دورية أكاديمية

Single-cell multi-omics sequencing of human spermatogenesis reveals a DNA demethylation event associated with male meiotic recombination.

التفاصيل البيبلوغرافية
العنوان: Single-cell multi-omics sequencing of human spermatogenesis reveals a DNA demethylation event associated with male meiotic recombination.
المؤلفون: Huang Y; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Li L; Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., An G; Department of Reproductive Medicine Center, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China., Yang X; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Cui M; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Song X; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Lin J; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Zhang X; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Yao Z; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Wan C; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Zhou C; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Zhao J; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Song K; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Ren S; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Xia X; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Fu X; Department of Reproductive Medicine Center, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China., Lan Y; Department of Reproductive Medicine Center, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, P. R. China., Hu X; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Wang W; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Wang M; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Zheng Y; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Miao K; Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau, P. R. China., Bai X; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China., Hutchins AP; Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P. R. China., Chang G; Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, P. R. China. changgang@szu.edu.cn., Gao S; State Key Laboratory of Animal Biotech Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction of the MARA, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China. gaoshuai1959@163.com., Zhao XY; State Key Laboratory of Organ Failure Research, Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China. zhaoxiaoyang@smu.edu.cn.; Guangdong Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, P. R. China. zhaoxiaoyang@smu.edu.cn.; Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, P. R. China. zhaoxiaoyang@smu.edu.cn.; Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, P. R. China. zhaoxiaoyang@smu.edu.cn.; National Clinical Research Center for Kidney Disease, Guangzhou, P. R. China. zhaoxiaoyang@smu.edu.cn.
المصدر: Nature cell biology [Nat Cell Biol] 2023 Oct; Vol. 25 (10), pp. 1520-1534. Date of Electronic Publication: 2023 Sep 18.
نوع المنشور: Journal Article
اللغة: English
بيانات الدورية: Publisher: Macmillan Magazines Ltd Country of Publication: England NLM ID: 100890575 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1476-4679 (Electronic) Linking ISSN: 14657392 NLM ISO Abbreviation: Nat Cell Biol Subsets: MEDLINE
أسماء مطبوعة: Original Publication: London : Macmillan Magazines Ltd., [1999-
مواضيع طبية MeSH: DNA Demethylation* , Multiomics*, Humans ; Male ; Animals ; Mice ; Spermatogenesis/genetics ; Meiosis/genetics ; Chromatin/genetics
مستخلص: Human spermatogenesis is a highly ordered process; however, the roles of DNA methylation and chromatin accessibility in this process remain largely unknown. Here by simultaneously investigating the chromatin accessibility, DNA methylome and transcriptome landscapes using the modified single-cell chromatin overall omic-scale landscape sequencing approach, we revealed that the transcriptional changes throughout human spermatogenesis were correlated with chromatin accessibility changes. In particular, we identified a set of transcription factors and cis elements with potential functions. A round of DNA demethylation was uncovered upon meiosis initiation in human spermatogenesis, which was associated with male meiotic recombination and conserved between human and mouse. Aberrant DNA hypermethylation could be detected in leptotene spermatocytes of certain nonobstructive azoospermia patients. Functionally, the intervention of DNA demethylation affected male meiotic recombination and fertility. Our work provides multi-omics landscapes of human spermatogenesis at single-cell resolution and offers insights into the association between DNA demethylation and male meiotic recombination.
(© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
References: Handel, M. A. & Schimenti, J. C. Genetics of mammalian meiosis: regulation, dynamics and impact on fertility. Nat. Rev. Genet. 11, 124–136 (2010). (PMID: 20051984)
Godmann, M., Lambrot, R. & Kimmins, S. The dynamic epigenetic program in male germ cells: Its role in spermatogenesis, testis cancer, and its response to the environment. Microsc. Res. Tech. 72, 603–619 (2009). (PMID: 19319879)
Jan, S. Z. et al. Molecular control of rodent spermatogenesis. Biochim. Biophys. Acta 1822, 1838–1850 (2012). (PMID: 22366765)
Baudat, F., Imai, Y. & de Massy, B. Meiotic recombination in mammals: localization and regulation. Nat. Rev. Genet. 14, 794–806 (2013). (PMID: 24136506)
Baudat, F. et al. PRDM9 is a major determinant of meiotic recombination hotspots in humans and mice. Science 327, 836–840 (2010). (PMID: 20044539)
Parvanov, E. D., Petkov, P. M. & Paigen, K. Prdm9 controls activation of mammalian recombination hotspots. Science 327, 835 (2010). (PMID: 20044538)
Diagouraga, B. et al. PRDM9 methyltransferase activity is essential for meiotic DNA double-strand break formation at its binding sites. Mol. Cell 69, 853–865.e6 (2018). (PMID: 29478809)
Hermann, B. P. et al. The mammalian spermatogenesis single-cell transcriptome, from spermatogonial stem cells to spermatids. Cell Rep. 25, 1650–1667.e8 (2018). (PMID: 304040166384825)
Sohni, A. et al. The neonatal and adult human testis defined at the single-cell level. Cell Rep. 26, 1501–1517.e4 (2019). (PMID: 307267346402825)
Wang, M. et al. Single-cell RNA sequencing analysis reveals sequential cell fate transition during human spermatogenesis. Cell Stem Cell 23, 599–614.e4 (2018). (PMID: 30174296)
Dai, H. Q. et al. TET-mediated DNA demethylation controls gastrulation by regulating Lefty-Nodal signalling. Nature 538, 528–532 (2016). (PMID: 27760115)
Markenscoff-Papadimitriou, E. et al. A chromatin accessibility atlas of the developing human telencephalon. Cell 182, 754–769.e8 (2020). (PMID: 326100827415678)
Izzo, F. et al. DNA methylation disruption reshapes the hematopoietic differentiation landscape. Nat. Genet. 52, 378–387 (2020). (PMID: 322034687216752)
Klemm, S. L., Shipony, Z. & Greenleaf, W. J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20, 207–220 (2019). (PMID: 30675018)
Maezawa, S., Yukawa, M., Alavattam, K. G., Barski, A. & Namekawa, S. H. Dynamic reorganization of open chromatin underlies diverse transcriptomes during spermatogenesis. Nucleic Acids Res. 46, 593–608 (2018). (PMID: 29126117)
Gaysinskaya, V. et al. Transient reduction of DNA methylation at the onset of meiosis in male mice. Epigenetics Chromatin 11, 15 (2018). (PMID: 296183745883305)
Chen, Y. et al. Refined spatial temporal epigenomic profiling reveals intrinsic connection between PRDM9-mediated H3K4me3 and the fate of double-stranded breaks. Cell Res. 30, 256–268 (2020).
Guo, J. et al. Chromatin and single-cell RNA-seq profiling reveal dynamic signaling and metabolic transitions during human spermatogonial stem cell development. Cell Stem Cell 21, 533–546.e6 (2017). (PMID: 289855285832720)
Guo, J. et al. The adult human testis transcriptional cell atlas. Cell Res. 28, 1141–1157 (2018). (PMID: 303152786274646)
Guo, F. et al. Single-cell multi-omics sequencing of mouse early embryos and embryonic stem cells. Cell Res. 27, 967–988 (2017). (PMID: 286213295539349)
Clark, S. J. et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun. 9, 781 (2018). (PMID: 294726105823944)
Li, L. et al. Single-cell multi-omics sequencing of human early embryos. Nat. Cell Biol. 20, 847–858 (2018). (PMID: 29915357)
Pott, S. Simultaneous measurement of chromatin accessibility, DNA methylation, and nucleosome phasing in single cells. eLife 6, e23203 (2017). (PMID: 286536225487215)
Wang, J. et al. Single-cell multiomics sequencing reveals the reprogramming defects in embryos generated by round spermatid injection. Sci. Adv. 8, eabm3976 (2022). (PMID: 359476549365279)
Dong, J. et al. Single-cell RNA-seq analysis unveils a prevalent epithelial/mesenchymal hybrid state during mouse organogenesis. Genome Biol. 19, 31 (2018). (PMID: 295402035853091)
Kelly, T. K. et al. Genome-wide mapping of nucleosome positioning and DNA methylation within individual DNA molecules. Genome Res. 22, 2497–2506 (2012). (PMID: 229603753514679)
Li, L. et al. Single-cell RNA-seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell 20, 858–873.e4 (2017). (PMID: 28457750)
Guo, H. et al. DNA methylation and chromatin accessibility profiling of mouse and human fetal germ cells. Cell Res. 27, 165–183 (2017). (PMID: 27824029)
Ren, B. & Yue, F. Transcriptional enhancers: bridging the genome and phenome. Cold Spring Harb. Symp. Quant. Biol. 80, 17–26 (2015). (PMID: 26582789)
Tan, K. et al. Transcriptome profiling reveals signaling conditions dictating human spermatogonia fate in vitro. Proc. Natl Acad. Sci. USA 117, 17832–17841 (2020). (PMID: 326611787395452)
Tassinari, V. et al. Fgf9 inhibition of meiotic differentiation in spermatogonia is mediated by Erk-dependent activation of Nodal-Smad2/3 signaling and is antagonized by Kit Ligand. Cell Death Dis. 6, e1688 (2015). (PMID: 257663274385934)
Matson, C. K. et al. The mammalian doublesex homolog DMRT1 is a transcriptional gatekeeper that controls the mitosis versus meiosis decision in male germ cells. Dev. Cell 19, 612–624 (2010). (PMID: 209513512996490)
Zhang, T., Murphy, M. W., Gearhart, M. D., Bardwell, V. J. & Zarkower, D. The mammalian Doublesex homolog DMRT6 coordinates the transition between mitotic and meiotic developmental programs during spermatogenesis. Development 141, 3662–3671 (2014). (PMID: 252494584197572)
Kistler, W. S. et al. RFX2 is a major transcriptional regulator of spermiogenesis. PLoS Genet. 11, e1005368 (2015). (PMID: 261621024498915)
Hai, T. & Curran, T. Cross-family dimerization of transcription factors Fos/Jun and ATF/CREB alters DNA binding specificity. Proc. Natl Acad. Sci. USA 88, 3720–3724 (1991). (PMID: 182720351524)
De Cesare, D. & Sassone-Corsi, P. Transcriptional regulation by cyclic AMP-responsive factors. Prog. Nucleic Acid Res. Mol. Biol. 64, 343–369 (2000). (PMID: 10697414)
Smith, Z. D. & Meissner, A. DNA methylation: roles in mammalian development. Nat. Rev. Genet. 14, 204–220 (2013). (PMID: 23400093)
Kagiwada, S., Kurimoto, K., Hirota, T., Yamaji, M. & Saitou, M. Replication-coupled passive DNA demethylation for the erasure of genome imprints in mice. EMBO J. 32, 340–353 (2013). (PMID: 23241950)
Sharif, J. et al. The SRA protein Np95 mediates epigenetic inheritance by recruiting Dnmt1 to methylated DNA. Nature 450, 908–912 (2007). (PMID: 17994007)
Bostick, M. et al. UHRF1 plays a role in maintaining DNA methylation in mammalian cells. Science 317, 1760–1764 (2007). (PMID: 17673620)
Zhang, J. et al. S phase-dependent interaction with DNMT1 dictates the role of UHRF1 but not UHRF2 in DNA methylation maintenance. Cell Res. 21, 1723–1739 (2011). (PMID: 220647033357991)
Li, T. et al. Structural and mechanistic insights into UHRF1-mediated DNMT1 activation in the maintenance DNA methylation. Nucleic Acids Res. 46, 3218–3231 (2018). (PMID: 294713505887372)
Dong, J. et al. UHRF1 suppresses retrotransposons and cooperates with PRMT5 and PIWI proteins in male germ cells. Nat. Commun. 10, 4705 (2019). (PMID: 316242446797737)
Pan, H. et al. UHRF1-repressed 5’-hydroxymethylcytosine is essential for the male meiotic prophase I. Cell Death Dis. 11, 142 (2020). (PMID: 320818447035279)
Pratto, F. et al. DNA recombination. Recombination initiation maps of individual human genomes. Science 346, 1256442 (2014). (PMID: 253955425588152)
The 1000 Genomes Project Consortium A map of human genome variation from population-scale sequencing. Nature 467, 1061–1073 (2010). (PMID: 3042601)
Halldorsson, B. V. et al. Characterizing mutagenic effects of recombination through a sequence-level genetic map. Science 363, eaau1043 (2019). (PMID: 30679340)
Myers, S., Freeman, C., Auton, A., Donnelly, P. & McVean, G. A common sequence motif associated with recombination hot spots and genome instability in humans. Nat. Genet. 40, 1124–1129 (2008). (PMID: 19165926)
Arnheim, N., Calabrese, P. & Tiemann-Boege, I. Mammalian meiotic recombination hot spots. Annu. Rev. Genet 41, 369–399 (2007).
McLeay, R. C. & Bailey, T. L. Motif enrichment analysis: a unified framework and an evaluation on ChIP data. BMC Bioinf. 11, 165 (2010).
Dreau, A., Venu, V., Avdievich, E., Gaspar, L. & Jones, F. C. Genome-wide recombination map construction from single individuals using linked-read sequencing. Nat. Commun. 10, 4309 (2019). (PMID: 315410916754380)
Brick, K., Smagulova, F., Khil, P., Camerini-Otero, R. D. & Petukhova, G. V. Genetic recombination is directed away from functional genomic elements in mice. Nature 485, 642–645 (2012). (PMID: 226603273367396)
Lange, J. et al. The landscape of mouse meiotic double-strand break formation, processing, and repair. Cell 167, 695–708.e6 (2016). (PMID: 277459715117687)
Walker, M. et al. Affinity-seq detects genome-wide PRDM9 binding sites and reveals the impact of prior chromatin modifications on mammalian recombination hotspot usage. Epigenetics Chromatin 8, 31 (2015). (PMID: 263515204562113)
Jiao, Y. et al. A TOP6BL mutation abolishes meiotic DNA double-strand break formation and causes human infertility. Sci. Bull. 65, 2120–2129 (2020).
Yuan, Y. et al. Generation of fertile offspring from Kit w /Kit wv mice through differentiation of gene corrected nuclear transfer embryonic stem cells. Cell Res. 25, 851–863 (2015). (PMID: 260884174493281)
Crespo, M. et al. Multi-omic analysis of gametogenesis reveals a novel signature at the promoters and distal enhancers of active genes. Nucleic Acids Res. 48, 4115–4138 (2020). (PMID: 321823407192594)
Saitou, M., Kagiwada, S. & Kurimoto, K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development 139, 15–31 (2012). (PMID: 22147951)
Melamed-Bessudo, C. & Levy, A. A. Deficiency in DNA methylation increases meiotic crossover rates in euchromatic but not in heterochromatic regions in Arabidopsis. Proc. Natl Acad. Sci. USA 109, E981–E988 (2012). (PMID: 224607913341010)
Zamudio, N. et al. DNA methylation restrains transposons from adopting a chromatin signature permissive for meiotic recombination. Genes Dev. 29, 1256–1270 (2015). (PMID: 261090494495397)
Gonzalo, S. et al. DNA methyltransferases control telomere length and telomere recombination in mammalian cells. Nat. Cell Biol. 8, 416–424 (2006). (PMID: 16565708)
Liu, Y., Sarkar, A., Kheradpour, P., Ernst, J. & Kellis, M. Evidence of reduced recombination rate in human regulatory domains. Genome Biol. 18, 193 (2017). (PMID: 290585995651596)
Altemose, N. et al. A map of human PRDM9 binding provides evidence for novel behaviors of PRDM9 and other zinc-finger proteins in meiosis. eLife 6, e28383 (2017). (PMID: 290725755705219)
Wang, M. et al. Deciphering the autophagy regulatory network via single-cell transcriptome analysis reveals a requirement for autophagy homeostasis in spermatogenesis. Theranostics 11, 5010–5027 (2021). (PMID: 337540417978313)
Sadate-Ngatchou, P. I., Payne, C. J., Dearth, A. T. & Braun, R. E. Cre recombinase activity specific to postnatal, premeiotic male germ cells in transgenic mice. Genesis 46, 738–742 (2008). (PMID: 188505942837914)
Zhao, J. et al. Cell-fate transition and determination analysis of mouse male germ cells throughout development. Nat. Commun. 12, 6839 (2021). (PMID: 348242378617176)
Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019). (PMID: 313758077605509)
Anders, S., Pyl, P. T. & Huber, W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015). (PMID: 25260700)
Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011). (PMID: 214936563102221)
Eichten, S. R., Stuart, T., Srivastava, A., Lister, R. & Borevitz, J. O. DNA methylation profiles of diverse Brachypodium distachyon align with underlying genetic diversity. Genome Res. 26, 1520–1531 (2016). (PMID: 276136115088594)
Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009). (PMID: 195059432723002)
Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019). (PMID: 311781186687398)
Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019). (PMID: 307874376434952)
Stacklies, W., Redestig, H., Scholz, M., Walther, D. & Selbig, J. pcaMethods–a bioconductor package providing PCA methods for incomplete data. Bioinformatics 23, 1164–1167 (2007). (PMID: 17344241)
McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).
Liu, Y., Siegmund, K. D., Laird, P. W. & Berman, B. P. Bis-SNP: combined DNA methylation and SNP calling for Bisulfite-seq data. Genome Biol. 13, R61 (2012). (PMID: 227843813491382)
Danecek, P. et al. The variant call format and VCFtools. Bioinformatics 27, 2156–2158 (2011). (PMID: 216535223137218)
Bansal, V. & Bafna, V. HapCUT: an efficient and accurate algorithm for the haplotype assembly problem. Bioinformatics 24, i153–i159 (2008). (PMID: 18689818)
Van der Auwera, G. A. et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinforma. 43, 11.10.11–11.10.33 (2013).
Gaysinskaya, V., Soh, I. Y., van der Heijden, G. W. & Bortvin, A. Optimized flow cytometry isolation of murine spermatocytes. Cytom. A 85, 556–565 (2014).
Lima, A. C. et al. A standardized approach for multispecies purification of mammalian male germ cells by mechanical tissue dissociation and flow cytometry. J. Vis. Exp. https://doi.org/10.3791/55913 (2017).
Chen, Y. et al. Single-cell RNA-seq uncovers dynamic processes and critical regulators in mouse spermatogenesis. Cell Res. 28, 879–896 (2018). (PMID: 300617426123400)
Hogarth, C. A. et al. Turning a spermatogenic wave into a tsunami: synchronizing murine spermatogenesis using WIN 18,446. Biol Reprod. 88, 40 (2013).
Bilichak, A. & Kovalchuk, I. The combined bisulfite restriction analysis (COBRA) assay for the analysis of locus-specific changes in methylation patterns. Methods Mol Biol. 1456, 63–71 (2017).
Yao, X. et al. Homology-mediated end joining-based targeted integration using CRISPR/Cas9. Cell Res. 27, 801–814 (2017).
Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013). (PMID: 241575483969860)
Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014). (PMID: 250427864117646)
Wang, X. et al. The chromatin accessibility landscape reveals distinct transcriptional regulation in the induction of human primordial germ cell-like cells from pluripotent stem cells. Stem Cell Rep. 16, 1245–1261 (2021).
Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014). (PMID: 246954044103590)
Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012). (PMID: 223882863322381)
Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016). (PMID: 270799754987876)
Acquaviva, L. et al. Ensuring meiotic DNA break formation in the mouse pseudoautosomal region. Nature 582, 426–431 (2020). (PMID: 324616907337327)
Li, M. et al. The histone modification reader ZCWPW1 is required for meiosis prophase I in male but not in female mice. Sci. Adv. 5, eaax1101 (2019). (PMID: 314533356693912)
Wang, Y. et al. Single-cell multiomics sequencing reveals the functional regulatory landscape of early embryos. Nat. Commun. 12, 1247 (2021). (PMID: 336230217902657)
Yan, R. et al. Decoding dynamic epigenetic landscapes in human oocytes using single-cell multi-omics sequencing. Cell Stem Cell 28, 1641–1656 (2021).
Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).
المشرفين على المادة: 0 (Chromatin)
تواريخ الأحداث: Date Created: 20230918 Date Completed: 20231221 Latest Revision: 20231221
رمز التحديث: 20231221
DOI: 10.1038/s41556-023-01232-7
PMID: 37723297
قاعدة البيانات: MEDLINE
الوصف
تدمد:1476-4679
DOI:10.1038/s41556-023-01232-7