Summary

Высокая пропускная способность Single-камеру и несколько клеток микро-инкапсуляции

Published: June 15, 2012
doi:

Summary

Объединение монодисперсных падение поколения с инерционным порядком клеток и частиц, описывается метод инкапсуляции нужное количество клеток или частиц в одной капле в кГц ставки. Мы демонстрируем эффективность в два раза превышающие неупорядоченных инкапсуляции для одно-и двух-частиц падает.

Abstract

Microfluidic encapsulation methods have been previously utilized to capture cells in picoliter-scale aqueous, monodisperse drops, providing confinement from a bulk fluid environment with applications in high throughput screening, cytometry, and mass spectrometry. We describe a method to not only encapsulate single cells, but to repeatedly capture a set number of cells (here we demonstrate one- and two-cell encapsulation) to study both isolation and the interactions between cells in groups of controlled sizes. By combining drop generation techniques with cell and particle ordering, we demonstrate controlled encapsulation of cell-sized particles for efficient, continuous encapsulation. Using an aqueous particle suspension and immiscible fluorocarbon oil, we generate aqueous drops in oil with a flow focusing nozzle. The aqueous flow rate is sufficiently high to create ordering of particles which reach the nozzle at integer multiple frequencies of the drop generation frequency, encapsulating a controlled number of cells in each drop. For representative results, 9.9 μm polystyrene particles are used as cell surrogates. This study shows a single-particle encapsulation efficiency Pk=1 of 83.7% and a double-particle encapsulation efficiency Pk=2 of 79.5% as compared to their respective Poisson efficiencies of 39.3% and 33.3%, respectively. The effect of consistent cell and particle concentration is demonstrated to be of major importance for efficient encapsulation, and dripping to jetting transitions are also addressed.

Introduction

Continuous media aqueous cell suspensions share a common fluid environment which allows cells to interact in parallel and also homogenizes the effects of specific cells in measurements from the media. High-throughput encapsulation of cells into picoliter-scale drops confines the samples to protect drops from cross-contamination, enable a measure of cellular diversity within samples, prevent dilution of reagents and expressed biomarkers, and amplify signals from bioreactor products. Drops also provide the ability to re-merge drops into larger aqueous samples or with other drops for intercellular signaling studies.1,2 The reduction in dilution implies stronger detection signals for higher accuracy measurements as well as the ability to reduce potentially costly sample and reagent volumes.3 Encapsulation of cells in drops has been utilized to improve detection of protein expression,4 antibodies,5,6 enzymes,7 and metabolic activity8 for high throughput screening, and could be used to improve high throughput cytometry.9 Additional studies present applications in bio-electrospraying of cell containing drops for mass spectrometry10 and targeted surface cell coatings.11 Some applications, however, have been limited by the lack of ability to control the number of cells encapsulated in drops. Here we present a method of ordered encapsulation12 which increases the demonstrated encapsulation efficiencies for one and two cells and may be extrapolated for encapsulation of a larger number of cells.

To achieve monodisperse drop generation, microfluidic “flow focusing” enables the creation of controllable-size drops of one fluid (an aqueous cell mixture) within another (a continuous oil phase) by using a nozzle at which the streams converge.13 For a given nozzle geometry, the drop generation frequency f and drop size can be altered by adjusting oil and aqueous flow rates Qoil and Qaq. As the flow rates increase, the flows may transition from drop generation to unstable jetting of aqueous fluid from the nozzle.14

When the aqueous solution contains suspended particles, particles become encapsulated and isolated from one another at the nozzle. For drop generation using a randomly distributed aqueous cell suspension, the average fraction of drops Dk containing k cells is dictated by Poisson statistics, where Dk = λk exp(-λ)/(k!) and λ is the average number of cells per drop. The fraction of cells which end up in the “correctly” encapsulated drops is calculated using Pk = (k x Dk)/Σ(k’ x Dk). The subtle difference between the two metrics is that Dk relates to the utilization of aqueous fluid and the amount of drop sorting that must be completed following encapsulation, and Pk relates to the utilization of the cell sample. As an example, one could use a dilute cell suspension (low λ) to encapsulate drops where most drops containing cells would contain just one cell. While the efficiency metric Pk would be high, the majority of drops would be empty (low Dk), thus requiring a sorting mechanism to remove empty drops, also reducing throughput.15

Combining drop generation with inertial ordering provides the ability to encapsulate drops with more predictable numbers of cells per drop and higher throughputs than random encapsulation. Inertial focusing was first discovered by Segre and Silberberg16 and refers to the tendency of finite-sized particles to migrate to lateral equilibrium positions in channel flow. Inertial ordering refers to the tendency of the particles and cells to passively organize into equally spaced, staggered, constant velocity trains. Both focusing and ordering require sufficiently high flow rates (high Reynolds number) and particle sizes (high Particle Reynolds number).17,18 Here, the Reynolds number Re =uDh and particle Reynolds number Rep =Re(a/Dh)2, where u is a characteristic flow velocity, Dh [=2wh/(w+h)] is the hydraulic diameter, ν is the kinematic viscosity, a is the particle diameter, w is the channel width, and h is the channel height. Empirically, the length required to achieve fully ordered trains decreases as Re and Rep increase. Note that the high Re and Rep requirements (for this study on the order of 5 and 0.5, respectively) may conflict with the need to keep aqueous flow rates low to avoid jetting at the drop generation nozzle. Additionally, high flow rates lead to higher shear stresses on cells, which are not addressed in this protocol. The previous ordered encapsulation study demonstrated that over 90% of singly encapsulated HL60 cells under similar flow conditions to those in this study maintained cell membrane integrity.12 However, the effect of the magnitude and time scales of shear stresses will need to be carefully considered when extrapolating to different cell types and flow parameters. The overlapping of the cell ordering, drop generation, and cell viability aqueous flow rate constraints provides an ideal operational regime for controlled encapsulation of single and multiple cells.

Because very few studies address inter-particle train spacing,19,20 determining the spacing is most easily done empirically and will depend on channel geometry, flow rate, particle size, and particle concentration. Nonetheless, the equal lateral spacing between trains implies that cells arrive at predictable, consistent time intervals. When drop generation occurs at the same rate at which ordered cells arrive at the nozzle, the cells become encapsulated within the drop in a controlled manner. This technique has been utilized to encapsulate single cells with throughputs on the order of 15 kHz,12 a significant improvement over previous studies reporting encapsulation rates on the order of 60-160 Hz.4,15 In the controlled encapsulation work, over 80% of drops contained one and only one cell, a significant efficiency improvement over Poisson (random) statistics, which predicts less than 40% efficiency on average.12

In previous controlled encapsulation work,12 the average number of particles per drop λ was tuned to provide single-cell encapsulation. We hypothesize that through tuning of flow rates, we can efficiently encapsulate any number of cells per drop when λ is equal or close to the number of desired cells per drop. While single-cell encapsulation is valuable in determining individual cell responses from stimuli, multiple-cell encapsulation provides information relating to the interaction of controlled numbers and types of cells. Here we present a protocol, representative results using polystyrene microspheres, and discussion for controlled encapsulation of multiple cells using a passive inertial ordering channel and drop generation nozzle.

Protocol

Протоколы в этом разделе описываются материалов и оборудования, используемого в частности получить экспериментальные результаты. Обратите внимание, что альтернативные поставщики для химикатов и оборудования могут быть использованы. 1. Изготовление устройств и мягкие ?…

Discussion

Несмотря на относительно высокую степень упорядоченности, не все капли будет содержать нужное количество частиц или клеток. Инкапсуляции эффективность может рассчитываться как число клеток или частиц, которые становятся заключены в каплях с желаемой занятости разделить их общего чи…

Divulgations

The authors have nothing to disclose.

Acknowledgements

Мы благодарим Raindance технологии для выборки PFPE-PEG поверхностно-активных веществ, используемых в данном исследовании, и мы благодарим BioMEMS ресурсный центр (Mehmet Toner, директор) для формы кремниевой пластины используются для создания реплик PDMS канала.

Materials

Name of the reagent Company Catalogue number Comments
AutoCAD AutoDesk    
Transparency Mask Fineline Imaging Inc.    
SU-8 Photoresist MicroChem 2050  
Dektak Profilometer Veeco    
Petri Dish BD Falcon 351058  
PDMS Silicone Elastomer Kit Dow Corning Corp. Sylgard 184, Material Number (240)4019862  
Vacuum Desiccator Jencons 250-030  
Vacuum Pump Alcatel Vacuum Technology 2010 C2  
Vacuum Regulator Cole-Parmer EW-00910-10  
Oven Thermo Scientific Lindberg Blue M, OV800F  
Biopsy Punch, 0.75 mm Harris Uni-Core 15072  
Laboratory Corona Treater Electro-Technic Products Inc. BD-20AC, SKU 12051A  
Glass Slides Gold Seal 3010  
Aquapel PPG Industries   Alternative Strategy
Polystyrene Microspheres, 9.9 μm Thermo G1000  
OptiPrep Sigma-Aldrich D1556 Not Demonstrated
Luer-Lok Syringes BD 1 mL: 309628
3 mL: 309585
 
FC-40 Fluorocarbon Oil 3M Inc. Sigma Aldrich, F9755  
PFPE-PEG Fluorosurfactant RainDance Technologies    
Light Mineral Oil PTI Process Chemicals 08042-47-5 Alternative Strategy
Mineral Oil Surfactant Evonik Goldschmidt Corporation ABIL EM 90 Alternative Strategy
Tygon PVC Tubing SmallParts TGY-010  
30 Gauge Luer-Lok Syringe Needle, 1/2″ SmallParts NE-301PL-C  
Inverted Microscope Carl Zeiss Imaging Axio Observer.Z1  
High Speed Camera Vision Research Phantom V310  
Syringe Pumps (2) Chemyx Inc. Nexus 3000  
Silicone Oil Dow Corning 200 fluid, 10 cSt Optional for Emulsion Storage

References

  1. Zagnoni, M., Lain, G. L. e., Cooper, J. M. Electrocoalescence mechanisms of microdroplets using localized electric fields in microfluidic channels. Langmuir : the ACS journal of surfaces and colloids. 26, 14443-14449 (2010).
  2. Niu, X. Z., Gielen, F., Edel, J. B., deMello, A. J. A microdroplet dilutor for high-throughput screening. Nat. Chem. 3, 437-442 (2011).
  3. Vincent, M. E., Liu, W., Haney, E. B., Ismagilov, R. F. Microfluidic stochastic confinement enhances analysis of rare cells by isolating cells and creating high density environments for control of diffusible signals. Chemical Society reviews. 39, 974-984 (2010).
  4. Huebner, A. Quantitative detection of protein expression in single cells using droplet microfluidics. Chemical communications. , 1218-1220 (2007).
  5. Love, J. C., Ronan, J. L., Grotenbreg, G. M., van der Veen, A. G., Ploegh, H. L. A microengraving method for rapid selection of single cells producing antigen-specific antibodies. Nature biotechnology. 24, 703-707 (2006).
  6. Bradshaw, E. M. Concurrent detection of secreted products from human lymphocytes by microengraving: Cytokines and antigen-reactive antibodies. Clin. Immunol. 129, 10-18 (2008).
  7. Liu, W. S., Kim, H. J., Lucchetta, E. M., Du, W. B., Ismagilov, R. F. Isolation, incubation, and parallel functional testing and identification by FISH of rare microbial single-copy cells from multi-species mixtures using the combination of chemistrode and stochastic confinement. Lab on a chip. 9, 2153-2162 (2009).
  8. Boedicker, J. Q., Li, L., Kline, T. R., Ismagilov, R. F. Detecting bacteria and determining their susceptibility to antibiotics by stochastic confinement in nanoliter droplets using plug-based microfluidics. Lab on a chip. 8, 1265-1272 (2008).
  9. Koster, S. Drop-based microfluidic devices for encapsulation of single cells. Lab on a chip. 8, 1110-1115 (2008).
  10. Kelly, R. T., Page, J. S., Marginean, I., Tang, K., Smith, R. D. Dilution-free analysis from picoliter droplets by nano-electrospray ionization mass spectrometry. Angew Chem. Int. Ed. Engl. 48, 6832-6835 (2009).
  11. Hong, J., deMello, A. J., Jayasinghe, S. N. Bio-electrospraying and droplet-based microfluidics: control of cell numbers within living residues. Biomedical materials. 5, 21001 (2010).
  12. Edd, J. F. Controlled encapsulation of single-cells into monodisperse picolitre drops. Lab on a chip. 8, 1262-1264 (2008).
  13. Anna, S. L., Bontoux, N., Stone, H. A. Formation of dispersions using “flow focusing” in microchannels. Applied Physics Letters. 82, 364 (2003).
  14. Utada, A., Fernandez-Nieves, A., Stone, H., Weitz, D. Dripping to Jetting Transitions in Coflowing Liquid Streams. Physical Review Letters. 99, (2007).
  15. Chabert, M., Viovy, J. L. Microfluidic high-throughput encapsulation and hydrodynamic self-sorting of single cells. Proceedings of the National Academy of Sciences of the United States of America. 105, 3191-3196 (2008).
  16. Segrí, G., Silberberg, A. Radial Particle Displacements in Poiseuille Flow of Suspensions. Nature. 189, 209-210 (1961).
  17. Carlo, D. D. i. Inertial microfluidics. Lab on a chip. 9, 3038-3046 (2009).
  18. Carlo, D. D. i., Edd, J., Humphry, K., Stone, H., Toner, M. Particle Segregation and Dynamics in Confined Flows. Physical Review Letters. 102, (2009).
  19. Humphry, K. J., Kulkarni, P. M., Weitz, D. A., Morris, J. F., Stone, H. A. Axial and lateral particle ordering in finite Reynolds number channel flows. Physics of Fluids. 22, 081703 (2010).
  20. Lee, W., Amini, H., Stone, H. A., Carlo, D. D. i. Dynamic self-assembly and control of microfluidic particle crystals. Proceedings of the National Academy of Sciences of the United States of America. 107, 22413 (2010).
  21. Duffy, D. C., McDonald, J. C., Schueller, O. J. A., Whitesides, G. M. Rapid prototyping of microfluidic systems in poly(dimethylsiloxane. Anal. Chem. 70, 4974-4984 (1998).
  22. Kotz, K., Cheng, X., Toner, M. PDMS Device Fabrication and Surface Modification. J. Vis. Exp. (8), e319 (2007).
  23. Haubert, K., Drier, T., Beebe, D. PDMS bonding by means of a portable, low-cost corona system. Lab on a chip. 6, 1548-1549 (2006).
  24. Hatch, A. C. 1-Million droplet array with wide-field fluorescence imaging for digital PCR. Lab on a chip. , 3838-3845 (2011).
  25. Holtze, C. Biocompatible surfactants for water-in-fluorocarbon emulsions. Lab on a chip. 8, 1632-1639 (2008).
  26. Garstecki, P., Stone, H., Whitesides, G. Mechanism for Flow-Rate Controlled Breakup in Confined Geometries: A Route to Monodisperse Emulsions. Physical Review Letters. 94, (2005).
  27. Garstecki, P., Fuerstman, M. J., Stone, H. A., Whitesides, G. M. Formation of droplets and bubbles in a microfluidic T-junction-scaling and mechanism of break-up. Lab on a chip. 6, 437-446 (2006).
  28. Nie, Z. Emulsification in a microfluidic flow-focusing device: effect of the viscosities of the liquids. Microfluidics and Nanofluidics. , (2008).
  29. Holt, D. J., Payne, R. J., Chow, W. Y., Abell, C. Fluorosurfactants for microdroplets: interfacial tension analysis. Journal of colloid and interface science. 350, 205-211 (2010).
  30. Holt, D. J., Payne, R. J., Abell, C. Synthesis of novel fluorous surfactants for microdroplet stabilisation in fluorous oil streams. Journal of Fluorine Chemistry. 131, 398-407 (2010).
  31. Hatch, A. C., Fisher, J. S., Pentoney, S. L., Yang, D. L., Lee, A. P. Tunable 3D droplet self-assembly for ultra-high-density digital micro-reactor arrays. Lab on a chip. 11, 2509-2517 (2011).
  32. Baret, J. C. Surfactants in droplet-based microfluidics. Lab on a chip. 12, 422-433 (2012).
check_url/fr/4096?article_type=t

Play Video

Citer Cet Article
Lagus, T. P., Edd, J. F. High Throughput Single-cell and Multiple-cell Micro-encapsulation. J. Vis. Exp. (64), e4096, doi:10.3791/4096 (2012).

View Video