Levine et al., Core Transcriptional Regulatory Circuitry in Human Embryonic Stem Cells. Cell, vol.122, pp.947-956, 2005.

J. L. Brodsky, J. Goeckeler, and R. Schekman, BiP and Sec63p are required for both co-and posttranslational protein translocation into the yeast endoplasmic reticulum, Proc. Natl. Acad. Sci, vol.92, pp.9643-9646, 1995.

S. M. Buckley, B. Aranda-orgilles, A. Strikoudis, E. Apostolou, E. Loizou et al., Regulation of Pluripotency and Cellular Reprogramming by the Ubiquitin-Proteasome System, Cell Stem Cell, vol.11, pp.783-798, 2012.

Y. Buganim, D. A. Faddah, A. W. Cheng, E. Itskovich, S. Markoulaki et al., Single-Cell Expression Analyses during Cellular Reprogramming Reveal an Early Stochastic and a Late Hierarchic Phase, Cell, vol.150, pp.1209-1222, 2012.

Y. Buganim, D. A. Faddah, J. , and R. , Mechanisms and models of somatic cell reprogramming, Nat. Rev. Genet, vol.14, pp.427-439, 2013.

T. Burdon, C. Stracey, I. Chambers, J. Nichols, and A. Smith, Suppression of SHP-2 and ERK Signalling Promotes Self-Renewal of Mouse Embryonic Stem Cells, Dev. Biol, vol.210, pp.30-43, 1999.
DOI : 10.1006/dbio.1999.9265

URL : https://doi.org/10.1006/dbio.1999.9265

T. Burdon, I. Chambers, C. Stracey, H. Niwa, and A. Smith, Signaling Mechanisms Regulating Self-Renewal and Differentiation of Pluripotent Embryonic Stem Cells, Cells Tissues Organs, vol.165, pp.131-143, 1999.
DOI : 10.1159/000016693

D. Cacchiarelli, C. Trapnell, M. J. Ziller, M. Soumillon, M. Cesana et al., Integrative Analyses of Human Reprogramming Reveal Dynamic Nature of Induced Pluripotency, Cell, vol.162, pp.412-424, 2015.
DOI : 10.1016/j.cell.2015.06.016

URL : https://doi.org/10.1016/j.cell.2015.06.016

M. Calfon, H. Zeng, F. Urano, J. H. Till, S. R. Hubbard et al., IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA, Nature, vol.415, pp.92-96, 2002.
DOI : 10.1038/415092a

P. Cartwright, C. Mclean, A. Sheppard, D. Rivett, K. Jones et al., , 2005.

, LIF/STAT3 controls ES cell self-renewal and pluripotency by a Myc-dependent mechanism, Development, vol.132, pp.885-896

E. M. Chan, S. Ratanasirintrawoot, I. Park, P. D. Manos, Y. Loh et al., Live cell imaging distinguishes bona fide human iPS cells from partially reprogrammed cells, Nat. Biotechnol, vol.27, pp.1033-1037, 2009.

J. Chen, Z. Zhang, L. Li, B. Chen, A. Revyakin et al., Single-Molecule Dynamics of Enhanceosome Assembly in Embryonic Stem Cells, Cell, vol.156, pp.1274-1285, 2014.

T. Chen, L. Shen, J. Yu, H. Wan, A. Guo et al., Rapamycin and other longevity-promoting compounds enhance the generation of mouse induced pluripotent stem cells, Aging Cell, vol.10, pp.908-911, 2011.

A. M. Cheng, T. M. Saxton, R. Sakai, S. Kulkarni, G. Mbamalu et al., Mammalian Grb2 Regulates Multiple Steps in Embryonic Development and Malignant Transformation, Cell, vol.95, pp.793-803, 1998.

L. Cheng, N. F. Hansen, L. Zhao, Y. Du, C. Zou et al., Low Incidence of DNA Sequence Variation in Human Induced Pluripotent Stem Cells Generated by Nonintegrating Plasmid Expression, Cell Stem Cell, vol.10, pp.337-344, 2012.

C. A. Cowan, J. Atienza, D. A. Melton, and K. Eggan, Nuclear reprogramming of somatic cells after fusion with human embryonic stem cells, Science, vol.309, pp.1369-1373, 2005.

J. S. Cox and P. Walter, A Novel Mechanism for Regulating Activity of a Transcription Factor That Controls the Unfolded Protein Response, Cell, vol.87, pp.391-404, 1996.

J. S. Cox, C. E. Shamu, and P. Walter, Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase, Cell, vol.73, pp.1197-1206, 1993.

J. J. Credle, J. S. Finer-moore, F. R. Papa, R. M. Stroud, and P. Walter, On the mechanism of sensing unfolded protein in the endoplasmic reticulum, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.18773-18784, 2005.

R. L. Davis, H. Weintraub, and A. B. Lassar, Expression of a single transfected cDNA converts fibroblasts to myoblasts, Cell, vol.51, pp.987-1000, 1987.

A. J. Dorner, L. C. Wasley, P. Raney, S. Haugejorden, M. Green et al., The stress response in Chinese hamster ovary cells. Regulation of ERp72 and protein disulfide isomerase expression and secretion, J. Biol. Chem, vol.265, pp.22029-22034, 1990.

E. Duplan, E. Giaime, J. Viotti, J. Sévalle, O. Corti et al., ER-stress-associated functional link between Parkin and DJ-1 via a transcriptional cascade involving the tumor suppressor p53 and the spliced X-box binding protein XBP-1, J Cell Sci, vol.126, pp.2124-2133, 2013.

M. J. Edel, C. Menchon, S. Menendez, A. Consiglio, A. Raya et al., Rem2 GTPase maintains survival of human embryonic stem cells as well as enhancing reprogramming by regulating p53 and cyclin D1, Genes Dev, vol.24, pp.561-573, 2010.

M. J. Evans and M. H. Kaufman, Establishment in culture of pluripotential cells from mouse embryos, Nature, vol.292, pp.154-156, 1981.

B. Feng, J. Jiang, P. Kraus, J. Ng, J. D. Heng et al., Reprogramming of fibroblasts into induced pluripotent stem cells with orphan nuclear receptor Esrrb, Nat. Cell Biol, vol.11, pp.197-203, 2009.

N. Festuccia, R. Osorno, F. Halbritter, V. Karwacki-neisius, P. Navarro et al., Esrrb Is a Direct Nanog Target Gene that Can Substitute for Nanog Function in Pluripotent Cells, Cell Stem Cell, vol.11, pp.477-490, 2012.

C. D. Folmes, T. J. Nelson, A. Martinez-fernandez, D. K. Arrell, J. Z. Lindor et al., Somatic Oxidative Bioenergetics Transitions into Pluripotency-Dependent Glycolysis to Facilitate Nuclear Reprogramming, Cell Metab, vol.14, pp.264-271, 2011.

A. J. Fornace, I. Alamo, and M. C. Hollander, DNA damage-inducible transcripts in mammalian cells, Proc. Natl. Acad. Sci, vol.85, pp.8800-8804, 1988.

J. R. Friedman and G. K. Voeltz, The ER in 3D: a multifunctional dynamic membrane network, Trends Cell Biol, vol.21, pp.709-717, 2011.

N. Fusaki, H. Ban, A. Nishiyama, K. Saeki, and M. Hasegawa, Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome, Proc. Jpn. Acad. Ser. B, vol.85, pp.348-362, 2009.

F. González, S. Boué, and J. C. Belmonte, Methods for making induced pluripotent stem cells: reprogramming à la carte, Nat. Rev. Genet, vol.12, pp.231-242, 2011.

F. González, D. Georgieva, F. Vanoli, Z. Shi, M. Stadtfeld et al., Homologous Recombination DNA Repair Genes Play a Critical Role in Reprogramming to a Pluripotent State, Cell Rep, vol.3, pp.651-660, 2013.

A. Gore, Z. Li, H. Fung, J. E. Young, S. Agarwal et al., Somatic coding mutations in human induced pluripotent stem cells, Nature, vol.471, pp.63-67, 2011.

A. M. Gorman, S. J. Healy, R. Jäger, and A. Samali, Stress management at the ER: Regulators of ER stress-induced apoptosis, Pharmacol. Ther, vol.134, pp.306-316, 2012.

M. G. Guenther, G. M. Frampton, F. Soldner, D. Hockemeyer, M. Mitalipova et al., Chromatin Structure and Gene Expression Programs of Human Embryonic and Induced Pluripotent Stem Cells, Cell Stem Cell, vol.7, pp.249-257, 2010.

J. B. Gurdon, The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles, J. Embryol. Exp. Morphol, vol.10, pp.622-640, 1962.

L. Hackler, B. Ózsvári, M. Gyuris, P. Sipos, G. Fábián et al., The Curcumin Analog C-150, Influencing NF-?B, UPR and Akt/Notch Pathways Has Potent Anticancer Activity In Vitro and In Vivo, PloS One, vol.11, p.149832, 2016.

J. Han, P. Yuan, H. Yang, J. Zhang, B. S. Soh et al., Tbx3 improves the germ-line competency of induced pluripotent stem cells, Nature, vol.463, pp.1096-1100, 2010.

J. Hansson, M. R. Rafiee, S. Reiland, J. M. Polo, J. Gehring et al., Highly Coordinated Proteome Dynamics during Reprogramming of Somatic Cells to Pluripotency, Cell Rep, vol.2, pp.1579-1592, 2012.

H. P. Harding, Y. Zhang, R. , and D. , ER Stress Causes Rapid Loss of Intestinal Epithelial Stemness through Activation of the Unfolded Protein Response, References, vol.397, pp.1128-1139, 1999.

J. D. Heng, B. Feng, J. Han, J. Jiang, P. Kraus et al., The Nuclear Receptor Nr5a2 Can Replace Oct4 in the Reprogramming of Murine Somatic Cells to Pluripotent Cells, Cell Stem Cell, vol.6, pp.167-174, 2010.

C. Hetz, E. Chevet, and H. P. Harding, Targeting the unfolded protein response in disease, Nat. Rev. Drug Discov, vol.12, pp.703-719, 2013.

M. W. Hetzer, T. C. Walther, and I. W. Mattaj, PUSHING THE ENVELOPE: Structure, Function, and Dynamics of the Nuclear Periphery, Annu. Rev. Cell Dev. Biol, vol.21, pp.347-380, 2005.

K. Hochedlinger, J. , and R. , Monoclonal mice generated by nuclear transfer from mature B and T donor cells, Nature, vol.415, pp.1035-1038, 2002.

D. Hockemeyer, J. , and R. , Induced Pluripotent Stem Cells Meet Genome Editing, Cell Stem Cell, vol.18, pp.573-586, 2016.
DOI : 10.1016/j.stem.2016.04.013

URL : https://doi.org/10.1016/j.stem.2016.04.013

D. Hockemeyer, H. Wang, S. Kiani, C. S. Lai, Q. Gao et al., Genetic engineering of human pluripotent cells using TALE nucleases, Nat. Biotechnol, vol.29, pp.731-734, 2011.
DOI : 10.1038/nbt.1927

URL : http://europepmc.org/articles/pmc3152587?pdf=render

J. Hollien and J. S. Weissman, Decay of Endoplasmic Reticulum-Localized mRNAs During the Unfolded Protein Response, Science, vol.313, pp.104-107, 2006.
DOI : 10.1126/science.1129631

H. Hong, K. Takahashi, T. Ichisaka, T. Aoi, O. Kanagawa et al., Suppression of induced pluripotent stem cell generation by the p53-p21 pathway, Nature, vol.460, pp.1132-1135, 2009.
DOI : 10.1038/nature08235

URL : http://europepmc.org/articles/pmc2917235?pdf=render

C. A. Hu, S. K. Dougan, A. M. Mcgehee, J. C. Love, and H. L. Ploegh, XBP-1 regulates signal transduction, transcription factors and bone marrow colonization in B cells, EMBO J, vol.28, pp.1624-1636, 2009.
DOI : 10.1038/emboj.2009.117

URL : http://emboj.embopress.org/content/embojnl/28/11/1624.full.pdf

S. M. Hussein, N. N. Batada, S. Vuoristo, R. W. Ching, R. Autio et al., Copy number variation and selection during reprogramming to pluripotency, Nature, vol.471, pp.58-62, 2011.
DOI : 10.1038/nature09871

M. Ieda, J. Fu, P. Delgado-olguin, V. Vedantham, Y. Hayashi et al., Direct Reprogramming of Fibroblasts into Functional Cardiomyocytes by Defined Factors, Cell, vol.142, pp.375-386, 2010.
DOI : 10.1016/j.cell.2010.07.002

URL : https://doi.org/10.1016/j.cell.2010.07.002

N. N. Iwakoshi, A. Lee, and L. H. Glimcher, The X-box binding protein-1 transcription factor is required for plasma cell differentiation and the unfolded protein response, Immunol. Rev, vol.194, pp.29-38, 2003.
DOI : 10.1034/j.1600-065x.2003.00057.x

T. Iwawaki, R. Akai, S. Yamanaka, and K. Kohno, Function of IRE1 alpha in the placenta is essential for placental development and embryonic viability, Proc. Natl. Acad. Sci, vol.106, pp.16657-16662, 2009.

S. Jagannathan, J. C. Hsu, D. W. Reid, Q. Chen, W. J. Thompson et al., Multifunctional Roles for the Protein Translocation Machinery in RNA Anchoring to the Endoplasmic Reticulum, J. Biol. Chem, vol.289, pp.25907-25924, 2014.

M. Jeanne, C. Labelle-dumais, J. Jorgensen, W. B. Kauffman, G. M. Mancini et al., COL4A2 mutations impair COL4A1 and COL4A2 secretion and cause hemorrhagic stroke, Am. J. Hum. Genet, vol.90, pp.91-101, 2012.
DOI : 10.1016/j.ajhg.2011.11.022

URL : https://doi.org/10.1016/j.ajhg.2011.11.022

J. Ji, V. Sharma, S. Qi, M. E. Guarch, P. Zhao et al., Antioxidant Supplementation Reduces Genomic Aberrations in Human Induced Pluripotent Stem Cells, Stem Cell Rep, vol.2, pp.44-51, 2014.
DOI : 10.1016/j.stemcr.2013.11.004

URL : https://doi.org/10.1016/j.stemcr.2013.11.004

D. J. Kahler, F. S. Ahmad, A. Ritz, H. Hua, D. N. Moroziewicz et al., Improved Methods for Reprogramming Human Dermal Fibroblasts Using Fluorescence Activated Cell Sorting, PLoS ONE, vol.8, p.59867, 2013.
DOI : 10.1371/journal.pone.0059867

URL : https://doi.org/10.1371/journal.pone.0059867

K. Kaji, K. Norrby, A. Paca, M. Mileikovsky, P. Mohseni et al., , 2009.

, Virus-free induction of pluripotency and subsequent excision of reprogramming factors, Nature, vol.458, pp.771-775

S. Kang, N. S. Rane, S. J. Kim, J. L. Garrison, J. Taunton et al., , 2006.

, Substrate-Specific Translocational Attenuation during ER Stress Defines a Pre-Emptive Quality Control Pathway, Cell, vol.127, pp.999-1013

H. Kato, S. Nakajima, Y. Saito, S. Takahashi, R. Katoh et al., mTORC1 serves ER stress-triggered apoptosis via selective activation of the IRE1-JNK pathway, Cell Death Differ, vol.19, pp.310-320, 2012.

R. P. Koche, Z. D. Smith, M. Adli, H. Gu, M. Ku et al., Reprogramming Factor Expression Initiates Widespread Targeted Chromatin Remodeling, Cell Stem Cell, vol.8, pp.96-105, 2011.

Y. Kozutsumi, M. Segal, K. Normington, M. Gething, and J. Sambrook, The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose-regulated proteins, Nature, vol.332, pp.462-464, 1988.

W. M. Kuwabara, L. Zhang, I. Schuiki, R. Curi, A. Volchuk et al., NADPH oxidase-dependent production of reactive oxygen species induces endoplasmatic reticulum stress in neutrophil-like HL60 cells, PloS One, vol.10, 2015.

L. C. Laurent, I. Ulitsky, I. Slavin, H. Tran, A. Schork et al., Dynamic Changes in the Copy Number of Pluripotency and Cell Proliferation Genes in Human ESCs and iPSCs during Reprogramming and Time in Culture, Cell Stem Cell, vol.8, pp.106-118, 2011.

A. Lee, N. N. Iwakoshi, and L. H. Glimcher, XBP-1 Regulates a Subset of Endoplasmic Reticulum Resident Chaperone Genes in the Unfolded Protein Response, Mol. Cell. Biol, vol.23, pp.7448-7459, 2003.

H. Li, M. Collado, A. Villasante, K. Strati, S. Ortega et al., The Ink4/Arf locus is a barrier for iPS cell reprogramming, Nature, vol.460, pp.1136-1139, 2009.
DOI : 10.1038/nature08290

URL : http://europepmc.org/articles/pmc3578184?pdf=render

R. Li, J. Liang, S. Ni, T. Zhou, X. Qing et al., A Mesenchymal-to-Epithelial Transition Initiates and Is Required for the Nuclear Reprogramming of Mouse Fibroblasts, Cell Stem Cell, vol.7, pp.51-63, 2010.

J. H. Lin, H. Li, Y. Zhang, D. Ron, and P. Walter, Divergent Effects of PERK and IRE1 Signaling on Cell Viability, PLOS ONE, vol.4, 2009.

N. Maherali, T. Ahfeldt, A. Rigamonti, J. Utikal, C. Cowan et al., A High-Efficiency System for the Generation and Study of Human Induced Pluripotent Stem Cells, Cell Stem Cell, vol.3, pp.340-345, 2008.

S. J. Marciniak, C. Y. Yun, S. Oyadomari, I. Novoa, Y. Zhang et al., CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum, Genes Dev, vol.18, pp.3066-3077, 2004.

R. M. Marión, K. Strati, H. Li, M. Murga, R. Blanco et al., A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity, Nature, vol.460, pp.1149-1153, 2009.

G. R. Martin, Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells, Proc. Natl. Acad. Sci, vol.78, pp.7634-7638, 1981.

T. Matsuda, STAT3 activation is sufficient to maintain an undifferentiated state of mouse embryonic stem cells, EMBO J, vol.18, pp.4261-4269, 1999.
DOI : 10.1093/emboj/18.15.4261

URL : http://europepmc.org/articles/pmc1171502?pdf=render

P. I. Merksamer, A. Trusina, and F. R. Papa, Real-Time Redox Measurements during Endoplasmic Reticulum Stress Reveal Interlinked Protein Folding Functions, Cell, vol.135, pp.933-947, 2008.
DOI : 10.1016/j.cell.2008.10.011

URL : https://doi.org/10.1016/j.cell.2008.10.011

N. Mesaeli, K. Nakamura, E. Zvaritch, P. Dickie, E. Dziak et al., Calreticulin Is Essential for Cardiac Development, J. Cell Biol, vol.144, pp.857-868, 1999.
DOI : 10.1083/jcb.144.5.857

URL : http://jcb.rupress.org/content/144/5/857.full.pdf

T. S. Mikkelsen, J. Hanna, X. Zhang, M. Ku, M. Wernig et al., Dissecting direct reprogramming through integrative genomic analysis, Nature, vol.454, pp.49-55, 2008.
DOI : 10.1038/nature07056

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2754827

J. Ming, S. Ruan, M. Wang, D. Ye, N. Fan et al., , 2015.

, STF-083010, reverses tamoxifen-related drug resistance in breast cancer by inhibiting IRE1/XBP1, A novel chemical, vol.6, pp.40692-40703

K. Mitsui, Y. Tokuzawa, H. Itoh, K. Segawa, M. Murakami et al., The Homeoprotein Nanog Is Required for Maintenance of Pluripotency in Mouse Epiblast and ES Cells, Cell, vol.113, pp.631-642, 2003.

K. Mori, T. Kawahara, H. Yoshida, H. Yanagi, Y. et al., Signalling from endoplasmic reticulum to nucleus: transcription factor with a basic-leucine zipper motif is required for the unfolded protein-response pathway, Genes Cells, vol.1, pp.803-817, 1996.

A. M. Newman, C. , and J. B. , Lab-Specific Gene Expression Signatures in Pluripotent Stem Cells, Cell Stem Cell, vol.7, pp.258-262, 2010.
DOI : 10.1016/j.stem.2010.06.016

URL : https://doi.org/10.1016/j.stem.2010.06.016

J. Nichols and A. Smith, Naive and Primed Pluripotent States, Cell Stem Cell, vol.4, pp.487-492, 2009.
DOI : 10.1016/j.stem.2009.05.015

URL : https://doi.org/10.1016/j.stem.2009.05.015

H. Niwa, T. Burdon, I. Chambers, and A. Smith, Self-renewal of pluripotent embryonic stem cells is mediated via activation of STAT3, Genes Dev, vol.12, pp.2048-2060, 1998.

H. Niwa, K. Ogawa, D. Shimosato, A. , and K. , A parallel circuit of LIF signalling pathways maintains pluripotency of mouse ES cells, Nature, vol.460, pp.118-122, 2009.

M. Ogata, S. Hino, A. Saito, K. Morikawa, S. Kondo et al., Autophagy Is Activated for Cell Survival after Endoplasmic Reticulum Stress, Mol. Cell. Biol, vol.26, pp.9220-9231, 2006.

K. Okamura, Y. Kimata, H. Higashio, A. Tsuru, and K. Kohno, Dissociation of Kar2p/BiP from an ER Sensory Molecule, Ire1p, Triggers the Unfolded Protein Response in Yeast, Biochem. Biophys. Res. Commun, vol.279, pp.445-450, 2000.

K. Okita, Y. Matsumura, Y. Sato, A. Okada, A. Morizane et al., A more efficient method to generate integration-free human iPS cells, Nat. Methods, vol.8, pp.409-412, 2011.

T. T. Onder, N. Kara, A. Cherry, A. U. Sinha, N. Zhu et al., Chromatin-modifying enzymes as modulators of reprogramming, Nature, vol.483, pp.598-602, 2012.

A. D. Panopoulos, O. Yanes, S. Ruiz, Y. S. Kida, D. Diep et al., The metabolome of induced pluripotent stem cells reveals metabolic changes occurring in somatic cell reprogramming, Cell Res, vol.22, pp.168-177, 2012.

R. C. Pereira, A. M. Delany, C. , and E. , CCAAT/Enhancer Binding Protein Homologous Protein (DDIT3) Induces Osteoblastic Cell Differentiation, Endocrinology, vol.145, pp.1952-1960, 2004.

V. Picanço-castro, E. Russo-carbolante, L. C. Reis, A. M. Fraga, D. A. De-magalhães et al., , 2010.

C. and T. , Pluripotent Reprogramming of Fibroblasts by Lentiviralmediated Insertion of SOX2, Stem Cells Dev, vol.20, pp.169-180

O. Pluquet, A. Pourtier, A. , and C. , The unfolded protein response and cellular senescence. A Review in the Theme: Cellular Mechanisms of Endoplasmic Reticulum Stress Signaling in Health and Disease, Am. J. Physiol.-Cell Physiol, vol.308, pp.415-425, 2015.

J. M. Polo, E. Anderssen, R. M. Walsh, B. A. Schwarz, C. M. Nefzger et al., A Molecular Roadmap of Reprogramming Somatic Cells into iPS Cells, Cell, vol.151, pp.1617-1632, 2012.

H. Qin, A. Diaz, L. Blouin, R. J. Lebbink, W. Patena et al., Systematic Identification of Barriers to Human iPSC Generation, Cell, vol.158, pp.449-461, 2014.

A. M. Reimold, A. Etkin, I. Clauss, A. Perkins, D. S. Friend et al., An essential role in liver development for transcription factor XBP-1, Genes Dev, vol.14, pp.152-157, 2000.

D. A. Robinton and G. Q. Daley, The promise of induced pluripotent stem cells in research and therapy, Nature, vol.481, pp.295-305, 2012.

D. Ron and P. Walter, Signal integration in the endoplasmic reticulum unfolded protein response, Nat. Rev. Mol. Cell Biol, vol.8, pp.519-529, 2007.

C. Ruberti and F. Brandizzi, Conserved and plant-unique strategies for overcoming endoplasmic reticulum stress, Plant Cell Biol, vol.5, p.69, 2014.

S. Ruiz, A. D. Panopoulos, N. Montserrat, M. Multon, A. Daury et al., Generation of a Drug-inducible Reporter System to Study Cell Reprogramming in Human Cells, J. Biol. Chem, vol.287, pp.40767-40778, 2012.

S. Ruiz, A. J. Lopez-contreras, M. Gabut, R. M. Marion, P. Gutierrez-martinez et al., Limiting replication stress during somatic cell reprogramming reduces genomic instability in induced pluripotent stem cells, Nat. Commun, vol.6, 2015.

D. T. Rutkowski and R. S. Hegde, Regulation of basal cellular physiology by the homeostatic unfolded protein response, J. Cell Biol, vol.189, pp.783-794, 2010.

P. Samavarchi-tehrani, A. Golipour, L. David, H. Sung, T. A. Beyer et al., Functional Genomics Reveals a BMPDriven Mesenchymal-to-Epithelial Transition in the Initiation of Somatic Cell Reprogramming, Cell Stem Cell, vol.7, pp.64-77, 2010.

H. Sathananthan, M. Pera, T. , and A. , The fine structure of human embryonic stem cells, Reprod. Biomed. Online, vol.4, pp.56-61, 2002.

N. Sato, L. Meijer, L. Skaltsounis, P. Greengard, and A. H. Brivanlou, Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor, Nat. Med, vol.10, pp.55-63, 2004.
URL : https://hal.archives-ouvertes.fr/hal-00020135

S. Schuck, W. A. Prinz, K. S. Thorn, C. Voss, and P. Walter, Membrane expansion alleviates endoplasmic reticulum stress independently of the unfolded protein response, J. Cell Biol, vol.187, pp.525-536, 2009.
DOI : 10.1083/jcb.200907074

URL : http://jcb.rupress.org/content/187/4/525.full.pdf

D. Senft and Z. A. Ronai, UPR, autophagy, and mitochondria crosstalk underlies the ER stress response, Trends Biochem. Sci, vol.40, pp.141-148, 2015.
DOI : 10.1016/j.tibs.2015.01.002

URL : http://europepmc.org/articles/pmc4340752?pdf=render

C. E. Shamu and P. Walter, Oligomerization and phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus, EMBO J, vol.15, pp.3028-3039, 1996.

Y. Shi, Y. Yang, B. Hoang, C. Bardeleben, B. Holmes et al., Therapeutic potential of targeting IRES-dependent c-myc translation in multiple myeloma cells during ER stress, Oncogene, vol.35, pp.1015-1024, 2016.
DOI : 10.1038/onc.2015.156

URL : http://europepmc.org/articles/pmc5104155?pdf=render

Y. Shibata, C. Voss, J. M. Rist, J. Hu, T. A. Rapoport et al., The Reticulon and Dp1/Yop1p Proteins Form Immobile Oligomers in the Tubular Endoplasmic Reticulum, J. Biol. Chem, vol.283, pp.18892-18904, 2008.
DOI : 10.1074/jbc.m800986200

URL : http://www.jbc.org/content/283/27/18892.full.pdf

Y. Shibata, T. Shemesh, W. A. Prinz, A. F. Palazzo, M. M. Kozlov et al., Mechanisms Determining the Morphology of the Peripheral ER, Cell, vol.143, pp.774-788, 2010.
DOI : 10.1016/j.cell.2010.11.007

URL : https://doi.org/10.1016/j.cell.2010.11.007

C. Sidrauski and P. Walter, The Transmembrane Kinase Ire1p Is a Site-Specific Endonuclease That Initiates mRNA Splicing in the Unfolded Protein Response, Cell, vol.90, pp.1031-1039, 1997.

C. Sidrauski, R. Chapman, and P. Walter, The unfolded protein response: an intracellular signalling pathway with many surprising features, Trends Cell Biol, vol.8, pp.245-249, 1998.
DOI : 10.1016/s0962-8924(98)01267-7

A. H. Skalet, J. A. Isler, L. B. King, H. P. Harding, D. Ron et al., , 2005.

, Rapid B Cell Receptor-induced Unfolded Protein Response in Nonsecretory B Cells Correlates with Pro-Versus Antiapoptotic Cell Fate, J. Biol. Chem, vol.280, pp.39762-39771

A. G. Smith, J. K. Heath, D. D. Donaldson, G. G. Wong, J. Moreau et al., Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides, Nature, vol.336, pp.688-690, 1988.

F. Soldner, D. Hockemeyer, C. Beard, Q. Gao, G. W. Bell et al., Parkinson's Disease Patient-Derived Induced Pluripotent Stem Cells Free of Viral Reprogramming Factors, Cell, vol.136, pp.964-977, 2009.

R. Sridharan, J. Tchieu, M. J. Mason, R. Yachechko, E. Kuoy et al., Role of the Murine Reprogramming Factors in the Induction of Pluripotency, Cell, vol.136, pp.364-377, 2009.

K. Sugiura, Y. Muro, K. Futamura, K. Matsumoto, N. Hashimoto et al., The Unfolded Protein Response Is Activated in Differentiating Epidermal Keratinocytes, J. Invest. Dermatol, vol.129, pp.2126-2135, 2009.

S. T. Suhr, E. A. Chang, J. Tjong, N. Alcasid, G. A. Perkins et al., Mitochondrial Rejuvenation After Induced Pluripotency, PLoS ONE, vol.5, 2010.

M. Tada, Y. Takahama, K. Abe, N. Nakatsuji, and T. Tada, Nuclear reprogramming of somatic cells by in vitro hybridization with ES cells, Curr. Biol, vol.11, pp.1553-1558, 2001.

N. Takagi, M. A. Yoshida, O. Sugawara, and M. Sasaki, Reversal of Xinactivation in female mouse somatic cells hybridized with murine teratocarcinoma stem cells in vitro, Cell, vol.34, pp.1053-1062, 1983.

K. Takahashi, Y. , and S. , Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors, Cell, vol.126, pp.663-676, 2006.

K. Takahashi, Y. , and S. , A decade of transcription factor-mediated reprogramming to pluripotency, Nat. Rev. Mol. Cell Biol, 2016.

K. Takahashi, K. Tanabe, M. Ohnuki, M. Narita, T. Ichisaka et al., Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors, Cell, vol.131, pp.861-872, 2007.

R. C. Taylor and A. Dillin, XBP-1 Is a Cell-Nonautonomous Regulator of Stress Resistance and Longevity, Cell, vol.153, pp.1435-1447, 2013.

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.

Y. Tokuzawa, E. Kaiho, M. Maruyama, K. Takahashi, K. Mitsui et al., Fbx15 Is a Novel Target of Oct3/4 but Is Dispensable for Embryonic Stem Cell Self-Renewal and Mouse Development, Mol. Cell. Biol, vol.23, pp.2699-2708, 2003.

D. Tomar, P. Prajapati, L. Sripada, K. Singh, R. Singh et al., TRIM13 regulates caspase-8 ubiquitination, translocation to autophagosomes and activation during ER stress induced cell death, Biochim. Biophys. Acta BBA-Mol. Cell Res, vol.1833, pp.3134-3144, 2013.

M. Tomioka, M. Nishimoto, S. Miyagi, T. Katayanagi, N. Fukui et al., Identification of Sox-2 regulatory region which is under the control of Oct-3/4-Sox-2 complex, Nucleic Acids Res, vol.30, pp.3202-3213, 2002.

K. J. Travers, C. K. Patil, L. Wodicka, D. J. Lockhart, J. S. Weissman et al., Functional and Genomic Analyses Reveal an Essential Coordination between the Unfolded Protein Response and ER-Associated Degradation, Cell, vol.101, pp.249-258, 2000.

P. A. Tsonis, M. Madhavan, E. E. Tancous, D. Rio-tsonis, and K. , A newt's eye view of lens regeneration, Int. J. Dev. Biol, vol.48, pp.975-980, 2004.

N. Tsubooka, T. Ichisaka, K. Okita, K. Takahashi, M. Nakagawa et al., Roles of Sall4 in the generation of pluripotent stem cells from blastocysts and fibroblasts, Genes Cells, vol.14, pp.683-694, 2009.

J. Utikal, J. M. Polo, M. Stadtfeld, N. Maherali, W. Kulalert et al., Immortalization eliminates a roadblock during cellular reprogramming into iPS cells, Nature, vol.460, pp.1145-1148, 2009.

L. Van-haute, C. Spits, M. Geens, S. Seneca, and K. Sermon, Human embryonic stem cells commonly display large mitochondrial DNA deletions, Nat. Biotechnol, vol.31, pp.20-23, 2013.

K. M. Vattem and R. C. Wek, Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.11269-11274, 2004.

T. Vierbuchen and M. Wernig, Molecular Roadblocks for Cellular Reprogramming, Mol. Cell, vol.47, pp.827-838, 2012.

T. Vierbuchen, A. Ostermeier, Z. P. Pang, Y. Kokubu, T. C. Südhof et al., Direct conversion of fibroblasts to functional neurons by defined factors, Nature, vol.463, pp.1035-1041, 2010.

D. Vilchez, L. Boyer, I. Morantte, M. Lutz, C. Merkwirth et al., Increased proteasome activity in human embryonic stem cells is regulated by PSMD11, Nature, vol.489, pp.304-308, 2012.

D. Vilchez, M. S. Simic, and A. Dillin, Proteostasis and aging of stem cells, Trends Cell Biol, vol.24, pp.161-170, 2014.

A. M. Vitale, N. A. Matigian, S. Ravishankar, B. Bellette, S. A. Wood et al., Variability in the Generation of Induced Pluripotent Stem Cells: Importance for Disease Modeling, Stem Cells Transl. Med, vol.1, pp.641-650, 2012.

C. H. Waddington, The strategy of the genes: a discussion of some aspects of theoretical biology, 1957.

P. Walter, R. , and D. , The Unfolded Protein Response: From Stress Pathway to Homeostatic Regulation, Science, vol.334, pp.1081-1086, 2011.
DOI : 10.1126/science.1209038

S. Wanderling, B. B. Simen, O. Ostrovsky, N. T. Ahmed, S. M. Vogen et al., GRP94 Is Essential for Mesoderm Induction and Muscle Development Because It Regulates Insulin-like Growth Factor Secretion, Mol. Biol. Cell, vol.18, pp.3764-3775, 2007.
DOI : 10.1091/mbc.e07-03-0275

URL : http://www.molbiolcell.org/content/18/10/3764.full.pdf

S. Wang, P. Xia, B. Ye, G. Huang, J. Liu et al., Transient Activation of Autophagy via Sox2-Mediated Suppression of mTOR Is an Important Early Step in Reprogramming to Pluripotency, Cell Stem Cell, vol.13, pp.617-625, 2013.

T. Wang, K. Chen, X. Zeng, J. Yang, Y. Wu et al., The Histone Demethylases Jhdm1a/1b Enhance Somatic Cell Reprogramming in a Vitamin-C-Dependent Manner, Cell Stem Cell, vol.9, pp.575-587, 2011.
DOI : 10.1016/j.stem.2011.10.005

URL : https://doi.org/10.1016/j.stem.2011.10.005

L. Warren, P. D. Manos, T. Ahfeldt, Y. Loh, H. Li et al., Highly Efficient Reprogramming to Pluripotency and Directed Differentiation of Human Cells with Synthetic Modified mRNA, Cell Stem Cell, vol.7, pp.618-630, 2010.
DOI : 10.1016/j.stem.2010.08.012

URL : https://doi.org/10.1016/j.stem.2010.08.012

A. Weismann, W. N. Parker, N. William, R. , and H. , The germ-plasm; a theory of heredity, 's), 1893.
DOI : 10.2307/2842101

I. Wilmut, A. E. Schnieke, J. Mcwhir, A. J. Kind, and K. H. Campbell, Viable offspring derived from fetal and adult mammalian cells, Nature, vol.385, pp.810-813, 1997.
DOI : 10.1038/385810a0

Y. Wu, Y. Li, H. Zhang, Y. Huang, P. Zhao et al., Autophagy and mTORC1 regulate the stochastic phase of somatic cell reprogramming, Nat. Cell Biol, vol.17, pp.715-725, 2015.
DOI : 10.1038/ncb3172

X. Xia, Y. Zhang, C. R. Zieth, and S. Zhang, Transgenes Delivered by Lentiviral Vector are Suppressed in Human Embryonic Stem Cells in A PromoterDependent Manner, Stem Cells Dev, vol.16, pp.167-176, 2007.
DOI : 10.1089/scd.

K. Yamamoto, T. Sato, T. Matsui, M. Sato, T. Okada et al., Transcriptional Induction of Mammalian ER Quality Control Proteins Is Mediated by Single or Combined Action of ATF6? and XBP1, Dev. Cell, vol.13, pp.365-376, 2007.
DOI : 10.1016/j.devcel.2007.07.018

URL : https://doi.org/10.1016/j.devcel.2007.07.018

W. Yan, C. L. Frank, M. J. Korth, B. L. Sopher, I. Novoa et al., Control of PERK eIF2? kinase activity by the endoplasmic reticulum stressinduced molecular chaperone P58IPK, Proc. Natl. Acad. Sci, vol.99, pp.15920-15925, 2002.
DOI : 10.1073/pnas.252341799

URL : http://www.pnas.org/content/99/25/15920.full.pdf

L. Yang, S. G. Carlson, D. Mcburney, and W. E. Horton, Multiple Signals Induce Endoplasmic Reticulum Stress in Both Primary and Immortalized Chondrocytes Resulting in Loss of Differentiation, Impaired Cell Growth, and Apoptosis, J. Biol. Chem, vol.280, pp.31156-31165, 2005.

H. Yoshida, T. Okada, K. Haze, H. Yanagi, T. Yura et al., ATF6 Activated by Proteolysis Binds in the Presence of NF-Y (CBF) Directly to the cis-Acting Element Responsible for the Mammalian Unfolded Protein Response, Mol. Cell. Biol, vol.20, pp.6755-6767, 2000.

H. Yoshida, T. Matsui, A. Yamamoto, T. Okada, and K. Mori, XBP1 mRNA Is Induced by ATF6 and Spliced by IRE1 in Response to ER Stress to Produce a Highly Active Transcription Factor, Cell, vol.107, pp.881-891, 2001.
DOI : 10.1016/s0092-8674(01)00611-0

URL : https://doi.org/10.1016/s0092-8674(01)00611-0

H. Yoshida, M. Oku, M. Suzuki, and K. Mori, pXBP1(U) encoded in XBP1 pre-mRNA negatively regulates unfolded protein response activator pXBP1(S) in mammalian ER stress response, J. Cell Biol, vol.172, pp.565-575, 2006.
DOI : 10.1083/jcb.200508145

URL : http://jcb.rupress.org/content/jcb/172/4/565.full.pdf

J. Yu, M. A. Vodyanik, K. Smuga-otto, J. Antosiewicz-bourget, J. L. Frane et al., Induced Pluripotent Stem Cell Lines Derived from Human Somatic Cells, Science, vol.318, pp.1917-1920, 2007.
DOI : 10.1097/01.ogx.0000305193.72586.39

J. Yu, K. Hu, K. Smuga-otto, S. Tian, R. Stewart et al., Human Induced Pluripotent Stem Cells Free of Vector and Transgene Sequences, Science, vol.324, pp.797-801, 2009.

J. Zhang, E. Nuebel, G. Q. Daley, C. M. Koehler, and M. A. Teitell, Metabolic Regulation in Pluripotent Stem Cells during Reprogramming and Self-Renewal, Cell Stem Cell, vol.11, pp.589-595, 2012.
DOI : 10.1016/j.stem.2012.10.005

URL : https://doi.org/10.1016/j.stem.2012.10.005

K. Zhang, H. N. Wong, B. Song, C. N. Miller, D. Scheuner et al., , 2005.

, The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis, J. Clin. Invest, vol.115, pp.268-281

Y. Zhao, X. Yin, H. Qin, F. Zhu, H. Liu et al., Two Supporting Factors Greatly Improve the Efficiency of Human iPSC Generation, Cell Stem Cell, vol.3, pp.475-479, 2008.
DOI : 10.1016/j.stem.2008.10.002

URL : https://doi.org/10.1016/j.stem.2008.10.002

, HSF-1-mediated cytoskeletal integrity determines thermotolerance and life span

N. A. Baird, P. M. Douglas, M. S. Simic, A. R. Grant, J. J. Moresco et al., HSF-1-mediated cytoskeletal integrity determines thermotolerance and life span, Science, vol.346, pp.360-363, 2014.

P. M. Douglas, N. A. Baird, M. S. Simic, S. Uhlein, M. A. Mccormick et al., Heterotypic Signals from Neural HSF-1 Separate Thermotolerance from Longevity, vol.12, pp.1196-1204, 2015.

, * equal contributions

. Appendix-heterotypic, Signals from Neural HSF-1 Separate Thermotolerance from Longevity