Skip to main content
Log in

Development of microfluidic platform capable of characterizing cytoplasmic viscosity, cytoplasmic conductivity and specific membrane capacitance of single cells

  • Research Paper
  • Published:
Microfluidics and Nanofluidics Aims and scope Submit manuscript

Abstract

This paper presents a microfluidic platform capable of characterizing cytoplasmic viscosity μcy, cytoplasmic conductivity σcy and specific membrane capacitance Csm of single cells continuously. A travelling cell is forced to squeeze through a major microfluidic constriction channel and aspirated into a side microfluidic constriction channel with cell aspiration length and impedance variations captured and translated into intrinsic markers of μcy, σcy and Csm based on an equivalent biophysical model. As a demonstration, μcy, σcy and Csm of hundreds of HL-60 cells that were native or treated by Cytochalasin D (CD for cytoskeleton modulation) or Concanavalin A (ConA for membrane regulation) were quantified where high success rates of cell type classification were found, which were 88.0% for HL-60 cells vs. HL-60 + CD cells and 75.6% for HL-60 cells vs. HL-60 + ConA cells. Furthermore, the microfluidic system was used to process granulocytes from two healthy donors where comparable distributions of μcy, σcy and Csm and low success rates of cell type classification (< 60%) were found, indicating that there may exist ranges of μcy (10–20 Pa•s), σcy (0.4–0.6 S/m) and Csm (2.0–3.0 μF/cm2) for normal granulocytes. In summary, the developed microfluidic system can collect cytoplasmic viscosity, cytoplasmic conductivity and specific membrane capacitance from hundreds of single cells simultaneously and may provide new perspective for future developments of hematology analyzers.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Data availability

Cytoplasmic viscosity, cytoplasmic conductivity and specific membrane capacitance collected during this study are available as a supplementary file “Data of Single Cells”. The Excel-based file is composed of five sheets named after five experimental groups which are HL-60, HL-60 + CD, HL-60 + ConA, Healthy Donor 1 and Healthy Donor 2, respectively. In each sheet, there are four columns which are cell number (column A), cytoplasmic viscosity (column B), cytoplasmic conductivity (column C) and specific membrane capacitance (column D).

References

  • Ahmmed SM et al (2018) Multi-sample deformability cytometry of cancer cells. APL Bioeng 2(3):032002–032002

    Article  Google Scholar 

  • Ahuja K et al (2019) Toward point-of-care assessment of patient response: a portable tool for rapidly assessing cancer drug efficacy using multifrequency impedance cytometry and supervised machine learning. Microsyst Nanoeng 5:34

    Article  Google Scholar 

  • Armistead FJ, Gala De Pablo J, Gadelha H, Peyman SA, Evans SD (2019) Cells under stress: an inertial-shear microfluidic determination of cell behavior. Biophys J 116:1127–1135

    Article  Google Scholar 

  • Bashant KR et al (2019) Real-time deformability cytometry reveals sequential contraction and expansion during neutrophil priming. J Leukoc Biol 105(6):1143–1153

    Article  Google Scholar 

  • Becker FF, Wang XB, Huang Y, Pethig R, Vykoukal J, Gascoyne PRC (1995) Separation of human breast-cancer cells from blood by differential dielectric affinity. Proc Natl Acad Sci USA 92(3):860–864

    Article  Google Scholar 

  • Che J, Yu V, Garon EB, Goldman JW, Di Carlo D (2017) Biophysical isolation and identification of circulating tumor cells. Lab Chip 17(8):1452–1461

    Article  Google Scholar 

  • Cross SE, Jin YS, Rao J, Gimzewski JK (2007) Nanomechanical analysis of cells from cancer patients. Nat Nanotechnol 2(12):780–783

    Article  Google Scholar 

  • Darling EM, Carlo DD (2015) High-throughput assessment of cellular mechanical properties. Annu Rev Biomed Eng 17(1):35–62

    Article  Google Scholar 

  • Davidson PM et al (2019) High-throughput microfluidic micropipette aspiration device to probe time-scale dependent nuclear mechanics in intact cells. Lab Chip 19(21):3652–3663

    Article  Google Scholar 

  • De Ninno A et al (2019) High-throughput label-free characterization of viable, necrotic and apoptotic human lymphoma cells in a coplanar-electrode microfluidic impedance chip. Biosens Bioelectron 15:111887

    Google Scholar 

  • Deng Y et al (2017) Inertial microfluidic cell stretcher (iMCS): fully automated, high-throughput, and near real-time cell mechanotyping. Small 13:1700705

    Article  Google Scholar 

  • Fan W, Chen X, Ge Y, Jin Y, Jin Q, Zhao J (2019) Single-cell impedance analysis of osteogenic differentiation by droplet-based microfluidics. Biosens Bioelectron 145:111730

    Article  Google Scholar 

  • Feng Y, Huang L, Zhao P, Liang F, Wang W (2019) A microfluidic device integrating impedance flow cytometry and electric impedance spectroscopy for high-efficiency single-cell electrical property measurement. Anal Chem 91(23):15204–15212

    Article  Google Scholar 

  • Fregin B et al (2019) High-throughput single-cell rheology in complex samples by dynamic real-time deformability cytometry. Nat Commun 10(1):415

    Article  Google Scholar 

  • Guruprasad P et al (2019) Integrated automated particle tracking microfluidic enables high-throughput cell deformability cytometry for red cell disorders. Am J Hematol 94(2):189–199

    Article  Google Scholar 

  • Hassan U et al (2017) A point-of-care microfluidic biochip for quantification of CD64 expression from whole blood for sepsis stratification. Nature Commun 8:15949

    Article  Google Scholar 

  • Honrado C, Ciuffreda L, Spencer D, Ranford-Cartwright L, Morgan H (2018) Dielectric characterization of Plasmodium falciparum-infected red blood cells using microfluidic impedance cytometry. J R Soc Interf. https://doi.org/10.1098/rsif.2018.0416

    Article  Google Scholar 

  • Jacobi A, Rosendahl P, Krater M, Urbanska M, Herbig M, Guck J (2019) Analysis of biomechanical properties of hematopoietic stem and progenitor cells using real-time fluorescence and deformability cytometry. Methods Mol Biol 2017:135–148

    Article  Google Scholar 

  • Jaffe A, Voldman J (2018) Multi-frequency dielectrophoretic characterization of single cells. Microsyst Nanoeng 4:23–23

    Article  Google Scholar 

  • Kang JH et al (2019) Noninvasive monitoring of single-cell mechanics by acoustic scattering. Nat Methods 16(3):263–269

    Article  Google Scholar 

  • Leblanc-Hotte A et al (2019) On-chip refractive index cytometry for whole-cell deformability discrimination. Lab Chip 19(3):464–474

    Article  Google Scholar 

  • Liang H, Tan H, Chen D, Wang J, Chen J, Wu M-H (2019) Single-cell impedance flow cytometry. In: Santra TS, Tseng F-G (eds) Handbook of single cell technologies. Springer, Singapore, pp 1–31

    Google Scholar 

  • Liu J, Qiang Y, Alvarez O, Du E (2018) "Electrical impedance microflow cytometry with oxygen control for detection of sickle cells. Sens Actuators B 255(Pt 2):2392–2398

    Article  Google Scholar 

  • MATLAB (2016). https://www.mathworks.com/

  • Nyberg KD, Hu KH, Kleinman SH, Khismatullin DB, Butte MJ, Rowat AC (2017) Quantitative deformability cytometry: rapid, calibrated measurements of cell mechanical properties. Biophys J 113(7):1574–1584

    Article  Google Scholar 

  • Prieto JL et al (2016) Monitoring sepsis using electrical cell profiling. Lab Chip 16(22):4333–4340

    Article  Google Scholar 

  • Reale R, De Ninno A, Businaro L, Bisegna P, Caselli F (2019) High-throughput electrical position detection of single flowing particles/cells with non-spherical shape. Lab Chip 19(10):1818–1827

    Article  Google Scholar 

  • Reichel F, Mauer J, Nawaz AA, Gompper G, Guck J, Fedosov DA (2019) High-throughput microfluidic characterization of erythrocyte shapes and mechanical variability. Biophys J 117(1):14–24

    Article  Google Scholar 

  • Ren X, Ghassemi P, Strobl JS, Agah M (2019) Biophysical phenotyping of cells via impedance spectroscopy in parallel cyclic deformability channels. Biomicrofluidics 13(4):044103

    Article  Google Scholar 

  • Rosendahl P et al (2018) Real-time fluorescence and deformability cytometry. Nat Methods 15:355

    Article  Google Scholar 

  • Serhatlioglu M, Asghari M, Tahsin GM, Elbuken C (2019) Impedance-based viscoelastic flow cytometry. Electrophoresis 40(6):906–913

    Article  Google Scholar 

  • Toepfner N et al (2018) Detection of human disease conditions by single-cell morpho-rheological phenotyping of blood. Elife. https://doi.org/10.7554/eLife.29213

    Article  Google Scholar 

  • Wang H, Sobahi N, Han A (2017) Impedance spectroscopy-based cell/particle position detection in microfluidic systems. Lab Chip 17(7):1264–1269

    Article  Google Scholar 

  • Wang K et al (2017) Specific membrane capacitance, cytoplasm conductivity and instantaneous Young's modulus of single tumour cells. Sci Data 4:170015

    Article  Google Scholar 

  • Wang K et al (2019) Microfluidic cytometry for high-throughput characterization of single cell cytoplasmic viscosity using crossing constriction channels. Cytometry A. https://doi.org/10.1002/cyto.a.23921

    Article  Google Scholar 

  • Wu PH et al (2018) A comparison of methods to assess cell mechanical properties. Nat Methods 15(7):491–498

    Article  Google Scholar 

  • Xavier M et al (2016) Mechanical phenotyping of primary human skeletal stem cells in heterogeneous populations by real-time deformability cytometry. Integr Biol 8(5):616–623

    Article  Google Scholar 

  • Xavier M, de Andres MC, Spencer D, Oreffo ROC, Morgan H (2017) Size and dielectric properties of skeletal stem cells change critically after enrichment and expansion from human bone marrow: consequences for microfluidic cell sorting. J R Soc Interf 14:133

    Article  Google Scholar 

  • Xu Y, Xie X, Duan Y, Wang L, Cheng Z, Cheng J (2016) A review of impedance measurements of whole cells. Biosens Bioelectron 77:824–836

    Article  Google Scholar 

  • Yang D, Ai Y (2019) Microfluidic impedance cytometry device with N-shaped electrodes for lateral position measurement of single cells/particles. Lab Chip 19(21):3609–3617

    Article  Google Scholar 

  • Yang X, Chen Z, Miao J, Cui L, Guan W (2017) High-throughput and label-free parasitemia quantification and stage differentiation for malaria-infected red blood cells. Biosens Bioelectron 98:408–414

    Article  Google Scholar 

  • Yang D, Zhou Y, Zhou Y, Han J, Ai Y (2019) Biophysical phenotyping of single cells using a differential multiconstriction microfluidic device with self-aligned 3D electrodes. Biosens Bioelectron 133:16–23

    Article  Google Scholar 

  • Zhang Y et al (2019) Crossing constriction channel-based microfluidic cytometry capable of electrically phenotyping large populations of single cells. Analyst 144(3):1008–1015

    Article  Google Scholar 

  • Zhao Y et al (2018) Development of microfluidic impedance cytometry enabling the quantification of specific membrane capacitance and cytoplasm conductivity from 100,000 single cells. Biosens Bioelectron 111:138–143

    Article  Google Scholar 

  • Zheng Y, Nguyen J, Wei Y, Sun Y (2013) Recent advances in microfluidic techniques for single-cell biophysical characterization. Lab Chip 13(13):2464–2483

    Article  Google Scholar 

  • Zhou Y, Basu S, Laue E, Seshia AA (2016) Single cell studies of mouse embryonic stem cell (mESC) differentiation by electrical impedance measurements in a microfluidic device. Biosens Bioelectron 81:249–258

    Article  Google Scholar 

  • Zhou Y, Yang D, Zhou Y, Khoo BL, Han J, Ai Y (2018) Characterizing deformability and electrical impedance of cancer cells in a microfluidic device. Anal Chem 90(1):912–919

    Article  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge financial supports from the National Natural Science Foundation of China (Grant No. 61431019, 61825107, 61671430, 61922079); Key Project (QYZDB-SSW-JSC011), Youth Innovation Promotion Association and Interdisciplinary Innovation Team of Chinese Academy of Sciences; Research Start-up Foundation for Talent Introduction of Beijing University of Posts and Telecommunications (505019125); New Teacher Basic Scientific Research Foundation of Beijing University of Posts and Telecommunications.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Junbo Wang or Jian Chen.

Additional information

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary file1 (XLSX 80 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Y., Wang, K., Sun, X. et al. Development of microfluidic platform capable of characterizing cytoplasmic viscosity, cytoplasmic conductivity and specific membrane capacitance of single cells. Microfluid Nanofluid 24, 45 (2020). https://doi.org/10.1007/s10404-020-02350-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s10404-020-02350-6

Keywords

Navigation