Next Article in Journal
New Perspectives on the Use of Phytochemicals as an Emergent Strategy to Control Bacterial Infections Including Biofilms
Next Article in Special Issue
A PDMS-Based Microfluidic Hanging Drop Chip for Embryoid Body Formation
Previous Article in Journal
Design and Stereochemical Research (DFT, ECD and Crystal Structure) of Novel Bedaquiline Analogs as Potent Antituberculosis Agents
Previous Article in Special Issue
The Deformation of Polydimethylsiloxane (PDMS) Microfluidic Channels Filled with Embedded Circular Obstacles under Certain Circumstances
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Development of Droplet Microfluidics Enabling High-Throughput Single-Cell Analysis

1
Institute of Electronics, Chinese Academy of Sciences, Beijing 100190, China
2
Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
*
Authors to whom correspondence should be addressed.
Molecules 2016, 21(7), 881; https://doi.org/10.3390/molecules21070881
Submission received: 30 May 2016 / Revised: 27 June 2016 / Accepted: 28 June 2016 / Published: 5 July 2016
(This article belongs to the Special Issue Micro/Nano Fluidics and Bio-MEMS)

Abstract

:
This article reviews recent developments in droplet microfluidics enabling high-throughput single-cell analysis. Five key aspects in this field are included in this review: (1) prototype demonstration of single-cell encapsulation in microfluidic droplets; (2) technical improvements of single-cell encapsulation in microfluidic droplets; (3) microfluidic droplets enabling single-cell proteomic analysis; (4) microfluidic droplets enabling single-cell genomic analysis; and (5) integrated microfluidic droplet systems enabling single-cell screening. We examine the advantages and limitations of each technique and discuss future research opportunities by focusing on key performances of throughput, multifunctionality, and absolute quantification.

Graphical Abstract

1. Introduction

Cellular heterogeneity arising from stochastic expressions of genes, proteins and metabolites is a key component of cell biology and, thus, analyzing individual cells can better reveal cell-to-cell variations which are masked by bulk measurements [1,2]. Technique developments in single-cell separation (e.g., liquid chromatography and electrophoresis) and detection (e.g., laser induced fluorescence, mass spectroscopy imaging, electrochemistry, and chemiluminescence) enable single-cell analysis at both the intracellular levels (e.g., genomic, transcriptomic, and proteomic studies) and at the levels of secretory responses, microenvironments, and cell-cell interactions [3,4,5,6].
One emerging tool with the potential to provide new opportunities of single-cell analysis is microfluidics [7,8,9]. As a technology on the processing and manipulation of small amounts of fluids (10−9 to 10−18 liters) in channels with dimensions of tens of micrometers [10,11,12], microfluidics matches with the sizes of biological cells and, thus, functions as a promising platform for cellular analysis [12,13,14], enabling the characterization of biochemical (e.g., gene [15] and protein [16,17,18]) and/or biophysical properties (mechanical [19,20,21] and electrical properties [19,22,23]) of single cells.
As a subset of microfluidics, droplet microfluidics involves the production of microscale droplets (typically on the order of tens to hundreds of µm in diameter) of one fluid within a second immiscible carrier fluid in a high-throughput manner, leading to promising applications in a variety of fields including directed evolution, tissue printing, and polymerase chain reaction (PCR) [24,25,26,27,28,29]. In the field of single-cell analysis, droplet microfluidics is also promising since each cell can be individually confined within its own droplet, and cell-secreted molecules can rapidly reach detectable concentrations in the confined fluid surrounding the encapsulated single cells [30,31,32,33].
Rather than a comprehensive review, this mini review is aimed to briefly summarize key developments in the applications of droplet microfluidics in single-cell analysis, which includes (1) prototype demonstration of single-cell encapsulation in microfluidic droplets; (2) technical improvements of single-cell encapsulation in microfluidic droplets; (3) microfluidic droplets enabling single-cell proteomic analysis; (4) microfluidic droplets enabling single-cell genomic analysis; and (5) integrated microfluidic droplet systems enabling single-cell screening. Furthermore, we examine the advantages and limitations of each technique and discuss future research opportunities by focusing on three key parameters: throughput, multifunctionality, and absolute quantification.

2. Prototype Demonstration of Single-Cell Encapsulation in Microfluidic Droplets

As pioneers in the field of single-cell encapsulation in microfluidic droplets, in 2005, Chiu et al. combined optical trapping and microfluidic T-junctions to encapsulate single cells in picoliter- or femtoliter-volume aqueous droplets surrounded by an immiscible phase (see Figure 1). More specifically, optical trapping was used to transport and position single cells close to the water/oil interface, followed by a pressure pulse, which sheared off the aqueous phase into a single droplet with single cells encapsulated. In addition, laser-induced lysis of cells within droplets was demonstrated and the activities of an intracellular enzyme were assayed using corresponding fluorogenic substrates [34].
In addition to confining single cells in aqueous droplets surrounded by a continuous oil phase, microfluidic flow-focusing structures were also used to (1) encapsulate single cells (e.g., Hela, MCF7, and yeast cells) in lipid vesicles surrounded by a continuous aqueous phase [35] and to (2) generate cell-enclosing agarose microcapsules in a continuous oil phase [36]. Although these prototype devices suffered from key shortcomings, such as low throughputs and low single-cell trapping efficiencies, these pioneering studies indeed open new possibilities for carrying out single-cell studies in droplet microfluidics, and pave the foundations for the upcoming studies focusing on single-cell proteomic analysis, genetic analysis, and high-throughput screening based on droplet microfluidics.

3. Technical Improvements in Single-Cell Encapsulation in Microfluidic Droplets

Accurate control of the number of cells per droplet is a challenging issue due to the nature of Poisson (random) encapsulation where the Poisson probability of a droplet containing one and only one cell is only 36.8%, and the probability of pairing two distinct cell types in a droplet is reduced to 13.5%. In practice, in order to make sure that no two cells are confined within one droplet, cell suspensions are further diluted, leading to a large number of empty droplets, which is wasteful [33].
The first approach to address this issue was to remove empty droplets after single-cell confinements [37,38,39,40]. As the first demonstration, Viovy et al. presented a purely hydrodynamic method for encapsulation of single cells into picoliter droplets, followed by spontaneous self-sorting based on the sizes. As shown in Figure 2, encapsulation was realized based on a cell-triggered Rayleigh–Plateau instability in a flow-focusing geometry, and self-sorting relied on two extra hydrodynamic mechanisms, which are lateral drift of deformable objects in a shear flow, and sterically-driven dispersion in a compressional flow, respectively. Successful encapsulation and sorting of 70%–80% of the droplets containing one and only one cell was reported, demonstrating a significant improvement in comparison to random cell encapsulation [37].
Furthermore, passive separation of microfluidic droplets by size was also enabled by deterministic lateral displacement where a tilted pillar array allows droplets smaller than a certain critical diameter to follow the direction of the incoming fluid flow while larger droplets are constrained to follow the tilted lanes of the pillar array [39,40]. Based on this microfluidic structure, recent studies (1) sorted out shrunken yeast-cell containing droplets from 31% larger diameter droplets which were generated at the same time containing only media [39]; and (2) separated large droplets encapsulating tumor cells (diameter, ~25 µm) and small empty droplets (diameter, ~14 µm), enriching the single-cell encapsulated droplets to roughly 78% [40].
Meanwhile, inertial focusing was used to evenly space single cells before the emulsification process, which can significantly improve the single-cell encapsulation efficiency in droplet microfluidics [41,42,43,44,45]. As pioneers in this field, Toner and coworkers forced a high-density suspension of cells to travel rapidly through a high aspect-ratio straight microchannel to evenly space cells, reporting a single-cell encapsulation efficiency of 80% (see Figure 3a) [41]. Furthermore, a curved microchannel was introduced to bring a second Dean force to focus cells into a single equilibrium position, with the single-cell encapsulation efficiency quantified as ~77% (see Figure 3b) [42]. Recently, a short pinched flow channel consisting of contracting and expanding chambers was also used to conduct inertial focusing along the center of the channel, enabling the encapsulation of single cells with >55% single-cell efficiencies (see Figure 3c) [43].
Although several studies have been conducted to address the issue of low efficiency of single-cell encapsulation, the optimal encapsulation efficiency was only about 80%. Thus, more studies are suggested to further address this issue from the point of technical development. However, if we view this issue from the application perspective, whether further improvements in the efficiency of single-cell encapsulation is a must is questionable. For instance, in the following sections of single-cell proteomic/genomic analysis, the relatively low trapping efficiency of single-cell confinement is indeed not a top concern.

4. Microfluidic Droplets Enabling Single-Cell Proteomic Analysis

Following technical developments capable of encapsulating single cells with high throughput and efficiency, the research focus was then shifted to applications in the field of single-cell proteomic and genomic analysis, which may function as integrated platforms for high-throughput screening.
In the droplet microfluidics, single cells are confined within small volumes, allowing rapid accumulations of secreted metabolites to detectable levels [34,43,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70] (see Table 1). The first approach was demonstrated by Hollfelder et al. to assay the activities of the enzyme alkaline phosphatase expressed by Escherichia coli cells. As shown in Figure 4a, individual E coli and substrate 3-O-methylfluorescein-phosphates were encapsulated within single droplets where the substrates were enzymatically hydrolyzed by the target enzyme alkaline phosphatase expressed by E coli cells, leading to fluorescent detections [47]. In a follow-up study, the activities of glucuronidase was quantified by encapsulating single E. coli with a fluorogenic reporter of 4-Methylumbelliferyl β-d-glucuronide [59]. Furthermore, the activity of receptor tyrosine kinases (RTKs) in lung cancer cells was assayed by binding surface ligands of 8-hydroxy-5-(N,N-dimethylsulfonamido)-2-methylquinoline), generating fluorescent signals [43].
As to the detection of the secreted antibodies of single cells, both microspheres conjugated with capture antibodies and detection fluorescence labeled antibodies are encapsulated with single cells. The secreted substances are captured on the microsphere surfaces, leading to the further binding of fluorescence-labeled detection antibodies, which generates localized fluorescent signals on microsphere surfaces (see Figure 4b) [58,60,64]. Using this approach, (1) secreted IL-10 of CD4 + CD25 + regulatory T cells [58]; (2) intracellular HRas-mCitrine of HEK-293 cells and actin-EGFP of MCF-7 cells [60]; and (3) secreted IL-2, IFN-γ, and TNF-α of activated T-cells [64] were assayed, respectively.
Although powerful, there are two critical concerns for single-cell proteomic analysis in droplet microfluidics. (1) The local microenvironments (e.g., nutrient levels and gas permeability) for single-cell encapsulation may be significantly different from in vivo situations and if single cells are challenged by harsh environments, the metabolites may not indicate normal activities of these cells; (2) the current approaches can only report fluorescent intensities without quantifying the absolute number of metabolites under interest due to the lack of effective calibration approaches. Since fluorescent intensities are deeply influenced by experimental setups (excitation laser intensities and geometries of the droplets), results collected by different groups cannot be effectively compared.

5. Microfluidic Droplets Enabling Single-Cell Genomic Analysis

In addition to applications in single-cell proteomic analysis, droplet microfluidics has also functioned as an effective tool enabling single-cell genomic analysis [71,72,73,74,75,76,77,78,79,80,81,82,83,84,85] (see Table 2). As pioneers in this field, Mathies et al. utilized a microfluidic droplet generator to encapsulate individual cells together with primer functionalized microbeads in uniform PCR mix droplets. After bulk PCR amplification, the droplets were lysed and the micro beads were recovered and rapidly analyzed via flow cytometry. Successful single-cell analysis of the glyceraldehyde 3 phosphate dehydrogenase gene in human lymphocyte cells and of the gyr B gene in bacterial E. coli K12 cells validated the proposed approach for performing high-throughput genetic analysis on single cells (see Figure 5a) [71].
This approach was then scaled up to an ultra-high throughput system with 96 channels in parallel to generate up to 3.4 × 106 nanoliter-volume droplets per hour. Leveraging this platform, pathogenic E. coli O157 cells were identified in a high background of normal K12 cells, with a detection limit on the order of 1:105 [75]. Furthermore, single cells and primer functionalized microbeads were confined with agarose droplets rather than aqueous droplets, which can help (1) maintain single genome fidelity during cell lysis and DNA purification; (2) improve the efficiency of emulsion PCR. Using this approach, multi-locus single-cell sequencing of the control gene β-actin and across the chromosomal translocation t (14;18), a mutation associated with 85%–90% of follicular lymphoma cases was demonstrated [79].
However, this aforementioned method requires the precision pairing of a single cell and a microbead within a single droplet, which is a challenging issue. To address this issue, Yang et al. developed a bead-free agarose droplet-based microfluidic method for emulsification PCR, where reverse primers were covalently conjugated to agarose (see Figure 5b) [74]. Since agarose functioned as the trapping matrix to replace conventional primer functionalized microbeads, the efficiency of droplet generation was increased by one to two orders of magnitude. In addition, during all of the PCR cycling temperatures, the agarose was always in the liquid form, which can effectively address the drawbacks of PCR at the solid surface of the microbeads, leading to high PCR efficiencies (~95%).
Leveraging this approach, single-cell PCR was conducted to identify a single pathogen E. coli O157:H7 in the high background of 100,000 excess normal K12 cells [82]. In addition, this approach was optimized to realize single-cell RT-PCR with two aqueous inlets flushed with cells and RT-PCR reagents/cell lysis buffer, respectively. Single-cell RT-PCR was successfully performed, recording a clear difference in gene expression levels of EpCAM, a cancer biomarker gene, at the single-cell level between different types of cancer cells [81].
Currently, the reported single-cell PCR in droplet microfluidics always requires the off-line amplification of interested gene sections and then droplets are flushed in the flow cytometry for further characterization. Further technical developments may integrate functional units of on-chip droplet formation, PCR or RT-PCR, as well as fluorescence detection, maximizing the multifunctional capabilities of microfluidics.

6. Integrated Microfluidic Droplet Systems Enabling Single-Cell Screening

The integrated microfluidic droplet systems were also developed for single-cell screening, which integrates key steps of cell encapsulation, incubation, fluorescence detection of metabolic materials, and droplet sorting relying on the fluorescent intensities [53,86,87,88,89,90,91,92,93]. As a robust working platform, this type of the integrated microfluidic system has been used to screen (1) the β-galactosidase activity of E. coli [86]; (2) cytotoxic effects against U937 cells [87]; (3) the horseradish peroxidase activity of yeasts [88]; (4) the effect of rifampicin against E. coli [89]; (5) antibody secretion capabilities of mouse hybridoma cells; and 6) xylose-overconsuming capabilities of Saccharomyces cerevisiae cells and L-lactate–producing capabilities of E. coli [92].
As a proof-of-concept demonstration, Griffiths et al. proposed a droplet microfluidic platform enabling the fluorescence-activated cell sorting based on enzyme activities of encapsulated single cells (see Figure 6) [86]. In this study, mixtures of E. coli cells, expressing either the reporter enzyme b-galactosidase or an inactive variant, were compartmentalized with a fluorogenic substrate, which were further sorted using dielectrophoresis in a fluorescence-activated manner at rates up to 300 droplets s−1. When the cells were encapsulated at a low density (approximately one cell for every 50 droplets), sorting was very efficient and all of the recovered cells were the active strain.
Furthermore, this approach was used to screen antibody secretion capabilities of single mouse hybridoma cells [91]. Single mouse hybridoma cells, fluorescent probes, and single beads coated with anti-mouse IgG antibodies were encapsulated in individual 50-pl droplets. Following the capture of the secreted antibodies by the micro beads, which further bind to the fluorescent probes, the fluorescence becomes localized on the beads, generating a clearly distinguishable fluorescence signal that enables droplet sorting at ~200 Hz.

7. Conclusions and Future Work

In this study, we summarize key developments of droplet microfluidics enabling single-cell analysis. Although significant improvements have been made within the last decade, there is still significant room left for studies in terms of throughput, multifunctionality, and absolute quantification.
Throughput is always a concern in droplet microfluidics, especially when the limited cross-sectional area of the droplet generating microchannels was taken into consideration. In the current design, multiple channels can be scaled up to generate droplets in parallel, leading to ultrahigh throughput in the formation of microdroplets. However, in the step of fluorescent detection, droplets under measurement are loaded into the microfluidic channels and detected in a serial manner, compromising the overall throughput. Future work may focus on the scaling up of the fluorescent detection units of droplet microfluidics, further improving the throughput of droplet microfluidics.
The second issue is the multi-functional capabilities of droplet microfluidics. Confinement of single cells within individual droplets is beneficial in cellular analysis without the concern of cross-contamination. However, multi-step sampling mixture and rinsing cannot be effectively conducted in droplet microfluidics, limiting functional extensions of the current microfluidic platforms. Thus, currently, only limited functional units, including cellular encapsulation, incubation, as well as fluorescent generation and detection, have been effectively integrated, and future studies may extend the functions of the current microfluidic systems by integrating more key steps including droplet fusion, division, and manipulation to the field of single-cell analysis.
In the field of single-cell droplet microfluidics, absolute quantification is a key issue, which definitely deserves more attention. Taking single-cell proteomic analysis as an example. The use of cytokine-capture beads which function as fluorescent microspots in individual droplets leads to uneven fluorescent signals in droplets, resulting in the issue of poor calibration. Without an effective calibration approach, the secreted molecules of single cells cannot be absolutely quantified and, thus, experiments from different groups cannot be compared and discussed. Thus, more studies are required to enable the absolute quantification in the field of single-cell droplets.

Acknowledgments

The authors would like to acknowledge financial support from National Natural Science Foundation of China (Grant No. 61431019), National Basic Research Program of China (973 Program, Grant No. 2014CB744600), Natural Science Foundation of Beijing (4152056), Instrument Development Program of the Chinese Academy of Sciences, and Beijing NOVA Program of Science and Technology.

Author Contributions

N.W. contributed to the introduction section; Z.Z. reviewed the prototype demonstration of single-cell encapsulation in microfluidic droplets; B.F. reviewed the technical improvements in single-cell encapsulation in microfluidic droplets; D.C. reviewed the microfluidic droplets enabling single-cell proteomic analysis; D.M. reviewed the microfluidic droplets enabling single-cell genomic analysis; J.W. reviewed integrated microfluidic droplet systems enabling single-cell screening; J.C. contributed to the section of conclusion and future work. N.W. and J.C. drafted the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, D.; Bodovitz, S. Single cell analysis: The new frontier in ‘omics’. Trends Biotechnol. 2010, 28, 281–290. [Google Scholar] [CrossRef] [PubMed]
  2. Fritzsch, F.S.; Dusny, C.; Frick, O.; Schmid, A. Single-cell analysis in biotechnology, systems biology, and biocatalysis. Annu. Rev. Chem. Biomol. Eng. 2012, 3, 129–155. [Google Scholar] [CrossRef] [PubMed]
  3. Tsioris, K.; Torres, A.J.; Douce, T.B.; Love, J.C. A new toolbox for assessing single cells. Annu. Rev. Chem. Biomol. Eng. 2014, 5, 455–477. [Google Scholar] [CrossRef] [PubMed]
  4. Vasdekis, A.E.; Stephanopoulos, G. Review of methods to probe single cell metabolism and bioenergetics. Metab. Eng. 2015, 27, 115–135. [Google Scholar] [CrossRef] [PubMed]
  5. Klepárník, K.; Foret, F. Recent advances in the development of single cell analysis—A review. Anal. Chim. Acta 2013, 800, 12–21. [Google Scholar] [CrossRef] [PubMed]
  6. Haselgrübler, T.; Haider, M.; Ji, B.; Juhasz, K.; Sonnleitner, A.; Balogi, Z.; Hesse, J. High-throughput, multiparameter analysis of single cells. Anal. Bioanal. Chem. 2014, 406, 3279–3296. [Google Scholar] [CrossRef] [PubMed]
  7. Yin, H.; Marshall, D. Microfluidics for single cell analysis. Curr. Opin. Biotechnol. 2012, 23, 110–119. [Google Scholar] [CrossRef] [PubMed]
  8. Weaver, W.M.; Tseng, P.; Kunze, A.; Masaeli, M.; Chung, A.J.; Dudani, J.S.; Kittur, H.; Kulkarni, R.P.; di Carlo, D. Advances in high-throughput single-cell microtechnologies. Curr. Opin. Biotechnol. 2014, 25, 114–123. [Google Scholar] [CrossRef] [PubMed]
  9. Reece, A.; Xia, B.; Jiang, Z.; Noren, B.; McBride, R.; Oakey, J. Microfluidic techniques for high throughput single cell analysis. Curr. Opin. Biotechnol. 2016, 40, 90–96. [Google Scholar] [CrossRef] [PubMed]
  10. Squires, T.M.; Quake, S.R. Microfluidics: Fluid physics at the nanoliter scale. Rev. Mod. Phys. 2005, 77. [Google Scholar] [CrossRef]
  11. Whitesides, G.M. The origins and the future of microfluidics. Nature 2006, 442, 368–373. [Google Scholar] [CrossRef] [PubMed]
  12. Wootton, R.C.; de Mello, A.J. Microfluidics: Exploiting elephants in the room. Nature 2010, 464, 839–840. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, C.; van Noort, D. Cells in microfluidics. Top. Curr. Chem. 2011, 304, 295–321. [Google Scholar] [PubMed]
  14. Xiong, B.; Ren, K.; Shu, Y.; Chen, Y.; Shen, B.; Wu, H. Recent developments in microfluidics for cell studies. Adv. Mater. 2014, 26, 5525–5532. [Google Scholar] [CrossRef] [PubMed]
  15. Thompson, A.M.; Paguirigan, A.L.; Kreutz, J.E.; Radich, J.P.; Chiu, D.T. Microfluidics for single-cell genetic analysis. Lab Chip 2014, 14, 3135–3142. [Google Scholar] [CrossRef] [PubMed]
  16. Wei, W.; Shin, Y.S.; Ma, C.; Wang, J.; Elitas, M.; Fan, R.; Heath, J.R. Microchip platforms for multiplex single-cell functional proteomics with applications to immunology and cancer research. Genome Med. 2013, 5. [Google Scholar] [CrossRef] [PubMed]
  17. Yu, J.; Zhou, J.; Sutherland, A.; Wei, W.; Shin, Y.S.; Xue, M.; Heath, J.R. Microfluidics-based single-cell functional proteomics for fundamental and applied biomedical applications. Annu. Rev. Anal. Chem. 2014, 7, 275–295. [Google Scholar] [CrossRef] [PubMed]
  18. Fan, B.; Li, X.; Chen, D.; Peng, H.; Wang, J.; Chen, J. Development of microfluidic systems enabling high-throughput single-cell protein characterization. Sensors 2016, 16. [Google Scholar] [CrossRef] [PubMed]
  19. Zheng, Y.; Nguyen, J.; Wei, Y.; Sun, Y. Recent advances in microfluidic techniques for single-cell biophysical characterization. Lab Chip 2013, 13, 2464–2483. [Google Scholar] [CrossRef] [PubMed]
  20. Darling, E.M.; Carlo, D.D. High-throughput assessment of cellular mechanical properties. Annu. Rev. Biomed. Eng. 2015, 17, 35–62. [Google Scholar] [CrossRef] [PubMed]
  21. Xue, C.; Wang, J.; Zhao, Y.; Chen, D.; Yue, W.; Chen, J. Constriction channel based single-cell mechanical property characterization. Micromachines 2015, 6, 1794–1804. [Google Scholar] [CrossRef]
  22. Chen, J.; Xue, C.; Zhao, Y.; Chen, D.; Wu, M.H.; Wang, J. Microfluidic impedance flow cytometry enabling high-throughput single-cell electrical property characterization. Int. J. Mol. Sci. 2015, 16, 9804–9830. [Google Scholar] [CrossRef] [PubMed]
  23. Xu, Y.; Xie, X.; Duan, Y.; Wang, L.; Cheng, Z.; Cheng, J. A review of impedance measurements of whole cells. Biosens. Bioelectron. 2016, 77, 824–836. [Google Scholar] [CrossRef] [PubMed]
  24. Teh, S.Y.; Lin, R.; Hung, L.H.; Lee, A.P. Droplet microfluidics. Lab Chip 2008, 8, 198–220. [Google Scholar] [CrossRef] [PubMed]
  25. Theberge, A.B.; Courtois, F.; Schaerli, Y.; Fischlechner, M.; Abell, C.; Hollfelder, F.; Huck, W.T.S. Microdroplets in microfluidics: An evolving platform for discoveries in chemistry and biology. Angew. Chem. Int. Ed. 2010, 49, 5846–5868. [Google Scholar] [CrossRef] [PubMed]
  26. Ralf, S.; Martin, B.; Thomas, P.; Stephan, H. Droplet based microfluidics. Rep. Prog. Phys. 2012, 75, 207–230. [Google Scholar]
  27. Basova, E.Y.; Foret, F. Droplet microfluidics in (bio)chemical analysis. Analyst 2015, 140, 22–38. [Google Scholar] [CrossRef] [PubMed]
  28. Leman, M.; Abouakil, F.; Griffiths, A.D.; Tabeling, P. Droplet-based microfluidics at the femtolitre scale. Lab Chip 2015, 15, 753–765. [Google Scholar] [CrossRef] [PubMed]
  29. Shembekar, N.; Chaipan, C.; Utharala, R.; Merten, C.A. Droplet-based microfluidics in drug discovery, transcriptomics and high-throughput molecular genetics. Lab Chip 2016, 16, 1314–1331. [Google Scholar] [CrossRef] [PubMed]
  30. Brouzes, E. Droplet microfluidics for single-cell analysis. Methods Mol. Biol. 2012, 853, 105–139. [Google Scholar] [PubMed]
  31. Joensson, H.; Andersson Svahn, H. Droplet microfluidics-a tool for single-cell analysis. Angew. Chem. Int. Ed. 2012, 51, 12176–12192. [Google Scholar] [CrossRef] [PubMed]
  32. Lagus, T.P.; Edd, J.F. A review of the theory, methods and recent applications of high-throughput single-cell droplet microfluidics. J. Phys. D Appl. Phys. 2013, 46. [Google Scholar] [CrossRef]
  33. Collins, D.J.; Neild, A.; de Mello, A.; Liu, A.Q.; Ai, Y. The poisson distribution and beyond: Methods for microfluidic droplet production and single cell encapsulation. Lab Chip 2015, 15, 3439–3459. [Google Scholar] [CrossRef] [PubMed]
  34. He, M.; Edgar, J.S.; Jeffries, G.D.; Lorenz, R.M.; Shelby, J.P.; Chiu, D.T. Selective encapsulation of single cells and subcellular organelles into picoliter- and femtoliter-volume droplets. Anal. Chem. 2005, 77, 1539–1544. [Google Scholar] [CrossRef] [PubMed]
  35. Tan, Y.C.; Hettiarachchi, K.; Siu, M.; Pan, Y.R.; Lee, A.P. Controlled microfluidic encapsulation of cells, proteins, and microbeads in lipid vesicles. J. Am. Chem. Soc. 2006, 128, 5656–5658. [Google Scholar] [CrossRef] [PubMed]
  36. Luo, D.; Pullela, S.R.; Marquez, M.; Cheng, Z. Cell encapsules with tunable transport and mechanical properties. Biomicrofluidics 2007, 1. [Google Scholar] [CrossRef] [PubMed]
  37. Chabert, M.; Viovy, J.L. Microfluidic high-throughput encapsulation and hydrodynamic self-sorting of single cells. Proc. Natl. Acad. Sci. USA 2008, 105, 3191–3196. [Google Scholar] [CrossRef] [PubMed]
  38. Um, E.; Lee, S.G.; Park, J.K. Random breakup of microdroplets for single-cell encapsulation. Appl. Phys. Lett. 2010, 97. [Google Scholar] [CrossRef]
  39. Joensson, H.N.; Uhlen, M.; Svahn, H.A. Droplet size based separation by deterministic lateral displacement-separating droplets by cell-induced shrinking. Lab Chip 2011, 11, 1305–1310. [Google Scholar] [CrossRef] [PubMed]
  40. Jing, T.; Ramji, R.; Warkiani, M.E.; Han, J.; Lim, C.T.; Chen, C.-H. Jetting microfluidics with size-sorting capability for single-cell protease detection. Biosens. Bioelectron. 2015, 66, 19–23. [Google Scholar] [CrossRef] [PubMed]
  41. Edd, J.F.; di Carlo, D.; Humphry, K.J.; Koster, S.; Irimia, D.; Weitz, D.A.; Toner, M. Controlled encapsulation of single-cells into monodisperse picolitre drops. Lab Chip 2008, 8, 1262–1264. [Google Scholar] [CrossRef] [PubMed]
  42. Kemna, E.W.M.; Schoeman, R.M.; Wolbers, F.; Vermes, I.; Weitz, D.A.; van den Berg, A. High-yield cell ordering and deterministic cell-in-droplet encapsulation using dean flow in a curved microchannel. Lab Chip 2012, 12, 2881–2887. [Google Scholar] [CrossRef] [PubMed]
  43. Ramji, R.; Wang, M.; Bhagat, A.A.S.; Tan Shao Weng, D.; Thakor, N.V.; Teck Lim, C.; Chen, C.-H. Single cell kinase signaling assay using pinched flow coupled droplet microfluidics. Biomicrofluidics 2014, 8. [Google Scholar] [CrossRef] [PubMed]
  44. Lagus, T.P.; Edd, J.F. High-throughput co-encapsulation of self-ordered cell trains: Cell pair interactions in microdroplets. RSC Adv. 2013, 3, 20512–20522. [Google Scholar] [CrossRef]
  45. Lagus, T.P.; Edd, J.F. High throughput single-cell and multiple-cell micro-encapsulation. J. Vis. Exp. 2012, 64, e4096. [Google Scholar] [CrossRef] [PubMed]
  46. Huebner, A.; Srisa-Art, M.; Holt, D.; Abell, C.; Hollfelder, F.; de Mello, A.J.; Edel, J.B. Quantitative detection of protein expression in single cells using droplet microfluidics. Chem. Commun. 2007. [Google Scholar] [CrossRef] [PubMed]
  47. Huebner, A.; Olguin, L.F.; Bratton, D.; Whyte, G.; Huck, W.T.; de Mello, A.J.; Edel, J.B.; Abell, C.; Hollfelder, F. Development of quantitative cell-based enzyme assays in microdroplets. Anal. Chem. 2008, 80, 3890–3896. [Google Scholar] [CrossRef] [PubMed]
  48. Huebner, A.; Bratton, D.; Whyte, G.; Yang, M.; de Mello, A.J.; Abell, C.; Hollfelder, F. Static microdroplet arrays: A microfluidic device for droplet trapping, incubation and release for enzymatic and cell-based assays. Lab Chip 2009, 9, 692–698. [Google Scholar] [CrossRef] [PubMed]
  49. Schmitz, C.H.; Rowat, A.C.; Koster, S.; Weitz, D.A. Dropspots: A picoliter array in a microfluidic device. Lab Chip 2009, 9, 44–49. [Google Scholar] [CrossRef] [PubMed]
  50. Shim, J.-U.; Olguin, L.F.; Whyte, G.; Scott, D.; Babtie, A.; Abell, C.; Huck, W.T.S.; Hollfelder, F. Simultaneous determination of gene expression and enzymatic activity in individual bacterial cells in microdroplet compartments. J. Am. Chem. Soc. 2009, 131, 15251–15256. [Google Scholar] [CrossRef] [PubMed]
  51. Srisa-Art, M.; Bonzani, I.C.; Williams, A.; Stevens, M.M.; de Mello, A.J.; Edel, J.B. Identification of rare progenitor cells from human periosteal tissue using droplet microfluidics. Analyst 2009, 134, 2239–2245. [Google Scholar] [CrossRef] [PubMed]
  52. Wu, N.; Zhu, Y.; Brown, S.; Oakeshott, J.; Peat, T.S.; Surjadi, R.; Easton, C.; Leech, P.W.; Sexton, B.A. A pmma microfluidic droplet platform for in vitroprotein expression using crude e. Coli s30 extract. Lab Chip 2009, 9, 3391–3398. [Google Scholar] [CrossRef] [PubMed]
  53. Baret, J.C.; Beck, Y.; Billas-Massobrio, I.; Moras, D.; Griffiths, A.D. Quantitative cell-based reporter gene assays using droplet-based microfluidics. Chem. Biol. 2010, 17, 528–536. [Google Scholar] [CrossRef] [PubMed]
  54. Rane, T.D.; Puleo, C.M.; Liu, K.J.; Zhang, Y.; Lee, A.P.; Wang, T.H. Counting single molecules in sub-nanolitre droplets. Lab Chip 2010, 10, 161–164. [Google Scholar] [CrossRef] [PubMed]
  55. Casadevall i Solvas, X.; Niu, X.; Leeper, K.; Cho, S.; Chang, S.I.; Edel, J.B.; de Mello, A.J. Fluorescence detection methods for microfluidic droplet platforms. J. Vis. Exp. 2011. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, C.H.; Sarkar, A.; Song, Y.A.; Miller, M.A.; Kim, S.J.; Griffith, L.G.; Lauffenburger, D.A.; Han, J. Enhancing protease activity assay in droplet-based microfluidics using a biomolecule concentrator. J. Am. Chem. Soc. 2011, 133, 10368–10371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Gu, S.-Q.; Zhang, Y.-X.; Zhu, Y.; Du, W.-B.; Yao, B.; Fang, Q. Multifunctional picoliter droplet manipulation platform and its application in single cell analysis. Anal. Chem. 2011, 83, 7570–7576. [Google Scholar] [CrossRef] [PubMed]
  58. Konry, T.; Dominguez-Villar, M.; Baecher-Allan, C.; Hafler, D.A.; Yarmush, M.L. Droplet-based microfluidic platforms for single T cell secretion analysis of IL-10 cytokine. Biosens. Bioelectron. 2011, 26, 2707–2710. [Google Scholar] [CrossRef] [PubMed]
  59. Marcoux, P.R.; Dupoy, M.; Mathey, R.; Novelli-Rousseau, A.; Heran, V.; Morales, S.; Rivera, F.; Joly, P.L.; Moy, J.-P.; Mallard, F. Micro-confinement of bacteria into w/o emulsion droplets for rapid detection and enumeration. Colloids Surf. A: Physicochem. Eng. Asp. 2011, 377, 54–62. [Google Scholar] [CrossRef] [Green Version]
  60. Martino, C.; Zagnoni, M.; Sandison, M.E.; Chanasakulniyom, M.; Pitt, A.R.; Cooper, J.M. Intracellular protein determination using droplet-based immunoassays. Anal. Chem. 2011, 83, 5361–5368. [Google Scholar] [CrossRef] [PubMed]
  61. Dewan, A.; Kim, J.; McLean, R.H.; Vanapalli, S.A.; Karim, M.N. Growth kinetics of microalgae in microfluidic static droplet arrays. Biotechnol. Bioeng. 2012, 109, 2987–2996. [Google Scholar] [CrossRef] [PubMed]
  62. Joensson, H.N.; Zhang, C.; Uhlen, M.; Andersson-Svahn, H. A homogeneous assay for protein analysis in droplets by fluorescence polarization. Electrophoresis 2012, 33, 436–439. [Google Scholar] [CrossRef] [PubMed]
  63. Bai, Y.; Patil, S.N.; Bowden, S.D.; Poulter, S.; Pan, J.; Salmond, G.P.; Welch, M.; Huck, W.T.; Abell, C. Intra-species bacterial quorum sensing studied at single cell level in a double droplet trapping system. Int. J. Mol. Sci. 2013, 14, 10570–10581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chokkalingam, V.; Tel, J.; Wimmers, F.; Liu, X.; Semenov, S.; Thiele, J.; Figdor, C.G.; Huck, W.T. Probing cellular heterogeneity in cytokine-secreting immune cells using droplet-based microfluidics. Lab Chip 2013, 13, 4740–4744. [Google Scholar] [CrossRef] [PubMed]
  65. Konry, T.; Golberg, A.; Yarmush, M. Live single cell functional phenotyping in droplet nano-liter reactors. Sci. Rep. 2013, 3. [Google Scholar] [CrossRef] [PubMed]
  66. Abbaspourrad, A.; Zhang, H.; Tao, Y.; Cui, N.; Asahara, H.; Zhou, Y.; Yue, D.; Koehler, S.A.; Ung, L.W.; Heyman, J.; et al. Label-free single-cell protein quantification using a drop-based mix-and-read system. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [PubMed]
  67. Chiu, T.K.; Lei, K.F.; Hsieh, C.H.; Hsiao, H.B.; Wang, H.M.; Wu, M.H. Development of a microfluidic-based optical sensing device for label-free detection of circulating tumor cells (CTCs) through their lactic acid metabolism. Sensors 2015, 15, 6789–6806. [Google Scholar] [CrossRef] [PubMed]
  68. Larsen, A.C.; Dunn, M.R.; Hatch, A.; Sau, S.P.; Youngbull, C.; Chaput, J.C. A general strategy for expanding polymerase function by droplet microfluidics. Nat. Commun. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
  69. Nakamura, K.; Iizuka, R.; Nishi, S.; Yoshida, T.; Hatada, Y.; Takaki, Y.; Iguchi, A.; Yoon, D.H.; Sekiguchi, T.; Shoji, S.; et al. Culture-independent method for identification of microbial enzyme-encoding genes by activity-based single-cell sequencing using a water-in-oil microdroplet platform. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef] [PubMed]
  70. Ng, E.X.; Miller, M.A.; Jing, T.; Chen, C.-H. Single cell multiplexed assay for proteolytic activity using droplet microfluidics. Biosens. Bioelectron. 2016, 81, 408–414. [Google Scholar] [CrossRef] [PubMed]
  71. Kumaresan, P.; Yang, C.J.; Cronier, S.A.; Blazej, R.G.; Mathies, R.A. High-throughput single copy DNA amplification and cell analysis in engineered nanoliter droplets. Anal. Chem. 2008, 80, 3522–3529. [Google Scholar] [CrossRef] [PubMed]
  72. Mazutis, L.; Araghi, A.F.; Miller, O.J.; Baret, J.C.; Frenz, L.; Janoshazi, A.; Taly, V.; Miller, B.J.; Hutchison, J.B.; Link, D.; et al. Droplet-based microfluidic systems for high-throughput single DNA molecule isothermal amplification and analysis. Anal. Chem. 2009, 81, 4813–4821. [Google Scholar] [CrossRef] [PubMed]
  73. Schaerli, Y.; Wootton, R.C.; Robinson, T.; Stein, V.; Dunsby, C.; Neil, M.A.A.; French, P.M.W.; de Mello, A.J.; Abell, C.; Hollfelder, F. Continuous-flow polymerase chain reaction of single-copy DNA in microfluidic microdroplets. Anal. Chem. 2009, 81, 302–306. [Google Scholar] [CrossRef] [PubMed]
  74. Leng, X.; Zhang, W.; Wang, C.; Cui, L.; Yang, C.J. Agarose droplet microfluidics for highly parallel and efficient single molecule emulsion pcr. Lab Chip 2010, 10, 2841–2843. [Google Scholar] [CrossRef] [PubMed]
  75. Zeng, Y.; Novak, R.; Shuga, J.; Smith, M.T.; Mathies, R.A. High-performance single cell genetic analysis using microfluidic emulsion generator arrays. Anal. Chem. 2010, 82, 3183–3190. [Google Scholar] [CrossRef] [PubMed]
  76. Hatch, A.C.; Fisher, J.S.; Tovar, A.R.; Hsieh, A.T.; Lin, R.; Pentoney, S.L.; Yang, D.L.; Lee, A.P. 1-million droplet array with wide-field fluorescence imaging for digital pcr. Lab Chip 2011, 11, 3838–3845. [Google Scholar] [CrossRef] [PubMed]
  77. Hindson, B.J.; Ness, K.D.; Masquelier, D.A.; Belgrader, P.; Heredia, N.J.; Makarewicz, A.J.; Bright, I.J.; Lucero, M.Y.; Hiddessen, A.L.; Legler, T.C.; et al. High-throughput droplet digital pcr system for absolute quantitation of DNA copy number. Anal. Chem. 2011, 83, 8604–8610. [Google Scholar] [CrossRef] [PubMed]
  78. Mary, P.; Dauphinot, L.; Bois, N.; Potier, M.-C.; Studer, V.; Tabeling, P. Analysis of gene expression at the single-cell level using microdroplet-based microfluidic technology. Biomicrofluidics 2011, 5. [Google Scholar] [CrossRef] [PubMed]
  79. Novak, R.; Zeng, Y.; Shuga, J.; Venugopalan, G.; Fletcher, D.A.; Smith, M.T.; Mathies, R.A. Single cell multiplex gene detection and sequencing using microfluidically-generated agarose emulsions. Angew. Chem. Int. Ed. 2011, 50, 390–395. [Google Scholar] [CrossRef] [PubMed]
  80. Rane, T.D.; Zec, H.C.; Puleo, C.; Lee, A.P.; Wang, T.-H. Droplet microfluidics for amplification-free genetic detection of single cells. Lab Chip 2012, 12, 3341–3347. [Google Scholar] [CrossRef] [PubMed]
  81. Zhang, H.; Jenkins, G.; Zou, Y.; Zhu, Z.; Yang, C.J. Massively parallel single-molecule and single-cell emulsion reverse transcription polymerase chain reaction using agarose droplet microfluidics. Anal. Chem. 2012, 84, 3599–3606. [Google Scholar] [CrossRef] [PubMed]
  82. Zhu, Z.; Zhang, W.; Leng, X.; Zhang, M.; Guan, Z.; Lu, J.; Yang, C.J. Highly sensitive and quantitative detection of rare pathogens through agarose droplet microfluidic emulsion pcr at the single-cell level. Lab Chip 2012, 12, 3907–3913. [Google Scholar] [CrossRef] [PubMed]
  83. Fu, Y.; Li, C.; Lu, S.; Zhou, W.; Tang, F.; Xie, X.S.; Huang, Y. Uniform and accurate single-cell sequencing based on emulsion whole-genome amplification. Proc. Natl. Acad. Sci. USA 2015, 112, 11923–11928. [Google Scholar] [CrossRef] [PubMed]
  84. Klein, A.M.; Mazutis, L.; Akartuna, I.; Tallapragada, N.; Veres, A.; Li, V.; Peshkin, L.; Weitz, D.A.; Kirschner, M.W. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 2015, 161, 1187–1201. [Google Scholar] [CrossRef] [PubMed]
  85. Nishikawa, Y.; Hosokawa, M.; Maruyama, T.; Yamagishi, K.; Mori, T.; Takeyama, H. Monodisperse picoliter droplets for low-bias and contamination-free reactions in single-cell whole genome amplification. PLoS ONE 2015, 10, e0138733. [Google Scholar] [CrossRef] [PubMed]
  86. Baret, J.C.; Miller, O.J.; Taly, V.; Ryckelynck, M.; El-Harrak, A.; Frenz, L.; Rick, C.; Samuels, M.L.; Hutchison, J.B.; Agresti, J.J.; et al. Fluorescence-activated droplet sorting (fads): Efficient microfluidic cell sorting based on enzymatic activity. Lab Chip 2009, 9, 1850–1858. [Google Scholar] [CrossRef] [PubMed]
  87. Brouzes, E.; Medkova, M.; Savenelli, N.; Marran, D.; Twardowski, M.; Hutchison, J.B.; Rothberg, J.M.; Link, D.R.; Perrimon, N.; Samuels, M.L. Droplet microfluidic technology for single-cell high-throughput screening. Proc. Natl. Acad. Sci. USA 2009, 106, 14195–14200. [Google Scholar] [CrossRef] [PubMed]
  88. Agresti, J.J.; Antipov, E.; Abate, A.R.; Ahn, K.; Rowat, A.C.; Baret, J.C.; Marquez, M.; Klibanov, A.M.; Griffiths, A.D.; Weitz, D.A. Ultrahigh-throughput screening in drop-based microfluidics for directed evolution. Proc. Natl. Acad. Sci. USA 2010, 107, 4004–4009. [Google Scholar] [CrossRef] [PubMed]
  89. Eun, Y.-J.; Utada, A.; Copeland, M.F.; Takeuchi, S.; Weibel, D.B. Encapsulating bacteria in agarose microparticles using microfluidics for high-throughput cell analysis and isolation. ACS Chem. Biol. 2011, 6, 260–266. [Google Scholar] [CrossRef] [PubMed]
  90. Kintses, B.; Hein, C.; Mohamed, M.F.; Fischlechner, M.; Courtois, F.; Lainé, C.; Hollfelder, F. Picoliter cell lysate assays in microfluidic droplet compartments for directed enzyme evolution. Chem. Biol. 2012, 19, 1001–1009. [Google Scholar] [CrossRef] [PubMed]
  91. Mazutis, L.; Gilbert, J.; Ung, W.L.; Weitz, D.A.; Griffiths, A.D.; Heyman, J.A. Single-cell analysis and sorting using droplet-based microfluidics. Nat. Protoc. 2013, 8, 870–891. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, B.L.; Ghaderi, A.; Zhou, H.; Agresti, J.; Weitz, D.A.; Fink, G.R.; Stephanopoulos, G. Microfluidic high-throughput culturing of single cells for selection based on extracellular metabolite production or consumption. Nat. Biotechnol. 2014, 32, 473–478. [Google Scholar] [CrossRef] [PubMed]
  93. Hammar, P.; Angermayr, S.A.; Sjostrom, S.L.; van der Meer, J.; Hellingwerf, K.J.; Hudson, E.P.; Joensson, H.N. Single-cell screening of photosynthetic growth and lactate production by cyanobacteria. Biotechnol. Biofuels 2015, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. (a) Schematic of a microfluidic T channel and (b) sequences of images showing the encapsulation of a single B lymphocyte into an aqueous droplet in silicone oil. Optical trapping was used to transport and position the cell close to the water/oil interface. Upon application of a pressure pulse to the microchannels, the cell was carried away by the flow as the droplet was sheared off. Reproduction with permission from [34].
Figure 1. (a) Schematic of a microfluidic T channel and (b) sequences of images showing the encapsulation of a single B lymphocyte into an aqueous droplet in silicone oil. Optical trapping was used to transport and position the cell close to the water/oil interface. Upon application of a pressure pulse to the microchannels, the cell was carried away by the flow as the droplet was sheared off. Reproduction with permission from [34].
Molecules 21 00881 g001
Figure 2. Size-based droplet sorting after cellular encapsulation. A purely hydrodynamic approach for single-cell encapsulation, followed by spontaneous self-sorting of these droplets based on lateral drift of deformable objects in a shear flow, and sterically-driven dispersion in a compressional flow. Reproduction with permission from [37].
Figure 2. Size-based droplet sorting after cellular encapsulation. A purely hydrodynamic approach for single-cell encapsulation, followed by spontaneous self-sorting of these droplets based on lateral drift of deformable objects in a shear flow, and sterically-driven dispersion in a compressional flow. Reproduction with permission from [37].
Molecules 21 00881 g002
Figure 3. Inertial flow-based cell spacing and single-cell encapsulation using (a) a high aspect-ratio straight microchannel (reproduction with permission from [41]); (b) a curved microchannel (reproduction with permission from [42]); and (c) a short pinched flow channel (reproduction with permission from [43]).
Figure 3. Inertial flow-based cell spacing and single-cell encapsulation using (a) a high aspect-ratio straight microchannel (reproduction with permission from [41]); (b) a curved microchannel (reproduction with permission from [42]); and (c) a short pinched flow channel (reproduction with permission from [43]).
Molecules 21 00881 g003
Figure 4. Microfluidic droplets enabling single-cell proteomic analysis. (a) Individual E coli and substrate 3-O-methylfluorescein-phosphates were encapsulated within single droplets where the substrates were enzymatically hydrolyzed by the target enzyme alkaline phosphatase expressed by E coli, leading to fluorescent detections. Reproduction with permission from [47]; (b) Both microspheres conjugated with capture antibodies and detection fluorescence labeled antibodies were encapsulated with single cells and the secreted IL-10 of CD4 + CD25 + regulatory T cells was captured on the microsphere surface and detected via detection antibodies, generating localized fluorescent signals on microsphere surfaces. Reproduction with permission from [58].
Figure 4. Microfluidic droplets enabling single-cell proteomic analysis. (a) Individual E coli and substrate 3-O-methylfluorescein-phosphates were encapsulated within single droplets where the substrates were enzymatically hydrolyzed by the target enzyme alkaline phosphatase expressed by E coli, leading to fluorescent detections. Reproduction with permission from [47]; (b) Both microspheres conjugated with capture antibodies and detection fluorescence labeled antibodies were encapsulated with single cells and the secreted IL-10 of CD4 + CD25 + regulatory T cells was captured on the microsphere surface and detected via detection antibodies, generating localized fluorescent signals on microsphere surfaces. Reproduction with permission from [58].
Molecules 21 00881 g004
Figure 5. Microfluidic droplets enabling single-cell genomic analysis. (a) Individual cells together with primer-functionalized microbeads were encapsulated in uniform PCR mix droplets. After bulk PCR amplification, the droplets were lysed and the beads were recovered and rapidly analyzed via flow cytometry. Reproduction with permission from [71]; and (b) an agarose droplet-based microfluidic method for emulsification RT-PCR, where reverse primers were covalently conjugated to agarose, which functioned as the trapping matrix to replace conventional primer functionalized microbeads, resulting in high PCR efficiency (~95%). Reproduction with permission from [81].
Figure 5. Microfluidic droplets enabling single-cell genomic analysis. (a) Individual cells together with primer-functionalized microbeads were encapsulated in uniform PCR mix droplets. After bulk PCR amplification, the droplets were lysed and the beads were recovered and rapidly analyzed via flow cytometry. Reproduction with permission from [71]; and (b) an agarose droplet-based microfluidic method for emulsification RT-PCR, where reverse primers were covalently conjugated to agarose, which functioned as the trapping matrix to replace conventional primer functionalized microbeads, resulting in high PCR efficiency (~95%). Reproduction with permission from [81].
Molecules 21 00881 g005
Figure 6. Integrated microfluidic system for single-cell screening, including key steps of cell encapsulation, incubation, fluorescence detection of metabolic molecules, and droplet sorting relying on the fluorescent intensities. Reproduction with permission from [91].
Figure 6. Integrated microfluidic system for single-cell screening, including key steps of cell encapsulation, incubation, fluorescence detection of metabolic molecules, and droplet sorting relying on the fluorescent intensities. Reproduction with permission from [91].
Molecules 21 00881 g006
Table 1. Key developments of microfluidic droplets enabling single-cell proteomic analysis.
Table 1. Key developments of microfluidic droplets enabling single-cell proteomic analysis.
Interested ProteinsDetection MechanismsReferences
β-galactosidase of mast cellsFollowing cellular lysis, intracellular β-galactosidase catalyzed the substrate (fluorescein di-β-d-galactopyranoside) for fluorescence detection[34]
Yellow fluorescent protein mutant of E. coliThe expression of yellow fluorescent proteins was correlated with the growth status of encapsulated E. coli[46]
Alkaline phosphatase of E. coliExpresssed alkaline phosphatase in the cellular periplasm catalyzed the substrate (3-O-methylfluorescein-phosphates) for fluorescence detection[47]
Both red fluorescent protein and alkaline phosphatase of E. coliGene expression and enzymatic activity of E. coli were simultaneously and continuously monitored[50]
IL-10 of CD4+CD25+ regulatory T cellsThe secreted substance captured on the microsphere surface coated with capturing antibodies and detected via the further binding of fluorescence labled detection antibodies on microsphere surfaces[58]
Intracellular HRas-mCitrine of HEK-293 cells and actin-EGFP of MCF-7 cellsFollowing cell encapsulation and lysis, proteins under interest were captured on the microsphere surface coated with capturing antibodies and detected via the further binding of fluorescence labled detection antibodies on microsphere surfaces[60]
IL-2, IFN-γ, and TNF-α of activated T-cellsCells were encapsulated in agarose droplets together with functionalized cytokine-capture beads for subsequent binding and detection of secreted cytokines from single cells[64]
Receptor tyrosine kinases of PC-9 cellsBinding surface ligands of 8-hydroxy-5-(N,N-dimethylsulfonamido)-2-methylquinoline) with the receptor tyrosine kinases generates fluorescent signals[43]
Multiple proteases of MDA-MB-231, PC-9, and K-562 cellsProtease-catalyzed multi-color Förster resonance energy transfer based enzymatic substrates, enabling the simultaneous measurement of six proteases [70]
Table 2. Key developments of microfluidic droplets enabling single-cell genomic analysis.
Table 2. Key developments of microfluidic droplets enabling single-cell genomic analysis.
Interested Gene SectionsWorking MechanismsReferences
GAPDH gene of lymphocyte cells and gyr B gene of E. coliA single cell and a primer functionalized microbead were encapsulated in droplets, followed by bulk PCR, droplet lysis, and bead analysis in flow cytometry[71]
KI#128 island on the E. coli K12 and OI#43 island on the E. coli O157 cells96 channels were used to generate up to 3.4 × 106 nanoliter-volume droplets per hour, identifying rare pathogenic E. coli O157 cells (1:105 cells)[75]
Chromosomal translocation t(14;18) of follicular lymphoma cellsAgarose droplets were formed to encapsulate cells and primer-functionalized microbeads, maintaining genome fidelity during cell lysis and DNA purification, leading to efficient PCR and subsequent gene sequencing[79]
KI#128 island on the E. coli K12 and OI#43 island on the E. coli O157 cellsAn agarose droplet was formed to encapsulate single cells and PCR mix with reverse primers covalently conjugated to agarose[82]
Gene expression of EpCAM fromAn agarose droplet was formed to encapsulate single cells and RT-PCR mix with primers covalently conjugated to agarose[82]

Share and Cite

MDPI and ACS Style

Wen, N.; Zhao, Z.; Fan, B.; Chen, D.; Men, D.; Wang, J.; Chen, J. Development of Droplet Microfluidics Enabling High-Throughput Single-Cell Analysis. Molecules 2016, 21, 881. https://doi.org/10.3390/molecules21070881

AMA Style

Wen N, Zhao Z, Fan B, Chen D, Men D, Wang J, Chen J. Development of Droplet Microfluidics Enabling High-Throughput Single-Cell Analysis. Molecules. 2016; 21(7):881. https://doi.org/10.3390/molecules21070881

Chicago/Turabian Style

Wen, Na, Zhan Zhao, Beiyuan Fan, Deyong Chen, Dong Men, Junbo Wang, and Jian Chen. 2016. "Development of Droplet Microfluidics Enabling High-Throughput Single-Cell Analysis" Molecules 21, no. 7: 881. https://doi.org/10.3390/molecules21070881

Article Metrics

Back to TopTop