Perspectives on integrating artificial intelligence and single-cell omics for cellular plasticity research

Ahmed Ghobashi , Qin Ma

Quant. Biol. ›› 2025, Vol. 13 ›› Issue (4) : e70004

PDF (195KB)
Quant. Biol. ›› 2025, Vol. 13 ›› Issue (4) : e70004 DOI: 10.1002/qub2.70004
PERSPECTIVE

Perspectives on integrating artificial intelligence and single-cell omics for cellular plasticity research

Author information +
History +
PDF (195KB)

Abstract

Cellular plasticity enables cells to dynamically adapt to environmental changes by altering their phenotype. This plasticity plays a crucial role in tissue repair and regeneration and contributes to pathological processes such as cancer metastasis. Advances in single-cell omics have significantly advanced the study of cellular states and provided new opportunities for accurate cell classification and uncovering cellular transitions. In this perspective, we emphasize integrating chromatin accessibility data and extrinsic factors, such as microenvironmental cues, with single-cell transcriptomic data to develop holistic models for identifying plastic cell states. Additionally, coupling artificial intelligence with single-cell omics offers transformative potential to address existing challenges and fill gaps in identifying and characterizing plastic cells. We envision the development of a universal plasticity metric, a standardized metric for quantifying cellular plasticity. This metric would enable consistent measurement across diverse studies, creating a unified framework that bridges fields such as developmental biology, cancer research, and regenerative medicine. Fostering innovative approaches to identifying and analyzing cellular plasticity promises not only to deepen our understanding of cellular plasticity but also to accelerate therapeutic advancements, paving the way for novel precision medicine strategies to treat complex diseases such as cancer.

Keywords

cell plasticity / epigenetics / foundation models / single-cell omics

Cite this article

Download citation ▾
Ahmed Ghobashi, Qin Ma. Perspectives on integrating artificial intelligence and single-cell omics for cellular plasticity research. Quant. Biol., 2025, 13(4): e70004 DOI:10.1002/qub2.70004

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

TataA, ChowRD, TataPR. Epithelial cell plasticity: breaking boundaries and changing landscapes. EMBO Rep. 2021;22(7):e51921.

[2]

QinX, TapeCJ. Functional analysis of cell plasticity using single-cell technologies. Trends Cell Biol. 2024;34(10):854–64.

[3]

Adkins-ThreatsM, MillsJC. Cell plasticity in regeneration in the stomach and beyond. Curr Opin Genet Dev. 2022;75:101948.

[4]

JessenKR, MirskyR, Arthur-FarrajP. The role of cell plasticity in tissue repair: adaptive cellular reprogramming. Dev Cell. 2015;34(6):613–20.

[5]

ChafferCL, SanJBP, LimE, Weinberg RA. EMT, cell plasticity and metastasis. Cancer Metastasis Rev. 2016;35(4):645–54.

[6]

YeX, Weinberg RA. Epithelial-mesenchymal plasticity: a central regulator of cancer progression. Trends Cell Biol. 2015;25(11):675–86.

[7]

CerneckisJ, CaiH, ShiY. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Ther. 2024;9(1):112.

[8]

KalluriR, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119(6):1420–8.

[9]

SequistLV, Waltman BA, Dias-SantagataD, DigumarthyS, TurkeAB, FidiasP, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3(75):75ra26.

[10]

SunL, Burnett J, GasparyanM, XuF, JiangH, LinCC, et al. Novel cancer stem cell targets during epithelial to mesenchymal transition in PTEN-deficient trastuzumab-resistant breast cancer. Oncotarget. 2016;7(32):51408–22.

[11]

KretzschmarK, WattFM. Lineage tracing. Cell. 2012;148(1–2):33–45.

[12]

LangeM, BergenV, KleinM, Setty M, ReuterB, BakhtiM, et al. CellRank for directed single-cell fate mapping. Nat Methods. 2022;19(2):159–70.

[13]

ZhouP, WangS, LiT, NieQ. Dissecting transition cells from single-cell transcriptome data through multiscale stochastic dynamics. Nat Commun. 2021;12(1):5609.

[14]

SaelensW, Cannoodt R, TodorovH, SaeysY. A comparison of single-cell trajectory inference methods. Nat Biotechnol. 2019;37(5):547–54.

[15]

LiJ, PanX, YuanY, Shen HB. TFvelo: gene regulation inspired RNA velocity estimation. Nat Commun. 2024;15(1):1387.

[16]

BergenV, LangeM, PeidliS, Wolf FA, TheisFJ. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat Biotechnol. 2020;38(12):1408–14.

[17]

SchiebingerG, ShuJ, TabakaM, Cleary B, SubramanianV, SolomonA, et al. Optimal-transport analysis of single-cell gene expression identifies developmental trajectories in reprogramming. Cell. 2019;176(4):928–43.e22.

[18]

PanD, JiaD. Application of single-cell multi-omics in dissecting cancer cell plasticity and tumor heterogeneity. Front Mol Biosci. 2021;8:757024.

[19]

MarjanovicND, HofreeM, ChanJE, Canner D, WuK, TrakalaM, et al. Emergence of a high-plasticity cell state during lung cancer evolution. Cancer Cell. 2020;38(2):229–46.e13.

[20]

MalagoliTG, WiecekAJ, WithnellE, Secrier M. Genomic and microenvironmental heterogeneity shaping epithelial-tomesenchymal trajectories in cancer. Nat Commun. 2023;14(1):789.

[21]

KongW, FuYC, HollowayEM, Garipler G, YangX, MazzoniEO, et al. Capybara: a computational tool to measure cell identity and fate transitions. Cell Stem Cell. 2022;29(4):635–49.e11.

[22]

KangM, Armenteros JJA, GulatiGS, GleyzerR, Avagyan S, BrownEL, et al. Mapping single-cell developmental potential in health and disease with interpretable deep learning. 2024. Preprint at bioRxiv: 2024.03.19.585637.

[23]

KlemmSL, Shipony Z, GreenleafWJ. Chromatin accessibility and the regulatory epigenome. Nat Rev Genet. 2019;20(4): 207–20.

[24]

Gaspar-MaiaA, AlajemA, MeshorerE, Ramalho-Santos M. Open chromatin in pluripotency and reprogramming. Nat Rev Mol Cell Biol. 2011;12(1):36–47.

[25]

MartinEW, Rodriguez YBA, ReggiardoRE, WorthingtonAK, Mattingly CS, PoscabloDM, et al. Dynamics of chromatin accessibility during hematopoietic stem cell differentiation into progressively lineage-committed progeny. Stem Cells. 2023;41(5):520–39.

[26]

GhobashiAH, KimaniJW, LadaikaCA, O’Hagan HM. PTEN depletion reduces H3K27me3 levels to promote epithelial-tomesenchymal transition in epithelial colorectal cancer cells. PLoS One. 2024;19(11):e0313769.

[27]

SunL, FangJ. Epigenetic regulation of epithelial-mesenchymal transition. Cell Mol Life Sci. 2016;73(23):4493–515.

[28]

VazM, HwangSY, KagiampakisI, PhallenJ, PatilA, O’HaganHM, et al. Chronic cigarette smoke-induced epigenomic changes precede sensitization of bronchial epithelial cells to single-step transformation by KRAS mutations. Cancer Cell. 2017;32(3):360–76.e6.

[29]

CordaniM, DandoI, AmbrosiniG, Gonzalez-Menendez P. Signaling, cancer cell plasticity, and intratumor heterogeneity. Cell Commun Signal. 2024;22(1):255.

[30]

FeinbergAP, Levchenko A. Epigenetics as a mediator of plasticity in cancer. Science. 2023;379(6632):eaaw3835.

[31]

RingNAR, Valdivieso K, GrillariJ, RedlH, Ogrodnik M. The role of senescence in cellular plasticity: lessons from regeneration and development and implications for age-related diseases. Dev Cell. 2022;57(9):1083–101.

[32]

LabergeRM, AwadP, CampisiJ, Desprez PY. Epithelialmesenchymal transition induced by senescent fibroblasts. Cancer Microenviron. 2012;5(1):39–44.

[33]

MarxV. Method of the Year: spatially resolved transcriptomics. Nat Methods. 2021;18(1):9–14.

[34]

ChanJM, ZaidiS, LoveJR, Zhao JL, SettyM, WadoskyKM, et al. Lineage plasticity in prostate cancer depends on JAK/STAT inflammatory signaling. Science. 2022;377(6611): 1180–91.

[35]

BurdziakC, Alonso-Curbelo D, WalleT, ReyesJ, Barriga FM, HavivD, et al. Epigenetic plasticity cooperates with cell-cell interactions to direct pancreatic tumorigenesis. Science. 2023;380(6645):eadd5327.

[36]

ZhangK, HockerJD, MillerM, Hou X, ChiouJ, PoirionOB, et al. A single-cell atlas of chromatin accessibility in the human genome. Cell. 2021;184(24):5985–6001.e19.

[37]

HeimbergG, KuoT, DePiantoDJ, Salem O, HeiglT, DiamantN, et al. A cell atlas foundation model for scalable search of similar human cells. Nature. 2024;638(8052): 1085–94.

[38]

TabulaSC, JonesRC, KarkaniasJ, Krasnow MA, PiscoAO, QuakeSR, et al. The Tabula Sapiens: a multiple-organ, singlecell transcriptomic atlas of humans. Science. 2022;376(6594):eabl4896.

[39]

TheodorisCV, XiaoL, ChopraA, Chaffin MD, Al SayedZR, HillMC, et al. Transfer learning enables predictions in network biology. Nature. 2023;618(7965):616–24.

[40]

RosenY, BrbicM, RoohaniY, Swanson K, LiZ, LeskovecJ. Toward universal cell embeddings: integrating single-cell RNAseq datasets across species with SATURN. Nat Methods. 2024;21(8):1492–500.

[41]

MaQ, JiangY, ChengH, Xu D. Harnessing the deep learning power of foundation models in single-cell omics. Nat Rev Mol Cell Biol. 2024;25(8):593–4.

[42]

WangX, DuanM, LiJ, MaA, XinG, XuD, et al. MarsGT: multi-omics analysis for rare population inference using single-cell graph transformer. Nat Commun. 2024;15(1):338.

[43]

NasirN, Alshabi M, Al-YateemN, RahmanSA, Arsyad Subu M, HijaziHH, et al. Review of machine learning advancements for single-cell analysis. In: 2023 IEEE 47th annual computers, software, and applications conference (COMPSAC);2023. p. 1383–7.

[44]

MaQ, XuD. Deep learning shapes single-cell data analysis. Nat Rev Mol Cell Biol. 2022;23(5):303–4.

[45]

LongY, AngKS, SethiR, Liao S, HengY, van OlstL, et al. Deciphering spatial domains from spatial multi-omics with SpatialGlue. Nat Methods. 2024;21(9):1658–67.

[46]

MaA, WangX, LiJ, WangC, XiaoT, Liu Y, et al. Single-cell biological network inference using a heterogeneous graph transformer. Nat Commun. 2023;14(1):964.

[47]

WagleMM, LongS, ChenC, Liu C, YangP. Interpretable deep learning in single-cell omics. Bioinformatics. 2024;40(6):btae374.

[48]

Huang J, Wolf G, Van Dijk D, Krishnaswamy S, editors. Trajectorynet: a dynamic optimal transport network for modeling cellular dynamics. International conference on machine learning. PMLR; 2020.

[49]

BunneC, Roohani Y, RosenY, GuptaA, ZhangX, RoedM, et al. How to build the virtual cell with artificial intelligence: priorities and opportunities.2024. Preprint at arXiv:2409 11654.

[50]

ArgelaguetR, CuomoASE, StegleO, Marioni JC. Computational principles and challenges in single-cell data integration. Nat Biotechnol. 2021;39(10):1202–15.

[51]

LahnemannD, KosterJ, SzczurekE, McCarthy DJ, HicksSC, RobinsonMD, et al. Eleven grand challenges in single-cell data science. Genome Biol. 2020;21(1):31.

[52]

AngererP, SimonL, TritschlerS, Wolf FA, FischerD, TheisFJ. Single cells make big data: new challenges and opportunities in transcriptomics. Curr Opin Syst Biol. 2017;4:85–91.

[53]

LobentanzerS, FengS, BrudererN, Maier A, DíazAG, StrangeA, et al. A platform for the biomedical application of large language models. Nat Biotechnol. 2025;43(2):166–9.

[54]

NingY, Teixayavong S, ShangY, SavulescuJ, Nagaraj V, MiaoD, et al. Generative artificial intelligence and ethical considerations in health care: a scoping review and ethics checklist. Lancet Digit Health. 2024;6(11):e848–56.

RIGHTS & PERMISSIONS

2025 The Author(s). Quantitative Biology published by John Wiley & Sons Australia, Ltd on behalf of Higher Education Press.

AI Summary AI Mindmap
PDF (195KB)

478

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/