リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Inherent genomic properties underlie the epigenomic heterogeneity of human induced pluripotent stem cells」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Inherent genomic properties underlie the epigenomic heterogeneity of human induced pluripotent stem cells

Yokobayashi, Shihori Yabuta, Yukihiro Nakagawa, Masato Okita, Keisuke Hu, Bo Murase, Yusuke Nakamura, Tomonori Bourque, Guillaume Majewski, Jacek Yamamoto, Takuya Saitou, Mitinori 京都大学 DOI:10.1016/j.celrep.2021.109909

2021.11.02

概要

Human induced pluripotent stem cells (hiPSCs) show variable differentiation potential due to their epigenomic heterogeneity, whose extent/attributes remain unclear, except for well-studied elements/chromosomes such as imprints and the X chromosomes. Here, we show that seven hiPSC lines with variable germline potential exhibit substantial epigenomic heterogeneity, despite their uniform transcriptomes. Nearly a quarter of autosomal regions bear potentially differential chromatin modifications, with promoters/CpG islands for H3K27me3/H2AK119ub1 and evolutionarily young retrotransposons for H3K4me3. We identify 145 large autosomal blocks (≥100 kb) with differential H3K9me3 enrichment, many of which are lamina-associated domains (LADs) in somatic but not in embryonic stem cells. A majority of these epigenomic heterogeneities are independent of genetic variations. We identify an X chromosome state with chromosome-wide H3K9me3 that stably prevents X chromosome erosion. Importantly, the germline potential of female hiPSCs correlates with X chromosome inactivation. We propose that inherent genomic properties, including CpG density, transposons, and LADs, engender epigenomic heterogeneity in hiPSCs.

この論文で使われている画像

参考文献

Adewumi, O., Aflatoonian, B., Ahrlund-Richter, L., Amit, M., Andrews, P.W., Beighton, G., Bello, P.A., Benvenisty, N., Berry, L.S., Bevan, S., et al.; Interna- tional Stem Cell Initiative (2007). Characterization of human embryonic stem cell lines by the International Stem Cell Initiative. Nat. Biotechnol. 25, 803–816.

Balaton, B.P., Cotton, A.M., and Brown, C.J. (2015). Derivation of consensus inactivation status for X-linked genes from genome-wide studies. Biol. Sex Differ. 6, 35.

Bansal, P., Ahern, D.T., Kondaveeti, Y., Qiu, C.W., and Pinter, S.F. (2021). Contiguous erosion of the inactive X in human pluripotency concludes with global DNA hypomethylation. Cell Rep. 35, 109215.

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.

Bock, C., Kiskinis, E., Verstappen, G., Gu, H., Boulting, G., Smith, Z.D., Ziller, M., Croft, G.F., Amoroso, M.W., Oakley, D.H., et al. (2011). Reference Maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144, 439–452.

Boeva, V., Popova, T., Bleakley, K., Chiche, P., Cappo, J., Schleiermacher, G., Janoueix-Lerosey, I., Delattre, O., and Barillot, E. (2012). Control-FREEC: a tool for assessing copy number and allelic content using next-generation sequencing data. Bioinformatics 28, 423–425.

Borgel, J., Guibert, S., Li, Y., Chiba, H., Schu€beler, D., Sasaki, H., Forne´ , T., and Weber, M. (2010). Targets and dynamics of promoter DNA methylation during early mouse development. Nat. Genet. 42, 1093–1100.

Brenes, A.J., Yoshikawa, H., Bensaddek, D., Mirauta, B., Seaton, D., Hukel- mann, J.L., Jiang, H., Stegle, O., and Lamond, A.I. (2021). Erosion of human X chromosome inactivation causes major remodeling of the iPSC proteome. Cell Rep. 35, 109032.

Carcamo-Orive, I., Hoffman, G.E., Cundiff, P., Beckmann, N.D., D’Souza, S.L., Knowles, J.W., Patel, A., Papatsenko, D., Abbasi, F., Reaven, G.M., et al. (2017). Analysis of Transcriptional Variability in a Large Human iPSC Library Reveals Genetic and Non-genetic Determinants of Heterogeneity. Cell Stem Cell 20, 518–532.e9.

Chaligne´ , R., Popova, T., Mendoza-Parra, M.A., Saleem, M.A., Gentien, D., Ban, K., Piolot, T., Leroy, O., Mariani, O., Gronemeyer, H., et al. (2015). The inactive X chromosome is epigenetically unstable and transcriptionally labile in breast cancer. Genome Res. 25, 488–503.

Chen, D., Liu, W., Lukianchikov, A., Hancock, G.V., Zimmerman, J., Lowe, M.G., Kim, R., Galic, Z., Irie, N., Surani, M.A., et al. (2017). Germline compe- tency of human embryonic stem cells depends on eomesodermin. Biol. Re- prod. 97, 850–861.

Choi, J., Lee, S., Mallard, W., Clement, K., Tagliazucchi, G.M., Lim, H., Choi, I.Y., Ferrari, F., Tsankov, A.M., Pop, R., et al. (2015). A comparison of geneti- cally matched cell lines reveals the equivalence of human iPSCs and ESCs. Nat. Biotechnol. 33, 1173–1181.

Court, F., Tayama, C., Romanelli, V., Martin-Trujillo, A., Iglesias-Platas, I., Oka- mura, K., Sugahara, N., Simo´ n, C., Moore, H., Harness, J.V., et al. (2014). Genome-wide parent-of-origin DNA methylation analysis reveals the intri- cacies of human imprinting and suggests a germline methylation-independent mechanism of establishment. Genome Res. 24, 554–569.

Darrow, E.M., Huntley, M.H., Dudchenko, O., Stamenova, E.K., Durand, N.C., Sun, Z., Huang, S.C., Sanborn, A.L., Machol, I., Shamim, M., et al. (2016). Dele- tion of DXZ4 on the human inactive X chromosome alters higher-order genome architecture. Proc. Natl. Acad. Sci. USA 113, E4504–E4512.

DeBoever, C., Li, H., Jakubosky, D., Benaglio, P., Reyna, J., Olson, K.M., Huang, H., Biggs, W., Sandoval, E., D’Antonio, M., et al. (2017). Large-Scale Profiling Reveals the Influence of Genetic Variation on Gene Expression in Hu- man Induced Pluripotent Stem Cells. Cell Stem Cell 20, 533–546.e7.

Dekker, J., Belmont, A.S., Guttman, M., Leshyk, V.O., Lis, J.T., Lomvardas, S., Mirny, L.A., O’Shea, C.C., Park, P.J., Ren, B., et al.; 4D Nucleome Network (2017). The 4D nucleome project. Nature 549, 219–226.

Ernst, J., and Kellis, M. (2017). Chromatin-state discovery and genome anno- tation with ChromHMM. Nat. Protoc. 12, 2478–2492.

Fishilevich, S., Nudel, R., Rappaport, N., Hadar, R., Plaschkes, I., Iny Stein, T., Rosen, N., Kohn, A., Twik, M., Safran, M., et al. (2017). GeneHancer: genome- wide integration of enhancers and target genes in GeneCards. Database (Ox- ford) 2017, bax028.

Galupa, R., and Heard, E. (2018). X-Chromosome Inactivation: A Crossroads Between Chromosome Architecture and Gene Regulation. Annu. Rev. Genet. 52, 535–566.

Guenther, M.G., Frampton, G.M., Soldner, F., Hockemeyer, D., Mitalipova, M., Jaenisch, R., and Young, R.A. (2010). Chromatin structure and gene expres- sion programs of human embryonic and induced pluripotent stem cells. Cell Stem Cell 7, 249–257.

Hayashi-Takanaka, Y., Yamagata, K., Wakayama, T., Stasevich, T.J., Kai- numa, T., Tsurimoto, T., Tachibana, M., Shinkai, Y., Kurumizaka, H., Nozaki, N., and Kimura, H. (2011). Tracking epigenetic histone modifications in single cells using Fab-based live endogenous modification labeling. Nucleic Acids Res. 39, 6475–6488.

Heinz, S., Benner, C., Spann, N., Bertolino, E., Lin, Y.C., Laslo, P., Cheng, J.X., Murre, C., Singh, H., and Glass, C.K. (2010). Simple combinations of lineage- determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589.

Huang, W., Sherman, B.T., and Lempicki, R.A. (2009). Systematic and integra- tive analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57.

Kajiwara, M., Aoi, T., Okita, K., Takahashi, R., Inoue, H., Takayama, N., Endo, H., Eto, K., Toguchida, J., Uemoto, S., and Yamanaka, S. (2012). Donor- dependent variations in hepatic differentiation from human-induced pluripo- tent stem cells. Proc. Natl. Acad. Sci. USA 109, 12538–12543.

Kim, D., Pertea, G., Trapnell, C., Pimentel, H., Kelley, R., and Salzberg, S.L. (2013). TopHat2: accurate alignment of transcriptomes in the presence of in- sertions, deletions and gene fusions. Genome Biol. 14, R36.

Kimura, H., Hayashi-Takanaka, Y., Goto, Y., Takizawa, N., and Nozaki, N. (2008). The organization of histone H3 modifications as revealed by a panel of specific monoclonal antibodies. Cell Struct. Funct. 33, 61–73.

Kojima, Y., Sasaki, K., Yokobayashi, S., Sakai, Y., Nakamura, T., Yabuta, Y., Nakaki, F., Nagaoka, S., Woltjen, K., Hotta, A., et al. (2017). Evolutionarily Distinctive Transcriptional and Signaling Programs Drive Human Germ Cell Lineage Specification from Pluripotent Stem Cells. Cell Stem Cell 21, 517– 532.e5.

Koyanagi-Aoi, M., Ohnuki, M., Takahashi, K., Okita, K., Noma, H., Sawamura, Y., Teramoto, I., Narita, M., Sato, Y., Ichisaka, T., et al. (2013). Differentiation- defective phenotypes revealed by large-scale analyses of human pluripotent stem cells. Proc. Natl. Acad. Sci. USA 110, 20569–20574.

Krueger, F., and Andrews, S.R. (2011). Bismark: a flexible aligner and methyl- ation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572.

Langmead, B., and Salzberg, S.L. (2012). Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359.

Larson, A.G., Elnatan, D., Keenen, M.M., Trnka, M.J., Johnston, J.B., Burlin- game, A.L., Agard, D.A., Redding, S., and Narlikar, G.J. (2017). Liquid droplet formation by HP1a suggests a role for phase separation in heterochromatin. Nature 547, 236–240.

Lawrence, M., Huber, W., Page` s, H., Aboyoun, P., Carlson, M., Gentleman, R., Morgan, M.T., and Carey, V.J. (2013). Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118.

Lee, T.I., Johnstone, S.E., and Young, R.A. (2006). Chromatin immunoprecip- itation and microarray-based analysis of protein location. Nat. Protoc. 1, 729–748.

Li, H., and Durbin, R. (2010). Fast and accurate long-read alignment with Bur- rows-Wheeler transform. Bioinformatics 26, 589–595.

Li, H., Handsaker, B., Wysoker, A., Fennell, T., Ruan, J., Homer, N., Marth, G., Abecasis, G., and Durbin, R.; 1000 Genome Project Data Processing Sub- group (2009). The Sequence Alignment/Map format and SAMtools. Bioinfor- matics 25, 2078–2079.

Lister, R., Pelizzola, M., Kida, Y.S., Hawkins, R.D., Nery, J.R., Hon, G., Antosie- wicz-Bourget, J., O’Malley, R., Castanon, R., Klugman, S., et al. (2011). Hot- spots of aberrant epigenomic reprogramming in human induced pluripotent stem cells. Nature 471, 68–73.

Ma, H., Morey, R., O’Neil, R.C., He, Y., Daughtry, B., Schultz, M.D., Hariharan, M., Nery, J.R., Castanon, R., Sabatini, K., et al. (2014). Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature 511, 177–183.

Mekhoubad, S., Bock, C., de Boer, A.S., Kiskinis, E., Meissner, A., and Eggan, K. (2012). Erosion of dosage compensation impacts human iPSC disease modeling. Cell Stem Cell 10, 595–609.

Milagre, I., Stubbs, T.M., King, M.R., Spindel, J., Santos, F., Krueger, F., Bach- man, M., Segonds-Pichon, A., Balasubramanian, S., Andrews, S.R., et al. (2017). Gender Differences in Global but Not Targeted Demethylation in iPSC Reprogramming. Cell Rep. 18, 1079–1089.

Miura, F., Enomoto, Y., Dairiki, R., and Ito, T. (2012). Amplification-free whole- genome bisulfite sequencing by post-bisulfite adaptor tagging. Nucleic Acids Res. 40, e136.

Nakagawa, M., Taniguchi, Y., Senda, S., Takizawa, N., Ichisaka, T., Asano, K., Morizane, A., Doi, D., Takahashi, J., Nishizawa, M., et al. (2014). A novel effi- cient feeder-free culture system for the derivation of human induced pluripo- tent stem cells. Sci. Rep. 4, 3594.

Nazor, K.L., Altun, G., Lynch, C., Tran, H., Harness, J.V., Slavin, I., Garitaonan- dia, I., Mu€ller, F.J., Wang, Y.C., Boscolo, F.S., et al. (2012). Recurrent varia- tions in DNA methylation in human pluripotent stem cells and their differenti- ated derivatives. Cell Stem Cell 10, 620–634.

Nishizawa, M., Chonabayashi, K., Nomura, M., Tanaka, A., Nakamura, M., In- agaki, A., Nishikawa, M., Takei, I., Oishi, A., Tanabe, K., et al. (2016). Epigenetic Variation between Human Induced Pluripotent Stem Cell Lines Is an Indicator of Differentiation Capacity. Cell Stem Cell 19, 341–354.

Nozawa, R.S., Nagao, K., Igami, K.T., Shibata, S., Shirai, N., Nozaki, N., Sado, T., Kimura, H., and Obuse, C. (2013). Human inactive X chromosome is com- pacted through a PRC2-independent SMCHD1-HBiX1 pathway. Nat. Struct. Mol. Biol. 20, 566–573.

Ohi, Y., Qin, H., Hong, C., Blouin, L., Polo, J.M., Guo, T., Qi, Z., Downey, S.L., Manos, P.D., Rossi, D.J., et al. (2011). Incomplete DNA methylation underlies a transcriptional memory of somatic cells in human iPS cells. Nat. Cell Biol. 13, 541–549.

Okae, H., Chiba, H., Hiura, H., Hamada, H., Sato, A., Utsunomiya, T., Kikuchi, H., Yoshida, H., Tanaka, A., Suyama, M., and Arima, T. (2014). Genome-wide analysis of DNA methylation dynamics during early human development. PLoS Genet. 10, e1004868.

Okita, K., Yamakawa, T., Matsumura, Y., Sato, Y., Amano, N., Watanabe, A., Goshima, N., and Yamanaka, S. (2013). An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells 31, 458–466.

Ortmann, D., Brown, S., Czechanski, A., Aydin, S., Muraro, D., Huang, Y., Tomaz, R.A., Osnato, A., Canu, G., Wesley, B.T., et al. (2020). Naive Pluripotent Stem Cells Exhibit Phenotypic Variability that Is Driven by Genetic Variation. Cell Stem Cell 27, 470–481.e6.

Pasque, V., Karnik, R., Chronis, C., Petrella, P., Langerman, J., Bonora, G., Song, J., Vanheer, L., Sadhu Dimashkie, A., Meissner, A., and Plath, K. (2018). X Chromosome Dosage Influences DNA Methylation Dynamics during Reprogramming to Mouse iPSCs. Stem Cell Reports 10, 1537–1550.

Patel, S., Bonora, G., Sahakyan, A., Kim, R., Chronis, C., Langerman, J., Fitz- Gibbon, S., Rubbi, L., Skelton, R.J.P., Ardehali, R., et al. (2017). Human Embry- onic Stem Cells Do Not Change Their X Inactivation Status during Differentia- tion. Cell Rep. 18, 54–67.

Patrat, C., Ouimette, J.F., and Rougeulle, C. (2020). X chromosome inactiva- tion in human development. Development 147, dev183095.

Quinlan, A.R., and Hall, I.M. (2010). BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842.

Ramı´rez, F., Ryan, D.P., Gru€ning, B., Bhardwaj, V., Kilpert, F., Richter, A.S., Heyne, S., Du€ndar, F., and Manke, T. (2016). deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165.

Rao, S.S., Huntley, M.H., Durand, N.C., Stamenova, E.K., Bochkov, I.D., Rob-

inson, J.T., Sanborn, A.L., Machol, I., Omer, A.D., Lander, E.S., and Aiden, E.L. (2014). A 3D map of the human genome at kilobase resolution reveals princi- ples of chromatin looping. Cell 159, 1665–1680.

Robinson, J.T., Thorvaldsdo´ ttir, H., Winckler, W., Guttman, M., Lander, E.S., Getz, G., and Mesirov, J.P. (2011). Integrative genomics viewer. Nat. Bio- technol. 29, 24–26.

Roost, M.S., Slieker, R.C., Bialecka, M., van Iperen, L., Gomes Fernandes, M.M., He, N., Suchiman, H.E.D., Szuhai, K., Carlotti, F., de Koning, E.J.P., et al. (2017). DNA methylation and transcriptional trajectories during human development and reprogramming of isogenic pluripotent stem cells. Nat. Commun. 8, 908.

Ruiz, S., Diep, D., Gore, A., Panopoulos, A.D., Montserrat, N., Plongthongkum, N., Kumar, S., Fung, H.L., Giorgetti, A., Bilic, J., et al. (2012). Identification of a specific reprogramming-associated epigenetic signature in human induced pluripotent stem cells. Proc. Natl. Acad. Sci. USA 109, 16196–16201.

Salomonis, N., Dexheimer, P.J., Omberg, L., Schroll, R., Bush, S., Huo, J., Schriml, L., Ho Sui, S., Keddache, M., Mayhew, C., et al. (2016). Integrated Genomic Analysis of Diverse Induced Pluripotent Stem Cells from the Progen- itor Cell Biology Consortium. Stem Cell Reports 7, 110–125.

Sasaki, K., Yokobayashi, S., Nakamura, T., Okamoto, I., Yabuta, Y., Kurimoto, K., Ohta, H., Moritoki, Y., Iwatani, C., Tsuchiya, H., et al. (2015). Robust In Vitro Induction of Human Germ Cell Fate from Pluripotent Stem Cells. Cell Stem Cell 17, 178–194.

Schulz, E.G., Meisig, J., Nakamura, T., Okamoto, I., Sieber, A., Picard, C., Bor- ensztein, M., Saitou, M., Blu€thgen, N., and Heard, E. (2014). The two active X chromosomes in female ESCs block exit from the pluripotent state by modu- lating the ESC signaling network. Cell Stem Cell 14, 203–216.

Shirane, K., Kurimoto, K., Yabuta, Y., Yamaji, M., Satoh, J., Ito, S., Watanabe, A., Hayashi, K., Saitou, M., and Sasaki, H. (2016). Global Landscape and Reg- ulatory Principles of DNA Methylation Reprogramming for Germ Cell Specifi- cation by Mouse Pluripotent Stem Cells. Dev. Cell 39, 87–103.

Skelly, D.A., Czechanski, A., Byers, C., Aydin, S., Spruce, C., Olivier, C., Choi, K., Gatti, D.M., Raghupathy, N., Keele, G.R., et al. (2020). Mapping the Effects of Genetic Variation on Chromatin State and Gene Expression Reveals Loci That Control Ground State Pluripotency. Cell Stem Cell 27, 459–469.e8.

Sloan, C.A., Chan, E.T., Davidson, J.M., Malladi, V.S., Strattan, J.S., Hitz, B.C., Gabdank, I., Narayanan, A.K., Ho, M., Lee, B.T., et al. (2016). ENCODE data at the ENCODE portal. Nucleic Acids Res. 44, D726–D732.

Song, J., Janiszewski, A., De Geest, N., Vanheer, L., Talon, I., El Bakkali, M., Oh, T., and Pasque, V. (2019). X-Chromosome Dosage Modulates Multiple Molecular and Cellular Properties of Mouse Pluripotent Stem Cells Indepen- dently of Global DNA Methylation Levels. Stem Cell Reports 12, 333–350.

Storer, J., Hubley, R., Rosen, J., Wheeler, T.J., and Smit, A.F. (2021). The Dfam community resource of transposable element families, sequence models, and genome annotations. Mob. DNA 12, 2.

Stovner, E.B., and Sætrom, P. (2019). epic2 efficiently finds diffuse domains in ChIP-seq data. Bioinformatics 35, 4392–4393.

Strom, A.R., Emelyanov, A.V., Mir, M., Fyodorov, D.V., Darzacq, X., and Karpen, G.H. (2017). Phase separation drives heterochromatin domain forma- tion. Nature 547, 241–245.

Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K., and Yamanaka, S. (2007). Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872.

Terranova, R., Yokobayashi, S., Stadler, M.B., Otte, A.P., van Lohuizen, M., Orkin, S.H., and Peters, A.H. (2008). Polycomb group proteins Ezh2 and Rnf2 direct genomic contraction and imprinted repression in early mouse em- bryos. Dev. Cell 15, 668–679.

Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A., Swiergiel, J.J., Marshall, V.S., and Jones, J.M. (1998). Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147.

Tomoda, K., Takahashi, K., Leung, K., Okada, A., Narita, M., Yamada, N.A., Ei- lertson, K.E., Tsang, P., Baba, S., White, M.P., et al. (2012). Derivation condi- tions impact X-inactivation status in female human induced pluripotent stem cells. Cell Stem Cell 11, 91–99.

Trapnell, C., Williams, B.A., Pertea, G., Mortazavi, A., Kwan, G., van Baren, M.J., Salzberg, S.L., Wold, B.J., and Pachter, L. (2010). Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515.

Tsankov, A.M., Gu, H., Akopian, V., Ziller, M.J., Donaghey, J., Amit, I., Gnirke, A., and Meissner, A. (2015). Transcription factor binding dynamics during hu- man ES cell differentiation. Nature 518, 344–349.

Vallot, C., Ouimette, J.F., Makhlouf, M., Fe´ raud, O., Pontis, J., Co^me, J., Mar- tinat, C., Bennaceur-Griscelli, A., Lalande, M., and Rougeulle, C. (2015). Erosion of X Chromosome Inactivation in Human Pluripotent Cells Initiates with XACT Coating and Depends on a Specific Heterochromatin Landscape. Cell Stem Cell 16, 533–546.

van Arensbergen, J., FitzPatrick, V.D., de Haas, M., Pagie, L., Sluimer, J., Bus- semaker, H.J., and van Steensel, B. (2017). Genome-wide mapping of auton- omous promoter activity in human cells. Nat. Biotechnol. 35, 145–153.

van Schaik, T., Vos, M., Peric-Hupkes, D., Hn Celie, P., and van Steensel, B. (2020). Cell cycle dynamics of lamina-associated DNA. EMBO Rep. 21, e50636.

Voigt, P., Tee, W.W., and Reinberg, D. (2013). A double take on bivalent pro- moters. Genes Dev. 27, 1318–1338.

Yamanaka, S. (2020). Pluripotent Stem Cell-Based Cell Therapy-Promise and Challenges. Cell Stem Cell 27, 523–531.

Yamashiro, C., Sasaki, K., Yabuta, Y., Kojima, Y., Nakamura, T., Okamoto, I., Yokobayashi, S., Murase, Y., Ishikura, Y., Shirane, K., et al. (2018). Generation of human oogonia from induced pluripotent stem cells in vitro. Science 362, 356–360.

Yin, T., Cook, D., and Lawrence, M. (2012). ggbio: an R package for extending the grammar of graphics for genomic data. Genome Biol. 13, R77.

Yokobayashi, S., and Saitou, M. (2018). Reconstitution of germ cell develop- ment in vitro. In Cell Biology of the Ovary (Springer).

Yokobayashi, S., Okita, K., Nakagawa, M., Nakamura, T., Yabuta, Y., Yama- moto, T., and Saitou, M. (2017). Clonal variation of human induced pluripotent stem cells for induction into the germ cell fate. Biol. Reprod. 96, 1154–1166.

Zhang, Y., Liu, T., Meyer, C.A., Eeckhoute, J., Johnson, D.S., Bernstein, B.E.,

Nusbaum, C., Myers, R.M., Brown, M., Li, W., and Liu, X.S. (2008). Model- based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137.

Zhang, Y., Li, T., Preissl, S., Amaral, M.L., Grinstein, J.D., Farah, E.N., Destici, E., Qiu, Y., Hu, R., Lee, A.Y., et al. (2019). Transcriptionally active HERV-H ret- rotransposons demarcate topologically associating domains in human plurip- otent stem cells. Nat. Genet. 51, 1380–1388.

Zhu, L., Gomez-Duran, A., Saretzki, G., Jin, S., Tilgner, K., Melguizo-Sanchis, D., Anyfantis, G., Al-Aama, J., Vallier, L., Chinnery, P., et al. (2016). The mito- chondrial protein CHCHD2 primes the differentiation potential of human induced pluripotent stem cells to neuroectodermal lineages. J. Cell Biol. 215, 187–202.

参考文献をもっと見る