JoVE Logo
Faculty Resource Center

Sign In

Summary

Abstract

Introduction

Protocol

Representative Results

Discussion

Acknowledgements

Materials

References

Bioengineering

On-Chip Octanol-Assisted Liposome Assembly for Bioengineering

Published: March 17th, 2023

DOI:

10.3791/65032

1Laboratory of Physical Chemistry and Soft Matter, Wageningen University & Research
* These authors contributed equally

The present protocol describes octanol-assisted liposome assembly (OLA), a microfluidic technique to generate biocompatible liposomes. OLA produces monodispersed, micron-sized liposomes with efficient encapsulation, allowing immediate on-chip experimentation. This protocol is anticipated to be particularly suitable for synthetic biology and synthetic cell research.

Microfluidics is a widely used tool to generate droplets and vesicles of various kinds in a controlled and high-throughput manner. Liposomes are simplistic cellular mimics composed of an aqueous interior surrounded by a lipid bilayer; they are valuable in designing synthetic cells and understanding the fundamentals of biological cells in an in vitro fashion and are important for applied sciences, such as cargo delivery for therapeutic applications. This article describes a detailed working protocol for an on-chip microfluidic technique, octanol-assisted liposome assembly (OLA), to produce monodispersed, micron-sized, biocompatible liposomes. OLA functions similarly to bubble blowing, where an inner aqueous (IA) phase and a surrounding lipid-carrying 1-octanol phase are pinched off by surfactant-containing outer fluid streams. This readily generates double-emulsion droplets with protruding octanol pockets. As the lipid bilayer assembles at the droplet interface, the pocket spontaneously detaches to give rise to a unilamellar liposome that is ready for further manipulation and experimentation. OLA provides several advantages, such as steady liposome generation (>10 Hz), efficient encapsulation of biomaterials, and monodispersed liposome populations, and requires very small sample volumes (~50 µL), which can be crucial when working with precious biologicals. The study includes details on microfabrication, soft-lithography, and surface passivation, which are needed to establish OLA technology in the lab. A proof-of-principle synthetic biology application is also shown by inducing the formation of biomolecular condensates inside the liposomes via transmembrane proton flux. It is anticipated that this accompanying video protocol will facilitate the readers to establish and troubleshoot OLA in their labs.

All cells have a plasma membrane as their physical boundary, and this membrane is essentially a scaffold in the form of a lipid bilayer formed by the self-assembly of amphiphilic lipid molecules. Liposomes are the minimal synthetic counterparts of biological cells; they have an aqueous lumen surrounded by phospholipids, which form a lipid bilayer with the hydrophilic head groups facing the aqueous phase and the hydrophobic tails buried inward. The stability of liposomes is governed by the hydrophobic effect, as well as the hydrophilicity between the polar groups, van der Waals forces between the hydrophobic carbon tails, and the hydrogen bonding between water molecule....

Log in or to access full content. Learn more about your institution’s access to JoVE content here

1. Fabricating the master wafer

  1. Take a 4 in (10 cm) diameter clean silicon wafer (see Table of Materials). Clean it further using pressurized air to remove any dust particles.
  2. Mount the wafer on a spin coater, and gently dispense ~5 mL of a negative photoresist (see Table of Materials) in the center of the wafer. Try to avoid air bubbles, as they might interfere with the downstream printing process of the wafer.
  3. To obtain a 10 µm thic.......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

This study demonstrates the formation of membraneless condensates via the process of liquid-liquid phase separation (LLPS) inside liposomes as a representative experiment.

Sample preparation
The IA, OA, ES, and feed solution (FS) are prepared as follows:

IA: 12% glycerol, 5 mM dextran, 150 mM KCl, 5 mg/mL poly-L-lysine (PLL), 0.05 mg/mL poly-L-lysine-FITC labeled (PLL-FITC), 8 mM adenosine triphosphate (ATP), 15 mM citrate-HCl (pH 4)<.......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

Cellular complexity makes it extremely difficult to understand living cells when studied as a whole. Reducing the redundancy and interconnectivity of cells by reconstituting the key components in vitro is necessary to further our understanding of biological systems and create artificial cellular mimics for biotechnological applications22,23,24. Liposomes serve as an excellent minimal system to understand cellular phenomena. A no.......

Log in or to access full content. Learn more about your institution’s access to JoVE content here

We would like to acknowledge Dolf Weijers, Vera Gorelova, and Mark Roosjen for kindly providing us with YFP. S.D. acknowledges financial support from the Dutch Research Council (grant number: OCENW.KLEIN.465).

....

Log in or to access full content. Learn more about your institution’s access to JoVE content here

NameCompanyCatalog NumberComments
1-OctanolSigma-AldrichNo. 297887
1.5 mL tubesFisher scientific10451043Eppendorf 3810X Polypropylene microcentrifuge tubes
ATPSigma-AldrichNo. A2383
Biopsy punchDarwin microfluidicsPT-T983-050.5 mm and 3 mm diameter
Citrate-baseSigma-AldrichNo. 71405
DextranSigma-AldrichNo. 31388Mr~6,000
Direct-write optical lithography machineDurham Magneto Optics LtdMicroWriter ML3 Babysetup and software
DOPC lipidAvantiSKU:850375C
F68Sigma-AldrichNo. 24040032
Glass cover slipCorning#1, 24 x 40 mm
GlycerolSigma-AldrichNo. G2025
Hydrochloric acidThermo Scientific AcrosNo. 124630010
Liss Rhod PE lipidAvantiSKU:810150C
ParafilmSigma-AldrichNo. P7793
PhotoresistMicro resist technology GmbHEpoCore 10
Photoresist developermicro resist technology GmbHmr-Dev 600
Plasma cleanerHarrick plasmaPDC-32G
PolydimethylsiloxaneDowSylgard 184PDMS and curing agent
Poly-L-lysineSigma-AldrichNo. P7890
Poly-L-lysine–FITC LabeledSigma-AldrichNo. P3543
Polyvinyl alcoholSigma-Aldrichno. P8136molecular weight 30,000–70,000, 87%–90% hydrolyzed
Pressure controllerElveflow OBK1 Mk3+Flow controller
Scotch tapeMagic Tape Invisible Matt Tape
Silicon waferSilicon Materials0620R16002
Spin coater Laurell Technologies CorporationModel WS-650MZ-23NPP
Stainless Steel 90° Bent PDMS CouplersDarwin microfluidicsPN-BEN-23G
Tris-baseSigma-AldrichNo. 252859
Tygon tubingDarwin microfluidics1/16" OD x 0.02" ID
UV laser 365 nm wavelength

  1. Frezard, F. Liposomes: From biophysics to the design of peptide vaccines. Brazilian Journal of Medical and Biological Research. 32 (2), 181-189 (1999).
  2. Monteiro, N., Martins, A., Reis, R. L., Neves, N. M. Liposomes in tissue engineering and regenerative medicine. Journal of the Royal Society Interface. 11 (101), 20140459 (2014).
  3. Mishra, H., Chauhan, V., Kumar, K., Teotia, D. A comprehensive review on liposomes: A novel drug delivery system. Journal of Drug Delivery and Therapeutics. 8 (6), 400-404 (2018).
  4. Liu, W., et al. Research progress on liposomes: Application in food, digestion behavior and absorption mechanism. Trends in Food Science & Technology. 104, 177-189 (2020).
  5. Kamiya, K., Takeuchi, S. Giant liposome formation toward the synthesis of well-defined artificial cells. Journal of Materials Chemistry B. 5 (30), 5911-5923 (2017).
  6. van Swaay, D., DeMello, A. Microfluidic methods for forming liposomes. Lab on a Chip. 13 (5), 752-767 (2013).
  7. Lombardo, D., Kiselev, M. A. Methods of liposomes preparation: Formation and control factors of versatile nanocarriers for biomedical and nanomedicine application. Pharmaceutics. 14 (3), 543 (2022).
  8. Zhang, H., D’Souza, G. G. M. Thin-film hydration followed by extrusion method for liposome preparation. Liposomes: Methods and Protocols. , 17-22 (2017).
  9. Filipczak, N., Pan, J., Yalamarty, S. S. K., Torchilin, V. P. Recent advancements in liposome technology. Advanced Drug Delivery Reviews. 156, 4-22 (2020).
  10. Has, C., Sunthar, P. A comprehensive review on recent preparation techniques of liposomes. Journal of Liposome Research. 30 (4), 336-365 (2020).
  11. Large, D. E., Abdelmessih, R. G., Fink, E. A., Auguste, D. T. Liposome composition in drug delivery design, synthesis, characterization, and clinical application. Advanced Drug Delivery Reviews. 176, 113851 (2021).
  12. Abkarian, M., Loiseau, E., Massiera, G. Continuous droplet interface crossing encapsulation (cDICE) for high throughput monodisperse vesicle design. Soft Matter. 7 (10), 4610-4614 (2011).
  13. Morita, M., et al. Droplet-shooting and size-filtration (DSSF) method for synthesis of cell-sized liposomes with controlled lipid compositions. ChemBioChem. 16 (14), 2029-2035 (2015).
  14. Convery, N., Gadegaard, N. 30 years of microfluidics. Micro and Nano Engineering. 2, 76-91 (2019).
  15. Stachowiak, J. C., et al. Unilamellar vesicle formation and encapsulation by microfluidic jetting. Proceedings of the National Academy of Sciences of the United States of America. 105 (12), 4697-4702 (2008).
  16. Chu, L. Y., Utada, A. S., Shah, R. K., Kim, J. W., Weitz, D. A. Controllable monodisperse multiple emulsions. Angewandte Chemie. 119 (47), 9128-9132 (2007).
  17. Pautot, S., Frisken, B. J., Weitz, D. Engineering asymmetric vesicles. Proceedings of the National Academy of Sciences of the United States of America. 100 (19), 10718-10721 (2003).
  18. Ota, S., Yoshizawa, S., Takeuchi, S. Microfluidic formation of monodisperse, cell-sized, and unilamellar vesicles. Angewandte Chemie International Edition. 48 (35), 6533-6537 (2009).
  19. Carugo, D., Bottaro, E., Owen, J., Stride, E., Nastruzzi, C. Liposome production by microfluidics: Potential and limiting factors. Scientific Reports. 6, 25876 (2016).
  20. Deshpande, S., Dekker, C. On-chip microfluidic production of cell-sized liposomes. Nature Protocols. 13 (5), 856-874 (2018).
  21. Last, M. G., Deshpande, S., Dekker, C. pH-controlled coacervate-membrane interactions within liposomes. ACS Nano. 14 (4), 4487-4498 (2020).
  22. Jia, H., Schwille, P. Bottom-up synthetic biology: reconstitution in space and time. Current Opinion in Biotechnology. 60, 179-187 (2019).
  23. Ganar, K. A., Leijten, L., Deshpande, S. Actinosomes: Condensate-Templated Containers for Engineering Synthetic Cells. ACE Synthetic Biology. 11 (8), 2869-2879 (2022).
  24. Gaut, N. T., Adamala, K. P. Reconstituting Natural Cell Elements in Synthetic Cells. Advanced Biology. 5 (3), 2000188 (2021).
  25. Ganar, K. A., Honaker, L. W., Deshpande, S. Shaping synthetic cells through cytoskeleton-condensate-membrane interactions. Current Opinion in Colloid & Interface Science. 54, 101459 (2021).
  26. Bashirzadeh, Y., Liu, A. P. Encapsulation of the cytoskeleton: Towards mimicking the mechanics of a cell. Soft Matter. 15 (42), 8425-8436 (2019).
  27. Deshpande, S., et al. Spatiotemporal control of coacervate formation within liposomes. Nature Communications. 10, 1800 (2019).
  28. Love, C., et al. Reversible pH-responsive coacervate formation in lipid vesicles activates dormant enzymatic reactions. Angewandte Chemie. 132 (15), 6006-6013 (2020).
  29. Lu, T., et al. Endocytosis of coacervates into liposomes. Journal of the American Chemical Society. 144 (30), 13451-13455 (2022).
  30. Van de Cauter, L., et al. Optimized cDICE for efficient reconstitution of biological systems in giant unilamellar vesicles. ACS Synthetic Biology. 10 (7), 1690-1702 (2021).
  31. Blanken, D., Foschepoth, D., Serrão, A. C., Danelon, C. Genetically controlled membrane synthesis in liposomes. Nature Communications. 11, 4317 (2020).
  32. Bouzetos, E., Ganar, K. A., Mastrobattista, E., Deshpande, S., vander Oost, J. (R) evolution-on-a-chip. Trends in Biotechnology. 40 (1), 60-76 (2022).
  33. Kamalinia, G., Grindel, B. J., Takahashi, T. T., Millward, S. W., Roberts, R. W. Directing evolution of novel ligands by mRNA display. Chemical Society Reviews. 50 (16), 9055-9103 (2021).
  34. Godino, E., et al. De novo synthesized Min proteins drive oscillatory liposome deformation and regulate FtsA-FtsZ cytoskeletal patterns. Nature Communications. 10, 4969 (2019).
  35. Tenchov, R., Bird, R., Curtze, A. E., Zhou, Q. Lipid nanoparticles─From liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano. 15 (11), 16982-17015 (2021).
  36. Deshpande, S., Caspi, Y., Meijering, A. E., Dekker, C. Octanol-assisted liposome assembly on chip. Nature Communications. 7, 10447 (2016).
  37. Schaich, M., et al. An integrated microfluidic platform for quantifying drug permeation across biomimetic vesicle membranes. Molecular Pharmaceutics. 16 (6), 2494-2501 (2019).
  38. Schaich, M., Sobota, D., Sleath, H., Cama, J., Keyser, U. F. Characterization of lipid composition and diffusivity in OLA generated vesicles. Biochimica et Biophysica Acta (BBA)-Biomembranes. 1862 (9), 183359 (2020).
  39. Deshpande, S., Spoelstra, W. K., Van Doorn, M., Kerssemakers, J., Dekker, C. Mechanical division of cell-sized liposomes. ACS Nano. 12 (3), 2560-2568 (2018).
  40. Jusková, P., et al. 34;Basicles&#34;: Microbial growth and production monitoring in giant lipid vesicles. ACS Applied Materials & Interfaces. 11 (38), 34698-34706 (2019).
  41. Fletcher, M., et al. DNA-based optical quantification of ion transport across giant vesicles. ACS Nano. 16 (10), 17128-17138 (2022).
  42. Vaezi, Z., et al. Investigation of the programmed cell death by encapsulated cytoskeleton drug liposomes using a microfluidic platform. Microfluidics and Nanofluidics. 24 (7), 48 (2020).
  43. Al Nahas, K., et al. A microfluidic platform for the characterisation of membrane active antimicrobials. Lab on a Chip. 19 (5), 837-844 (2019).
  44. Bao, P., et al. Production of giant unilamellar vesicles and encapsulation of lyotropic nematic liquid crystals. Soft Matter. 17 (8), 2234-2241 (2021).
  45. Yandrapalli, N., Petit, J., Bäumchen, O., Robinson, T. Surfactant-free production of biomimetic giant unilamellar vesicles using PDMS-based microfluidics. Communications Chemistry. 4, 100 (2021).
  46. Cama, J., et al. An ultrasensitive microfluidic approach reveals correlations between the physico-chemical and biological activity of experimental peptide antibiotics. Scientific Reports. 12, 4005 (2022).
  47. Guerzoni, L. P., et al. High macromolecular crowding in liposomes from microfluidics. Advanced Science. 9 (27), 2201169 (2022).
  48. Gonzales, D. T., Yandrapalli, N., Robinson, T., Zechner, C., Tang, T. D. Cell-free gene expression dynamics in synthetic cell populations. ACS Synthetic Biology. 11 (1), 205-215 (2022).
  49. Ushiyama, R., Koiwai, K., Suzuki, H. Plug-and-play microfluidic production of monodisperse giant unilamellar vesicles using droplet transfer across water-oil interface. Sensors and Actuators B: Chemical. 355, 131281 (2022).
  50. Banlaki, I., Lehr, F. -. X., Niederholtmeyer, H., Karim, A. S., Jewett, M. C. Microfluidic production of porous polymer cell-mimics capable of gene expression. Cell-Free Gene Expression. , 237-255 (2022).

This article has been published

Video Coming Soon

JoVE Logo

Privacy

Terms of Use

Policies

Research

Education

ABOUT JoVE

Copyright © 2024 MyJoVE Corporation. All rights reserved