Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Primer
  • Published:

Imaging flow cytometry

Abstract

Imaging flow cytometry combines the high-event-rate nature of flow cytometry with the advantages of single-cell image acquisition associated with microscopy. The measurement of large numbers of features from the resulting images provides rich data sets that have resulted in a wide range of novel biomedical applications. In this Primer, we discuss the typical imaging flow instrumentation, the form of data acquired and the typical analysis tools that can be applied to these data. Focusing on the first commercially available imaging flow cytometer, the ImageStream (Luminex), we use examples from the literature to discuss the progression of the analysis methods used in imaging flow cytometry. These methods start from the use of simple single-image features and multiple channel gating strategies, followed by the design and use of custom features for phenotype classification, through to powerful machine and deep-learning methods. For each of these methods, we outline the processes involved in analysing typical data sets and provide details of example applications. Finally, we discuss the current limitations of imaging flow cytometry and the innovations and new instruments that are addressing these challenges.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of imaging flow cytometry and examples of images generated.
Fig. 2: Process flow used to select in-focus, single-cell images from an acquired event set.
Fig. 3: Data analysis based on spatial information.
Fig. 4: Spatial analysis based on area masking.
Fig. 5: Spatial analysis based on morphology.

Similar content being viewed by others

References

  1. Brown, M. & Wittwer, C. Flow cytometry: principles and clinical applications in hematology. Clin. Chem. 46, 1221–1229 (2000).

    Article  Google Scholar 

  2. De Rosa, S. C., Brenchley, J. M. & Roederer, M. Beyond six colors: a new era in flow cytometry. Nat. Med. 9, 112–117 (2003).

    Article  Google Scholar 

  3. Basiji, D. A., Ortyn, W. E., Liang, L., Venkatachalam, V. & Morrissey, P. Cellular image analysis and imaging by flow cytometry. Clin. Lab. Med. 27, 653–670 (2007). Introduction to ImageStream.

    Article  Google Scholar 

  4. Maguire, O., Collins, C., O’Loughlin, K., Miecznikowski, J. & Minderman, H. Quantifying nuclear p65 as a parameter for NF-κB activation: correlation between ImageStream cytometry, microscopy, and Western blot. Cytometry A 79, 461–469 (2011).

    Article  Google Scholar 

  5. Kaur, M. & Esau, L. Two-step protocol for preparing adherent cells for high-throughput flow cytometry. Biotechniques 59, 119–126 (2015).

    Article  Google Scholar 

  6. Stavrakis, S., Holzner, G., Choo, J. & deMello, A. High-throughput microfluidic imaging flow cytometry. Curr. Opin. Biotechnol. 55, 36–43 (2019).

    Article  Google Scholar 

  7. Holzner, G. et al. High-throughput multiparametric imaging flow cytometry: toward diffraction-limited sub-cellular detection and monitoring of sub-cellular processes. Cell Rep. 34, 108824 (2021).

    Article  Google Scholar 

  8. Mikami, H. et al. Virtual-freezing fluorescence imaging flow cytometry. Nat. Commun. 11, 1162 (2020).

    Article  ADS  Google Scholar 

  9. Diebold, E. D. et al. Digitally synthesized beat frequency multiplexing for sub-millisecond fluorescence microscopy. Nat. Photonics 7, 806–810 (2013).

    Article  ADS  Google Scholar 

  10. George, T. C. et al. Distinguishing modes of cell death using the ImageStream multispectral imaging flow cytometer. Cytometry A 59, 237–245 (2004).

    Article  Google Scholar 

  11. Ortyn, W. E. et al. Extended depth of field imaging for high speed cell analysis. Cytometry A 71, 215–231 (2007).

    Article  Google Scholar 

  12. Haley Renee, P. & Raymond, K. K. Demonstration of high gain mode in combination with imaging flow cytometry for improved EV analysis. Mian Yi Xue Za Zhi 204, 15 (2020).

    Google Scholar 

  13. Vogt, R. F. Jr., Whitfield, W. E., Henderson, L. O. & Hannon, W. H. Fluorescence intensity calibration for immunophenotyping by flow cytometry. Methods 21, 289–296 (2000).

    Article  Google Scholar 

  14. Ortyn, W. E. et al. Sensitivity measurement and compensation in spectral imaging. Cytometry A 69, 852–862 (2006). Compensation of imaging flow cytometry data.

    Article  Google Scholar 

  15. Fazekas de St Groth, B., Zhu, E., Asad, S. & Lee, L. Flow cytometric detection of human regulatory T cells. Methods Mol. Biol. 707, 263–279 (2011).

    Article  Google Scholar 

  16. Holmberg-Thyden, S., Grønbæk, K., Gang, A. O., El Fassi, D. & Hadrup, S. R. A user’s guide to multicolor flow cytometry panels for comprehensive immune profiling. Anal. Biochem. 627, 114210 (2021).

    Article  Google Scholar 

  17. Dominical, V., Samsel, L. & McCoy, J. P. Jr. Masks in imaging flow cytometry. Methods 112, 917 (2017). Discussion of masks in imaging flow cytometry.

    Article  Google Scholar 

  18. Barteneva, N. S. & Vorobjev, I. A. Imaging Flow Cytometry: Methods and Protocols (Humana Press, 2015).

  19. Patterson, J. O., Swaffer, M. & Filby, A. An imaging flow cytometry-based approach to analyse the fission yeast cell cycle in fixed cells. Methods 82, 74–84 (2015).

    Article  Google Scholar 

  20. Patterson, J. O., Basu, S., Rees, P. & Nurse, P. CDK control pathways integrate cell size and ploidy information to control cell division. eLife 10, e64592 (2021).

    Article  Google Scholar 

  21. Patterson, J. O., Rees, P. & Nurse, P. Noisy cell-size-correlated expression of cyclin B drives probabilistic cell-size homeostasis in fission yeast. Curr. Biol. 29, 1379–1386.e1374 (2019).

    Article  Google Scholar 

  22. Calvert, M. E. K., Lannigan, J. A. & Pemberton, L. F. Optimization of yeast cell cycle analysis and morphological characterization by multispectral imaging flow cytometry. Cytometry A 73, 825–833 (2008). Cell cycle analysis using imaging flow cytometry using image properties.

    Article  Google Scholar 

  23. Filby, A. et al. An imaging flow cytometric method for measuring cell division history and molecular symmetry during mitosis. Cytometry A 79, 496–506 (2011).

    Article  Google Scholar 

  24. Summers, H. D. et al. Statistical analysis of nanoparticle dosing in a dynamic cellular system. Nat. Nanotechnol. 6, 170–174 (2011).

    Article  ADS  Google Scholar 

  25. Bourton, E. C. et al. Multispectral imaging flow cytometry reveals distinct frequencies of γ-H2AX foci induction in DNA double strand break repair defective human cell lines. Cytometry A 81, 130–137 (2012).

    Article  Google Scholar 

  26. Jurgielewicz, B. J., Yao, Y. & Stice, S. L. Kinetics and specificity of HEK293T extracellular vesicle uptake using imaging flow cytometry. Nanoscale Res. Lett. 15, 170 (2020).

    Article  ADS  Google Scholar 

  27. Fei, C., Lillico, D. M. E., Hall, B., Rieger, A. M. & Stafford, J. L. Connected component masking accurately identifies the ratio of phagocytosed and cell-bound particles in individual cells by imaging flow cytometry. Cytometry A 91, 372–381 (2017).

    Article  Google Scholar 

  28. Vranic, S. et al. Deciphering the mechanisms of cellular uptake of engineered nanoparticles by accurate evaluation of internalization using imaging flow cytometry. Part. Fibre Toxicol. 10, 2 (2013).

    Article  Google Scholar 

  29. Chivukula, R. R. et al. A human ciliopathy reveals essential functions for NEK10 in airway mucociliary clearance. Nat. Med. 26, 244–251 (2020).

    Article  Google Scholar 

  30. Blasi, T. et al. Label-free cell cycle analysis for high-throughput imaging flow cytometry. Nat. Commun. 7, 10256 (2016). Application of machine learning to imaging flow data.

    Article  ADS  Google Scholar 

  31. Nassar, M. et al. Label-free identification of white blood cells using machine learning. Cytometry A 95, 836–842 (2019).

    Article  Google Scholar 

  32. Lippeveld, M. et al. Classification of human white blood cells using machine learning for stain-free imaging flow cytometry. Cytometry A 97, 308–319 (2020).

    Article  Google Scholar 

  33. Carpenter, A. E. et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 7, R100 (2006).

    Article  Google Scholar 

  34. Hennig, H. et al. An open-source solution for advanced imaging flow cytometry data analysis using machine learning. Methods 112, 201–210 (2017).

    Article  Google Scholar 

  35. Ding, C. & Peng, H. Minimum redundancy feature selection from microarray gene expression data. in Computational Systems Bioinformatics. CSB2003. Proceedings of the 2003 IEEE Bioinformatics Conference (IEEE, 2003).

  36. Mandal, M. & Mukhopadhyay, A. An improved minimum redundancy maximum relevance approach for feature selection in gene expression data. Proc. Technol. 10, 20–27 (2013).

    Article  Google Scholar 

  37. Rees, P., Wills, J. W., Brown, M. R., Barnes, C. M. & Summers, H. D. The origin of heterogeneous nanoparticle uptake by cells. Nat. Commun. 10, 2341 (2019).

    Article  ADS  Google Scholar 

  38. Cerveira, J., Begum, J., Di Marco Barros, R., van der Veen, A. G. & Filby, A. An imaging flow cytometry-based approach to measuring the spatiotemporal calcium mobilisation in activated T cells. J. Immunol. Methods 423, 120–130 (2015).

    Article  Google Scholar 

  39. Piasecka, J. et al. Diffusion mapping of eosinophil-activation state. Cytometry A 97, 253–258 (2019).

    Article  Google Scholar 

  40. Eulenberg, P. et al. Reconstructing cell cycle and disease progression using deep learning. Nat. Commun. 8, 463 (2017). Application of deep learning to imaging flow data.

    Article  ADS  Google Scholar 

  41. Dunker, S., Boho, D., Wäldchen, J. & Mäder, P. Combining high-throughput imaging flow cytometry and deep learning for efficient species and life-cycle stage identification of phytoplankton. BMC Ecol. 18, 51 (2018).

    Article  Google Scholar 

  42. Dunker, S. et al. Pollen analysis using multispectral imaging flow cytometry and deep learning. New Phytol. 229, 593–606 (2021).

    Article  Google Scholar 

  43. Luo, S. et al. Deep learning-enabled imaging flow cytometry for high-speed Cryptosporidium and Giardia detection. Cytometry A 99, 1123–1133 (2021).

    Article  Google Scholar 

  44. Rodrigues, M. A., Beaton-Green, L. A., Kutzner, B. C. & Wilkins, R. C. Automated analysis of the cytokinesis-block micronucleus assay for radiation biodosimetry using imaging flow cytometry. Radiat. Environ. Biophys. 53, 273–282 (2014).

    Article  Google Scholar 

  45. Rodrigues, M. A., Probst, C. E., Beaton-Green, L. A. & Wilkins, R. C. Optimized automated data analysis for the cytokinesis-block micronucleus assay using imaging flow cytometry for high throughput radiation biodosimetry. Cytometry A 89, 653–662 (2016).

    Article  Google Scholar 

  46. Verma, J. R. et al. Evaluation of the automated MicroFlow® and MetaferTM platforms for high-throughput micronucleus scoring and dose response analysis in human lymphoblastoid TK6 cells. Arch. Toxicol. 91, 2689–2698 (2017).

    Article  Google Scholar 

  47. Rodrigues, M. A. Automation of the in vitro micronucleus assay using the Imagestream imaging flow cytometer. Cytometry A 93, 706–726 (2018).

    Article  Google Scholar 

  48. Rodrigues, M. A. et al. The in vitro micronucleus assay using imaging flow cytometry and deep learning. npj Syst. Biol. Appl. 7, 20 (2021).

    Article  Google Scholar 

  49. Wills, J. W. et al. Inter-laboratory automation of the in vitro micronucleus assay using imaging flow cytometry and deep learning. Arch. Toxicol. 95, 3101–3115 (2021).

    Article  Google Scholar 

  50. Krizhevsky, A., Sutskever, I. & Hinton, G. E. ImageNet classification with deep convolutional neural networks. Commun. ACM 60, 84–90 (2017).

    Article  Google Scholar 

  51. He, K., Zhang, X., Ren, S. & Sun, J. Deep residual learning for image recognition. in 2016 IEEE Conference on Computer Vision and Pattern Recognition (CVPR) (IEEE, 2016).

  52. Simonyan, K. & Zisserman, A. Very deep convolutional networks for large-scale image recognition. Preprint at https://doi.org/10.48550/arXiv.1409.1556 (2014).

  53. Mochalova, E. N., Kotov, I. A., Lifanov, D. A., Chakraborti, S. & Nikitin, M. P. Imaging flow cytometry data analysis using convolutional neural network for quantitative investigation of phagocytosis. Biotechnol. Bioeng. 119, 626–635 (2022).

    Article  Google Scholar 

  54. Probst, C., Zayats, A., Venkatachalam, V. & Davidson, B. Advanced characterization of silicone oil droplets in protein therapeutics using artificial intelligence analysis of imaging flow cytometry data. J. Pharm. Sci. 109, 2996–3005 (2020).

    Article  Google Scholar 

  55. Spidlen, J., Breuer, K. & Brinkman, R. Preparing a minimum information about a flow cytometry experiment (MIFlowCyt) compliant manuscript using the International Society for Advancement of Cytometry (ISAC) FCS file repository (FlowRepository.org). Curr. Protoc. Cytom. 61, 10.18.1–10.18.26 (2012).

    Google Scholar 

  56. Filby, A. & Davies, D. Reporting imaging flow cytometry data for publication: why mask the detail? Cytometry A 81, 637–642 (2012).

    Article  Google Scholar 

  57. Caicedo, J. C. et al. Data-analysis strategies for image-based cell profiling. Nat. Methods 14, 849–863 (2017).

    Article  Google Scholar 

  58. Montero Llopis, P. et al. Best practices and tools for reporting reproducible fluorescence microscopy methods. Nat Methods 18, 1463–1476 (2021).

    Article  Google Scholar 

  59. Doan, M. et al. Deepometry, a framework for applying supervised and weakly supervised deep learning to imaging cytometry. Nat. Protoc. 16, 3572–3595 (2021).

    Article  Google Scholar 

  60. Demont, Y. Tools for Imaging Flow Cytometry [R package IFC version 0.1.2] (2021).

  61. Schraivogel, D. et al. High-speed fluorescence image–enabled cell sorting. Science 375, 315320 (2022).

    Article  Google Scholar 

  62. Gu, Y. et al. Machine learning based real-time image-guided cell sorting and classification. Cytometry A 95, 499–509 (2019).

    Article  Google Scholar 

  63. Nitta, N. et al. Intelligent image-activated cell sorting. Cell 175, 266–276.e213 (2018). Image-based flow cytometry sorting.

    Article  Google Scholar 

  64. Nawaz, A. A. et al. Intelligent image-based deformation-assisted cell sorting with molecular specificity. Nat. Methods 17, 595–599 (2020).

    Article  MathSciNet  Google Scholar 

  65. Merola, F. et al. Tomographic flow cytometry by digital holography. Light Sci. Appl. 6, e16241 (2017).

    Article  Google Scholar 

  66. Kebschull, J. M. & Zador, A. M. Cellular barcoding: lineage tracing, screening and beyond. Nat. Methods 15, 871–879 (2018).

    Article  Google Scholar 

  67. Rees, P. et al. Nanoparticle vesicle encoding for imaging and tracking cell populations. Nat. Methods 11, 1177–1181 (2014).

    Article  Google Scholar 

  68. Han, Y. & Lo, W.-H. Imaging cells in flow cytometer using spatial-temporal transformation. Sci. Rep. 5, 13267 (2015).

    Article  ADS  Google Scholar 

  69. Lee, K. C. M. et al. Quantitative phase imaging flow cytometry for ultra-large-scale single-cell biophysical phenotyping. Cytometry A 95, 510–520 (2019).

    Article  Google Scholar 

  70. Ouk, C., Jayat-Vignoles, C., Donnard, M. & Feuillard, J. Both CD62 and CD162 antibodies prevent formation of CD36-dependent platelets, rosettes, and artefactual pseudoexpression of platelet markers on white blood cells: a study with ImageStream®. Cytometry A 79, 477–484 (2011).

    Article  Google Scholar 

  71. Ugawa, M. et al. In silico-labeled ghost cytometry. eLife 10, e67660 (2021).

    Article  Google Scholar 

  72. Rosenburg, C. A. Exploring dyserythropoiesis in patients with myelodysplasticsyndrome by imaging flow cytometry and machine-learning assisted morphometrics. Cytometry 100, 554–567 (2021).

    Article  Google Scholar 

Download references

Acknowledgements

P.R. and H.D.S. acknowledge the UK Engineering and Physical Sciences Research Council (EP/N013506/1) and UK Biotechnology and Biological Sciences Research Council (BB/P026818/1) for supporting this work. A.E.C. acknowledges the National Science Foundation (DBI 1458626) and the NIH (R35 GM122547) for supporting this work.

Author information

Authors and Affiliations

Authors

Contributions

Introduction (H.D.S., A.E.C., M.D. and P.R.); Experimentation (H.D.S., A.E.C. and P.R.); Results (H.D.S., A.F., A.E.C. and P.R.); Applications (H.D.S., A.F., A.E.C. and P.R.); Reproducibility and data deposition (H.D.S., A.F., A.E.C. and P.R.); Limitations and optimizations (H.D.S., A.E.C., M.D. and P.R.); Outlook (H.D.S., A.E.C., M.D. and P.R.).

Corresponding author

Correspondence to Paul Rees.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Reviews Method Primers thanks Maik Herbig and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

FigShare: https://figshare.com/

FlowRepository: https://flowrepository.org/

GitHub: https://github.com/

Luminex: https://www.luminexcorp.com/eu/imaging-flow-cytometry/

Supplementary information

Glossary

Brightfield

The simplest form of microscopy, in which the image is formed by white light that is transmitted through the sample and then captured on a detector.

Darkfield

In the context of imaging flow cytometry, the darkfield image is formed when light scattered from the cell is collected on the detector perpendicular to the excitation direction.

Gating

A range of bins for the histogram or a polygon for the scatter plot. This process selects cells for further analysis. The gating process can be repeated to define phenotypes that require more than two markers for identification.

Multi-spectral images

An image data set in which the same field of view is imaged in different spectral bands.

Masks

A mask is a binary image that defines the extent of the object in an image; the pixel values in the image are 1 inside the object perimeter and 0 elsewhere to represent the background.

Raw maximum pixel

A feature in the ImageStream Data Exploration and Analysis Software that returns the maximum pixel value in an image acquired by the detector before any compensation. This is often used to set the laser excitation intensity to ensure that the pixel values are not saturated.

Building block

Suggested feature scatter plots and gating strategies to help the user with simple analysis and preprocessing tasks, such as determining in-focus cells in the ImageStream Data Exploration and Analysis software.

Aspect ratio

The ratio of the minor axis and the major axis. The major axis is the longest line that can be drawn through the shape, and the minor axis is the shortest line that can be drawn through the shape at right angles to the major axis.

AND mask operation

The AND operator applied to two masks delivers the overlapped shared area between the masks.

NOT mask operation

The NOT operator is a logic operator that delivers the inverse of a mask, that is, 0s become 1s.

Confusion matrix

A confusion matrix is used to compare the predicted outcome of a machine-learning algorithm with the known classes of the data. Diagonal elements represent the number of correct classifications. Off-diagonal elements can be used to assess misclassifications.

t-Distributed stochastic neighbour embedding

An algorithm used to visualize high-dimensional data sets in two or three dimensions. Nonlinear dimensional reduction of the data to the 2D–3D coordinate system is used to preserve the distances between similar and dissimilar data points.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rees, P., Summers, H.D., Filby, A. et al. Imaging flow cytometry. Nat Rev Methods Primers 2, 86 (2022). https://doi.org/10.1038/s43586-022-00167-x

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43586-022-00167-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing