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.

  • Brief Communication
  • Published:

Molecular mechanocytometry using tension-activated cell tagging

Abstract

Flow cytometry is used routinely to measure single-cell gene expression by staining cells with fluorescent antibodies and nucleic acids. Here, we present tension-activated cell tagging (TaCT) to label cells fluorescently based on the magnitude of molecular force transmitted through cell adhesion receptors. As a proof-of-concept, we analyzed fibroblasts and mouse platelets after TaCT using conventional flow cytometry.

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: Tension-activated cell tagging.
Fig. 2: TaCT detecting mechanically active cells in a mixed population.

Similar content being viewed by others

Data availability

A data summary file containing all individual replicate data from the main text, all individual replicate data from the supporting information and all P values from statistical analyses performed in this manuscript is provided at https://osf.io/hkrve/?view_only=cc90b513cb5547358c652e5ace836106. Source data are provided with this paper.

Code availability

Code used for oxDNA data analysis and graph generation is publicly available online at https://github.com/SalaitaLab/Tension_activated_cell_tagging.

References

  1. Di Carlo, D. A mechanical biomarker of cell state in medicine. J. Lab. Autom. 17, 32–42 (2012).

    Article  PubMed  Google Scholar 

  2. Torrino, S. & Bertero, T. Metabo-reciprocity in cell mechanics: feeling the demands/feeding the demand. Trends Cell Biol. 32, 624–636 (2022).

    Article  CAS  PubMed  Google Scholar 

  3. Evers, T. M. J. et al. Reciprocal regulation of cellular mechanics and metabolism. Nat. Metab. 3, 456–468 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Belotti, Y. et al. High-throughput, time-resolved mechanical phenotyping of prostate cancer cells. Sci. Rep. 9, 5742 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Islam, M. et al. Microfluidic cell sorting by stiffness to examine heterogenic responses of cancer cells to chemotherapy. Cell Death Dis. 9, 239 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Lee, K. C. M. et al. Toward deep biophysical cytometry: prospects and challenges. Trends Biotechnol. 39, 1249–1262 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Otto, O. et al. Real-time deformability cytometry: on-the-fly cell mechanical phenotyping. Nat. Methods 12, 199–202 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Kozminsky, M. & Sohn, L. L. The promise of single-cell mechanophenotyping for clinical applications. Biomicrofluidics 14, 031301 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Tse, H. T. et al. Quantitative diagnosis of malignant pleural effusions by single-cell mechanophenotyping. Sci. Transl. Med. 5, 212ra163 (2013).

    Article  PubMed  Google Scholar 

  10. Thery, M. & Bornens, M. Get round and stiff for mitosis. HFSP J. 2, 65–71 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Fletcher, D. A. & Mullins, R. D. Cell mechanics and the cytoskeleton. Nature 463, 485–492 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sanyour, H. J. et al. Membrane cholesterol and substrate stiffness co-ordinate to induce the remodelling of the cytoskeleton and the alteration in the biomechanics of vascular smooth muscle cells. Cardiovasc. Res. 115, 1369–1380 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Guo, M. et al. Cell volume change through water efflux impacts cell stiffness and stem cell fate. Proc. Natl Acad. Sci. USA 114, E8618–E8627 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Stabley, D. et al. Visualizing mechanical tension across membrane receptors with a fluorescent sensor. Nat. Methods. https://doi.org/10.1038/nmeth.1747 (2011).

  15. Brockman, J. M. Live-cell super-resolved PAINT imaging of piconewton cellular traction forces. Nat. Methods. https://doi.org/10.1038/s41592-020-0929-2 (2020).

  16. Ma, R. et al. DNA probes that store mechanical information reveal transient piconewton forces applied by T cells, Proc. Natl Acad. Sci. USA. https://doi.org/10.1073/pnas.1904034116 (2019).

  17. Brockman, J. M. et al. Mapping the 3D orientation of piconewton integrin traction forces. Nat. Methods https://doi.org/10.1038/nmeth.4536 (2018).

  18. Hang, X. et al. High-throughput DNA tensioner platform for interrogating mechanical heterogeneity of single living cells. Small 18, e2106196 (2022).

    Article  PubMed  Google Scholar 

  19. Pushkarsky, I. et al. Elastomeric sensor surfaces for high-throughput single-cell force cytometry. Nat. Biomed. Eng. 2, 124–137 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yang, Z. et al. The kinetics of force-dependent hybridization and strand-peeling of short DNA fragments. Sci. China Phys. Mech. 59, 192 (2016).

    Article  Google Scholar 

  21. Cocco, S. et al. Overstretching and force-driven strand separation of double-helix DNA. Phys. Rev. E 70, 011910 (2004).

    Article  Google Scholar 

  22. Zhang, X. et al. Two distinct overstretched DNA structures revealed by single-molecule thermodynamics measurements. Proc. Natl Acad. Sci. USA 109, 8103–8108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Snodin, B. E. et al. Introducing improved structural properties and salt dependence into a coarse-grained model of DNA. J. Chem. Phys. 142, 234901 (2015).

    Article  PubMed  Google Scholar 

  24. Zhang, Y. et al. DNA-based digital tension probes reveal integrin forces during early cell adhesion. Nat. Commun. 5, 5167 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Galush, W. J., Nye, J. A. & Groves, J. T. Quantitative fluorescence microscopy using supported lipid bilayer standards. Biophys. J. 95, 2512–2519 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Martino, F. et al. Cellular mechanotransduction: from tension to function. Front Physiol. 9, 824 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Arnott, P. M. et al. Dynamic interactions between lipid-tethered DNA and phospholipid membranes. Langmuir 34, 15084–15092 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. You, M. et al. DNA probes for monitoring dynamic and transient molecular encounters on live cell membranes. Nat. Nanotechnol. 12, 453–459 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhang, Y. et al. Platelet integrins exhibit anisotropic mechanosensing and harness piconewton forces to mediate platelet aggregation. Proc. Natl Acad. Sci. USA 115, 325–330 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Duan, Y. et al. Mechanically triggered hybridization chain reaction. Angew. Chem. Int. Ed. Engl. 60, 19974–19981 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ishizaki, T. et al. Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol. Pharmacol. 57, 976–983 (2000).

    CAS  PubMed  Google Scholar 

  32. Uehata, M. et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature 389, 990–994 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Austen, K. et al. Extracellular rigidity sensing by talin isoform-specific mechanical linkages. Nat. Cell Biol. 17, 1597–1606 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Engel, M. C. et al. Force-induced unravelling of DNA origami. ACS Nano 12, 6734–6747 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

K.S. acknowledges support from the National Institutes of Health through the National Institute of General Medical Sciences (NIGMS) grants RM1GM145394 and R01GM131099 and the National Institute of Allergy and Infectious Diseases grant no. AI172452.

Author information

Authors and Affiliations

Authors

Contributions

R.M. and K.S. conceived the idea and wrote the manuscript. R.M., A.V., S.A.R. and B.R.D. carried out experiments and data analysis. A.V. performed oxDNA simulation. W.C., R.L. and B.P. provided mouse platelets. All authors contributed to editing the manuscript.

Corresponding author

Correspondence to Khalid Salaita.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Madhura Mukhopadhyay, in collaboration with the Nature Methods team.

Additional information

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

Extended data

Extended Data Fig. 1 Mapping integrin forces with peeling probe.

(a) Scheme showing that when integrin is in the inactive state, the peeling probe remains in the duplex form. As integrin binds and pulls on the RGD ligand, if F < Fpeel, the peeling strand is intact, and if F > Fpeel, the duplex dehybridizes and the Cy3B fluorescence turns on. FA proteins such as vinculin, talin, and FAK, as well as actin cytoskeleton participate extensively during the integrin force generation and mechanotransduction. (b) Histogram of 227 NIH3T3 cells showing the distribution of average %peel/µm2 per cell after 1 h incubation on peeling probe substrate. Data acquired from 3 biological replicates, bin width = 0.1%. (c) Representative microscopy images show that GFP-vinculin colocalized with integrin tension signal. Images were acquired with MEF GFP-vinculin cells that were cultured on peeling probe substrate (n = 3). (d) Representative microscopy images show that the phosphorylated FAK (pY397) colocalized with fixed integrin tension signal. MEF cells were incubated on peeling probe substrate for 40–45min, fixed and stained with Rabbit anti-FAK pY397 and Alexa488 labeled secondary antibody, followed by imaging (n = 3). (e) Representative microscopy images show that the actin stress fibers colocalized with fixed tension signal (n = 3). MEF cells were incubated on peeling probe substrate for 60–90 min, fixed and stained with Alexa647-phalloidin, followed by imaging.

Source data

Extended Data Fig. 2 Peeling probe does not perturbate mechanotransduction unlike TGTs.

(a) Schematics comparing the mechanism of TaCT/peeling probe and TGTs. Peeling probe maps integrin tension greater than 41 pN as the BHQ2 strand is separated, and the RGD anchor remains despite the duplex dehybridization. In contrast, TGTs map integrin forces greater than 56 or 12 pN in the shearing or unzipping geometry. The force-induced rupture of the duplex generates a Cy3B turn-on fluorescence signal as the top BHQ2 strand separates from the Cy3B anchor strand. Unlike peeling strand, the loss of the RGD anchors in TGTs terminates mechanotransduction through integrins. (b) Representative microcopy images of NIH3T3 cells incubated on peeling probe or TGT substrates for ~45–60 min (n = 3). Second row of images show the zoom-in view of the ROIs marked with the yellow dashed box. Intensity bar for Cy3B image indicates the peeling/rupturing percentage of the probes. Scale bar = 10 µm. (c) Microscopy images of NIH3T3 cells incubated on peeling probe or TGT substrates for ~90 min, fixed and stained for nucleus (DAPI), actin (SirActin), and YAP (Alexa488 conjugated antibody). Scale bar = 5 µm. Intensity bar indicates the gray value. (d) Quantitative analysis of the spreading area, %peel or %rupture, and YAP translocation for NIH3T3 cells incubated on three substrates for 60 min. For spreading area and tension quantification, data was collected from 3 biological replicates (n = 227, 150, and 105 for cells on peeling probe, 56 pN TGT, and 12 pN TGT substrate). For analysis on YAP translocation, images in DAPI channel were used as masks to quantify the mean fluorescence intensity of nuclear YAP and cytoplasm YAP. Data was acquired from 3 biological replicates (total n = 134, 98, and 86 cells for peeling probe, 56 pN TGT, and 12 pN TGT substrate). Plots show lines at the median and interquartile values. Statistical analysis was performed with one-way ANOVA and Tukey’s multiple comparison test.

Source data

Extended Data Fig. 3 Lat B inhibition of cells.

(a) Scheme showing that Lat B early treatment inhibits integrin force generation by inhibiting actin polymerization. (b) Representative RICM and Cy3B microscopy images of MEF cells incubated on peeling probe substrate after early Lat B treatment. Cells were treated with 20 µM Lat B after 15 min of plating on the substrate and imaged after 45 min of incubation. Scale bar = 10 µm. (c) Scheme showing that when integrin force signals are already generated on peeling probe substrate, if the force transmission is terminated by late Lat B treatment, with additional BHQ2 peeling strand the peeling probe can rehybridize to the duplex form. (d) Representative RICM and raw Cy3B microscopy images of MEF cells treated with 20 µM Lat B 50 min after seeding. The tension signal remained after 10 min of Lat B treatment and diminished after the addition of excess BHQ2 peeling strand. Scale bar = 5 µm. (e) Quantitative analysis of tension signal changes after Lat B treatment and the addition of BHQ2 peeling strand in n = 30 cells. (f) Linescan of the ROI (yellow dashed line) in (D) before Lat B treatment, with Lat B treatment, and with excess BHQ2 peeling strand.

Source data

Extended Data Fig. 4 Cholesterol DNA strands association and dissociation in cells.

(a) Scheme showing flow cytometry measurements of the DNA strands uptake in solution. Mouse platelets were incubated with 50 nM of cholesterol peeling strand, TaCT duplex, load-bearing strand, TaCT duplex lacking RGD, and load-bearing strand lacking RGD in Tyrode’s buffer for 30 min and spun down three times to wash away the excess oligos. The association was measured in both Atto647N and Cy3B channels by a flow cytometer. (b) Cy3B and Atto647N median fluorescence intensity (MFI) of platelets incubated with different oligonucleotides. Data collected from 3 replicates (mean ± SEM). Statistical analysis was performed by one-way ANOVA and Tukey’s multiple comparison. (c) Scheme showing flow cytometry measurements of the concentration dependent incorporation of cholesterol peeling strand. MEF cells were incubated with 0, 1, 10, and 100 nM of cholesterol peeling strand for 1 h. The excess cholesterol peeling strand was washed away by spinning down in PBS three times, and the fluorescence intensity of cells was measured by a flow cytometer. (d) Representative histogram of cholesterol peeling strand association in cells. Atto647N MFI was plotted from 3 replicates (mean ± SD). Linear relationship between cholesterol strand concentration and cell association was found, R2 = 0.977. (e) Scheme showing the measurement of cholesterol peeling strand dissociation from the cell. NIH3T3 cells were incubated with 100 nM cholesterol peeling strand for 30 min and rinsed with PBS 3 times. Cells were divided into 6 aliquots and the remaining cholesterol strand in cells was measured every 30 min for 150 min by flow cytometry. (f) Representative histogram shows the decay of fluorescence in cells over time. The normalized MFI from 2 replicates was plotted to show the dissociation of cholesterol strand over time in NIH3T3 cells (mean ± SD).

Source data

Extended Data Fig. 5 Control experiments supporting the concept of TaCT.

(a) Representative microscopy images of NIH3T3 cells cultured on TaCT substrate, or control substrate lacking cholesterol, lacking RGD, or treated with Lat B (n = 3). Atto647N shows the signal normalized to the background intact probes. Scale bar = 10 µm. (b) Representative flow cytometry histograms show that NIH3T3 cells in TaCT, (−)cholesterol, (−)RGD, and (+)Lat B groups had indistinguishable Cy3B intensity. Quantitative analysis (mean ± SEM) shows that cells cultured on TaCT substrate had similar level of geometric mean fluorescence intensity (gMFI) compared to control groups in Cy3B channel, despite higher Atto647N gMFI. Data collected from 3 replicates for each control, and statistical analysis was performed using ratio paired two-tailed Student’s t-test. (c) Plots show the gMFI of cells after TaCT normalized to different controls in both Atto647N and Cy3B channels (n = 3, mean ± SEM). One sample t-test (two-tailed) was used for statistical analysis to test whether TaCT signal is significantly different in Atto647N and Cy3B compared to a hypothetical value of 1 (representing no TaCT signal in the corresponding control group). TaCT signal in Atto647N channel consistently provides 2 to 3 fold higher gMFI.

Source data

Extended Data Fig. 6 Spreading control for TaCT.

(a) Scheme and representative microscopy data showing that TaCT signal is not due to spreading of the cells on the substrate. NIH3T3 cells were incubated on a TaCT substrate doped with a non-fluorescent DNA duplex, or a TaCT substrate lacking RGD doped with a non-fluorescent DNA duplex presenting the RGD motif, and imaged 45 min–60 min after seeding. After confirming the cell spreading, cells were collected, rinsed, and added to non-fluorescent glass substrate to image the cholesterol peeling strand incorporation in the cells. Tension images were normalized to the background of intact probes. Scale bar = 10 µm. (b) Quantitative analysis of the cell spreading area (n = 82 and 74 cells) and cholesterol peeling strand incorporation (n = 58 and 56 cells) for cells incubated on TaCT (+)RGD or (−)RGD substrates. Data was collected from three replicates and statistical analysis was performed using two-tailed Student’s t-test.

Source data

Extended Data Fig. 7 TaCT in Platelets.

(a) Scheme and representative microscopy images showing tension mapping with peeling probe in mouse platelets (n = 3). Tension images were normalized to the background of intact peeling probes, scale bar = 5 µm. (b) Linescan of the ROI (yellow dashed line) shows anti-colocalization of Cy3B and Atto647N intensities from raw data. (c) Scheme and representative images show mouse platelets incubated on the TaCT substrate in different buffer conditions (n = 3). RGD, Ca2+, Mg2+, and ADP leads to platelets activation, and withholding any of them results in no/impaired activation. Tension images were normalized to the background of intact TaCT probes, scale bar = 5 µm.

Supplementary information

Supplementary Information

Supplementary Tables 1–4, Note and Figs. 1–11.

Reporting Summary

Peer Review File

Supplementary Video 1

Simulation of 24 bp DNA peeling with oxDNA.

Supplementary Video 2

Fibroblast cell producing integrin tension >41 pN.

Source data

Source Data Figs. 1 and 2

Source data file containing all source data with clearly named tabs for each figure.

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

Ma, R., Rashid, S.A., Velusamy, A. et al. Molecular mechanocytometry using tension-activated cell tagging. Nat Methods 20, 1666–1671 (2023). https://doi.org/10.1038/s41592-023-02030-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-02030-7

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research