Data availability
Sequencing data were deposited into the NCBI Gene Expression Omnibus under GSE273803.
Code availability
The open-source software, tools, and packages used for data analysis in this study, as well as the version of each program, were R (v3.6.1), PIPseeker (v1.0.0), Seurat R package (v4.3.0), scGate R package (v1.6), ScType R package (v1.0), and SingleR R package (v1.0)57. No custom software, tools, or packages were used.
References
Ju, S. et al.…
Data availability
Sequencing data were deposited into the NCBI Gene Expression Omnibus under GSE273803.
Code availability
The open-source software, tools, and packages used for data analysis in this study, as well as the version of each program, were R (v3.6.1), PIPseeker (v1.0.0), Seurat R package (v4.3.0), scGate R package (v1.6), ScType R package (v1.0), and SingleR R package (v1.0)57. No custom software, tools, or packages were used.
References
Ju, S. et al. Detection of circulating tumor cells: opportunities and challenges. Biomark. Res 10, 58 (2022).
Habli, Z., AlChamaa, W., Saab, R., Kadara, H. & Khraiche, M. L. Circulating tumor cell detection technologies and clinical utility: challenges and opportunities. Cancers (Basel) 12, 1930 (2020).
Conte, M. I., Fuentes-Trillo, A. & Domínguez Conde, C. Opportunities and tradeoffs in single-cell transcriptomic technologies. Trends Genet. 40, 83–93 (2024).
Cohn, L. B. et al. Clonal CD4+ T cells in the HIV-1 latent reservoir display a distinct gene profile upon reactivation. Nat. Med 24, 604–609 (2018).
Pauken, K. E. et al. Single-cell analyses identify circulating anti-tumor CD8 T cells and markers for their enrichment. J. Exp. Med. 218, e20200920 (2021).
Lindner, S. et al. Altered microbial bile acid metabolism exacerbates T cell-driven inflammation during graft-versus-host disease. Nat. Microbiol 9, 614–630 (2024).
Clark, I. C. et al. Microfluidics-free single-cell genomics with templated emulsification. Nat. Biotechnol. 41, 1557–1566 (2023).
Zhang, P. et al. Single-cell RNA sequencing to track novel perspectives in HSC heterogeneity. Stem Cell Res Ther. 13, 39 (2022).
Hérault, L., Poplineau, M., Remy, E. & Duprez, E. Single cell transcriptomics to understand HSC heterogeneity and its evolution upon aging. Cells 11, 3125 (2022).
Desterke, C., Bennaceur-Griscelli, A. & Turhan, A. G. EGR1 dysregulation defines an inflammatory and leukemic program in the cell trajectory of human-aged hematopoietic stem cells (HSC). Stem Cell Res Ther. 12, 419 (2021).
Adelman, E. R. et al. Aging human hematopoietic stem cells manifest profound epigenetic reprogramming of enhancers that may predispose to leukemia. Cancer Discov. 9, 1080–1101 (2019).
Kulkarni, R. et al. Early growth response factor 1 in aging hematopoietic stem cells and leukemia. Front. Cell Dev. Biol 10, 925761 (2022).
Lawrence, R., Watters, M., Davies, C. R., Pantel, K. & Lu, Y. J. Circulating tumour cells for early detection of clinically relevant cancer. Nat. Rev. Clin. Oncol. 20, 487–500 (2023).
Lin, D. et al. Circulating tumor cells: biology and clinical significance. Signal Transduct. Target Ther. 6, 404 (2021).
Fankhauser, R. et al. Single-cell identification of melanoma biomarkers in circulating tumor cells. Cancers 14, 4921 (2022).
Marsavela, G., Aya-Bonilla, C. A., Warkiani, M. E., Gray, E. S. & Ziman, M. Melanoma circulating tumor cells: benefits and challenges required for clinical application. Cancer Lett. 424, 1–8 (2018).
Grissa, D., Junge, A., Oprea, T. I. & Jensen, L. J. Diseases 2.0: a weekly updated database of disease-gene associations from text mining and data integration. Database 2022, baac019 (2022).
Franzoni, A. et al. A polymorphic GGC repeat in the NPAS2 gene and its association with melanoma. Exp. Biol. Med. 242, 1553–1558 (2017).
Van Den Eynde, B. J., Van Baren, N. & Baurain, J. F. Is there a clinical future for ido1 inhibitors after the failure of epacadostat in melanoma? Annu. Rev. Cancer Biol. 4, 241–256 (2020).
Xu, Y. et al. Identification of differentially expressed genes and functional annotations associated with metastases of the uveal melanoma. J. Cell Biochem. 120, 19202–19214 (2019).
Silina, K. et al. Sperm-associated antigens as targets for cancer immunotherapy: Expression pattern and humoral immune response in cancer patients. J. Immunother. 34, 28–44 (2011).
Tabach, Y. et al. Amplification of the 20q chromosomal arm occurs early in tumorigenic transformation and may initiate cancer. PLoS ONE 6, e14632 (2011).
Wilson, M. A. et al. Copy number changes are associated with response to treatment with carboplatin, paclitaxel, and sorafenib in melanoma. Clin. Cancer Res. 22, 374–382 (2016).
Goldberg, E. K. et al. Localization of multiple melanoma tumor–suppressor genes on chromosome 11 by use of homozygosity mapping-of-deletions analysis. Am. J. Hum. Genet. 67, 417–431 (2000).
Li, J. et al. Comprehensive genomic analysis of primary malignant melanoma of the esophagus reveals similar genetic patterns compared with epithelium-associated melanomas. Mod. Pathol. 35, 1596–1608 (2022).
Rossi, L. et al. Less is more: unveiling the functional core of hematopoietic stem cells through knockout mice. Cell Stem Cell 11, 302–317 (2012).
Challen, G. A., Pietras, E. M., Wallscheid, N. C. & Signer, R. A. J. Simplified murine multipotent progenitor isolation scheme: establishing a consensus approach for multipotent progenitor identification. Exp. Hematol. 104, 55–63 (2021).
Andreatta, M., Berenstein, A. J. & Carmona, S. J. scGate: marker-based purification of cell types from heterogeneous single-cell RNA-seq datasets. Bioinformatics 38, 2642–2644 (2022).
Gordiienko, I., Shlapatska, L., Kovalevska, L. & Sidorenko, S. P. SLAMF1/CD150 in hematologic malignancies: silent marker or active player?. Clin. Immunol. 204, 14–22 (2019).
Kim, K. M. et al. Taz protects hematopoietic stem cells from an aging-dependent decrease in PU.1 activity. Nat. Commun. 13, 5187 (2022).
de Haan, G., Nijhof, W. & Van Zant, G. Mouse strain-dependent changes in frequency and proliferation of hematopoietic stem cells during aging: correlation between lifespan and cycling activity. Blood 89, 1543–1550 (1997).
Kucinski, I. et al. A time- and single-cell-resolved model of murine bone marrow hematopoiesis. Cell Stem Cell 31, 244–259.e10 (2024).
Somuncular, E. et al. CD49b identifies functionally and epigenetically distinct subsets of lineage-biased hematopoietic stem cells. Stem Cell Rep. 17, 1546–1560 (2022).
Eich, M., Trumpp, A. & Schmitt, S. OMIP-059: identification of mouse hematopoietic stem and progenitor cells with simultaneous detection of CD45.1/2 and controllable green fluorescent protein expression by a single staining panel. Cytometry A 95, 1049–1052 (2019).
Hérault, L. et al. Single-cell RNA-seq reveals a concomitant delay in differentiation and cell cycle of aged hematopoietic stem cells. BMC Biol. 19, 19 (2021).
Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).
Zhang, L., Mack, R., Breslin, P. & Zhang, J. Molecular and cellular mechanisms of aging in hematopoietic stem cells and their niches. J. Hematol. Oncol. 13, 157 (2020).
Young, K. et al. Decline in IGF1 in the bone marrow microenvironment initiates hematopoietic stem cell aging. Cell Stem Cell 28, 1473–1482.e7 (2021).
Bogeska, R. et al. Inflammatory exposure drives long-lived impairment of hematopoietic stem cell self-renewal activity and accelerated aging. Cell Stem Cell 29, 1273–1284.e8 (2022).
Mann, M. et al. Heterogeneous responses of hematopoietic stem cells to inflammatory stimuli are altered with age. Cell Rep. 25, 2992–3005.e5 (2018).
Ge, S. X., Jung, D. & Yao, R. ShinyGO: a graphical gene-set enrichment tool for animals and plants. Bioinformatics 36, 2628–2629 (2020).
Sun, D. et al. Epigenomic profiling of young and aged hscs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 14, 673–688 (2014).
Keenan, A. B. et al. ChEA3: transcription factor enrichment analysis by orthogonal omics integration. Nucleic Acids Res. 47, W212–W224 (2019).
Min, I. M. et al. The transcription factor EGR1 controls both the proliferation and localization of hematopoietic stem cells. Cell Stem Cell 2, 380–391 (2008).
Bandura, D. R. et al. Mass cytometry: technique for real-time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem. 81, 6813–6822 (2009).
Shahi, P., Kim, S. C., Haliburton, J. R., Gartner, Z. J. & Abate, A. R. Abseq: Ultrahigh-throughput single cell protein profiling with droplet microfluidic barcoding. Sci. Rep. 7, 44447 (2017).
Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).
Keller, L. & Pantel, K. Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nat. Rev. Cancer 19, 553–567 (2019).
Descamps, L., Le Roy, D. & Deman, A.-L. Microfluidic-based technologies for ctc isolation: a review of 10 years of intense efforts towards liquid biopsy. Int J. Mol. Sci. 23, 1981 (2022).
Cheng, Y.-H. et al. Hydro-seq enables contamination-free high-throughput single-cell RNA-sequencing for circulating tumor cells. Nat. Commun. 10, 2163 (2019).
Stoddart, A., Fernald, A. A., Davis, E. M., McNerney, M. E. & Le Beau, M. M. EGR1 haploinsufficiency confers a fitness advantage to hematopoietic stem cells following chemotherapy. Exp. Hematol. 115, 54–67 (2022).
Joslin, J. M. et al. Haploinsufficiency of EGR1, a candidate gene in the del(5q), leads to the development of myeloid disorders. Blood 110, 719–726 (2007).
Abay, T. et al. Transcript-specific enrichment enables profiling of rare cell states via single-cell RNA sequencing. Nat. Genet. 57, 451–460 (2025).
Shin, S. W. et al. FIND-seq: high-throughput nucleic acid cytometry for rare single-cell transcriptomics. Nat. Protoc. 19, 3191–3218 (2024).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
Ianevski, A., Giri, A. K. & Aittokallio, T. Fully-automated and ultra-fast cell-type identification using specific marker combinations from single-cell transcriptomic data. Nat. Commun. 13, 1246 (2022).
Aran, D. et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 20, 163–172 (2019).
Acknowledgments
This work was supported by grants R01AI149699 and R01NS130876. I.F-M. was supported by a postgraduate fellowship from the La Caixa Foundation, a Momentum Fellowship from the Mark Foundation for Cancer Research, a Scholarship of Excellence Rafael del Pino, and an NCI F99 award (CA284253-01). R.L.B. was supported by a Damon Runyon-Sohn Fellowship and the NCI (K99CA248460). R.L.L. was supported by a Memorial Sloan Kettering Cancer Center Support Grant/Core Grant P30 CA088748, an R35 grant from the National Institute of Cancer (CA197594), and a collaborative U01 Research Project grant from the National Institute of Aging—the U01 grant was jointly received with the laboratory of Jennifer Trowbridge at the Jackson Laboratories (U01AG077925; 210374-0622-02). We are grateful to members of the Abate laboratory for helpful discussions. We thank Eric Chow and the staff of the UCSF Center for Advanced Technology for their technical support, the members of the Flow Cytometry and the Integrated Genomics Operation (IGO) cores at Memorial Sloan Kettering Cancer Center for their advice and technical help, Kristina Fontanez, Autumn Cholger and Bob Wikle from Fluent BioSciences for their advice and support.
Author information
Author notes
These authors contributed equally: Sixuan Pan, Inés Fernández-Maestre.
Authors and Affiliations
Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
Sixuan Pan, Kai-Chun Chang & Adam R. Abate 1.
Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Inés Fernández-Maestre, Matthew G. Wereski & Ross L. Levine 1.
Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Inés Fernández-Maestre 1.
Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Stéphane Van Haver 1.
Tow Center for Developmental Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Stéphane Van Haver 1.
Department of Medicine, Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA, USA
Alexandra M. Haugh, Katy K. Tsai & Adil I. Daud 1.
Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
Robert L. Bowman 1.
Departments of Radiation Oncology and Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
Harish N. Vasudevan 1.
Department of Medicine, Weill Cornell Medical College, New York, NY, USA
Ross L. Levine 1.
Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
Ross L. Levine
Authors
- Sixuan Pan
- Inés Fernández-Maestre
- Kai-Chun Chang
- Stéphane Van Haver
- Matthew G. Wereski
- Alexandra M. Haugh
- Katy K. Tsai
- Adil I. Daud
- Robert L. Bowman
- Harish N. Vasudevan
- Ross L. Levine
- Adam R. Abate
Contributions
S.P., K.C., and A.R.A. designed the study; S.P. and K.C. optimized the PURE-seq workflow; I.F.-M. designed and performed the experiments for all mouse studies; S.P., A.M.H, K.K.T, A.I.D., and H.N.V. conducted the CTC study; I.F.-M, K.C., and S.P. analyzed scRNA-seq data; S.V.H. provided bioinformatic and data curation support; M.G.W. assisted with mouse dissections and sample processing; R.L.B. provided input on data visualization; S.P. and I.F.-M. wrote the manuscript; A.R.A. and R.L.L. supervised the work and revised the manuscript; all authors read, reviewed, and approved the manuscript.
Corresponding authors
Correspondence to Ross L. Levine or Adam R. Abate.
Ethics declarations
Competing interests
R.L.L. is on the Supervisory board of Qiagen (compensation/equity), a co-founder/board member at Ajax (equity), and is a scientific advisor to Mission Bio, Syndax, Scorpion, Zentalis, Auron, Prelude, and C4 Therapeutics; for each of these entities, he receives equity/compensation. He has received research support from the Cure Breast Cancer Foundation, Calico, Zentalis and Ajax, and has consulted for Jubilant, Goldman Sachs, Incyte, AstraZeneca and Janssen. The remaining authors declare no competing interests.
Peer review
Peer review information
Nature Communications thanks Iannis Aifantis, Leif Ludwig and the other anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Pan, S., Fernández-Maestre, I., Chang, KC. et al. PURE-seq integrates FACS and PIP-seq for single-cell genomics of ultra-rare cells. Nat Commun (2026). https://doi.org/10.1038/s41467-025-68146-w
Received: 05 August 2024
Accepted: 19 December 2025
Published: 21 January 2026
DOI: https://doi.org/10.1038/s41467-025-68146-w