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.

  • Protocol
  • Published:

On-chip microfluidic production of cell-sized liposomes

Abstract

In this protocol, we describe a recently developed on-chip microfluidic method to form monodisperse, cell-sized, unilamellar, and biocompatible liposomes with excellent encapsulation efficiency. Termed octanol-assisted liposome assembly (OLA), it resembles bubble-blowing on a microscopic scale. Hydrodynamic flow focusing of two immiscible fluid streams (an aqueous phase and a lipid-containing 1-octanol phase) by orthogonal outer aqueous streams gives rise to double-emulsion droplets. As the lipid bilayer assembles along the interface, each emulsion droplet quickly evolves into a liposome and a 1-octanol droplet. OLA has several advantages as compared with other on-chip techniques, such as a very fast liposome maturation time (a few minutes), a relatively straightforward and completely on-chip setup, and a biologically relevant liposome size range (5–20 μm). Owing to the entire approach being on-chip, OLA enables high-throughput liposome production (typical rate of tens of Hz) using low sample volumes (10 μl). For prolonged on-chip experimentation, liposomes are subsequently purified by removing the 1-octanol droplets. For device fabrication, a reusable silicon template is produced in a clean room facility using electron-beam lithography followed by dry reactive ion etching, which takes 3 h. The patterned silicon template is used to prepare polydimethylsiloxane (PDMS)-based microfluidic devices in the wet lab, followed by a crucial surface treatment; the whole process takes 2 d. Liposomes can be produced in 1 h and further manipulated, depending on the experimental setup. OLA offers an ideal microfluidic platform for diverse bottom-up biotechnology studies by enabling creation of synthetic cells, microreactors and bioactive cargo delivery systems, and also has potential as an analytical tool.

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

Figure 1: Schematics showing OLA-based liposome production and purification.
Figure 2: Hydrophilic surface treatment of the microfluidic device.
Figure 3: Octanol-assisted liposome assembly.
Figure 4: On-chip purification of liposomes.

Similar content being viewed by others

References

  1. Balaram, P. Synthesizing life. Curr. Sci. 85, 1509–1510 (2003).

    Google Scholar 

  2. Pohorille, A. & Deamer, D. Artificial cells: prospects for biotechnology. Trends Biotechnol. 20, 123–128 (2002).

    CAS  PubMed  Google Scholar 

  3. Deamer, D. On the origin of systems. Systems biology, synthetic biology and the origin of life. EMBO Rep. 10, S1–S4 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Walde, P. Building artificial cells and protocell models: experimental approaches with lipid vesicles. BioEssays 32, 296–303 (2010).

    CAS  PubMed  Google Scholar 

  5. Dzieciol, A.J. & Mann, S. Designs for life: protocell models in the laboratory. Chem. Soc. Rev. 41, 79 (2012).

    CAS  PubMed  Google Scholar 

  6. Shang, L., Cheng, Y. & Zhao, Y. Emerging droplet microfluidics. Chem. Rev. 117, 7964–8040 (2017).

    CAS  PubMed  Google Scholar 

  7. van Swaay, D. & deMello, A. Microfluidic methods for forming liposomes. Lab Chip 13, 752–67 (2013).

    CAS  PubMed  Google Scholar 

  8. Stano, P., Carrara, P., Kuruma, Y., Pereira de Souza, T. & Luisi, P.L. Compartmentalized reactions as a case of soft-matter biotechnology: synthesis of proteins and nucleic acids inside lipid vesicles. J. Mater. Chem. 21, 18887 (2011).

    CAS  Google Scholar 

  9. Deshpande, S., Caspi, Y., Meijering, A.E. & Dekker, C. Octanol-assisted liposome assembly on chip. Nat. Commun. 7, 10447 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Deshpande, S., Birnie, A. & Dekker, C. On-chip density-based purification of liposomes. Biomicrofluidics 11, 34106 (2017).

    Google Scholar 

  11. Shum, H.C., Lee, D., Yoon, I., Kodger, T. & Weitz, D.A. Double emulsion templated monodisperse phospholipid vesicles. Langmuir 24, 7651–7653 (2008).

    CAS  PubMed  Google Scholar 

  12. Teh, S.Y., Khnouf, R., Fan, H. & Lee, A.P. Stable, biocompatible lipid vesicle generation by solvent extraction-based droplet microfluidics. Biomicrofluidics 5, 44113 (2011).

    PubMed  Google Scholar 

  13. Utada, A.S. et al. Monodisperse double emulsions generated from a microcapillary device. Science 308, 537–541 (2005).

    CAS  PubMed  Google Scholar 

  14. Walde, P., Cosentino, K., Engel, H. & Stano, P. Giant vesicles: preparations and applications. ChemBioChem 11, 848–865 (2010).

    CAS  PubMed  Google Scholar 

  15. Spoelstra, K.W., Deshpande, S. & Dekker, C. Tailoring the appearance: what will synthetic cells look like? Curr. Opin. Biotechnol. 51, 47–56 (2018).

    CAS  PubMed  Google Scholar 

  16. Forster, A.C. & Church, G.M. Towards synthesis of a minimal cell. Mol. Syst. Biol. 2, 45 (2006).

    PubMed  PubMed Central  Google Scholar 

  17. Elani, Y. Construction of membrane-bound artificial cells using microfluidics: a new frontier in bottom-up synthetic biology. Biochem. Soc. Trans. 44, 723–730 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Caspi, Y. & Dekker, C. Divided we stand: splitting synthetic cells for their proliferation. Syst. Synth. Biol. 8, 249–269 (2014).

    PubMed  PubMed Central  Google Scholar 

  19. Szwedziak, P., Wang, Q., Bharat, T.A.M., Tsim, M. & Löwe, J. Architecture of the ring formed by the tubulin homologue FtsZ in bacterial cell division. Elife 3, e04601 (2014).

    PubMed  PubMed Central  Google Scholar 

  20. Cabré, E.J. et al. Bacterial division proteins FtsZ and ZipA induce vesicle shrinkage and cell membrane invagination. J. Biol. Chem. 288, 26625–26634 (2013).

    PubMed  PubMed Central  Google Scholar 

  21. Osawa, M. & Erickson, H.P. Liposome division by a simple bacterial division machinery. Proc. Natl. Acad. Sci. USA 110, 11000–11004 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Keber, F.C. et al. Topology and dynamics of active nematic vesicles. Science 345, 1135–1139 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Carvalho, K. et al. Cell-sized liposomes reveal how actomyosin cortical tension drives shape change. Proc. Natl. Acad. Sci. USA 110, 16456–16461 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Cáceres, R., Abou-Ghali, M. & Plastino, J. Reconstituting the actin cytoskeleton at or near surfaces in vitro. Biochim. Biophys. Acta 1853, 3006–3014 (2015).

    PubMed  Google Scholar 

  25. van Nies, P. et al. Unbiased tracking of the progression of mRNA and protein synthesis in bulk and in liposome-confined reactions. ChemBioChem 14, 1963–1966 (2013).

    CAS  PubMed  Google Scholar 

  26. Scott, A. et al. Cell-free phospholipid biosynthesis by gene-encoded enzymes reconstituted in liposomes. PLoS One 11, e0163058 (2016).

    PubMed  PubMed Central  Google Scholar 

  27. Noireaux, V. & Libchaber, A. A vesicle bioreactor as a step toward an artificial cell assembly. Proc. Natl. Acad. Sci. USA 101, 17669–17674 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Gardner, P.M., Winzer, K. & Davis, B.G. Sugar synthesis in a protocellular model leads to a cell signalling response in bacteria. Nat. Chem. 1, 377–383 (2009).

    CAS  PubMed  Google Scholar 

  29. Lentini, R. et al. Two-way chemical communication between artificial and natural cells. ACS Cent. Sci. 3, 117–123 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Adamala, K.P., Martin-Alarcon, D.A., Guthrie-Honea, K.R. & Boyden, E.S. Engineering genetic circuit interactions within and between synthetic minimal cells. Nat. Chem. 9, 431–439 (2016).

    PubMed  PubMed Central  Google Scholar 

  31. Cama, J. et al. Direct optofluidic measurement of the lipid permeability of fluoroquinolones. Sci. Rep. 6, 32824 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Cama, J., Chimerel, C., Pagliara, S., Javer, A. & Keyser, U.F. A label-free microfluidic assay to quantitatively study antibiotic diffusion through lipid membranes. Lab Chip 14, 2303–8 (2014).

    CAS  PubMed  Google Scholar 

  33. Dezi, M., Di Cicco, A., Bassereau, P. & Lévy, D. Detergent-mediated incorporation of transmembrane proteins in giant unilamellar vesicles with controlled physiological contents. Proc. Natl. Acad. Sci. USA 110, 7276–81 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, H. & Lappalainen, P. A simple guide to biochemical approaches for analyzing protein-lipid interactions. Mol. Biol. Cell 23, 2823–2830 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Lemière, J., Valentino, F., Campillo, C. & Sykes, C. How cellular membrane properties are affected by the actin cytoskeleton. Biochimie 130, 33–40 (2016).

    PubMed  Google Scholar 

  36. Kahya, N. Protein-protein and protein-lipid interactions in domain-assembly: lessons from giant unilamellar vesicles. Biochim. Biophys. Acta 1798, 1392–1398 (2010).

    CAS  PubMed  Google Scholar 

  37. Estes, D.J., Lopez, S.R., Fuller, A.O. & Mayer, M. Triggering and visualizing the aggregation and fusion of lipid membranes in microfluidic chambers. Biophys. J. 91, 233–43 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Terasawa, H., Nishimura, K., Suzuki, H., Matsuura, T. & Yomo, T. Coupling of the fusion and budding of giant phospholipid vesicles containing macromolecules. Proc. Natl. Acad. Sci. USA 109, 5942–5947 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Karatekin, E. et al. Cascades of transient pores in giant vesicles: line tension and transport. Biophys. J. 84, 1734–1749 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Oglęcka, K., Rangamani, P., Liedberg, B., Kraut, R.S. & Parikh, A.N. Oscillatory phase separation in giant lipid vesicles induced by transmembrane osmotic differentials. Elife 3, e03695 (2014).

    PubMed  PubMed Central  Google Scholar 

  41. Carugo, D., Bottaro, E., Owen, J., Stride, E. & Nastruzzi, C. Liposome production by microfluidics: potential and limiting factors. Sci. Rep. 6, 25876 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Zylberberg, C. & Matosevic, S. Pharmaceutical liposomal drug delivery: a review of new delivery systems and a look at the regulatory landscape. Drug Deliv. 23, 3319–3329 (2016).

    CAS  PubMed  Google Scholar 

  43. Yang, J. et al. Drug delivery via cell membrane fusion using lipopeptide modified liposomes. ACS Cent. Sci. 2, 621–630 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Armada-Moreira, A., Taipaleenmäki, E., Itel, F., Zhang, Y. & Städler, B. Droplet-microfluidics towards the assembly of advanced building blocks in cell mimicry. Nanoscale 8, 19510–19522 (2016).

    CAS  PubMed  Google Scholar 

  45. Edwards, K.A., Bolduc, O.R. & Baeumner, A.J. Miniaturized bioanalytical systems: enhanced performance through liposomes. Curr. Opin. Chem. Biol. 16, 444–452 (2012).

    CAS  PubMed  Google Scholar 

  46. Jesorka, A. & Orwar, O. Liposomes: technologies and analytical applications. Annu. Rev. Anal. Chem. 1, 801–832 (2008).

    CAS  Google Scholar 

  47. Thiele, J. Polymer material design by microfluidics inspired by cell biology and cell-free biotechnology. Macromol. Chem. Phys. 218, 1600429 (2017).

    Google Scholar 

  48. Reeves, J.P. & Dowben, R.M. Formation and properties of thin-walled phospholipid vesicles. J. Cell. Physiol. 73, 49–60 (1969).

    CAS  PubMed  Google Scholar 

  49. Olson, F., Hunt, C.A., Szoka, F.C., Vail, W.J. & Papahadjopoulos, D. Preparation of liposomes of defined size distribution by extrusion through polycarbonate membranes. Biochim. Biophys. Acta 557, 9–23 (1979).

    CAS  PubMed  Google Scholar 

  50. Angelova, M.I. & Dimitrov, D.S. Liposome electroformation. Faraday Discuss. Chem. Soc. 81, 303 (1986).

    CAS  Google Scholar 

  51. Yu, B., Lee, R.J. & Lee, L.J. Microfluidic methods for production of liposomes. Methods Enzymol. 465, 129–141 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Deng, N.N., Yelleswarapu, M. & Huck, W.T.S. Monodisperse uni- and multicompartment liposomes. J. Am. Chem. Soc. 138, 7584–7591 (2016).

    CAS  PubMed  Google Scholar 

  53. Mozafari, M.R. Liposomes: an overview of manufacturing techniques. Cell. Mol. Biol. Lett. 10, 711–719 (2005).

    CAS  PubMed  Google Scholar 

  54. Lotter, J., Botes, A.L., Van Dyk, M.S. & Breytenbach, J.C. Correlation between the physicochemical properties of organic solvents and their biocompatibility toward epoxide hydrolase activity in whole-cells of a yeast, Rhodotorula sp.. Biotechnol. Lett. 26, 1191–1195 (2004).

    CAS  PubMed  Google Scholar 

  55. Jiménez, M., Martos, A., Cabré, E.J., Raso, A. & Rivas, G. Giant vesicles: a powerful tool to reconstruct bacterial division assemblies in cell-like compartments. Environ. Microbiol. 15, 3158–3168 (2013).

    PubMed  Google Scholar 

  56. Nunes, J.K., Tsai, S.S.H., Wan, J. & Stone, H.A. Dripping and jetting in microfluidic multiphase flows applied to particle and fibre synthesis. J. Phys. D Appl. Phys. 46, 114002 (2013).

    PubMed  PubMed Central  Google Scholar 

  57. Ho, C.S., Kim, J.W. & Weitz, D.A. Microfluidic fabrication of monodisperse biocompatible and biodegradable polymersomes with controlled permeability. J. Am. Chem. Soc. 130, 9543–9549 (2008).

    Google Scholar 

  58. Pautot, S., Frisken, B.J. & Weitz, D.A. Engineering asymmetric vesicles. Proc. Natl. Acad. Sci. USA 100, 10718–10721 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Richmond, D.L. et al. Forming giant vesicles with controlled membrane composition, asymmetry, and contents. Proc. Natl. Acad. Sci. USA 108, 9431–9436 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Matosevic, S. & Paegel, B.M. Layer-by-layer cell membrane assembly. Nat. Chem. 5, 958–63 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Knovel Critical Tables (2008) Available at: https://app.knovel.com/web/toc.v/cid:kpKCTE000X/viewerType:toc/root_slug:knovel-critical-tables (accessed 27th February, 2018).

  62. Campillo, C. et al. Unexpected membrane dynamics unveiled by membrane nanotube extrusion. Biophys. J. 104, 1248–1256 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Surewicz, W.K. Membrane actions of water-soluble fusogens: effect of dimethyl sulfoxide, glycerol and sucrose on lipid bilayer order and fluidity. Chem. Phys. Lipids 34, 363–372 (1984).

    CAS  Google Scholar 

  64. Cheng, C.Y., Wang, J.Y., Kausik, R., Lee, K.Y.C. & Han, S. Nature of interactions between PEO-PPO-PEO triblock copolymers and lipid membranes: (II) role of hydration dynamics revealed by dynamic nuclear polarization. Biomacromolecules 13, 2624–2633 (2012).

    CAS  PubMed  Google Scholar 

  65. Kazayama, Y., Teshima, T., Osaki, T., Takeuchi, S. & Toyota, T. Integrated microfluidic system for size-based selection and trapping of giant vesicles. Anal. Chem. 88, 1111–1116 (2016).

    CAS  PubMed  Google Scholar 

  66. Akagi, J. et al. Miniaturized embryo array for automated trapping, immobilization and microperfusion of zebrafish embryos. PLoS One 7 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Nishimura, K., Suzuki, H., Toyota, T. & Yomo, T. Size control of giant unilamellar vesicles prepared from inverted emulsion droplets. J. Colloid Interface Sci. 376, 119–125 (2012).

    CAS  PubMed  Google Scholar 

  68. Abkarian, M., Loiseau, E. & Massiera, G. Continuous droplet interface crossing encapsulation (cDICE) for high throughput monodisperse vesicle design. Soft Matter 7, 4610 (2011).

    CAS  Google Scholar 

  69. Jahn, A., Vreeland, W.N., Gaitan, M. & Locascio, L.E. Controlled vesicle self-assembly in microfluidic channels with hydrodynamic focusing. J. Am. Chem. Soc. 126, 2674–2675 (2004).

    CAS  PubMed  Google Scholar 

  70. Jahn, A. et al. Microfluidic mixing and the formation of nanoscale lipid vesicles. ACS Nano 4, 2077–2087 (2010).

    CAS  PubMed  Google Scholar 

  71. Stachowiak, J.C. et al. Unilamellar vesicle formation and encapsulation by microfluidic jetting. Proc. Natl. Acad. Sci. USA 105, 4697–4702 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Funakoshi, K., Suzuki, H. & Takeuchi, S. Formation of giant lipid vesiclelike compartments from a planar lipid membrane by a pulsed jet flow. J. Am. Chem. Soc. 129, 12608–12609 (2007).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge Y. Caspi, A. Birnie, F. Fanalista, K. Spoelstra, D. Hueting, M. van Doorn, M. Schaich, K. Al-Nahas, and S. Sachdev for their contributions to the development of the described protocol and valuable feedback. This work was supported by an NWO TOP-PUNT grant (no. 718014001), the Netherlands Organisation for Scientific Research (NWO/OCW) and a European Research Council Advanced Grant SynDiv (no. 669598).

Author information

Authors and Affiliations

Authors

Contributions

S.D. performed the experiments. S.D. and C.D. wrote the paper.

Corresponding author

Correspondence to Cees Dekker.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Schematic flow diagram summarizing the key steps involved in the preparation of a master wafer and subsequent steps leading to the production of a microfluidic device.

(a) Master wafer is prepared in the clean room. A silicon wafer is spin-coated with a suitable negative resist and pre-baked. The wafer is then selectively exposed to e-beam to write the desired pattern. The wafer is further post-baked and chemically developed to remove the non-patterned resist. The developed wafer is appropriately etched, cleaned, and silanized to obtain a master wafer. (b) The microfluidic device is prepared in the wet lab. PDMS is poured over the master and cured by baking. The cured PDMs is then carefully peeled off and holes are punched at appropriate locations. The PDMS surface is then activated using plasma and bonded to a PDMS-coated, surface-activated glass slide. The bonded device is surface-treated to render it partially hydrophilic. The device is now ready to perform OLA. The master can be used multiple times.

Supplementary Figure 2 Various steps involved in the making of a microfluidic device.

(a) Master wafer enclosed within a well-like structure made up of aluminium foil. The design patterns are faintly visible on the wafer surface. (b) Cut-out PDMS block having a single OLA design (faint channels can be seen). Inlets, exit and separation hole are punched. (c) Glass slides (seen as faint outlines) immersed in a thin layer of PDMS, which is spread on a blank wafer. (d) Cured PDMS layer is peeled off to obtain PDMS-coated glass slides. The left-most glass slide is damaged severely during the peeling-off process and thus will not be used, while the down-most glass slide is broken only in the corner and can be used further. The remaining two glass slides are intact and in good condition. One glass slide has been removed showing a clean, reusable wafer surface. (e) A microfluidic device prepared by bonding the PDMS-coated glass slide to the design-containing PDMS block, right after the plasma treatment. The microfluidic channels are clearly visible. (f) Thicker (outer tube) and thinner (outer tube) tubes, placed in fittings, with the corresponding metal connectors inserted at one end.

Supplementary Figure 3 Production junction with bumpers.

Bright-field image of the production junction having bumper-like protrusions, in order to facilitate the surface treatment.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–3. (PDF 579 kb)

Supplementary Data

.dxf file containing a standard design for OLA and subsequent liposome purification from 1-octanol droplets. (ZIP 91 kb)

Partial hydrophilic surface treatment of the microfluidic device.

1% (vol/vol) PVA solution is used. IA and LO channel (air) pressure: 50 mbar; OA channel (PVA) pressure: 120 mbar. (AVI 5817 kb)

Optimal double-emulsion droplet production.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 2960 kb)

Double-emulsion droplets with a prominent 1-octanol pocket flowing downstream.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 5173 kb)

Liposomes and budded-off 1-octanol droplets further downstream of the channel.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 1786 kb)

A top view across the separation hole, showing the separation process.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 9768 kb)

Optimal separation process, mainly yielding liposomes into the post-hole channel.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 383 kb)

Suboptimal production, yet double-emulsion droplets are still formed.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 9372 kb)

Suboptimal post-junction channel, showing aggregates of liposomes and 1-octanol droplets.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 474 kb)

Suboptimal purification process, showing unwanted 1-octanol aggregates entering the post-hole channel.

IA is 15% (vol/vol) glycerol; LO is 0.2% (wt/vol) lipids (DOPC/Rh-PE in a molar ratio of 1,000:1) in 1-octanol; OA is 15% (vol/vol) glycerol and 5% (wt/vol) P188. (AVI 434 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deshpande, S., Dekker, C. On-chip microfluidic production of cell-sized liposomes. Nat Protoc 13, 856–874 (2018). https://doi.org/10.1038/nprot.2017.160

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2017.160

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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