Search
2023 Volume 3
Article Contents
REVIEW   Open Access    

Application and prospects of single-cell and spatial omics technologies in woody plants

  • # These authors contributed equally: Shaoming Liang, Yiling Li

More Information
  • Over the past decade, high-throughput sequencing and high-resolution single-cell transcriptome sequencing technologies have undergone rapid development, leading to significant breakthroughs. Traditional molecular biology methods are limited in their ability to unravel cellular-level heterogeneity within woody plant tissues. Consequently, techniques such as single-cell transcriptomics, single-cell epigenetics, and spatial transcriptomics are rapidly gaining popularity in the study of woody plants. In this review, we provide a comprehensive overview of the development of these technologies, with a focus on their applications and the challenges they present in single-cell transcriptome research in woody plants. In particular, we delve into the similarities and differences among the results of current studies and analyze the reasons behind these differences. Furthermore, we put forth potential solutions to overcome the challenges encountered in single-cell transcriptome applications in woody plants. Finally, we discuss the application directions of these techniques to address key challenges in woody plant research in the future.
  • 加载中
  • Supplemental Table S1 Summary of the marker genes used in poplar single-cell studies. Genes highlighted in yellow indicate that they were used to identify a specific cell type in certain study, showing the relative orthologous gene in other studies.
    Supplemental Table S2 Summary of the marker genes used in single-cell studies of woody plants. Genes highlighted in yellow indicate that they were used to identify a specific cell type in a study, showing the best hits in Arabidopsis.
  • [1]

    Schaum N, Karkanias J, Neff NF, May AP, Quake SR, et al. 2018. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562:367−72

    doi: 10.1038/s41586-018-0590-4

    CrossRef   Google Scholar

    [2]

    Han L, Wei X, Liu C, Volpe G, Zhuang Z, et al. 2022. Cell transcriptomic atlas of the non-human primate Macaca fascicularis. Nature 604:723−31

    doi: 10.1038/s41586-022-04587-3

    CrossRef   Google Scholar

    [3]

    Li H, Janssens J, De Waegeneer M, Kolluru SS, Davie K, et al. 2022. Fly Cell Atlas: a single-nucleus transcriptomic atlas of the adult fruit fly. Science 375:eabk2432

    doi: 10.1126/science.abk2432

    CrossRef   Google Scholar

    [4]

    Wei H. 2021. Inaugural editorial. Forestry Research 1:1

    doi: 10.48130/fr-2021-0001

    CrossRef   Google Scholar

    [5]

    Li H, Yin S, Wang L, Xu N, Liu L. 2022. Transcription factor PagLBD21 functions as a repressor of secondary xylem development in Populus. Forestry Research 2:19

    doi: 10.48130/fr-2022-0019

    CrossRef   Google Scholar

    [6]

    Takata N, Awano T, Nakata MT, Sano Y, Sakamoto S, et al. 2019. Populus NST/SND orthologs are key regulators of secondary cell wall formation in wood fibers, phloem fibers and xylem ray parenchyma cells. Tree Physiology 39:514−25

    doi: 10.1093/treephys/tpz004

    CrossRef   Google Scholar

    [7]

    Tang X, Wang C, Chai G, Wang D, Xu H, et al. 2022. Ubiquitinated DA1 negatively regulates vascular cambium activity through modulating the stability of WOX4 in Populus. The Plant Cell 34:3364−82

    doi: 10.1093/plcell/koac178

    CrossRef   Google Scholar

    [8]

    Hu J, Su H, Cao H, Wei H, Fu X, et al. 2022. AUXIN RESPONSE FACTOR7 integrates gibberellin and auxin signaling via interactions between DELLA and AUX/IAA proteins to regulate cambial activity in poplar. The Plant Cell 34:2688−707

    doi: 10.1093/plcell/koac107

    CrossRef   Google Scholar

    [9]

    Dai X, Zhai R, Lin J, Wang Z, Meng D, et al. 2023. Cell-type-specific PtrWOX4a and PtrVCS2 form a regulatory nexus with a histone modification system for stem cambium development in Populus trichocarpa. Nature Plants 9:96−111

    doi: 10.1038/s41477-022-01315-7

    CrossRef   Google Scholar

    [10]

    Tong S, Wang Y, Chen N, Wang D, Liu B, et al. 2022. PtoNF-YC9-SRMT-PtoRD26 module regulates the high saline tolerance of a triploid poplar. Genome Biology 23:148

    doi: 10.1186/s13059-022-02718-7

    CrossRef   Google Scholar

    [11]

    Jiang Y, Tong S, Chen N, Liu B, Bai Q, et al. 2021. The PalWRKY77 transcription factor negatively regulates salt tolerance and abscisic acid signaling in Populus. The Plant Journal 105:1258−73

    doi: 10.1111/tpj.15109

    CrossRef   Google Scholar

    [12]

    Kong L, Song Q, Wei H, Wang Y, Lin M, et al. 2023. The AP2/ERF transcription factor PtoERF15 confers drought tolerance via JA-mediated signaling in Populus. New Phytologist 240:1848−67

    doi: 10.1111/nph.19251

    CrossRef   Google Scholar

    [13]

    Tong S, Chen N, Wang D, Ai F, Liu B, et al. 2021. The U-box E3 ubiquitin ligase PalPUB79 positively regulates ABA-dependent drought tolerance via ubiquitination of PalWRKY77 in Populus. Plant Biotechnology Journal 19:2561−75

    doi: 10.1111/pbi.13681

    CrossRef   Google Scholar

    [14]

    Tylewicz S, Petterle A, Marttila S, Miskolczi P, Azeez A, et al. 2018. Photoperiodic control of seasonal growth is mediated by ABA acting on cell-cell communication. Science 360:212−15

    doi: 10.1126/science.aan8576

    CrossRef   Google Scholar

    [15]

    Azeez A, Zhao YC, Singh RK, Yordanov YS, Dash M, et al. 2021. EARLY BUD-BREAK 1 and EARLY BUD-BREAK 3 control resumption of poplar growth after winter dormancy. Nature Communications 12:1123

    doi: 10.1038/s41467-021-21449-0

    CrossRef   Google Scholar

    [16]

    Singh RK, Svystun T, Aldahmash B, Jönsson AM, Bhalerao RP. 2017. Photoperiod- and temperature-mediated control of phenology in trees – a molecular perspective. New Phytologist 213:511−24

    doi: 10.1111/nph.14346

    CrossRef   Google Scholar

    [17]

    Ding J, Böhlenius H, Rühl MG, Chen P, Sane S, et al. 2018. GIGANTEA-like genes control seasonal growth cessation in Populus. New Phytologist 218:1491−503

    doi: 10.1111/nph.15087

    CrossRef   Google Scholar

    [18]

    Li Y, Wang D, Wang W, Yang W, Gao J, et al. 2023. A chromosome-level Populus qiongdaoensis genome assembly provides insights into tropical adaptation and a cryptic turnover of sex determination. Molecular Ecology 32:1366−80

    doi: 10.1111/mec.16566

    CrossRef   Google Scholar

    [19]

    Yang W, Wang D, Li Y, Zhang Z, Tong S, et al. 2021. A general model to explain repeated turnovers of sex determination in the Salicaceae. Molecular Biology and Evolution 38:968−80

    doi: 10.1093/molbev/msaa261

    CrossRef   Google Scholar

    [20]

    Xue L, Wu H, Chen Y, Li X, Hou J, et al. 2020. Evidences for a role of two Y-specific genes in sex determination in Populus deltoides. Nature Communications 11:5893

    doi: 10.1038/s41467-020-19559-2

    CrossRef   Google Scholar

    [21]

    Zhou R, Macaya-Sanz D, Carlson CH, Schmutz J, Jenkins JW, et al. 2020. A willow sex chromosome reveals convergent evolution of complex palindromic repeats. Genome Biology 21:38

    doi: 10.1186/s13059-020-1952-4

    CrossRef   Google Scholar

    [22]

    Efroni I, Mello A, Nawy T, Ip PL, Rahni R, et al. 2016. Root regeneration triggers an embryo-like sequence guided by hormonal interactions. Cell 165:1721−33

    doi: 10.1016/j.cell.2016.04.046

    CrossRef   Google Scholar

    [23]

    Denyer T, Ma X, Klesen S, Scacchi E, Nieselt K, et al. 2019. Spatiotemporal developmental trajectories in the Arabidopsis root revealed using high-throughput single-cell RNA sequencing. Developmental Cell 48:840−52

    doi: 10.1016/j.devcel.2019.02.022

    CrossRef   Google Scholar

    [24]

    Jean-Baptiste K, Mcfaline-Figueroa JL, Alexandre CM, Dorrity MW, Saunders L, et al. 2019. Dynamics of gene expression in single root cells of Arabidopsis thaliana. The Plant Cell 31:993−1011

    doi: 10.1105/tpc.18.00785

    CrossRef   Google Scholar

    [25]

    Ryu KH, Huang L, Kang HM, Schiefelbein J. 2019. Single-cell RNA sequencing resolves molecular relationships among individual plant cells. Plant Physiology 179:1444−56

    doi: 10.1104/pp.18.01482

    CrossRef   Google Scholar

    [26]

    Zhang T, Xu Z, Shang G, Wang J. 2019. A single-cell RNA sequencing profiles the developmental landscape of Arabidopsis root. Molecular Plant 12:648−60

    doi: 10.1016/j.molp.2019.04.004

    CrossRef   Google Scholar

    [27]

    Turco GM, Rodriguez-Medina J, Siebert S, Han D, Valderrama-Gómez MÁ, et al. 2019. Molecular mechanisms driving switch behavior in xylem cell differentiation. Cell Reports 28:342−351.E4

    doi: 10.1016/j.celrep.2019.06.041

    CrossRef   Google Scholar

    [28]

    Liu Z, Zhou Y, Guo J, Li J, Tian Z, et al. 2020. Global dynamic molecular profiling of stomatal lineage cell development by single-cell RNA sequencing. Molecular Plant 13:1178−93

    doi: 10.1016/j.molp.2020.06.010

    CrossRef   Google Scholar

    [29]

    Zhang T, Chen Y, Wang J. 2021. A single-cell analysis of the Arabidopsis vegetative shoot apex. Developmental Cell 56:1056−1074.E8

    doi: 10.1016/j.devcel.2021.02.021

    CrossRef   Google Scholar

    [30]

    Zhai N, Xu L. 2021. Pluripotency acquisition in the middle cell layer of callus is required for organ regeneration. Nature Plants 7:1453−60

    doi: 10.1038/s41477-021-01015-8

    CrossRef   Google Scholar

    [31]

    Liu Z, Wang J, Zhou Y, Zhang Y, Qin A, et al. 2022. Identification of novel regulators required for early development of vein pattern in the cotyledons by single-cell RNA-sequencing. The Plant Journal 110:7−22

    doi: 10.1111/tpj.15719

    CrossRef   Google Scholar

    [32]

    Wang Y, Huan Q, Li K, Qian W. 2021. Single-cell transcriptome atlas of the leaf and root of rice seedlings. Journal of Genetics and Genomics 48:881−98

    doi: 10.1016/j.jgg.2021.06.001

    CrossRef   Google Scholar

    [33]

    Liu Q, Liang Z, Feng D, Jiang S, Wang Y, et al. 2021. Transcriptional landscape of rice roots at the single-cell resolution. Molecular Plant 14:384−94

    doi: 10.1016/j.molp.2020.12.014

    CrossRef   Google Scholar

    [34]

    Zong J, Wang L, Zhu L, Bian L, Zhang B, et al. 2022. A rice single cell transcriptomic atlas defines the developmental trajectories of rice floret and inflorescence meristems. New Phytologist 234:494−512

    doi: 10.1111/nph.18008

    CrossRef   Google Scholar

    [35]

    Ortiz-Ramírez C, Guillotin B, Xu X, Rahni R, Zhang S, et al. 2021. Ground tissue circuitry regulates organ complexity in maize and Setaria. Science 374:1247−52

    doi: 10.1126/science.abj2327

    CrossRef   Google Scholar

    [36]

    Xu X, Crow M, Rice BR, Li F, Harris B, et al. 2021. Single-cell RNA sequencing of developing maize ears facilitates functional analysis and trait candidate gene discovery. Developmental Cell 56:557−568.E6

    doi: 10.1016/j.devcel.2020.12.015

    CrossRef   Google Scholar

    [37]

    Liu H, Hu D, Du P, Wang L, Liang X, et al. 2021. Single-cell RNA-seq describes the transcriptome landscape and identifies critical transcription factors in the leaf blade of the allotetraploid peanut (Arachis hypogaea L.). Plant Biotechnology Journal 19:2261−76

    doi: 10.1111/pbi.13656

    CrossRef   Google Scholar

    [38]

    Kang M, Choi Y, Kim H, Kim SG. 2022. Single-cell RNA-sequencing of Nicotiana attenuata corolla cells reveals the biosynthetic pathway of a floral scent. New Phytologist 234:527−44

    doi: 10.1111/nph.17992

    CrossRef   Google Scholar

    [39]

    Bai Y, Liu H, Lyu H, Su L, Xiong J, et al. 2022. Development of a single-cell atlas for woodland strawberry (Fragaria vesca) leaves during early Botrytis cinerea infection using single-cell RNA-seq. Horticulture Research 9:uhab055

    doi: 10.1093/hr/uhab055

    CrossRef   Google Scholar

    [40]

    Sun X, Feng D, Liu M, Qin R, Li Y, et al. 2022. Single-cell transcriptome reveals dominant subgenome expression and transcriptional response to heat stress in Chinese cabbage. Genome Biology 23:262

    doi: 10.1186/s13059-022-02834-4

    CrossRef   Google Scholar

    [41]

    Guo X, Liang J, Lin R, Zhang L, Zhang Z, et al. 2022. Single-cell transcriptome reveals differentiation between adaxial and abaxial mesophyll cells in Brassica rapa. Plant Biotechnology Journal 20:2233−35

    doi: 10.1111/pbi.13919

    CrossRef   Google Scholar

    [42]

    Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, et al. 2009. mRNA-Seq whole-transcriptome analysis of a single cell. Nature Methods 6:377−82

    doi: 10.1038/nmeth.1315

    CrossRef   Google Scholar

    [43]

    Picelli S, Faridani OR, Björklund ÅK, Winberg G, Sagasser S, et al. 2014. Full-length RNA-seq from single cells using Smart-seq2. Nature Protocols 9:171−81

    doi: 10.1038/nprot.2014.006

    CrossRef   Google Scholar

    [44]

    Hagemann-Jensen M, Ziegenhain C, Chen P, Ramsköld D, Hendriks GJ, et al. 2020. Single-cell RNA counting at allele and isoform resolution using Smart-seq3. Nature Biotechnology 38:708−14

    doi: 10.1038/s41587-020-0497-0

    CrossRef   Google Scholar

    [45]

    Hashimshony T, Senderovich N, Avital G, Klochendler A, De Leeuw Y, et al. 2016. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biology 17:77

    doi: 10.1186/s13059-016-0938-8

    CrossRef   Google Scholar

    [46]

    Chen H, Liao Y, Zhang G, Sun Z, Yang L, et al. 2021. High-throughput Microwell-seq 2.0 profiles massively multiplexed chemical perturbation. Cell Discovery 7:107

    doi: 10.1038/s41421-021-00333-7

    CrossRef   Google Scholar

    [47]

    Jaitin DA, Kenigsberg E, Keren-Shaul H, Elefant N, Paul F, et al. 2014. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343:776−79

    doi: 10.1126/science.1247651

    CrossRef   Google Scholar

    [48]

    Satija R, Farrell JA, Gennert D, Schier AF, Regev A. 2015. Spatial reconstruction of single-cell gene expression data. Nature Biotechnology 33:495−502

    doi: 10.1038/nbt.3192

    CrossRef   Google Scholar

    [49]

    McGinnis CS, Murrow LM, Gartner ZJ. 2019. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Systems 8:329−337.E4

    doi: 10.1016/j.cels.2019.03.003

    CrossRef   Google Scholar

    [50]

    Wolock SL, Lopez R, Klein AM. 2019. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Systems 8:281−291.E9

    doi: 10.1016/j.cels.2018.11.005

    CrossRef   Google Scholar

    [51]

    DePasquale EAK, Schnell DJ, Van Camp PJ, Valiente-Alandí Í, Blaxall BC, et al. 2019. DoubletDecon: deconvoluting doublets from single-cell RNA-sequencing data. Cell Reports 29:1718−1727.E8

    doi: 10.1016/j.celrep.2019.09.082

    CrossRef   Google Scholar

    [52]

    Xi NM, Li JJ. 2021. Benchmarking computational doublet-detection methods for single-cell RNA sequencing data. Cell Systems 12:176−194.E6

    doi: 10.1016/j.cels.2020.11.008

    CrossRef   Google Scholar

    [53]

    Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, et al. 2019. Fast, sensitive and accurate integration of single-cell data with Harmony. Nature Methods 16:1289−96

    doi: 10.1038/s41592-019-0619-0

    CrossRef   Google Scholar

    [54]

    Welch JD, Kozareva V, Ferreira A, Vanderburg C, Martin C, et al. 2019. Single-cell multi-omic integration compares and contrasts features of brain cell identity. Cell 177:1873−1887.E17

    doi: 10.1016/j.cell.2019.05.006

    CrossRef   Google Scholar

    [55]

    Lin Y, Ghazanfar S, Wang KYX, Gagnon-Bartsch JA, Lo KK, et al. 2019. scMerge leverages factor analysis, stable expression, and pseudoreplication to merge multiple single-cell RNA-seq datasets. Proceedings of the National Academy of Sciences of the United States of America 116:9775−84

    doi: 10.1073/pnas.1820006116

    CrossRef   Google Scholar

    [56]

    Lotfollahi M, Wolf FA, Theis FJ. 2019. scGen predicts single-cell perturbation responses. Nature Methods 16:715−21

    doi: 10.1038/s41592-019-0494-8

    CrossRef   Google Scholar

    [57]

    Tran HTN, Ang KS, Chevrier M, Zhang X, Lee NYS, et al. 2020. A benchmark of batch-effect correction methods for single-cell RNA sequencing data. Genome Biology 21:12

    doi: 10.1186/s13059-019-1850-9

    CrossRef   Google Scholar

    [58]

    Haghverdi L, Lun ATL, Morgan MD, Marioni JC. 2018. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nature Biotechnology 36:421−27

    doi: 10.1038/nbt.4091

    CrossRef   Google Scholar

    [59]

    Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. 2018. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nature Biotechnology 36:411−20

    doi: 10.1038/nbt.4096

    CrossRef   Google Scholar

    [60]

    Villani AC, Satija R, Reynolds G, Sarkizova S, Shekhar K, et al. 2017. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 356:eaah4573

    doi: 10.1126/science.aah4573

    CrossRef   Google Scholar

    [61]

    Van der Maaten L, Hinton G. 2008. Visualizing Data using t-SNE. Journal of Machine Learning Research 9:2579−605

    Google Scholar

    [62]

    Becht E, Mcinnes L, Healy J, Dutertre CA, Kwok IWH, et al. 2019. Dimensionality reduction for visualizing single-cell data using UMAP. Nature Biotechnology 37:38−44

    doi: 10.1038/nbt.4314

    CrossRef   Google Scholar

    [63]

    Kamiya T, Borghi M, Wang P, Danku JMC, Kalmbach L, et al. 2015. The MYB36 transcription factor orchestrates Casparian strip formation. Proceedings of the National Academy of Sciences of the United States of America 112:10533−38

    doi: 10.1073/pnas.1507691112

    CrossRef   Google Scholar

    [64]

    Sawchuk MG, Donner TJ, Head P, Scarpella E. 2008. Unique and overlapping expression patterns among members of photosynthesis-associated nuclear gene families in Arabidopsis. Plant Physiology 148:1908−24

    doi: 10.1104/pp.108.126946

    CrossRef   Google Scholar

    [65]

    Qiu X, Mao Q, Tang Y, Wang L, Chawla R, et al. 2017. Reversed graph embedding resolves complex single-cell trajectories. Nature Methods 14:979−82

    doi: 10.1038/nmeth.4402

    CrossRef   Google Scholar

    [66]

    Street K, Risso D, Fletcher RB, Das D, Ngai J, et al. 2018. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19:477

    doi: 10.1186/s12864-018-4772-0

    CrossRef   Google Scholar

    [67]

    Herring CA, Banerjee A, Mckinley ET, Simmons AJ, Ping J, et al. 2018. Unsupervised trajectory analysis of single-cell RNA-seq and imaging data reveals alternative tuft cell origins in the gut. Cell Systems 6:37−51.E9

    doi: 10.1016/j.cels.2017.10.012

    CrossRef   Google Scholar

    [68]

    Li H, Dai X, Huang X, Xu M, Wang Q, et al. 2021. Single-cell RNA sequencing reveals a high-resolution cell atlas of xylem in Populus. Journal of Integrative Plant Biology 63:1906−21

    doi: 10.1111/jipb.13159

    CrossRef   Google Scholar

    [69]

    Chen Y, Tong S, Jiang Y, Ai F, Feng Y, et al. 2021. Transcriptional landscape of highly lignified poplar stems at single-cell resolution. Genome Biology 22:319

    doi: 10.1186/s13059-021-02537-2

    CrossRef   Google Scholar

    [70]

    Xie J, Li M, Zeng J, Li X, Zhang D. 2022. Single-cell RNA sequencing profiles of stem-differentiating xylem in poplar. Plant Biotechnology Journal 20:417−19

    doi: 10.1111/pbi.13763

    CrossRef   Google Scholar

    [71]

    Wang Q, Wu Y, Peng A, Cui J, Zhao M, et al. 2022. Single-cell transcriptome atlas reveals developmental trajectories and a novel metabolic pathway of catechin esters in tea leaves. Plant Biotechnology Journal 20:2089−106

    doi: 10.1111/pbi.13891

    CrossRef   Google Scholar

    [72]

    Liang X, Ma Z, Ke Y, Wang J, Wang L, et al. 2023. Single-cell transcriptomic analyses reveal cellular and molecular patterns of rubber tree response to early powdery mildew infection. Plant, Cell & Environment 46:2222−37

    doi: 10.1111/pce.14585

    CrossRef   Google Scholar

    [73]

    Yu C, Hou K, Zhang H, Liang X, Chen C, et al. 2023. Integrated mass spectrometry imaging and single-cell transcriptome atlas strategies provide novel insights into taxoid biosynthesis and transport in Taxus mairei stems. The Plant Journal 115:1243−60

    doi: 10.1111/tpj.16315

    CrossRef   Google Scholar

    [74]

    Zhan X, Qiu T, Zhang H, Hou K, Liang X, et al. 2023. Mass spectrometry imaging and single-cell transcriptional profiling reveal the tissue-specific regulation of bioactive ingredient biosynthesis in Taxus leaves. Plant Communications 4:100630

    doi: 10.1016/j.xplc.2023.100630

    CrossRef   Google Scholar

    [75]

    Wolf FA, Hamey FK, Plass M, Solana J, Dahlin JS, et al. 2019. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biology 20:59

    doi: 10.1186/s13059-019-1663-x

    CrossRef   Google Scholar

    [76]

    Cao J, Spielmann M, Qiu X, Huang X, Ibrahim DM, et al. 2019. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566:496−502

    doi: 10.1038/s41586-019-0969-x

    CrossRef   Google Scholar

    [77]

    La Manno G, Soldatov R, Zeisel A, Braun E, Hochgerner H, et al. 2018. RNA velocity of single cells. Nature 560:494−98

    doi: 10.1038/s41586-018-0414-6

    CrossRef   Google Scholar

    [78]

    Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. 2020. Generalizing RNA velocity to transient cell states through dynamical modeling. Nature Biotechnology 38:1408−14

    doi: 10.1038/s41587-020-0591-3

    CrossRef   Google Scholar

    [79]

    Wang K, Hou L, Wang X, Zhai X, Lu Z, et al. 2023. PhyloVelo enhances transcriptomic velocity field mapping using monotonically expressed genes. Nature Biotechnology

    doi: 10.1038/s41587-023-01887-5

    CrossRef   Google Scholar

    [80]

    Jansson S, Douglas CJ. 2007. Populus: a model system for plant biology. Annual Review of Plant Biology 58:435−58

    doi: 10.1146/annurev.arplant.58.032806.103956

    CrossRef   Google Scholar

    [81]

    Douglas CJ. 2017. Populus as a model tree. In Comparative and Evolutionary Genomics of Angiosperm Trees, eds. Groover A, Cronk Q, PGG, volume 21. Cham: Springer. pp. 61−84. https://doi.org/10.1007/7397_2016_12

    [82]

    Taylor G. 2002. Populus: Arabidopsis for forestry. Do we need a model tree? Annals of Botany 90:681−89

    doi: 10.1093/aob/mcf255

    CrossRef   Google Scholar

    [83]

    Tuskan GA, Difazio S, Jansson S, Bohlmann J, Grigoriev I, et al. 2006. The genome of black cottonwood, Populus trichocarpa (Torr. & Gray). Science 313:1596−604

    doi: 10.1126/science.1128691

    CrossRef   Google Scholar

    [84]

    Ma T, Wang J, Zhou G, Yue Z, Hu Q, et al. 2014. Erratum: Genomic insights into salt adaptation in a desert poplar. Nature Communications 5:3454

    doi: 10.1038/ncomms4454

    CrossRef   Google Scholar

    [85]

    Zhang Z, Chen Y, Zhang J, Ma X, Li Y, et al. 2020. Improved genome assembly provides new insights into genome evolution in a desert poplar (Populus euphratica). Molecular Ecology Resources 20:781−94

    doi: 10.1111/1755-0998.13142

    CrossRef   Google Scholar

    [86]

    Yang W, Wang K, Zhang J, Ma J, Liu J, et al. 2017. The draft genome sequence of a desert tree Populus pruinosa. GigaScience 6:gix075

    doi: 10.1093/gigascience/gix075

    CrossRef   Google Scholar

    [87]

    Evert RF. 2006. Esau's plant anatomy: meristems, cells, and tissues of the plant body: their structure, function, and development. 3rd edn. New Jersey: John Wiley & Sons. 601 pp.

    [88]

    Nieminen KM, Kauppinen L, Helariutta Y. 2004. A weed for wood? Arabidopsis as a genetic model for xylem development Plant Physiology 135:653−59

    doi: 10.1104/pp.104.040212

    CrossRef   Google Scholar

    [89]

    Tung CC, Kuo SC, Yang CL, Yu JH, Huang CE, et al. 2023. Single-cell transcriptomics unveils xylem cell development and evolution. Genome Biology 24:3

    doi: 10.1186/s13059-022-02845-1

    CrossRef   Google Scholar

    [90]

    Li R, Wang Z, Wang J, Li L. 2023. Combining single-cell RNA sequencing with spatial transcriptome analysis reveals dynamic molecular maps of cambium differentiation in the primary and secondary growth of trees. Plant Communications 4:100665

    doi: 10.1016/j.xplc.2023.100665

    CrossRef   Google Scholar

    [91]

    Qin Y, Sun M, Li W, Xu M, Shao L, et al. 2022. Single-cell RNA-seq reveals fate determination control of an individual fibre cell initiation in cotton (Gossypium hirsutum). Plant Biotechnology Journal 20:2372−88

    doi: 10.1111/pbi.13918

    CrossRef   Google Scholar

    [92]

    Wang D, Hu X, Ye H, Wang Y, Yang Q, et al. 2023. Cell-specific clock-controlled gene expression program regulates rhythmic fiber cell growth in cotton. Genome Biology 24:49

    doi: 10.1186/s13059-023-02886-0

    CrossRef   Google Scholar

    [93]

    Sun Y, Han Y, Sheng K, Yang P, Cao Y, et al. 2023. Single-cell transcriptomic analysis reveals the developmental trajectory and transcriptional regulatory networks of pigment glands in Gossypium bickii. Molecular Plant 16:694−708

    doi: 10.1016/j.molp.2023.02.005

    CrossRef   Google Scholar

    [94]

    Long L, Xu F, Wang C, Zhao X, Yuan M, et al. 2023. Single-cell transcriptome atlas identified novel regulators for pigment gland morphogenesis in cotton. Plant Biotechnology Journal 21:1100−02

    doi: 10.1111/pbi.14035

    CrossRef   Google Scholar

    [95]

    Ding Y, Gao W, Qin Y, Li X, Zhang Z, et al. 2023. Single-cell RNA landscape of the special fiber initiation process in Bombax ceiba. Plant Communications 4:100554

    doi: 10.1016/j.xplc.2023.100554

    CrossRef   Google Scholar

    [96]

    Xia E, Li F, Tong W, Li P, Wu Q, et al. 2019. Tea Plant Information Archive: a comprehensive genomics and bioinformatics platform for tea plant. Plant Biotechnology Journal 17:1938−53

    doi: 10.1111/pbi.13111

    CrossRef   Google Scholar

    [97]

    Tang C, Yang M, Fang Y, Luo Y, Gao S, et al. 2016. The rubber tree genome reveals new insights into rubber production and species adaptation. Nature Plants 2:16073

    doi: 10.1038/nplants.2016.73

    CrossRef   Google Scholar

    [98]

    Hu W, Liu T, Zhu C, Wu Q, Chen L, et al. 2022. Physiological, proteomic analysis, and calcium-related gene expression reveal Taxus wallichiana var. mairei adaptability to acid rain stress under various calcium levels. Frontiers in Plant Science 13:845107

    doi: 10.3389/fpls.2022.845107

    CrossRef   Google Scholar

    [99]

    Guillotin B, Rahni R, Passalacqua M, Mohammed MA, Xu X, et al. 2023. A pan-grass transcriptome reveals patterns of cellular divergence in crops. Nature 617:785−91

    doi: 10.1038/s41586-023-06053-0

    CrossRef   Google Scholar

    [100]

    Conde D, Triozzi PM, Balmant KM, Doty AL, Miranda M, et al. 2021. A robust method of nuclei isolation for single-cell RNA sequencing of solid tissues from the plant genus Populus. PLoS ONE 16:e0251149

    doi: 10.1371/journal.pone.0251149

    CrossRef   Google Scholar

    [101]

    Conde D, Triozzi PM, Pereira WJ, Schmidt HW, Balmant KM, et al. 2022. Single-nuclei transcriptome analysis of the shoot apex vascular system differentiation in Populus. Development 149:dev200632

    doi: 10.1242/dev.200632

    CrossRef   Google Scholar

    [102]

    Fischer U, Kucukoglu M, Helariutta Y, Bhalerao RP. 2019. The dynamics of cambial stem cell activity. Annual Review of Plant Biology 70:293−319

    doi: 10.1146/annurev-arplant-050718-100402

    CrossRef   Google Scholar

    [103]

    Suer S, Agusti J, Sanchez P, Schwarz M, Greb T. 2011. WOX4 imparts auxin responsiveness to cambium cells in Arabidopsis. The Plant Cell 23:3247−59

    doi: 10.1105/tpc.111.087874

    CrossRef   Google Scholar

    [104]

    Kucukoglu M, Nilsson J, Zheng B, Chaabouni S, Nilsson O. 2017. WUSCHEL-RELATED HOMEOBOX4 (WOX4)-like genes regulate cambial cell division activity and secondary growth in Populus trees. New Phytologist 215:642−57

    doi: 10.1111/nph.14631

    CrossRef   Google Scholar

    [105]

    Etchells JP, Mishra LS, Kumar M, Campbell L, Turner SR. 2015. Wood formation in trees is increased by manipulating PXY-regulated cell division. Current Biology 25:1050−55

    doi: 10.1016/j.cub.2015.02.023

    CrossRef   Google Scholar

    [106]

    Xu Z, Wang Q, Zhu X, Wang G, Qin Y, et al. 2022. Plant Single Cell Transcriptome Hub (PsctH): an integrated online tool to explore the plant single-cell transcriptome landscape. Plant Biotechnology Journal 20:10−12

    doi: 10.1111/pbi.13725

    CrossRef   Google Scholar

    [107]

    Jin J, Lu P, Xu Y, Tao J, Li Z, et al. 2022. PCMDB: a curated and comprehensive resource of plant cell markers. Nucleic Acids Research 50:D1448−D1455

    doi: 10.1093/nar/gkab949

    CrossRef   Google Scholar

    [108]

    Chen H, Yin X, Guo L, Yao J, Ding Y, et al. 2021. PlantscRNAdb: a database for plant single-cell RNA analysis. Molecular Plant 14:855−57

    doi: 10.1016/j.molp.2021.05.002

    CrossRef   Google Scholar

    [109]

    Liu Z, Yu X, Qin A, Zhao Z, Liu Y, et al. 2022. Research strategies for single-cell transcriptome analysis in plant leaves. The Plant Journal 112:27−37

    doi: 10.1111/tpj.15927

    CrossRef   Google Scholar

    [110]

    Tarashansky AJ, Musser JM, Khariton M, Li P, Arendt D, et al. 2021. Mapping single-cell atlases throughout Metazoa unravels cell type evolution. eLife 10:e66747

    doi: 10.7554/eLife.66747

    CrossRef   Google Scholar

    [111]

    Liu X, Shen Q, Zhang S. 2023. Cross-species cell-type assignment from single-cell RNA-seq data by a heterogeneous graph neural network. Genome Research 33:96−111

    doi: 10.1101/gr.276868.122

    CrossRef   Google Scholar

    [112]

    Van de Peer Y, Mizrachi E, Marchal K. 2017. The evolutionary significance of polyploidy. Nature Reviews Genetics 18:411−24

    doi: 10.1038/nrg.2017.26

    CrossRef   Google Scholar

    [113]

    Jiao Y, Wickett NJ, Ayyampalayam S, Chanderbali AS, Landherr L, et al. 2011. Ancestral polyploidy in seed plants and angiosperms. Nature 473:97−100

    doi: 10.1038/nature09916

    CrossRef   Google Scholar

    [114]

    Shafer MER. 2019. Cross-species analysis of single-cell transcriptomic data. Frontiers in Cell and Developmental Biology 7:175

    doi: 10.3389/fcell.2019.00175

    CrossRef   Google Scholar

    [115]

    Shekhar K, Lapan SW, Whitney IE, Tran NM, Macosko EZ, et al. 2016. Comprehensive classification of retinal bipolar neurons by single-cell transcriptomics. Cell 166:1308−1323.E30

    doi: 10.1016/j.cell.2016.07.054

    CrossRef   Google Scholar

    [116]

    Pandey S, Shekhar K, Regev A, Schier AF. 2018. Comprehensive identification and spatial mapping of habenular neuronal types using single-cell RNA-seq. Current Biology 28:1052−1065.E7

    doi: 10.1016/j.cub.2018.02.040

    CrossRef   Google Scholar

    [117]

    Reinig J, Ruge F, Howard M, Ringrose L. 2020. A theoretical model of Polycomb/Trithorax action unites stable epigenetic memory and dynamic regulation. Nature Communications 11:4782

    doi: 10.1038/s41467-020-18507-4

    CrossRef   Google Scholar

    [118]

    Li X, Chen L, Zhang Q, Sun Y, Li Q, et al. 2019. BRIF-seq: bisulfite-converted randomly integrated fragments sequencing at the single-cell level. Molecular Plant 12:438−46

    doi: 10.1016/j.molp.2019.01.004

    CrossRef   Google Scholar

    [119]

    Buenostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, et al. 2015. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523:486−90

    doi: 10.1038/nature14590

    CrossRef   Google Scholar

    [120]

    Cao J, O'day DR, Pliner HA, Kingsley PD, Deng M, et al. 2020. A human cell atlas of fetal gene expression. Science 370:eaba7721

    doi: 10.1126/science.aba7721

    CrossRef   Google Scholar

    [121]

    Domcke S, Hill AJ, Daza RM, Cao J, O'day DR, et al. 2020. A human cell atlas of fetal chromatin accessibility. Science 370:eaba7612

    doi: 10.1126/science.aba7612

    CrossRef   Google Scholar

    [122]

    Wang W, Chen K, Chen N, Gao J, Zhang W, et al. 2023. Chromatin accessibility dynamics insight into crosstalk between regulatory landscapes in poplar responses to multiple treatments. Tree Physiology 43:1023−41

    doi: 10.1093/treephys/tpad023

    CrossRef   Google Scholar

    [123]

    Wang P, Jin S, Chen X, Wu L, Zheng Y, et al. 2021. Chromatin accessibility and translational landscapes of tea plants under chilling stress. Horticulture Research 8:96

    doi: 10.1038/s41438-021-00529-8

    CrossRef   Google Scholar

    [124]

    Brown K, Takawira LT, O'neill MM, Mizrachi E, Myburg AA, et al. 2019. Identification and functional evaluation of accessible chromatin associated with wood formation in Eucalyptus grandis. New Phytologist 223:1937−51

    doi: 10.1111/nph.15897

    CrossRef   Google Scholar

    [125]

    Marand AP, Chen Z, Gallavotti A, Schmitz RJ. 2021. A cis-regulatory atlas in maize at single-cell resolution. Cell 184:3041−3055.E21

    doi: 10.1016/j.cell.2021.04.014

    CrossRef   Google Scholar

    [126]

    Farmer A, Thibivilliers S, Ryu KH, Schiefelbein J, Libault M. 2021. Single-nucleus RNA and ATAC sequencing reveals the impact of chromatin accessibility on gene expression in Arabidopsis roots at the single-cell level. Molecular Plant 14:372−83

    doi: 10.1016/j.molp.2021.01.001

    CrossRef   Google Scholar

    [127]

    Zhang L, He C, Lai Y, Wang Y, Kang L, et al. 2023. Asymmetric gene expression and cell-type-specific regulatory networks in the root of bread wheat revealed by single-cell multiomics analysis. Genome Biology 24:65

    doi: 10.1186/s13059-023-02908-x

    CrossRef   Google Scholar

    [128]

    Ouyang W, Luan S, Xiang X, Guo M, Zhang Y, et al. 2022. Profiling plant histone modification at single-cell resolution using snCUT&Tag. Plant Biotechnology Journal 20:420−22

    doi: 10.1111/pbi.13768

    CrossRef   Google Scholar

    [129]

    Nagano T, Lubling Y, Yaffe E, Wingett SW, Dean W, et al. 2015. Single-cell Hi-C for genome-wide detection of chromatin interactions that occur simultaneously in a single cell. Nature Protocols 10:1986−2003

    doi: 10.1038/nprot.2015.127

    CrossRef   Google Scholar

    [130]

    Zhou S, Jiang W, Zhao Y, Zhou D. 2019. Single-cell three-dimensional genome structures of rice gametes and unicellular zygotes. Nature Plants 5:795−800

    doi: 10.1038/s41477-019-0471-3

    CrossRef   Google Scholar

    [131]

    Chen J, Suo S, Tam PPL, Han JDJ, Peng G, et al. 2017. Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nature Protocols 12:566−80

    doi: 10.1038/nprot.2017.003

    CrossRef   Google Scholar

    [132]

    Lubeck E, Coskun AF, Zhiyentayev T, Ahmad M, Cai L. 2014. Single-cell in situ RNA profiling by sequential hybridization. Nature Methods 11:360−61

    doi: 10.1038/nmeth.2892

    CrossRef   Google Scholar

    [133]

    Shah S, Takei Y, Zhou W, Lubeck E, Yun J, et al. 2018. Dynamics and spatial genomics of the nascent transcriptome by intron seqFISH. Cell 174:363−376.E16

    doi: 10.1016/j.cell.2018.05.035

    CrossRef   Google Scholar

    [134]

    Eng CHL, Lawson M, Zhu Q, Dries R, Koulena N, et al. 2019. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+. Nature 568:235−39

    doi: 10.1038/s41586-019-1049-y

    CrossRef   Google Scholar

    [135]

    Ke R, Mignardi M, Pacureanu A, Svedlund J, Botling J, et al. 2013. In situ sequencing for RNA analysis in preserved tissue and cells. Nature Methods 10:857−60

    doi: 10.1038/nmeth.2563

    CrossRef   Google Scholar

    [136]

    Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Yang JL, et al. 2014. Highly multiplexed subcellular RNA sequencing in situ. Science 343:1360−63

    doi: 10.1126/science.1250212

    CrossRef   Google Scholar

    [137]

    Lee JH, Daugharthy ER, Scheiman J, Kalhor R, Ferrante TC, et al. 2015. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nature Protocols 10:442−58

    doi: 10.1038/nprot.2014.191

    CrossRef   Google Scholar

    [138]

    Alon S, Goodwin DR, Sinha A, Wassie AT, Chen F, et al. 2021. Expansion sequencing: spatially precise in situ transcriptomics in intact biological systems. Science 371:eaax2656

    doi: 10.1126/science.aax2656

    CrossRef   Google Scholar

    [139]

    Ståhl PL, Salmén F, Vickovic S, Lundmark A, Navarro JF, et al. 2016. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353:78−82

    doi: 10.1126/science.aaf2403

    CrossRef   Google Scholar

    [140]

    Giacomello S, Salmén F, Terebieniec BK, Vickovic S, Navarro JF, et al. 2017. Spatially resolved transcriptome profiling in model plant species. Nature Plants 3:17061

    doi: 10.1038/nplants.2017.61

    CrossRef   Google Scholar

    [141]

    Du J, Wang Y, Chen W, Xu M, Zhou R, et al. 2023. High-resolution anatomical and spatial transcriptome analyses reveal two types of meristematic cell pools within the secondary vascular tissue of poplar stem. Molecular Plant 16:809−28

    doi: 10.1016/j.molp.2023.03.005

    CrossRef   Google Scholar

    [142]

    Chen A, Liao S, Cheng M, Ma K, Wu L, et al. 2022. Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell 185:1777−92

    doi: 10.1016/j.cell.2022.04.003

    CrossRef   Google Scholar

    [143]

    Xia K, Sun H, Li J, Li J, Zhao Y, et al. 2022. The single-cell stereo-seq reveals region-specific cell subtypes and transcriptome profiling in Arabidopsis leaves. Developmental Cell 57:1299−1310.E4

    doi: 10.1016/j.devcel.2022.04.011

    CrossRef   Google Scholar

    [144]

    Liu Y, Yang M, Deng Y, Su G, Enninful A, et al. 2020. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183:1665−1681.E18

    doi: 10.1016/j.cell.2020.10.026

    CrossRef   Google Scholar

    [145]

    Deng Y, Bartosovic M, Ma S, Zhang D, Kukanja P, et al. 2022. Spatial profiling of chromatin accessibility in mouse and human tissues. Nature 609:375−83

    doi: 10.1038/s41586-022-05094-1

    CrossRef   Google Scholar

    [146]

    Deng Y, Bartosovic M, Kukanja P, Zhang D, Liu Y, et al. 2022. Spatial-CUT&Tag: spatially resolved chromatin modification profiling at the cellular level. Science 375:681−86

    doi: 10.1126/science.abg7216

    CrossRef   Google Scholar

    [147]

    Wolf FA, Angerer P, Theis FJ. 2018. SCANPY: large-scale single-cell gene expression data analysis. Genome Biology 19:15

    doi: 10.1186/s13059-017-1382-0

    CrossRef   Google Scholar

    [148]

    Palla G, Spitzer H, Klein M, Fischer D, Schaar AC, et al. 2022. Squidpy: a scalable framework for spatial omics analysis. Nature Methods 19:171−78

    doi: 10.1038/s41592-021-01358-2

    CrossRef   Google Scholar

    [149]

    Bergenstråhle J, Larsson L, Lundeberg J. 2020. Seamless integration of image and molecular analysis for spatial transcriptomics workflows. BMC Genomics 21:482

    doi: 10.1186/s12864-020-06832-3

    CrossRef   Google Scholar

    [150]

    Dries R, Zhu Q, Dong R, Eng CHL, Li H, et al. 2021. Giotto: a toolbox for integrative analysis and visualization of spatial expression data. Genome Biology 22:78

    doi: 10.1186/s13059-021-02286-2

    CrossRef   Google Scholar

    [151]

    Jones RC, Karkanias J, Krasnow MA, Pisco AO, Quake SR, et al. 2022. The Tabula Sapiens: a multiple-organ, single-cell transcriptomic atlas of humans. Science 376:eabl4896

    doi: 10.1126/science.abl4896

    CrossRef   Google Scholar

    [152]

    Eraslan G, Drokhlyansky E, Anand S, Fiskin E, Subramanian A, et al. 2022. Single-nucleus cross-tissue molecular reference maps toward understanding disease gene function. Science 376:abl429

    doi: 10.1126/science.abl429

    CrossRef   Google Scholar

    [153]

    Conde CD, Xu C, Jarvis LB, Rainbow DB, Wells SB, et al. 2022. Cross-tissue immune cell analysis reveals tissue-specific features in humans. Science 376:eabl5197

    doi: 10.1126/science.abl5197

    CrossRef   Google Scholar

    [154]

    Suo C, Dann E, Goh I, Jardine L, Kleshchevnikov V, et al. 2022. Mapping the developing human immune system across organs. Science 376:eabo0510

    doi: 10.1126/science.abo0510

    CrossRef   Google Scholar

    [155]

    Liu Z, Zhang Z. 2022. Mapping cell types across human tissues. Science 376:695−96

    doi: 10.1126/science.abq2116

    CrossRef   Google Scholar

  • Cite this article

    Liang S, Li Y, Chen Y, Huang H, Zhou R, et al. 2023. Application and prospects of single-cell and spatial omics technologies in woody plants. Forestry Research 3:27 doi: 10.48130/FR-2023-0027
    Liang S, Li Y, Chen Y, Huang H, Zhou R, et al. 2023. Application and prospects of single-cell and spatial omics technologies in woody plants. Forestry Research 3:27 doi: 10.48130/FR-2023-0027

Figures(5)  /  Tables(1)

Article Metrics

Article views(2414) PDF downloads(417)

REVIEW   Open Access    

Application and prospects of single-cell and spatial omics technologies in woody plants

Forestry Research  3 Article number: 27  (2023)  |  Cite this article

Abstract: Over the past decade, high-throughput sequencing and high-resolution single-cell transcriptome sequencing technologies have undergone rapid development, leading to significant breakthroughs. Traditional molecular biology methods are limited in their ability to unravel cellular-level heterogeneity within woody plant tissues. Consequently, techniques such as single-cell transcriptomics, single-cell epigenetics, and spatial transcriptomics are rapidly gaining popularity in the study of woody plants. In this review, we provide a comprehensive overview of the development of these technologies, with a focus on their applications and the challenges they present in single-cell transcriptome research in woody plants. In particular, we delve into the similarities and differences among the results of current studies and analyze the reasons behind these differences. Furthermore, we put forth potential solutions to overcome the challenges encountered in single-cell transcriptome applications in woody plants. Finally, we discuss the application directions of these techniques to address key challenges in woody plant research in the future.

    • The common feature of multicellular organisms in nature is that they are comprised of tissues, organs, and systematic developmental programs that are coordinated and assembled by various cell types according to certain functional rules. In animals, more than 100 cell types, and even more cell subtypes, have been reported[13]. However, the different cell types and their heterogeneity in plants are still under investigation. Historically, the identification of plant cell types has mainly relied on the examination of cell morphology and the study of certain molecular markers identified in model plants such as Arabidopsis. Therefore, our understanding of the various cell types in plants, as well as the important scientific questions such as the heterogeneity among cell types, the composition of cell subtypes, the genetic developmental relationships, and the fate determination processes, is still limited, especially for woody plants characterized by highly lignified root and stem tissues.

      Woody plants, as important components of terrestrial ecosystems, have played key roles in human history, both from socio-economic and subsistence standpoints. They fulfill essential ecological functions, including oxygen production, soil and water conservation, and climate regulation, in addition to being valuable sources of timber and wood[4]. It is the world's most abundant renewable resource used for timber, pulp, and energy[5]. An understanding of wood development[69], environmental adaptability[1013], phenology[1417] and sex differentiation[1821] of woody plants has attracted significant attention. With the development of molecular biology and sequencing techniques, these studies have transitioned from using traditional quantitative polymerase chain reaction to characterize individual gene expression to analyzing the regulation of co-expressed genes using transcriptome data. However, transcriptome data alone cannot reveal the heterogeneity in gene expression between cells. As a result, our understanding of woody plants at the cellular level, including various cell types and molecular markers, remains poor.

      Single-cell RNA sequencing (scRNA-seq) is a revolutionary technology capable of providing transcript-wide information at the level of individual cells, thereby enabling the elucidation of cellular heterogeneity and the identification of novel molecular markers. The technique has been applied to the study of various plants, including Arabidopsis[2231], rice[3234], maize[35,36], peanut[37], tobacco[38], strawberry[39] and cabbage[40,41], and it has been instrumental in the characterization of distinct cell types in various tissues such as root, stem, leaf, and shoot apex (Fig. 1).

      Figure 1. 

      Cell types of roots, stems, leaves, and shoot apexes. The dotted lines indicate the absence of reported studies in woody plants.

      Until 2021, scRNA-seq was applied to the study of woody plants and showed great potential for development. Besides scRNA-seq, emerging technologies, such as single-cell epigenetics and spatial transcriptomics, have introduced new avenues for unraveling the mysteries that surround woody plants. This article offers an in-depth review of the development and data analysis pipeline of scRNA-seq, its applications and challenges in woody plant research, and the potential applications of single-cell epigenetics and spatial transcriptomics.

    • The fundamental concept of single-cell transcriptome sequencing is the ability to extract RNA from a single cell and to construct a transcriptome library. High-throughput sequencing is then used to read the library, thereby yielding the transcript information of a single cell. The library construction methods can be classified into two categories: full-length transcriptome-based library construction methods and tag-based transcriptome-based library construction methods (Fig. 2).

      Figure 2. 

      Development history of single-cell sequencing technologies and spatial multiomics technologies.

      The full-length transcriptome-based library construction methods are known for their robust gene detection capabilities. In 2009, Tang et al. pioneered a single-cell sequencing method[42], marking the beginning of the single-cell genomics era. The method encompasses five key steps: single-cell isolation and lysis, cDNA first-strand reverse transcription, polyA tail addition at the 3' end, cDNA library amplification, and library amplification. Building upon this foundation, Smart-seq2 and Smart-seq3 were developed. Smart-seq2 enhances enzyme thermostability by introducing betaine into the reverse transcription system, thereby increasing cDNA yield[43]. Meanwhile, Smart-seq3 introduces Unique Molecular Identifiers (UMIs) for individual transcripts, significantly enhancing the precision and reliability of gene expression detection[44].

      Conversely, the tag-based transcriptome-based library construction method offers higher detection throughput. The principle involves adding primers with cell-specific barcodes during reverse transcription and then using high-throughput sequencing to distinguish transcripts from different cells. Representative technologies utilizing this library construction method include CEL-Seq2, Microwell-seq, MARS-seq, and 10× Genomics Chromium. CEL-Seq2, for example, employs a dual barcode system to identify both cells and individual RNA molecules within the same cell, significantly enhancing sequencing data quality and accuracy[45]. Microwell-seq utilizes Microwells, specialized agarose microplates, as the single-cell capture platform. These Microwells are reusable, leading to cost-effective large-scale cell sample processing[46]. Chen et al. improved upon this approach and developed Microwell-seq2 by optimizing Microwell utilization and enhancing cell detection sensitivity[46]. MARS-seq and MARS-seq2 employ fluorescence-activated cell sorting, reducing the risk of sample contamination[46,47]. On the other hand, 10× Genomics Chromium integrates barcoding and microfluidics and uses the Illumina sequencing platform, allowing for the labeling of hundreds of thousands of cells within minutes. Notably, both MARS-seq2 and 10× Genomics Chromium have been applied to woody plant research.

    • Currently, woody plant research mainly utilizes the 10× Genomics Chromium platform. Therefore, we present an overview of the scRNA-seq data analysis workflow based on the 10× Genomics Chromium platform, which includes data quality control, data integration, data dimensionality reduction, cell type identification, and pseudo-time trajectory analysis (Fig. 3).

      Figure 3. 

      scRNA-seq data analysis pipeline.

    • To ensure the reliability of the subsequent data analysis, it is necessary to first perform a quality control step on the single-cell data. To achieve this, Seurat[48] is employed to eliminate low-quality cells and genes, while software options such as Doubletfinder[49], Scrublet[50], and DoubletDecon[51] are effective in removing double cells. Among these tools, Doubletfinder stands out as one of the most accurate for doublet removal[52]. Doubletfinder initially calculates the proportion of artificial nearest neighbors (pANN) within the nearest neighbor of each individual cell. It then assigns a probability score for doublets to each barcode based on the pANN value. Finally, using the Poisson distribution, Doubletfinder calculates the number of doublets present in each sample and efficiently filters out the doublets, all while taking into account the prior cell pANN value ranking.

    • For comprehensive analyses and comparisons, it is often necessary to integrate data from different samples or experimental batches, which can be accomplished by software such as Harmony[53], LIGER[54], scMerge[55], scGen[56], and Seurat3[48]. Among these options, Harmony stands out for its efficiency when handling large volumes of cellular data, while LIGER excels when the integrated samples show highly variable cell types[57]. Other popular methods for data integration and batch effects removal include mutual nearest neighbors (MNN)[58] and canonical correlation analysis (CCA)[59], both of which are available in Seurat3[48]. However, investigators should be cautious of overcorrection issues when using these methods. Following data integration, it is crucial to consider the distribution of cell cycle genes. If their distribution is uneven, their effects need to be eliminated, and this can be achieved through the SCTransform function in Seurat3[48].

    • scRNA-seq data presents a complex high-dimensional structure, involving a multitude of cells and genes, making it challenging to visualize the data in its raw form. Therefore, the data must be dimensionally reduced. Principal Component Analysis (PCA)[60] is the primary method for dimensionality reduction in single-cell data, and it can be further complemented by techniques such as t-Distributed Stochastic Neighbor Embedding (tSNE)[61] and Uniform Manifold Approximation and Projection (UMAP)[62] for data visualization. After the dimensionality reduction of data, spectral clustering based on shared nearest neighbor (SNN) and modular optimization of Seurat can be applied to identify the cell clusters.

    • Cell type identification is a critical step in the scRNA-seq analysis pipeline. In single-cell studies of Arabidopsis, the identification of cell types often relies on previously experimentally validated marker genes. Arabidopsis has many reliable marker genes such as the endodermal marker gene MYB36[63] in roots and the mesophyll marker gene CAB3[64] in leaves. However, given the limited availability of marker genes in woody plants, a common strategy is to leverage homologous genes from Arabidopsis as references for cell type annotations based on functional conservation. Additionally, in situ hybridization and laser capture microdissection (LCM) are candidate methods that can be used to validate the annotation of cell populations.

    • To understand the mechanism of organ formation in woody plants, it is important to comprehend the developmental trajectories of various cell types. Pseudo-time trajectory analysis reshapes the change process of cells over time by constructing the transition between cells. Common methods for pseudo-time trajectory analysis include Monocle DDRTree (Monocle2)[65], Slingshot[66], and pCreode[67]. Among these options, Monocle2 is frequently used to construct organ developmental trajectories in woody plants[6874]. Monocle2 utilizes a reverse graph embedding machine learning technique to construct cell developmental trajectories. Often, there are multiple branch points in the results of pseudo-time trajectory analysis, and these branch nodes represent cell state changes, so they are of great importance in the analysis of branch events. The BEAM (Branch Expression Analysis Modeling) function in Monocle2 is used to analyze the differential expression of genes at specified nodes, which can play important roles in cell development. However, the cell differentiation trajectory analyzed by Monocle2 is separated from the results of UMAP or tSNE that obtained by dimensionality reduction. Furthermore, when dealing with a large amount of cell data, Monocle2 may cluster cells with different developmental trajectories into the same trajectory. To address this issue, Cao et al. developed Monocle3 based on the partition-based graph abstraction (PAGA)[75], which can directly draw cell development trajectories on UMAP and efficiently analyze millions of single-cell data[76].

      In addition to pseudo-time trajectory analysis, the prediction of potential cell fate can also be achieved through RNA velocity analysis. This approach leverages splicing information to determine the directionality of cell differentiation[77]. Unlike pseudo-time trajectory analysis, RNA velocity analysis does not yield a continuous cell trajectory but instead provides insights into potential directions in cell differentiation, so there is no need to rely on prior biological experience to specify the start and end points in the analysis. Software tools, such as scVelo[78], Velocyto[77], and PhyloVelo[79], can be used for RNA velocity analysis and combined with pseudo-time trajectory analysis to infer the complex developmental trajectory of woody plant cells.

    • Poplar, renowned for its high genetic transformation and rapid growth, has long served as a model system in woody plant research[8082]. Several species of Populus have been sequenced, laying a solid foundation for in-depth research[8386]. Additionally, given its economic value as a tree species, poplar is a prime source of high-quality wood. However, enhancing wood properties requires a comprehensive understanding of the various processes that underlie cell formation and differentiation during wood formation, which are challenging to monitor through traditional molecular biology techniques. Thus, the initial application of scRNA-seq in woody plants has centered around wood development in poplar.

      According to an anatomical identification study, the xylem is primarily comprised of three cell types: fiber, vessel element, and ray[87]. As mentioned earlier, the characterization of cell types in most non-model species often relies on homologs of marker genes from Arabidopsis. While several molecular markers can be used to identify fiber and vessel elements, it is difficult to identify molecular markers for ray cells because they do not exist in Arabidopsis[88]. To our knowledge, five studies have recently used scRNA-seq to conduct preliminary analyses of cell types and growth dynamics in poplar woody tissues[6870,89,90]. Among these, Li et al.[68,90] and Chen et al.[69] used molecular markers derived from both poplar and Arabidopsis, respectively, while most of the cellular annotations of Xie et al. were based on molecular markers specific to poplar[70]. Tung et al. generated in situ cell transcriptomes using LCM and separated fibers, vessel elements, and ray cells by ranking the expression correlations between in situ cell transcriptomes and cell clusters[89]. Because the poplar species used in these studies were different, we integrated these gene markers for cell identity through their orthologous relationships (Supplemental Table S1). Unexpectedly, the results showed that the markers varied widely among these studies. For example, among the molecular markers used by Xie et al, only IRX1 (xylem cells marker) and AIL5 (cambium region) were found in other studies[70], while Tung et al. inferred only one gene encoding expansin (Potri.001G240900) as a candidate marker for identifying vessel elements[89]. Moreover, these studies also identified other cell types in the developing xylem, such as xylem mother cells, organizer cells, and xylem precursor cells, besides the three known types. These cells may be in transitional stages of their developmental fate, and their transcription levels may differ from those of the known cell types. Nevertheless, these studies consistently indicate that the cellular composition of woody tissue is far more complex than anatomically identified, and the characterization of these cells warrants further investigation. In addition to the differences in cell annotation, four out of the five studies also constructed xylem cell developmental trajectories, which showed variations (Fig. 4). Chen et al.[69] and Tung et al.[89] both suggested that fibers and vessel elements belong to the same lineage (Fig. 4b, d), while Li et al. showed that fibers and ray cells shared a common developmental trajectory branch[68] (Fig. 4a). In comparison, Xie et al. constructed two trajectories of cellular differentiation, with one differentiating into fibers and vessel elements and the other into fibers, vessel elements, and ray cells[70] (Fig. 4c). Notably, these differences in cell differentiation trajectories may be influenced by the identification of cell types.

      Figure 4. 

      scRNA-seq studies of poplar stem. The balls of different colors represent different types of cells and the arrows point to the direction of cell differentiation.

      Furthermore, Chen et al. and Li et al. performed analyses of phloem cells. In both studies, CalS7 and SEOR1 were used to identify sieve elements. These investigators also identified the phloem precursor cells (phloem mother cells), cortex cells, epidermal cells, and companion cells. Chen et al. also identified photosynthetic cells, phloem parenchyma cells, cork cells, and endodermal cells, as well as reconstructed the cell differentiation trajectories of the phloem with phloem mother cells, companion cells, and sieve elements[69], while Li et al. reconstructed the cell differentiation trajectories of the cambium to the phloem precursor and xylem precursor[90].

      In addition to their investigations on poplar xylem and phloem cell development, Chen et al. further used single-cell transcriptome data to predict potential gene redundancy resulting from whole-genome duplication (WGD) in Salicaceae[69]. Meanwhile, Tung et al. identified highly conserved ray lineages and variable fusiform lineages based on comparative single-cell mapping of four different woody angiosperms (Populus trichocarpa, Eucalyptus grandis, Trochodendron aralioides, and Liriodendron chinense)[89]. In summary, these studies underscore the high degree of heterogeneity within cell populations in wood, a phenomenon not previously observed in anatomical studies. These studies also highlight the importance of single-cell transcriptomics in the study of woody plants. The discrepancy between these studies may be attributed to the dynamic nature of cell development, where the data only shows a snapshot of the entire developmental process. Additionally, differences in growth conditions, experimental conditions, and expression variations between species may also lead to differences in cellular annotations and developmental trajectories. However, because different marker genes were used, it is difficult to assess whether these differences are due to artifacts in cell identification. One promising approach is to integrate these data and use the same criteria to characterize their cell types and perform comparative studies.

    • In addition to the various studies conducted on different poplar species, scRNA-seq has also been applied to other woody plants. For example, four reports have used scRNA-seq to study cotton, two of which identified the cell types in the ovule’s outer layer and the genes related to cotton fiber synthesis, while the other two focused on the cell types within the cotyledons and the genes affecting the formation of pigment glands[9194] (Table 1). Ding et al. analyzed the initiation process of Bombax ceiba fiber and compared it with single-cell mapping of cotton fiber to excavate genes associated with fiber development[95] (Table 1). Camellia sinensis, one of the most important woody crops cultivated globally, is known for its leaves, which produce various teas[96]. Wang et al. analyzed the spatiotemporal expression patterns of flavor-related genes in different cell types of C. sinensis leaves and discovered a new catechin glycosyltransferase (UGT72B23)[71] (Table 1). On the other hand, Hevea brasiliensis, an important source of natural rubber[97], was investigated by Liang et al., who constructed single-cell maps of normal and powdery mildew-inoculated H. brasiliensis leaves and identified a powdery mildew resistance gene HbCNL2 through pseudo-time trajectory and phylogenetic analyses. Its function was also verified by molecular biology methods[72] (Table 1). Taxol, a highly effective anticancer drug, is a component of Taxus chinensis[98], and Zhan et al. used scRNA-seq and mass spectrometry to reveal the molecular mechanism of the cell-specific secondary metabolism in T. chinensis leaves[74] (Table 1). In summary, these studies showed that scRNA-seq opens new avenues for us to explore the mysteries of woody plants.

      Table 1.  Application of scRNA-seq/snRNA-seq technologies in woody plants.

      SpeciesTissueCell typeSequencing method/
      platform
      Reference
      Populus alba × Populus glandulosaXylemXylem precursor cells, fiber cells, vessels cells, and ray parenchyma cells10× Genomics[68]
      Populus alba var.
      pyramidalis
      StemPhotosynthetic cells, cambium cells, phloem parenchyma cells, xylem cells, xylem mother cells, phloem mother cells, endodermal cells, xylem parenchyma cells, cork cells, epidermal cells, companion cells, cortex/endodermal cells, cortex/endodermis initial cells, and sieve elements10× Genomics[69]
      Populus trichocarpaXylemVessel cells, ray parenchyma cells, phloem cells, cambium cells, and fiber cells10× Genomics[70]
      Populus trichocarpaXylemFusiform organizer, fusiform early precursor, fusiform intermediate precursor, vessel cells, fiber cells, ray organizer, ray precursor, and ray parenchyma cells10× Genomics and MARS-seq2.0[89]
      Eucalyptus grandisXylemFusiform organizer, fusiform early precursor, fusiform intermediate precursor, vessel cells, fiber cells, ray organizer, ray precursor, and ray parenchyma cells
      Trochodendron aralioidesXylemFusiform organizer, fusiform early precursor, fusiform intermediate precursor, tracheid cells, ray organizer, ray precursor, and ray parenchyma cells
      Liriodendron chinenseXylemFusiform organizer, fusiform early precursor, fusiform intermediate precursor, vessel cells, fiber cells, ray organizer, ray precursor, and ray parenchyma cells
      Populus euramericana cv. ‘Nanlin895’StemPhloem precursor, xylem precursor, cambium, vessel, cortex and pith, ray, epidermis, sieve-companion, xylem fiber, and phloem parenchyma10× Genomics[90]
      Camellia sinensisLeafVascular bundle, protoxylem cells, protophloem cells, phloem cells, procambium cells, proliferating cells, epidermis cells, mesophyll cells, palisade mesophyll, and spongy mesophyll10× Genomics[71]
      Reyan73397(Hevea brasiliensis)LeafMeristem cells, latex cells, xylem cells, phloem cells, hydathode cells, bundle sheath cells, epidermis cells, and mesophyll cells10× Genomics[72]
      Bombax ceibaInner wall of the ovaryInitiated fiber cells and epidermal cells in the inner wall of the ovary10× Genomics[95]
      Populus tremula × albaShoot apex vascular systemTrichomes, mesophyll cells, epidermal cells, shoot meristematic cells, proliferating cells, vascular cells, companion cells, and ground meristem cells10× Genomics[101]
      Taxus maireiStemXylem parenchyma cells, xylem cells, epidermal cells, photosynthetic cells, vascular cells, xylem mother cells, companion cells, phloem cells, endodermal cells, cambium cells, and sieve elements10× Genomics[73]
      Taxus maireiLeafBundle sheath cells, mesophyll cells, stomatal complex cells, guard cells, epidermal cells, vascular cells, procambium cells, and pavement cells10× Genomics[74]
      The cotton Lint-Fuzz (Xu142_LF)Ovule outer integumentFiber, epidermis and outer pigment layer10× Genomics[91]
      G. hirsutum cv.
      Xuzhou 142
      Ovule outer integumentFiber and epidermis10× Genomics[92]
      Gland cotton 'CCRI12'
      and glandless cotton
      'CCRI12gl'
      CytoledonSpongy mesophyll cells, palisade mesophyll cells, epidermal cells, primordial cells, guard cells, xylem cells, parenchyma cells, phloem cells, pigment gland cells10× Genomics[94]
      Gossypium bickiiCytoledonMesophyll cells, pigment gland cells, epidermal cells, guard cells, xylem cells, procambium cells, phloem parenchyma cells, and companion cells10× Genomics[93]
    • While scRNA-seq has proven successful in woody plant research, it still faces several challenges, including cell separation, cell type annotation, and data integration. First, there are great difficulties in the preparation of woody plant protoplasts due to high lignification and active secondary metabolism, which may render some cells with thick cell walls unusable for analysis. This challenge is clearly evident in recent studies. For example, Li et al. failed to separate the protoplasts of phloem[68], whereas Chen et al. failed to detect cell types or state with strong expression of programmed cell death-related genes[69]. To circumvent these difficulties, single-nucleus transcriptome sequencing (snRNA-seq) can be used to isolate cell nuclei. While the gene detection capability of snRNA-seq may not be as robust as that of scRNA-seq, it excels at capturing cells that are not easily digested by enzymes. For example, Guillotin et al. identified columella cells in maize roots by snRNA-seq[99], a feat that had not been achieved in previous studies[35]. Recently, Conde et al. developed a nucleus isolation method suitable for tissues with thick secondary cell walls[100] and applied it to the study of shoot apex differentiation in Populus tremula×alba. Using snRNA-seq, they identified highly heterogeneous cell populations and performed comparative analyses of vascular development between Arabidopsis and poplar (Table 1)[101]. This approach not only helped in studying the transition from primary growth to secondary growth in perennial woody plants but also helped in establishing the foundation for the broad application of snRNA-seq to woody plants. These studies illustrate that snRNA-seq can mitigate transcriptome bias during protoplast preparation, thereby enabling the construction a more comprehensive single-cell atlas.

      Secondly, the annotation of cell types is equally challenging. The functions of homologous genes are not conserved across species, and this can significantly impact the accuracy of cell annotation. For example, prior studies identified WOX4 and PXY as marker genes of the cambium of Arabidopsis[102,103], and these genes were also reported in poplar[104,105]. However, Li et al. demonstrated that these two genes are also expressed in other cell types, indicating that they are not reliable molecular markers[90]. In this review, we compiled marker genes used in woody plant research. The results showed a wide variation in marker genes between different plant species, as well as between different tissues and even within the same tissue (Supplemental Table S2). Additionally, the manual annotation of cell types is both time-consuming and inefficient, so there is a pressing need to develop an automated process for identifying plant cell types. To date, the PsctH[106], PCMDB[107], and PlantscRNAdb[108] databases have provided us with a wealth of plant marker genes, but the number of marker genes available for woody plants remains limited. Liu et al. proposed a potential design process for creating automated annotation software for plants[109], which provides a theoretical basis for future software development. It is believed that with the mining of conserved marker genes, the establishment of marker gene databases, and the development of automated annotation software, cell type annotation in woody plants will become efficient and accurate.

      Thirdly, performing comparative transcriptome analyses provides invaluable insights into the evolutionary and developmental relationships between cell or tissue types of different species. The development of single-cell transcriptomic technologies has opened up new possibilities for investigating cell type phylogenies and inferring cell type-specific evolution. Comparative analyses of cell types require quantifying the similarity in gene expression profiles, which often relies on gene homologous relationships between species. Several tools, such as SAMap[110] and CAME[111], have been developed and are mostly used in animal studies where orthologous relationships are relatively clear. However, plants often undergone independent genome duplications[112,113], the complex genetic relationships make the integration of cross-species data extremely difficult. For example, Conde et al. integrated Arabidopsis and poplar data using a one-to-one orthologous gene approach[101], while Tung et al. employed many-to-many homologous clusters for cross-species integration and comparison[89]. It is important to note that the one-to-one approach introduces complexity when integrating distantly related species because there are fewer orthologous genes, and differences in WGD-derived duplicates may confound orthologous relationships. In contrast, the many-to-many approach may mask functional divergence and neofunctionalization of paralogous genes, making it difficult to accurately assess their diversification history. Comparative studies in plants are therefore relatively limited. A potential solution is thus to establish comparison methods that do not rely on homologous genes. For example, Random Forest Machine Learning (RFML) trains algorithms against the cell types of one species and then predicts interspecies cell type similarity[114]. This approach has been successfully applied in studies involving animals[115,116]. However, further studies are needed in the future to determine the best methods for comparing single-cell transcriptomes between plant species.

    • Heterogeneity in cellular transcriptional expression is often determined by heterogeneity in epigenetic modifications. Similar to transcriptomics, traditional epigenetic techniques examine entire tissues but overlook cellular heterogeneity[117]. In recent years, various single-cell sequencing technologies have emerged, each addressing different levels of epigenetic regulation (Fig. 2). These techniques mainly employ different enzymes to process chromatin and capture target information. Some techniques have been applied to plant research. For example, scBRIF-seq enables the study of DNA methylation in single cells. Its pipeline involves single-stranded ligation of small fragments generated through random amplification, MDA amplification, and Tn5-based library construction. This technique has been applied to investigate maize microspores, and significant methylation reprogramming and cellular heterogeneity during maize male gametophyte development were revealed[118]. For single-cell chromatin accessibility studies, two primary techniques have evolved: scATAC-seq and snATAC-seq[119121]. ATAC-seq, which assesses genome-wide chromatin accessibility by cutting DNA sequences using Tn5 transposase as a probe, has proven useful for identifying dynamic chromatin changes in response to stress and during the development of woody plants[122124]. ATAC-seq was subsequently modified by combining it with single-cell library construction methods to develop scATAC-seq and snATAC-seq. In plant research, these two methods are often paired with scRNA-seq or snRNA-seq to enable a more in-depth exploration of cellular heterogeneity[125127]. For example, Farmar et al. used snRNA-seq and snATAC-seq to reveal the impact of chromatin accessibility on gene expression in Arabidopsis root[126]. Wang et al. combined scATAC-seq and scRNA-seq to propose a model for the rhythmic regulation of early cotton fiber growth[92]. In terms of single-cell histone modifications, Ouyang et al. developed snCUT&Tag and applied it to the analysis of the characteristics of single-cell H3K4me3 histone modifications in rice seedlings[128]. This technology combines nCUT&Tag and single-cell barcode labeling, and its subsequent library construction bear similarity to ATAC-seq. Lastly, single-cell high-throughput chromosome conformation capture (scHi-C) is an important method for analyzing chromatin conformation at the single-cell level[129]. Zhou et al. developed the scHi-C technology, which is suitable for plant research, and revealed the changes in the chromatin spatial structure of rice gametes before and after fertilization at the single-cell level[130].

      While a variety of single-cell epigenomic technologies have been successfully employed in plant research, their application has been restricted to Arabidopsis and certain key crops such as rice and maize. Similar research involving woody plants is very limited. On the one hand, challenges in cell separation and sample preparation for woody plants persist. On the other hand, some technologies lack established analysis pipelines, and eliminating technical noise and batch effects are complex tasks. Therefore, the full potential of these technologies in woody plants has yet to be realized. However, as technologies and software tools continue to advance, there is promise that single-cell epigenetics will offer invaluable insights into the epigenetic landscape of woody plants.

    • One of the disadvantages of single-cell transcriptomics is that it destroys the spatial location information of tissues. Spatial transcriptome technologies have emerged to address this limitation, enabling the precise location of various cell types within plant tissues and facilitating the mapping of gene expression across different tissue regions. The current spatial transcriptome technologies primarily fall into four categories: laser microdissection, fluorescence in situ hybridization, fluorescence in situ sequencing, and in situ capture technology (Fig. 2).

      In recent years, researchers have combined laser microdissection and next-generation sequencing to develop spatial transcriptome technologies. For example, LCM-seq and Geo-seq can acquire cell transcriptome information while preserving the original location information of cells[131]. However, these technologies are time-consuming, and the cell separation process can increase the risk of cell damage. Fluorescence in situ hybridization (FISH) offers an alternative approach that uses fluorescently-labeled probes to quantify the abundance of RNA/DNA in cells or tissues without destroying cell morphology, and it has evolved to achieve single-molecule resolution, including sm FISH, seq FISH[132], and intron seq FISH[133,134]. While the detection, throughput, and accuracy of these technologies continue to improve, they still face challenges related to complex steps and high costs. Additionally, researchers have developed spatial transcriptome technologies with higher spatial resolution, including ISS[135], FISSEQ[136,137], and Exseq[138], which utilize fluorescently-labeled probes that hybridize with target sequences, allowing the determination of the location of the target sequence by observing the location and intensity of the fluorescent signals under a microscope. However, some issues still remain, including short sequencing read length and low detection efficiency.

      Compared with the above methods, spatial transcriptomics based on in situ capture technology offers several advantages, including the ability to achieve high throughput and large tissue areas. The principle is to capture transcripts in situ using primer microarrays with spatial tag sequences, thereby preserving the spatial information of the transcripts. Stahl et al. pioneered spatial transcriptomics[139], and it was rapidly applied to plant research. For example, Giacomello et al. constructed spatial transcriptome maps of the inflorescence meristems of Arabidopsis thaliana, the female autumn flowers of Picea abies, and the leaf buds of Populus tremula[140]. More importantly, these studies revealed the suitability of spatial transcriptomics for the study of woody plants. Subsequently, Du et al. used spatial transcriptomics to analyze the continuous development of poplar stems from primary growth to secondary growth, and they identified a new class of procambium-like cells that specifically develop into the phloem[141]. Li et al. constructed a spatial transcriptome map of poplar stems and analyzed gene expression in various cell types[90] . These studies demonstrate that spatial transcriptomics opens up new dimensions and avenues for discoveries in woody plant research. In addition, Stereo-seq[142], a new spatial transcriptome technology, was recently used to study Arabidopsis leaves[143]. This study successfully distinguished upper and lower epidermal cells of leaves and analyzed the expression changes of genes related to photosynthesis from leaf veins to leaf edges. The technique’s advantage lies in its nanoscale resolution and its precise identification of cell subtypes, which presents new opportunities for the study of woody plants. Besides spatial transcriptomics, other spatial multiomics technologies are also progressing rapidly. DBiT-seq realizes the simultaneous acquisition of transcriptomic and proteomic information while acquiring spatial information[144], whereas spatial-ATAC-seq[145] and spatial-CUT&Tag[146] enable the study of chromatin accessibility and histone modifications at spatial resolution. However, these techniques have yet to be applied to plant research.

      While spatial transcriptomics has experienced rapid advancements, technical difficulties still hinder its application in woody plant research. Currently, most spatial transcriptome studies are carried out on young tissues characterized by thin cell walls that are easily processed for freezing and embedding. However, tissues with a high degree of lignification may be damaged during processing, potentially compromising the integrity of reverse transcription products and the subsequent analysis. Therefore, future studies should focus on further optimizing the sample preparation protocol. On a more positive note, the subsequent data analysis process continues to mature, and software tools, including Seurat[48], scanpy[147], squidpy[148], STUtility[149], and Giotto[150], can be used for spatial data analysis and visualization. It is believed that with continued technological advancements, spatial transcriptome technologies will offer new perspectives on the growth and development, stress resistance, and species evolution of woody plants.

    • Studies involving single-cell transcriptomics, single-cell epigenetics, and spatial transcriptomics have been limited in woody plants compared to other organisms. Current research primarily focuses on a small number of species and young, fresh tissues. As a consequence, there is a pressing need to expand the application of these techniques to a broader range of woody plant species and diverse tissue types. In this regard, snRNA-seq has emerged as a powerful tool to help achieve these goals. In single-cell research, a comprehensive human pan-tissue single-cell atlas has been successfully established[151155]. Similarly, the creation of pan-tissue single-cell atlases covering different tissues, developmental stages, and environmental conditions in woody plants will significantly enhance our understanding of the diversity of cell types, and provide new perspectives for studying the evolution and origin of specific traits in woody plants. But before that, it will be important to develop and integrate species-specific and conserved molecular markers for precise identification of cell types (such as Supplemental Tables S1 & S2 in this review). However, current studies lack inter- and intra-species comparisons of single-cell atlases. Interspecific comparisons can illuminate the similarities, differences, and evolutionary relationships of cells among different plant phyla and classes. At the same time, intraspecies comparisons can be employed to investigate functional variations among different tissues (Fig. 5). Therefore, it is reasonable to expect that as single-cell and spatial transcriptome techniques become more widely employed, our knowledge of woody plants will significantly advance in the future.

      Figure 5. 

      Future application of single-cell transcriptomics, single-cell epigenetics, and spatial transcriptomics in woody plants.

    • The authors confirm contribution to the paper as follows: study conception and design: Ma T; draft manuscript preparation: Ma T, Liang S, Li Y; Figure creation: Liang S, Li Y, Chen Y, Huang H, Zhou R. All authors discussed the results and commented on the manuscript. All authors read and approved the final manuscript.

    • Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

      • This work was supported by the National Key Research and Development Program of China (2021YFD2201100), National Natural Science Foundation of China (31922061 and 32271828), and Fundamental Research Funds for the Central Universities (2020SCUNL103). We would like to thank A&L Scientific Editing (www.alpublish.com) for its linguistic assistance during the preparation of this manuscript.

      • The authors declare that they have no conflict of interest.

      • # These authors contributed equally: Shaoming Liang, Yiling Li

      • Supplemental Table S1 Summary of the marker genes used in poplar single-cell studies. Genes highlighted in yellow indicate that they were used to identify a specific cell type in certain study, showing the relative orthologous gene in other studies.
      • Supplemental Table S2 Summary of the marker genes used in single-cell studies of woody plants. Genes highlighted in yellow indicate that they were used to identify a specific cell type in a study, showing the best hits in Arabidopsis.
      • Copyright: © 2023 by the author(s). Published by Maximum Academic Press, Fayetteville, GA. This article is an open access article distributed under Creative Commons Attribution License (CC BY 4.0), visit https://creativecommons.org/licenses/by/4.0/.
    Figure (5)  Table (1) References (155)
  • About this article
    Cite this article
    Liang S, Li Y, Chen Y, Huang H, Zhou R, et al. 2023. Application and prospects of single-cell and spatial omics technologies in woody plants. Forestry Research 3:27 doi: 10.48130/FR-2023-0027
    Liang S, Li Y, Chen Y, Huang H, Zhou R, et al. 2023. Application and prospects of single-cell and spatial omics technologies in woody plants. Forestry Research 3:27 doi: 10.48130/FR-2023-0027

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return