DOI QR코드

DOI QR Code

Single cell heterogeneity in human pluripotent stem cells

  • Yang, Seungbok (Department of Life Sciences, Pohang University of Science and Technology (POSTECH)) ;
  • Cho, Yoonjae (Department of Life Sciences, Pohang University of Science and Technology (POSTECH)) ;
  • Jang, Jiwon (Department of Life Sciences, Pohang University of Science and Technology (POSTECH))
  • Received : 2021.06.18
  • Accepted : 2021.08.29
  • Published : 2021.10.31

Abstract

Human pluripotent stem cells (hPSCs) include human embryonic stem cells (hESCs) derived from blastocysts and human induced pluripotent stem cells (hiPSCs) generated from somatic cell reprogramming. Due to their self-renewal ability and pluripotent differentiation potential, hPSCs serve as an excellent experimental platform for human development, disease modeling, drug screening, and cell therapy. Traditionally, hPSCs were considered to form a homogenous population. However, recent advances in single cell technologies revealed a high degree of variability between individual cells within a hPSC population. Different types of heterogeneity can arise by genetic and epigenetic abnormalities associated with long-term in vitro culture and somatic cell reprogramming. These variations initially appear in a rare population of cells. However, some cancer-related variations can confer growth advantages to the affected cells and alter cellular phenotypes, which raises significant concerns in hPSC applications. In contrast, other types of heterogeneity are related to intrinsic features of hPSCs such as asynchronous cell cycle and spatial asymmetry in cell adhesion. A growing body of evidence suggests that hPSCs exploit the intrinsic heterogeneity to produce multiple lineages during differentiation. This idea offers a new concept of pluripotency with single cell heterogeneity as an integral element. Collectively, single cell heterogeneity is Janus-faced in hPSC function and application. Harmful heterogeneity has to be minimized by improving culture conditions and screening methods. However, other heterogeneity that is integral for pluripotency can be utilized to control hPSC proliferation and differentiation.

Keywords

Acknowledgement

This work has been supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF-2019R1C1C1002377, NRF-2020M3A9D8038184, and NRF-2021R1A4A1031754).

References

  1. Nichols J and Smith A (2009) Naive and primed pluripotent states. Cell Stem Cell 4, 487-492 https://doi.org/10.1016/j.stem.2009.05.015
  2. Weinberger L, Ayyash M, Novershtern N and Hanna JH (2016) Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat Rev Mol Cell Biol 17, 155-169 https://doi.org/10.1038/nrm.2015.28
  3. Niwa H, Ogawa K, Shimosato D and Adachi K (2009) A parallel circuit of LIF signalling pathways maintains pluripotency of mouse ES cells. Nature 460, 118-122 https://doi.org/10.1038/nature08113
  4. Toyooka Y, Shimosato D, Murakami K, Takahashi K and Niwa H (2008) Identification and characterization of subpopulations in undifferentiated ES cell culture. Development 135, 909-918 https://doi.org/10.1242/dev.017400
  5. Wang J, Xie G, Singh M et al (2014) Primate-specific endogenous retrovirus-driven transcription defines naivelike stem cells. Nature 516, 405-409 https://doi.org/10.1038/nature13804
  6. Liu Y, Mi Y, Mueller T et al (2019) Multi-omic measurements of heterogeneity in HeLa cells across laboratories. Nat Biotechnol 37, 314-322 https://doi.org/10.1038/s41587-019-0037-y
  7. Initiative ISC (2011) Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage. Nat Biotechnol 29, 1132-1144 https://doi.org/10.1038/nbt.2051
  8. Draper JS, Smith K, Gokhale P et al (2004) Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nat Biotechnol 22, 53-54 https://doi.org/10.1038/nbt922
  9. Taapken SM, Nisler BS, Newton MA et al (2011) Karyotypic abnormalities in human induced pluripotent stem cells and embryonic stem cells. Nat Biotechnol 29, 313-314 https://doi.org/10.1038/nbt.1835
  10. Baker D, Adam, Paul et al (2016) Detecting genetic mosaicism in cultures of human pluripotent stem cells. Stem Cell Reports 7, 998-1012 https://doi.org/10.1016/j.stemcr.2016.10.003
  11. Narva E, Autio R, Rahkonen N et al (2010) High-resolution DNA analysis of human embryonic stem cell lines reveals culture-induced copy number changes and loss of heterozygosity. Nat Biotechnol 28, 371-377 https://doi.org/10.1038/nbt.1615
  12. Merkle FT, Ghosh S, Kamitaki N et al (2017) Human pluripotent stem cells recurrently acquire and expand dominant negative P53 mutations. Nature 545, 229-233 https://doi.org/10.1038/nature22312
  13. Kucab JE, Zou X, Morganella S et al (2019) A compendium of mutational signatures of environmental agents. Cell 177, 821-836.e816 https://doi.org/10.1016/j.cell.2019.03.001
  14. Halliwell JA, Frith TJR, Laing O et al (2020) Nucleosides rescue replication-mediated genome instability of human pluripotent stem cells. Stem Cell Reports 14, 1009-1017 https://doi.org/10.1016/j.stemcr.2020.04.004
  15. Simara P, Tesarova L, Rehakova D et al (2017) DNA double-strand breaks in human induced pluripotent stem cell reprogramming and long-term in vitro culturing. Stem Cell Res Ther 8, 73 https://doi.org/10.1186/s13287-017-0522-5
  16. Vallabhaneni H, Lynch PJ, Chen G et al (2018) High basal levels of γH2AX in human induced pluripotent stem cells are linked to replication-associated DNA damage and repair. Stem Cells 36, 1501-1513 https://doi.org/10.1002/stem.2861
  17. Cannan WJ and Pederson DS (2016) Mechanisms and consequences of double-strand DNA break formation in chromatin. J Cell Physiol 231, 3-14 https://doi.org/10.1002/jcp.25048
  18. Zhang J, Hirst AJ, Duan F et al (2019) Anti-apoptotic mutations desensitize human pluripotent stem cells to mitotic stress and enable aneuploid cell survival. Stem Cell Reports 12, 557-571 https://doi.org/10.1016/j.stemcr.2019.01.013
  19. Lamm N, Ben-David U, Golan-Lev T, Storchova Z, Benvenisty N and Kerem B (2016) Genomic instability in human pluripotent stem cells arises from replicative stress and chromosome condensation defects. Cell Stem Cell 18, 253-261 https://doi.org/10.1016/j.stem.2015.11.003
  20. Ji J, Ng SH, Sharma V et al (2012) Elevated coding mutation rate during the reprogramming of human somatic cells into induced pluripotent stem cells. Stem Cells 30, 435-440 https://doi.org/10.1002/stem.1011
  21. Yoshihara M, Araki R, Kasama Y et al (2017) Hotspots of de novo point mutations in induced pluripotent stem cells. Cell Reports 21, 308-315 https://doi.org/10.1016/j.celrep.2017.09.060
  22. Rouhani FJ, Nik-Zainal S, Wuster A et al (2016) Mutational history of a human cell lineage from somatic to induced pluripotent stem cells. PLoS Genet 12, e1005932 https://doi.org/10.1371/journal.pgen.1005932
  23. Thompson O, Von Meyenn F, Hewitt Z et al (2020) Low rates of mutation in clinical grade human pluripotent stem cells under different culture conditions. Nat Commun 11, 1528 https://doi.org/10.1038/s41467-020-15271-3
  24. Milholland B, Dong X, Zhang L, Hao X, Suh Y and Vijg J (2017) Differences between germline and somatic mutation rates in humans and mice. Nat Commun 8, 15183 https://doi.org/10.1038/ncomms15183
  25. Jang J, Wang Y, Matthew et al (2016) Primary ciliumautophagy-nrf2 (pan) axis activation commits human embryonic stem cells to a neuroectoderm fate. Cell 165, 410-420 https://doi.org/10.1016/j.cell.2016.02.014
  26. Saretzki G, Walter T, Atkinson S et al (2008) Downregulation of multiple stress defense mechanisms during differentiation of human embryonic stem cells. Stem Cells 26, 455-464 https://doi.org/10.1634/stemcells.2007-0628
  27. Maynard S, Swistowska AM, Lee JW et al (2008) Human embryonic stem cells have enhanced repair of multiple forms of DNA damage. Stem Cells 26, 2266-2274 https://doi.org/10.1634/stemcells.2007-1041
  28. Hyka-Nouspikel N, Desmarais J, Gokhale PJ et al (2012) Deficient DNA damage response and cell cycle checkpoints lead to accumulation of point mutations in human embryonic stem cells. Stem Cells 30, 1901-1910 https://doi.org/10.1002/stem.1177
  29. Luo LZ, Gopalakrishna-Pillai S, Nay SL et al (2012) DNA repair in human pluripotent stem cells is distinct from that in non-pluripotent human cells. PLoS One 7, e30541 https://doi.org/10.1371/journal.pone.0030541
  30. Adams BR, Golding SE, Rao RR and Valerie K (2010) Dynamic dependence on ATR and ATM for doublestrand break repair in human embryonic stem cells and neural descendants. PLoS One 5, e10001 https://doi.org/10.1371/journal.pone.0010001
  31. Julia, Guan X, Jeremy et al (2013) High mitochondrial priming sensitizes hESCs to DNA-damage-induced apoptosis. Cell Stem Cell 13, 483-491 https://doi.org/10.1016/j.stem.2013.07.018
  32. Desmarais JA, Unger C, Damjanov I, Meuth M and Andrews P (2016) Apoptosis and failure of checkpoint kinase 1 activation in human induced pluripotent stem cells under replication stress. Stem Cell Res Ther 7, 17 https://doi.org/10.1186/s13287-016-0279-2
  33. Desmarais JA, Hoffmann MJ, Bingham G, Gagou ME, Meuth M and Andrews PW (2012) Human embryonic stem cells fail to activate CHK1 and commit to apoptosis in response to DNA replication stress. Stem Cells 30, 1385-1393 https://doi.org/10.1002/stem.1117
  34. Madden DT, Davila-Kruger D, Melov S and Bredesen DE (2011) Human embryonic stem cells express elevated levels of multiple pro-apoptotic BCL-2 family members. PLoS One 6, e28530 https://doi.org/10.1371/journal.pone.0028530
  35. Dumitru R, Gama V, Fagan BM et al (2012) Human embryonic stem cells have constitutively active Bax at the golgi and are primed to undergo rapid apoptosis. Molecular Cell 46, 573-583 https://doi.org/10.1016/j.molcel.2012.04.002
  36. Catalina P, Montes R, Ligero G et al (2008) Human ESCs predisposition to karyotypic instability: is a matter of culture adaptation or differential vulnerability among hESC lines due to inherent properties? Molecular Cancer 7, 76 https://doi.org/10.1186/1476-4598-7-76
  37. Imreh MP, Gertow K, Cedervall J et al (2006) In vitro culture conditions favoring selection of chromosomal abnormalities in human ES cells. J Cell Biochem 99, 508-516 https://doi.org/10.1002/jcb.20897
  38. Ben-David U, Arad G, Weissbein U et al (2014) Aneuploidy induces profound changes in gene expression, proliferation and tumorigenicity of human pluripotent stem cells. Nat Commun 5, 4825 https://doi.org/10.1038/ncomms5825
  39. Chambers I, Silva J, Colby D et al (2007) Nanog safeguards pluripotency and mediates germline development. Nature 450, 1230-1234 https://doi.org/10.1038/nature06403
  40. Darr H, Mayshar Y and Benvenisty N (2006) Overexpression of NANOG in human ES cells enables feeder-free growth while inducing primitive ectoderm features. Development 133, 1193-1201 https://doi.org/10.1242/dev.02286
  41. Korkola JE, Houldsworth J, Chadalavada RSV et al (2006) Down-regulation of stem cell genes, including those in a 200-kb gene cluster at 12p13.31, is associated with in vivo differentiation of human male germ cell tumors. Cancer Res 66, 820-827 https://doi.org/10.1158/0008-5472.CAN-05-2445
  42. Rodriguez S, Jafer O, Goker H et al (2003) Expression profile of genes from 12p in testicular germ cell tumors of adolescents and adults associated with i(12p) and amplification at 12p11.2-p12.1. Oncogene 22, 1880-1891 https://doi.org/10.1038/sj.onc.1206302
  43. Avery S, Adam, Baker D et al (2013) BCL-XL mediates the strong selective advantage of a 20q11.21 amplification commonly found in human embryonic stem cell cultures. Stem Cell Reports 1, 379-386 https://doi.org/10.1016/j.stemcr.2013.10.005
  44. Markouli C, Couvreu De Deckersberg E, Regin M et al (2019) Gain of 20q11.21 in human pluripotent stem cells impairs TGF-β-dependent neuroectodermal commitment. Stem Cell Reports 13, 163-176 https://doi.org/10.1016/j.stemcr.2019.05.005
  45. Jo HY, Lee Y, Ahn H et al (2020) Functional in vivo and in vitro effects of 20q11.21 genetic aberrations on hPSC differentiation. Sci Rep 10, 18582 https://doi.org/10.1038/s41598-020-75657-7
  46. Blum B, Bar-Nur O, Golan-Lev T and Benvenisty N (2009) The anti-apoptotic gene survivin contributes to teratoma formation by human embryonic stem cells. Nat Biotechnol 27, 281-287 https://doi.org/10.1038/nbt.1527
  47. Lee CT, Raphael, Abigail et al (2015) Functional consequences of 17q21.31/WNT3-WNT9B amplification in hPSCs with respect to neural differentiation. Cell Rep 10, 616-632 https://doi.org/10.1016/j.celrep.2014.12.050
  48. Jain AK and Barton MC (2018) p53: emerging roles in stem cells, development and beyond. Development 145, dev158360 https://doi.org/10.1242/dev.158360
  49. Suzuki MM and Bird A (2008) DNA methylation landscapes: provocative insights from epigenomics. Nat Rev Genet 9, 465-476 https://doi.org/10.1038/nrg2341
  50. Hackett JA and Surani MA (2013) DNA methylation dynamics during the mammalian life cycle. Philos Trans R Soc Lond B Biol Sci 368, 20110328 https://doi.org/10.1098/rstb.2011.0328
  51. Messerschmidt DM, Knowles BB and Solter D (2014) DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev 28, 812-828 https://doi.org/10.1101/gad.234294.113
  52. Liao J, Karnik R, Gu H et al (2015) Targeted disruption of DNMT1, DNMT3A and DNMT3B in human embryonic stem cells. Nat Genet 47, 469-478 https://doi.org/10.1038/ng.3258
  53. Bock C, Kiskinis E, Verstappen G et al (2011) Reference maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144, 439-452 https://doi.org/10.1016/j.cell.2010.12.032
  54. Kristopher, Altun G, Lynch C et al (2012) Recurrent variations in DNA methylation in human pluripotent stem cells and their differentiated derivatives. Cell Stem Cell 10, 620-634 https://doi.org/10.1016/j.stem.2012.02.013
  55. Calvanese V, Horrillo A, Hmadcha A et al (2008) Cancer genes hypermethylated in human embryonic stem cells. PLoS One 3, e3294 https://doi.org/10.1371/journal.pone.0003294
  56. Weissbein U, Plotnik O, Vershkov D and Benvenisty N (2017) Culture-induced recurrent epigenetic aberrations in human pluripotent stem cells. PLoS Genet 13, e1006979 https://doi.org/10.1371/journal.pgen.1006979
  57. Roost MS, Slieker RC, Bialecka M et al (2017) DNA methylation and transcriptional trajectories during human development and reprogramming of isogenic pluripotent stem cells. Nat Commun 8, 908 https://doi.org/10.1038/s41467-017-01077-3
  58. Bar-Nur O, Holger, Efrat S and Benvenisty N (2011) Epigenetic memory and preferential lineage-specific differentiation in induced pluripotent stem cells derived from human pancreatic islet beta cells. Cell Stem Cell 9, 17-23 https://doi.org/10.1016/j.stem.2011.06.007
  59. Bartolomei MS and Ferguson-Smith AC (2011) Mammalian genomic imprinting. Cold Spring Harb Perspect Biol 3, a002592-a002592 https://doi.org/10.1101/cshperspect.a002592
  60. McGrath J and Solter D (1984) Completion of mouse embryogenesis requires both the maternal and paternal genomes. Cell 37, 179-183 https://doi.org/10.1016/0092-8674(84)90313-1
  61. Surani MAH, Barton SC and Norris ML (1984) Development of reconstituted mouse eggs suggests imprinting of the genome during gametogenesis. Nature 308, 548-550 https://doi.org/10.1038/308548a0
  62. Lim DH and Maher ER (2010) Genomic imprinting syndromes and cancer. Adv Genet 70, 145-175 https://doi.org/10.1016/B978-0-12-380866-0.60006-X
  63. Weksberg R (2010) Imprinted genes and human disease. Am J Med Genet C Semin Med Genet 154C, 317-320 https://doi.org/10.1002/ajmg.c.30268
  64. Yamazawa K, Ogata T and Ferguson-Smith AC (2010) Uniparental disomy and human disease: An overview. Am J Med Genet C Semin Med Genet 154C, 329-334 https://doi.org/10.1002/ajmg.c.30270
  65. Initiative TISC (2007) Characterization of human embryonic stem cell lines by the International Stem Cell Initiative. Nat Biotechnol 25, 803-816 https://doi.org/10.1038/nbt1318
  66. Rugg-Gunn PJ, Ferguson-Smith AC and Pedersen RA (2007) Status of genomic imprinting in human embryonic stem cells as revealed by a large cohort of independently derived and maintained lines. Hum Mol Genet 16, R243-R251 https://doi.org/10.1093/hmg/ddm245
  67. Johannesson B, Sagi I, Gore A et al (2014) Comparable frequencies of coding mutations and loss of imprinting in human pluripotent cells derived by nuclear transfer and defined factors. Cell Stem Cell 15, 634-642 https://doi.org/10.1016/j.stem.2014.10.002
  68. Ma H, Morey R, O'Neil RC et al (2014) Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature 511, 177-183 https://doi.org/10.1038/nature13551
  69. Pick M, Stelzer Y, Bar-Nur O, Mayshar Y, Eden A and Benvenisty N (2009) Clone- and gene-specific aberrations of parental imprinting in human induced pluripotent stem cells. Stem Cells 27, 2686-2690 https://doi.org/10.1002/stem.205
  70. Thorold, Friedli M, He Y et al (2016) Molecular criteria for defining the naive human pluripotent state. Cell Stem Cell 19, 502-515 https://doi.org/10.1016/j.stem.2016.06.011
  71. William, Chen D, Liu W et al (2016) Naive human pluripotent cells feature a methylation landscape devoid of blastocyst or germline memory. Cell Stem Cell 18, 323-329 https://doi.org/10.1016/j.stem.2016.01.019
  72. Bar S, Schachter M, Eldar-Geva T and Benvenisty N (2017) Large-scale analysis of loss of imprinting in human pluripotent stem cells. Cell Rep 19, 957-968 https://doi.org/10.1016/j.celrep.2017.04.020
  73. Jelinic P and Shaw P (2007) Loss of imprinting and cancer. J Pathol 211, 261-268 https://doi.org/10.1002/path.2116
  74. Leick MB, Shoff CJ, Wang EC, Congress JL and Gallicano GI (2011) Loss of imprinting of IGF2 and the epigenetic progenitor model of cancer. Am J Stem Cells 1, 59-74
  75. Davies W, Isles AR and Wilkinson LS (2005) Imprinted gene expression in the brain. Neurosci Biobehav Rev 29, 421-430 https://doi.org/10.1016/j.neubiorev.2004.11.007
  76. Lyon MF (1961) Gene Action in the X-chromosome of the mouse (Mus musculus L.). Nature 190, 372-373 https://doi.org/10.1038/190372a0
  77. Petropoulos S, Edsgard D, Reinius B et al (2016) Single-cell RNA-Seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 165, 1012-1026 https://doi.org/10.1016/j.cell.2016.03.023
  78. Moreira de Mello JC, Fernandes GR, Vibranovski MD and Pereira LV (2017) Early X chromosome inactivation during human preimplantation development revealed by single-cell RNA-sequencing. Sci Rep 7, 10794 https://doi.org/10.1038/s41598-017-11044-z
  79. Bruck T and Benvenisty N (2011) Meta-analysis of the heterogeneity of X chromosome inactivation in human pluripotent stem cells. Stem Cell Research 6, 187-193 https://doi.org/10.1016/j.scr.2010.12.001
  80. Dvash T, Lavon N and Fan G (2010) Variations of X chromosome inactivation occur in early passages of female human embryonic stem cells. PLoS One 5, e11330 https://doi.org/10.1371/journal.pone.0011330
  81. Patel S, Bonora G, Sahakyan A et al (2017) Human embryonic stem cells do not change their X inactivation status during differentiation. Cell Rep 18, 54-67 https://doi.org/10.1016/j.celrep.2016.11.054
  82. Sahakyan A, Kim R, Chronis C et al (2017) Human naive pluripotent stem cells model X chromosome dampening and X inactivation. Cell Stem Cell 20, 87-101 https://doi.org/10.1016/j.stem.2016.10.006
  83. Shen Y, Matsuno Y, Fouse SD et al (2008) X-inactivation in female human embryonic stem cells is in a nonrandom pattern and prone to epigenetic alterations. Proc Natl Acad Sci U S A 105, 4709-4714 https://doi.org/10.1073/pnas.0712018105
  84. Silva SS, Rowntree RK, Mekhoubad S and Lee JT (2008) X-chromosome inactivation and epigenetic fluidity in human embryonic stem cells. Proc Natl Acad Sci U S A 105, 4820-4825 https://doi.org/10.1073/pnas.0712136105
  85. Vallot C, Patrat C, Collier AJ et al (2017) XACT noncoding RNA competes with XIST in the control of X chromosome activity during human early development. Cell Stem Cell 20, 102-111 https://doi.org/10.1016/j.stem.2016.10.014
  86. Lengner CJ, Gimelbrant AA, Erwin JA et al (2010) Derivation of pre-X inactivation human embryonic stem cells under physiological oxygen concentrations. Cell 141, 872-883 https://doi.org/10.1016/j.cell.2010.04.010
  87. Geens M, Seriola A, Barbe L et al (2016) Female human pluripotent stem cells rapidly lose X chromosome inactivation marks and progress to a skewed methylation pattern during culture. Mol Hum Reprod 22, 285-298 https://doi.org/10.1093/molehr/gaw004
  88. Becker KA, Ghule PN, Therrien JA et al (2006) Self-renewal of human embryonic stem cells is supported by a shortened G1 cell cycle phase. J Cell Physiol 209, 883-893 https://doi.org/10.1002/jcp.20776
  89. Singh AM and Dalton S (2009) The cell cycle and myc intersect with mechanisms that regulate pluripotency and reprogramming. Cell Stem Cell 5, 141-149 https://doi.org/10.1016/j.stem.2009.07.003
  90. Calder A, Roth-Albin I, Bhatia S et al (2013) Lengthened G1 phase indicates differentiation status in human embryonic stem cells. Stem Cells Dev 22, 279-295 https://doi.org/10.1089/scd.2012.0168
  91. Sakaue-Sawano A, Kurokawa H, Morimura T et al (2008) Visualizing spatiotemporal dynamics of multicellular cell-cycle progression. Cell 132, 487-498 https://doi.org/10.1016/j.cell.2007.12.033
  92. Pauklin S and Vallier L (2013) The cell-cycle state of stem cells determines cell fate propensity. Cell 155, 135-147 https://doi.org/10.1016/j.cell.2013.08.031
  93. Kevin, Liang H, Lim YS et al (2015) Deterministic restriction on pluripotent state dissolution by cell-cycle pathways. Cell 162, 564-579 https://doi.org/10.1016/j.cell.2015.07.001
  94. Pauklin S, Madrigal P, Bertero A and Vallier L (2016) Initiation of stem cell differentiation involves cell cycle-dependent regulation of developmental genes by cyclin D. Genes Dev 30, 421-433 https://doi.org/10.1101/gad.271452.115
  95. Tahiliani M, Koh KP, Shen Y et al (2009) Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324, 930-935 https://doi.org/10.1126/science.1170116
  96. Amar, Chappell J, Trost R et al (2013) Cell-cycle control of developmentally regulated transcription factors accounts for heterogeneity in human pluripotent cells. Stem Cell Reports 1, 532-544 https://doi.org/10.1016/j.stemcr.2013.10.009
  97. Jang J, Han D, Golkaram M et al (2019) Control over single-cell distribution of G1 lengths by WNT governs pluripotency. PLoS Biol 17, e3000453 https://doi.org/10.1371/journal.pbio.3000453
  98. Rossant J and Tam PPL (2017) New insights into early human development: lessons for stem cell derivation and differentiation. Cell Stem Cell 20, 18-28 https://doi.org/10.1016/j.stem.2016.12.004
  99. Thomson JA (1998) Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1147 https://doi.org/10.1126/science.282.5391.1145
  100. Warmflash A, Sorre B, Etoc F, Siggia ED and Brivanlou AH (2014) A method to recapitulate early embryonic spatial patterning in human embryonic stem cells. Nat Methods 11, 847-854 https://doi.org/10.1038/nmeth.3016
  101. Peerani R, Rao BM, Bauwens C et al (2007) Niche-mediated control of human embryonic stem cell self-renewal and differentiation. EMBO Rep 26, 4744-4755 https://doi.org/10.1038/sj.emboj.7601896
  102. Etoc F, Metzger J, Ruzo A et al (2016) A Balance between secreted inhibitors and edge sensing controls gastruloid self-organization. Dev Cell 39, 302-315 https://doi.org/10.1016/j.devcel.2016.09.016
  103. Fleming TP et al (2001) Cell junctions and cell interactions in animal and human blastocyst development. ART and the Human Blastocyst. Springer, New York, NY, 2001. 91-102.
  104. Bedzhov I and Zernicka-Goetz M (2014) Self-organizing properties of mouse pluripotent cells initiate morphogenesis upon implantation. Cell 156, 1032-1044 https://doi.org/10.1016/j.cell.2014.01.023
  105. Kallas-Kivi A, Trei A, Stepanjuk A et al (2018) The role of integrin β1 in the heterogeneity of human embryonic stem cells culture. Biol 7, bio034355
  106. Xu C, Inokuma MS, Denham J et al (2001) Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol 19, 971-974 https://doi.org/10.1038/nbt1001-971
  107. Toh YC, Xing J and Yu H (2015) Modulation of integrin and E-cadherin-mediated adhesions to spatially control heterogeneity in human pluripotent stem cell differentiation. Biomaterials 50, 87-97 https://doi.org/10.1016/j.biomaterials.2015.01.019
  108. Shelley, Thornton M, Mason E, Jessica, Christine and Martin (2014) Single-cell gene expression profiles define self-renewing, pluripotent, and lineage primed states of human pluripotent stem cells. Stem Cell Reports 2, 881-895 https://doi.org/10.1016/j.stemcr.2014.04.014
  109. Nakanishi M, Mitchell RR, Benoit YD et al (2019) Human pluripotency is initiated and preserved by a unique subset of founder cells. Cell 177, 910-924.e922 https://doi.org/10.1016/j.cell.2019.03.013
  110. Lee J, Hyeon DY and Hwang D (2020) Single-cell multiomics: technologies and data analysis methods. Exp Mol Med 52, 1428-1442 https://doi.org/10.1038/s12276-020-0420-2