Parallel derivation of isogenic human primed and naive induced pluripotent stem cells

Stéphanie Kilens, Dimitri Meistermann, Diego Moreno, Caroline Chariau, Anne Gaignerie, Arnaud Reignier, Yohann Lelièvre, Miguel Casanova, Céline Vallot, Steven Nedellec, Léa Flippe, Julie Firmin, Juan Song, Eric Charpentier, Jenna Lammers, Audrey Donnart, Nadège Marec, Wallid Deb, Audrey Bihouée, Cédric Le Caignec, Claire Pecqueur, Richard Redon, Paul Barrière, Jérémie Bourdon, Vincent Pasque, Magali Soumillon, Tarjei S Mikkelsen, Claire Rougeulle, Thomas Fréour, Laurent David, Milieu Intérieur Consortium, Laurent Abel, Andres Alcover, Kalla Astrom, Philippe Bousso, Pierre Bruhns, Ana Cumano, Darragh Duffy, Caroline Demangel, Ludovic Deriano, James Di Santo, Françoise Dromer, Gérard Eberl, Jost Enninga, Jacques Fellay, Antonio Freitas, Odile Gelpi, Ivo Gomperts-Boneca, Serge Hercberg, Olivier Lantz, Claude Leclerc, Hugo Mouquet, Etienne Patin, Sandra Pellegrini, Stanislas Pol, Lars Rogge, Anavaj Sakuntabhai, Olivier Schwartz, Benno Schwikowski, Spencer Shorte, Vassili Soumelis, Frédéric Tangy, Eric Tartour, Antoine Toubert, Marie-Noëlle Ungeheuer, Lluis Quintana-Murci, Matthew L Albert, Stéphanie Kilens, Dimitri Meistermann, Diego Moreno, Caroline Chariau, Anne Gaignerie, Arnaud Reignier, Yohann Lelièvre, Miguel Casanova, Céline Vallot, Steven Nedellec, Léa Flippe, Julie Firmin, Juan Song, Eric Charpentier, Jenna Lammers, Audrey Donnart, Nadège Marec, Wallid Deb, Audrey Bihouée, Cédric Le Caignec, Claire Pecqueur, Richard Redon, Paul Barrière, Jérémie Bourdon, Vincent Pasque, Magali Soumillon, Tarjei S Mikkelsen, Claire Rougeulle, Thomas Fréour, Laurent David, Milieu Intérieur Consortium, Laurent Abel, Andres Alcover, Kalla Astrom, Philippe Bousso, Pierre Bruhns, Ana Cumano, Darragh Duffy, Caroline Demangel, Ludovic Deriano, James Di Santo, Françoise Dromer, Gérard Eberl, Jost Enninga, Jacques Fellay, Antonio Freitas, Odile Gelpi, Ivo Gomperts-Boneca, Serge Hercberg, Olivier Lantz, Claude Leclerc, Hugo Mouquet, Etienne Patin, Sandra Pellegrini, Stanislas Pol, Lars Rogge, Anavaj Sakuntabhai, Olivier Schwartz, Benno Schwikowski, Spencer Shorte, Vassili Soumelis, Frédéric Tangy, Eric Tartour, Antoine Toubert, Marie-Noëlle Ungeheuer, Lluis Quintana-Murci, Matthew L Albert

Abstract

Induced pluripotent stem cells (iPSCs) have considerably impacted human developmental biology and regenerative medicine, notably because they circumvent the use of cells of embryonic origin and offer the potential to generate patient-specific pluripotent stem cells. However, conventional reprogramming protocols produce developmentally advanced, or primed, human iPSCs (hiPSCs), restricting their use to post-implantation human development modeling. Hence, there is a need for hiPSCs resembling preimplantation naive epiblast. Here, we develop a method to generate naive hiPSCs directly from somatic cells, using OKMS overexpression and specific culture conditions, further enabling parallel generation of their isogenic primed counterparts. We benchmark naive hiPSCs against human preimplantation epiblast and reveal remarkable concordance in their transcriptome, dependency on mitochondrial respiration and X-chromosome status. Collectively, our results are essential for the understanding of pluripotency regulation throughout preimplantation development and generate new opportunities for disease modeling and regenerative medicine.

Conflict of interest statement

The authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
Direct reprogramming of somatic cells into hiNPSCs. a Direct generation of isogenic naive and primed hiPSCs. Fibroblasts were transduced with 3 Sendai viruses expressing a polycistron KLF4/OCT4/SOX2, MYC and KLF4 at a ratio of 5:5:3, respectively. Cells were split on feeders at day 7, and placed in the indicated media at day 9. Scale bar = 100 µm. b Summary of lines generated for this study in primed (KSR+FGF2, yellow) or naive culture media (RSeT, blue or T2iLGö, pink) originated from 5 different donors. c Different pluripotent states are induced depending on culture media. Transcriptomes of hiPSCs and hiNPSCs, control primed hESC lines H1 and H9 or the naive hESC line HNES1 were analyzed by PCA. Symbols represent donor lines, and size of the symbols represents the passage. Arrows have been drawn to highlight the reprogramming trajectories. d T2iLGö hiNPSCs are the closest to human epiblast cells. PCA of single-cell RNA-seq data sets from preimplantation embryo samples, compared to primed hPSCs from ref. and to primed/naive hiPSCs/hESCs from this study
Fig. 2
Fig. 2
hiNPSCs express markers specific to human epiblast cells. a Specific naive pluripotency markers display identical profiles in T2iLGö hiNPSCs and preimplantation epiblast cells. Individual differentially expressed genes plotted as RPKM for single-cell RNA-seq or mRNA molecules per million of total mRNA molecules for DGE-seq. Upper panel: all genes are differentially expressed (Epi vs primed), except SOX2; lower panel: all genes are differentially expressed (T2iLGö vs primed), except POU5F1(OCT4) and NANOG. Error bars are defined as s.e.m. Statistical tests used to compute differentially expressed genes are defined in the “Differential Expression profiling” section of the Methods. b Schematic representation of the human preimplantation development comparing clinical staging (Morula, B2, B3, B4 and B5) with corresponding embryonic days (E). EPI epiblast cells in red, PE primitive endoderm cells in green; TE trophectoderm cells in blue. c KLF17 protein is expressed in all morula cells before being restricted to epiblast cells in the blastocyst. Human embryos were cultivated in a time-lapse microscope and fixed at indicated stages (morula, B2 or B4 blastocysts). Immunofluorescence for KLF17 (red), GATA2 (blue) and SOX17 (green) was performed. For each indicated embryonic stage, immunofluorescence was performed on 3 biological replicates. Scale bar = 50 µm. d KLF17 protein is specifically expressed in T2iLGö hiNPSCs (pink) and not in isogenic lines cultivated in RSeT (blue) and KSR-FGF2 (yellow). Indicated cell lines were analyzed by immunofluorescence for NANOG (yellow), KLF17 (red) and DNMT3L (cyan). This figure is representative of 8 biological replicates. Scale bar = 50 µm
Fig. 3
Fig. 3
T2iLGö hiNPSCs metabolic profile is closely related to preimplantation epiblast. a Genes coding proteins of the electron transport chain, located in the inner membrane of the mitochondria, are upregulated in human epiblast cells and T2iLGö hiNPSCs in comparison to their primed counterparts. Relative expression of genes related to oxidative phosphorylation pathway for hESC, morula and epiblast samples analyzed by single-cell RNA-seq (left), and analyzed by DGE-seq for primed or naive hPSCs (right). Genes were classified by mitochondrion complex and hierarchically clustered. b T2iLGö hiNPSCs have higher metabolic activity than their isogenic counterparts in RSeT and KSR+FGF2. A SeaHorse apparatus was used to measure the oxygen consumption rate and the extracellular acidification rate of hiNPSC and hiPSC lines, maintained in indicated culture conditions. This figure presents six biological replicates. Each symbol in the panel is the average of a technical triplicate. c T2iLGö hiNPSCs have a higher resistance to inhibition of glycolysis. Quantification of colony numbers obtained after culture with the indicated concentrations of 2-deoxy-D-glucose. Primed cells were seeded in StemMACS™ iPS Brew XF, and naive cells were seeded in the indicated medium. Error bars indicate s.d. of three technical replicates. The presented experiment is representative of four independent experiments
Fig. 4
Fig. 4
T2iLGö hiNPSCs are hypomethylated. a T2iLGö hiNPSCs are hypomethylated in comparison to their RSeT and KSR+FGF2 counterparts. 5mC content is expressed as the percentage of 5mC in the total pool of cytosine for the indicated cell lines. Significance level was determined using Kruskal–Wallis test **p < 0.01. b Expression of indicated epigenetic-related genes is plotted as RPKM for single-cell RNA-seq or mRNA molecules per million of total mRNA molecules for DGE-seq. Error bars are defined as s.e.m. *Differentially expressed gene, as defined in Methods
Fig. 5
Fig. 5
T2iLGö hiNPSCs have a X-chromosome status related to preimplantation epiblast. a T2iLGö hiNPSCs or high-passage KSR-FGF2 do not display H3K27me3 foci. Indicated cell lines were analyzed by immunofluorescence for H3K27me3. This experiment is representative of three technical replicates performed at different passages of the cell lines. Scale bar = 50 µm. b, c T2iLGö hiNPSCs show signs of X-chromosome reactivation. mRNA FISH analysis for bATRX and XACT or c XIST and XACT. For each cell line represented, more than 100 cells were investigated for their nuclear expression of the indicated mRNA. Quantifications for each combination are indicated below pictures. For each table, samples distribution between FISH counting were found statistically different (p < 0.01) by a homogeneity χ2 test. Scale bar = 20 µm
Fig. 6
Fig. 6
Clonal analysis of X-chromosome reactivation in T2iLGö hiNPSCs. a Representative karyotypes of T2iLGö subclones. b Karyotype and mRNA FISH analysis for ATRX and XACT (left) or XIST and XACT (right) of indicated subclones. For each subclone represented, more than 50 cells were investigated for their nuclear expression of the indicated mRNA. Quantifications for each combination are indicated below pictures
Fig. 7
Fig. 7
hiNPSCs in T2iLGö achieve the most naive pluripotency hallmarks. The presented reprogramming method enables to simultaneously generate isogenic hiPSCs in KSR+FGF2, RSeT and T2iLGö media. The naive pluripotency level of the generated cell lines can be assessed with specific markers, X-chromosome activity status, DNA methylation level and capacity to tolerate glycolysis inhibition

References

    1. Nichols J, Smith A. Naive and primed pluripotent states. Cell Stem Cell. 2009;4:487–492. doi: 10.1016/j.stem.2009.05.015.
    1. Weinberger L, Ayyash M, Novershtern N, Hanna JH. Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat. Rev. Mol. Cell. Biol. 2016;17:155–169. doi: 10.1038/nrm.2015.28.
    1. Chen H, et al. Reinforcement of STAT3 activity reprogrammes human embryonic stem cells to naive-like pluripotency. Nat. Commun. 2015;6:7095. doi: 10.1038/ncomms8095.
    1. Chan YS, et al. Induction of a human pluripotent state with distinct regulatory circuitry that resembles preimplantation epiblast. Cell Stem Cell. 2013;13:663–675. doi: 10.1016/j.stem.2013.11.015.
    1. Gafni O, et al. Derivation of novel human ground state naive pluripotent stem cells. Nature. 2013;504:282–286. doi: 10.1038/nature12745.
    1. Valamehr B, et al. Platform for induction and maintenance of transgene-free hiPSCs resembling ground state pluripotent stem cells. Stem Cell Rep. 2014;2:366–381. doi: 10.1016/j.stemcr.2014.01.014.
    1. Takashima Y, et al. Resetting transcription factor control circuitry toward ground-state pluripotency in human. Cell. 2014;158:1254–1269. doi: 10.1016/j.cell.2014.08.029.
    1. Theunissen TW, et al. Systematic Identification of culture conditions for induction and maintenance of naive human pluripotency. Cell Stem Cell. 2014;15:471–487. doi: 10.1016/j.stem.2014.07.002.
    1. Ware CB, et al. Derivation of naive human embryonic stem cells. Proc. Natl. Acad. Sci. USA. 2014;111:4484–4489. doi: 10.1073/pnas.1319738111.
    1. Blakeley P, et al. Defining the three cell lineages of the human blastocyst by single-cell RNA-seq. Development. 2015;142:3151–3165. doi: 10.1242/dev.123547.
    1. Yan L, et al. Single-cell RNA-Seq profiling of human preimplantation embryos and embryonic stem cells. Nat. Struct. Mol. Biol. 2013;20:1131–1139. doi: 10.1038/nsmb.2660.
    1. Petropoulos S, et al. Single-Cell RNA-Seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell. 2016;165:1012–1026. doi: 10.1016/j.cell.2016.03.023.
    1. Guo H, et al. The DNA methylation landscape of human early embryos. Nature. 2014;511:606–610. doi: 10.1038/nature13544.
    1. Smith ZD, et al. DNA methylation dynamics of the human preimplantation embryo. Nature. 2014;511:611–615. doi: 10.1038/nature13581.
    1. Vallot C, et al. XACT noncoding RNA competes with XIST in the control of X chromosome activity during human early development. Cell Stem Cell. 2017;20:102–111. doi: 10.1016/j.stem.2016.10.014.
    1. De Los Angeles A, et al. Hallmarks of pluripotency. Nature. 2015;525:469–478. doi: 10.1038/nature15515.
    1. Pastor WA, et al. Naive human pluripotent cells feature a methylation landscape devoid of blastocyst or germline memory. Cell Stem Cell. 2016;18:323–329. doi: 10.1016/j.stem.2016.01.019.
    1. Sahakyan A, et al. Human naive pluripotent stem cells model X chromosome dampening and X inactivation. Cell Stem Cell. 2016;20:87–101. doi: 10.1016/j.stem.2016.10.006.
    1. Guo G, et al. Naive pluripotent stem cells derived directly from isolated cells of the human inner cell mass. Stem Cell Rep. 2016;6:437–446. doi: 10.1016/j.stemcr.2016.02.005.
    1. Collier AJ, et al. Comprehensive cell surface protein profiling identifies specific markers of human naive and primed pluripotent states. Cell Stem Cell. 2017;20:874–890 e877. doi: 10.1016/j.stem.2017.02.014.
    1. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448:313–317. doi: 10.1038/nature05934.
    1. Maherali N, et al. Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell. 2007;1:55–70. doi: 10.1016/j.stem.2007.05.014.
    1. Wernig M, et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature. 2007;448:318–324. doi: 10.1038/nature05944.
    1. Ying QL, et al. The ground state of embryonic stem cell self-renewal. Nature. 2008;453:519–523. doi: 10.1038/nature06968.
    1. Guo G, et al. Epigenetic resetting of human pluripotency. Development. 2017;144:2748–2763. doi: 10.1242/dev.146811.
    1. Choi J, et al. DUSP9 modulates DNA hypomethylation in female mouse pluripotent stem cells. Cell Stem Cell. 2017;20:706–719 e707. doi: 10.1016/j.stem.2017.03.002.
    1. Cacchiarelli D, et al. Integrative analyses of human reprogramming reveal dynamic nature of induced pluripotency. Cell. 2015;162:412–424. doi: 10.1016/j.cell.2015.06.016.
    1. Soumillon, M., Cacchiarelli, D. & Semrau, S. Characterization of directed differentiation by high-throughput single-cell RNA-Seq. Preprint at bioRxiv (2014)
    1. Niakan KK, Han J, Pedersen RA, Simon C, Pera RA. Human pre-implantation embryo development. Development. 2012;139:829–841. doi: 10.1242/dev.060426.
    1. Theunissen TW, et al. Molecular criteria for defining the naive human pluripotent state. Cell Stem Cell. 2016;19:502–515. doi: 10.1016/j.stem.2016.06.011.
    1. Ogata H, et al. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 1999;27:29–34. doi: 10.1093/nar/27.1.29.
    1. Gu W, et al. Glycolytic metabolism plays a functional role in regulating human pluripotent stem cell state. Cell Stem Cell. 2016;19:476–490. doi: 10.1016/j.stem.2016.08.008.
    1. Durruthy-Durruthy J, et al. Spatiotemporal reconstruction of the human blastocyst by single-cell gene-expression analysis informs induction of naive pluripotency. Dev. Cell. 2016;38:100–115. doi: 10.1016/j.devcel.2016.06.014.
    1. Okamoto I, et al. Eutherian mammals use diverse strategies to initiate X-chromosome inactivation during development. Nature. 2011;472:370–374. doi: 10.1038/nature09872.
    1. Vallot C, et al. Erosion of X chromosome inactivation in human pluripotent cells initiates with XACT coating and depends on a specific heterochromatin landscape. Cell Stem Cell. 2015;16:533–546. doi: 10.1016/j.stem.2015.03.016.
    1. Mekhoubad S, et al. Erosion of dosage compensation impacts human iPSC disease modeling. Cell Stem Cell. 2012;10:595–609. doi: 10.1016/j.stem.2012.02.014.
    1. Patel S, et al. Human embryonic stem cells do not change their X inactivation status during differentiation. Cell Rep. 2017;18:54–67. doi: 10.1016/j.celrep.2016.11.054.
    1. Smith A. Formative pluripotency: the executive phase in a developmental continuum. Development. 2017;144:365–373. doi: 10.1242/dev.142679.
    1. Buecker C, et al. A murine ESC-like state facilitates transgenesis and homologous recombination in human pluripotent stem cells. Cell Stem Cell. 2010;6:535–546. doi: 10.1016/j.stem.2010.05.003.
    1. Wu J, et al. Interspecies chimerism with mammalian pluripotent stem cells. Cell. 2017;168:473–486 e415. doi: 10.1016/j.cell.2016.12.036.
    1. Yang Y, et al. Derivation of pluripotent stem cells with in vivo embryonic and extraembryonic potency. Cell. 2017;169:243–257 e225. doi: 10.1016/j.cell.2017.02.005.
    1. von Meyenn F, et al. Comparative principles of DNA methylation reprogramming during human and mouse in vitro primordial germ cell specification. Dev. Cell. 2016;39:104–115. doi: 10.1016/j.devcel.2016.09.015.
    1. Daley GQ, et al. Setting global standards for stem cell research and clinical translation: the 2016 ISSCR guidelines. Stem Cell Rep. 2016;6:787–797. doi: 10.1016/j.stemcr.2016.05.001.
    1. Alpha Scientists in Reproductive Medicine and ESHRE Special Interest Group of Embryology. The Istanbul consensus workshop on embryo assessment: proceedings of an expert meeting. Hum. Reprod.26, 1270–1283 (2011).
    1. Samavarchi-Tehrani P, et al. Functional genomics reveals a BMP-driven mesenchymal-to-epithelial transition in the initiation of somatic cell reprogramming. Cell Stem Cell. 2010;7:64–77. doi: 10.1016/j.stem.2010.04.015.
    1. Picelli S, et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 2014;9:171–181. doi: 10.1038/nprot.2014.006.
    1. Picelli S, et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods. 2013;10:1096–1098. doi: 10.1038/nmeth.2639.
    1. Trombetta JJ, et al. Preparation of single-cell RNA-Seq libraries for next generation sequencing. Curr. Protoc. Mol. Biol. 2014;107:4 22 21–17.
    1. Trapnell C, et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 2012;7:562–578. doi: 10.1038/nprot.2012.016.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat. Methods. 2012;9:357–359. doi: 10.1038/nmeth.1923.
    1. Anders S, Pyl PT, Huber W. HTSeq--a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–169. doi: 10.1093/bioinformatics/btu638.
    1. Lun AT, McCarthy DJ, Marioni JC. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Res. 2016;5:2122.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Suomi T, Seyednasrollah F, Jaakkola MK, Faux T, Elo LL. PLoS Comput. Biol. 2017. ROTS: An R package for reproducibility-optimized statistical testing; p. e1005562.
    1. Suzuki R, Shimodaira H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics. 2006;22:1540–1542. doi: 10.1093/bioinformatics/btl117.
    1. Alexa A, Rahnenfuhrer J, Lengauer T. Improved scoring of functional groups from gene expression data by decorrelating GO graph structure. Bioinformatics. 2006;22:1600–1607. doi: 10.1093/bioinformatics/btl140.
    1. Luo W, Friedman MS, Shedden K, Hankenson KD, Woolf PJ. BMC Bioinformatics. 2009. GAGE: generally applicable gene set enrichment for pathway analysis; p. 161.

Source: PubMed

3
Prenumerera