You are on page 1of 22

Home Search Collections Journals About Contact us My IOPscience

A review of the theory, methods and recent applications of high-throughput single-cell droplet

microfluidics

This article has been downloaded from IOPscience. Please scroll down to see the full text article.

2013 J. Phys. D: Appl. Phys. 46 114005

(http://iopscience.iop.org/0022-3727/46/11/114005)

View the table of contents for this issue, or go to the journal homepage for more

Download details:
IP Address: 132.174.255.49
The article was downloaded on 17/05/2013 at 06:59

Please note that terms and conditions apply.


IOP PUBLISHING JOURNAL OF PHYSICS D: APPLIED PHYSICS
J. Phys. D: Appl. Phys. 46 (2013) 114005 (21pp) doi:10.1088/0022-3727/46/11/114005

A review of the theory, methods and


recent applications of high-throughput
single-cell droplet microfluidics
Todd P Lagus and Jon F Edd
Vanderbilt University, Department of Mechanical Engineering, 2400 Highland Avenue, Nashville,
TN, 37212, USA
E-mail: jon.f.edd@vanderbilt.edu

Received 31 July 2012, in final form 18 September 2012


Published 22 February 2013
Online at stacks.iop.org/JPhysD/46/114005

Abstract
Most cell biology experiments are performed in bulk cell suspensions where cell secretions
become diluted and mixed in a contiguous sample. Confinement of single cells to small,
picoliter-sized droplets within a continuous phase of oil provides chemical isolation of each
cell, creating individual microreactors where rare cell qualities are highlighted and otherwise
undetectable signals can be concentrated to measurable levels. Recent work in microfluidics
has yielded methods for the encapsulation of cells in aqueous droplets and hydrogels at
kilohertz rates, creating the potential for millions of parallel single-cell experiments. However,
commercial applications of high-throughput microdroplet generation and downstream sensing
and actuation methods are still emerging for cells. Using fluorescence-activated cell sorting
(FACS) as a benchmark for commercially available high-throughput screening, this focused
review discusses the fluid physics of droplet formation, methods for cell encapsulation in
liquids and hydrogels, sensors and actuators and notable biological applications of
high-throughput single-cell droplet microfluidics.
(Some figures may appear in colour only in the online journal)

1. Introduction microfluidic materials list also includes various polymers,


hydrogels, semiconductors and metals as needed to achieve
The intense activity and progress in microfluidics over the last specific properties. Mass transport, thermal transport,
several years is a result of combined efforts of researchers chemical reactions and biological interactions must all be
with expertise in diverse fields such as fluid mechanics, considered as microfluidics widens to a large number of fields
thermodynamics, applied physics, chemistry, materials and applications.
science, manufacturing, biochemistry, biotechnology and As a subset of microfluidics, droplet microfluidics
medicine. Microfluidic systems manipulate small volumes involves devices that produce microscale diameter (typically
of fluid inside microscale channels. Microfluidic channels on the order of tens to hundreds of µm) droplets of one fluid
may be integrated into macroscale systems or may be within a second immiscible carrier fluid. Droplet-producing
standalone miniaturized devices, often referred to as lab-on- micronozzles are capable of formation rates from slow dripping
a-chip systems [1–3]. Self-contained microfluidic devices to over 10 kHz. Monodisperse water-in-oil (w/o) droplet
necessarily include channel networks, fluid reservoirs and emulsions provide nearly identically sized (typically 1–5%
integrated sensors and actuators which are interfaced to deviation from the mean) femto-, pico- or nanoliter aqueous
users or automated programs by multiple means. In simple compartments, each a microreactor with a specified chemical
forms, microfluidics involves single-phase flow of liquids, composition and encapsulated cargo such as particles, cells,
but complex two-phase flows of multiple liquids, gases and or biomolecules (proteins, DNA and metabolites). Within
suspended solids are encountered. Although the primary each droplet, a separate experiment takes place that can
materials are silicone elastomers [4], glass and silicon, the reveal sample heterogeneity, otherwise obscured in bulk, to

0022-3727/13/114005+21$33.00 1 © 2013 IOP Publishing Ltd Printed in the UK & the USA
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

characterize the effects of rare cells [5, 6]. Alternatively, technologies for traditional applications such as FACS and
identical samples can be exposed to distinct stimuli by varying for smaller and potentially more mobile systems. A number
droplet conditions for applications such as drug discovery of excellent reviews highlight recent progress in droplet-
[7, 8]. Moreover, volumetric confinement avoids dilution of based microfluidics for single-cell analysis [24–27]. Rather
products, improving detectable signals and reducing reagent than replicate the scope of those reviews, we discuss droplet
volumes, and the large droplet surface area to volume ratios microfluidics with a focus on high-throughput applications.
can be exploited for enhanced heat and mass transport. This includes a summary of two-phase fluid physics, recent
To avoid confusion with a closely related field, it is advances in cell encapsulation, sensing and actuation methods
important to distinguish between droplet microfluidics and which are at least potentially adaptable for high-throughput
digital microfluidics. Specifically, we limit this review applications. Finally, we discuss in detail a select number of
of ‘droplet microfluidics’ to pressure-driven devices where recent droplet microfluidics publications which provide unique
droplets are enclosed within channels and immersed in a contributions in techniques and methods for high-throughput
continuous phase oil carrier fluid. On the other hand, single-cell analysis.
‘digital microfluidics’ utilizes microfabricated electrode arrays
programmed to move, mix, react, analyse and split droplets
2. Droplet physics
through electrowetting on a dielectric-coated substrate [9, 10].
Despite many digital microfluidic methods in lab-on-a-chip 2.1. Droplet formation fundamentals
systems, expansion of digital microfluidics into massively
parallel, high-throughput systems is limited by the size, A number of focused reviews [28–32] provide detailed insight
scalability and dynamic response of the electrodes, so it is into droplet generation and flow dynamics. Here, we
not widely discussed in this review. briefly summarize them to highlight how droplet dynamics
Pressure-driven droplet microfluidics has been success- both facilitate and limit high-throughput droplet microfluidic
fully implemented for molecular biology applications such applications. While droplet generation may be driven
as amplifying single-copy DNA molecules using the poly- using active methods such as piezoelectric actuators [33]
merase chain reaction (PCR) [11–14]. Microscale droplets and electric fields [34], aqueous monodisperse droplets
can also provide encapsulation of single cells [8, 15–18] for are most commonly formed in continuous pressure-driven
high throughput screening (HTS) applications involving drug nozzles with flow focusing [35] (figure 1(a)), co-flowing [36]
discovery, toxicity testing, various ‘omics’ studies and rare (figure 1(b)), or T-junction [37] (figure 2(a)) designs. These
cell analysis. This review highlights recent advances in high- passive droplet generation methods benefit from the ability
throughput, single-cell droplet microfluidic methods and appli- to increase throughput simply by adding parallel channels
cations which have a potential to be economically competitive since they do not require individually addressable actuation.
in the commercial marketplace. By cascading droplet generation interfaces in series, more
As a testament to the recent push to high-throughput complex emulsions within emulsions are also possible [38, 39]
microfluidic research, a Web of Science citation report (figure 1(c)).
for the topic phrase ‘high throughput’ microfluidic(s)’ Hydrophobic channel surfaces are required to prevent
indicated publication of around 250 papers in both unstable, polydisperse w/o droplet formation due to wetting
2010 and 2011 [19]. Despite this high interest level, effects [40]. For multiple emulsions [38, 39] (figure 1(c)),
the definition of ‘high throughput’ remains nebulous. nozzle surface properties must alternate between hydrophobic
As a working definition for this review, we select and hydrophilic. Polydimethylsiloxane (PDMS) is a
a commercial method to quantify our high-throughput commonly used natively hydrophobic, transparent (to both
benchmark. Specifically, commercial fluorescence-activated UV and visible light) elastomer used in microfluidic devices.
cell sorters (FACS) utilize laser illumination and fluorescence The high elasticity and gas permeability of PDMS, while
sensing with photomultiplication followed by electrostatic beneficial in some cases, are not amenable to all applications.
deflection of cell-containing droplets suspended in air. Additionally, the typical laboratory-scale fabrication method,
This allows them to count and automatically sort cells replica moulding with thermal curing [4], does not lend itself
to microwells at rates on the order of 10 000 cells s−1 to mass production due to long process times [41]. Additional
[20, 21]. Although FACS machines can in fact process materials such as polymethyl methacrylate (PMMA) and
on the order of 70 000–100 000 droplets s−1 [22, 23], single- polyether ether ketone (PEEK) thermoplastics have also been
cell encapsulation requires the use of dilute cell suspensions, studied as alternatives (discussed in depth elsewhere [41–45])
causing many empty droplets and a reduction in the effective using methods such as hot embossing and injection moulding,
cell throughput. State-of-the-art FACS units are also equipped but manufacturing costs have been too high to justify replacing
with on-board ‘index sorting’ software that tracks and indexes existing screening methods with microfluidic devices.
droplet properties by microwell, allowing for correlation For flow-focusing and co-flowing nozzles, water-in-oil
of sorted cell group properties, with downstream transient droplets break off from the aqueous fluid stream when oil
response tracking of cell populations, in any given well [20]. shear stress overcomes interfacial tension that keeps droplets
Given their ability to efficiently encapsulate molecules attached to the aqueous neck. For T-junction nozzles at low
and cells at kHz rates, droplet microfluidic devices are well- oil flow rates, the oil flow ‘pinches’ droplets when the aqueous
poised to have a major impact on high-throughput screening stream blocks the continuous flow channel, thereby creating a

2
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

OIL

WATER

OIL

(a)

Oil
Aqueous
Oil

50 m
(b) (c)
Figure 1. (a) Flow-focusing geometry implemented in a microfluidic device (left) with orifice with of 43.5 µm. The droplet generation
(right) demonstrates the ability to create monodisperse droplets of controlled size [35]. Copyright 2003, American Institute of Physics.
(b) Co-flowing device with tapered aqueous flow inner capillary nested inside an oil flow outer capillary (top), co-flowing droplet generation
(middle) and a widening jet (bottom) [36]. The widening jet reflects the transition from dripping to inertial jetting due to Weber number
greater than one as the aqueous flow rate increases. Copyright 2007 by the American Physical Society. (c) Precisely controlled
monodisperse double emulsions using nested co-flowing capillaries. Each scale bar equals 200 µm [38]. Parts (a), (b) and (c) reprinted with
permission from [35, 36, 38], respectively.

pressure buildup which leads to droplet generation (figure 2(a)) viscous and interfacial tension forces. Plotting the aqueous
[46]. At higher oil flow rates, oil shear acts to detach droplets phase Weber number against the oil-phase Capillary number
in a mechanism similar to that shown in flow-focusing and provides a droplet formation regime map and highlights the
co-flowing nozzles [47]. Droplet size and formation rates critical Weber and Capillary numbers marking the transition
depend on the combinations of aqueous and oil-phase flow from steady droplets to jetting [36]. At small aqueous and
rates, nozzle geometry, respective dynamic viscosities µ and oil flow rates (W e  1 and Ca  1), the aqueous fluid
densities ρ and interfacial tension between phases. forms pulls apart an aqueous neck to minimize surface energy,
The balance of inertial, viscous and interfacial tension yielding monodisperse droplets. At larger aqueous flow rates
forces govern droplet formation and confined droplet flow. The (W e ∼ 1 or greater), inertial forces begin to dominate
relationship between the inertial and interfacial tension forces interfacial tension forces. At a critically high Weber number,
of the aqueous phase is quantified by the Weber number We, the aqueous neck moves downstream as a wide unstable ‘jet’
given by of aqueous fluid from the nozzle (see figure 1(b)). A transition
ρU 2 Dh from dripping to jetting also occurs as oil flow rates increase
We = , (1)
σ (Ca ∼ 1 or greater). At a critically large Capillary number,
where ρ, U and Dh represent the aqueous fluid density, mean viscous drag forces from the outer carrier oil pull on the
velocity, channel hydraulic diameter and σ represents the aqueous flow enough to overcome interfacial tension forces,
interfacial tension between the two immiscible fluids. In causing the inner aqueous fluid to stretch into long, thin streams
determining droplet formation dynamics, the Weber number is [36, 47, 48].
often paired with the Capillary number Ca, which represents In T-junctions, slightly different breakup mechanisms lead
the ratio of viscous forces to interfacial tension forces and is to dripping/jetting transitions [47]. At low Capillary numbers
given by (low oil flow rates), the aqueous fluid interface moves into the
µU continuous fluid stream and creates a blockage in the channel
Ca = , (2)
σ (figure 2(a)). The upstream pressure builds until the pressure
where µ and U represent the oil fluid viscosity and mean force overcomes the interfacial tension forces, and the droplet
velocity, respectively, and σ again represents the interfacial is ‘pinched’ off the main stream. As the Capillary number
tension between the two immiscible fluids [36]. increases (higher oil flow rates), the continuous stream cross-
While obtaining a clear-cut scaling law is challenging [30], flow deforms the droplet such that the aqueous stream does
We and Ca still illustrate the relative importance of inertial, not fully block the continuous flow channel. Similar to the

3
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

given nozzle. Thus, increasing throughput must be attained


by adding parallel channels, and not by simply increasing
flow rates.
It is important to note that dripping to jetting transition
maps [36] indicate that interfacial tension forces should be
significant in comparison with aqueous inertia or oil-induced
shear forces in order to ensure steady dripping. However,
high interfacial tension conflicts with another important design
constraint for droplet microfluidics: emulsion stability. While
droplet formation may occur at a nozzle, the emulsion is
at best meta-stable. For any non-zero interfacial tension,
droplet surface area is minimized to decrease the surface
energy of the emulsion such that two separate droplets are
more stable as a single droplet. Decreasing interfacial
tension using surfactants provides additional stability such
that some emulsions remain stable for several years [50].
However, decreased surface tension also increases the Weber
and Capillary numbers, increasing the likelihood of jetting at
drop generation nozzles. These competing design constraints
necessitate careful consideration of surfactant types and
concentrations for optimal droplet generation and emulsion
stability regimes.
For water-in-oil emulsions, surfactants such as ABIL
EM 90 and Span 80 are typically mixed in organic carrier
phases such as mineral or silicone oils [51]. Fluorocarbon
oils such as FC-40 mixed with fluorosurfactants are popular
carrier fluids for biological droplet applications. Since
fluorocarbon oils are both hydrophobic and lipophobic, they
are considered more biocompatible than lipophilic organic oils,
as fluorocarbon oils are not likely to interact with aqueous
solutes, biomolecules and cell membrane lipids [52]. Non-
ionic fluorosurfactants which provide both emulsion stability
and an inert inner droplet surface are, however, difficult to
obtain commercially but may be synthesized using protocols
Figure 2. (a) Multiple inlet fluid mixing in droplets generated at a presented in the literature [52–54]. Pickering emulsions
T-junction. Note the swirling and folding patterns indicative of can also be created using nanoparticle-stabilized interfaces
internal advection [72]. (b) Mixing schematic in smooth and sharp [55–57]. More complete reviews of surfactants [51] and
turning devices with arrows demonstrating mixing flow
patterns [72]. (c) Example method for generating droplets with nanoparticle pickering emulsions [58] are available elsewhere.
unique concentrations using multiple aqueous inlets upstream of the
flow-focusing nozzle. Supply reservoirs are mechanically actuated 2.2. Modelling droplets: analytical, numerical and empirical
to provide unique reagents to expand the experimental parameter
approaches
space [75]. Copyright 2012, American Chemical Society. Parts (a)
and (b) are reprinted with permission from [72], and part (c) is At the core of droplet microfluidics are the fundamental
reprinted with permission from [75].
transport equations for mass, momentum and energy
conservation. When increasing flow rates to increase device
dripping regime for flow-focusing and co-flowing nozzles, throughput, one necessarily must consider and quantify
monodisperse droplets form as the oil shear pulls droplets the effects of pressure and shear forces on droplets and
from the main aqueous stream. Recent experimental pressure encapsulated samples, especially for biological applications.
measurements in T-junctions also have found that pressure Because exact solutions to the Navier–Stokes equations
oscillations still persist even in the dripping regime, suggesting exist only for relatively simple flow geometries, most droplet-
that the dripping mechanism may not be entirely shear-driven based analyses utilize numerical solution methods to recover
[49]. Intuitively, as Ca further increases, the oil shear stretches pressures and shear forces for separate solution domains (water
the aqueous fluid into long, thin streams with unstable droplet and oil). In addition to the already complicated geometries and
detachment. meshes for the continuous and dispersed phases, it is necessary
To ensure steady droplet generation in flow focusing, co- to resolve the phase interface. Here, some properties, such as
flowing or T-junction schemes, aqueous and oil flow rates must pressure, viscosity and density are not continuous between
therefore be sufficiently small to avoid inertial and shear-based phases. Reviews of numerical solutions exist which highlight
jetting. This imposes a strict throughput limitation on the methods such as diffuse-interface [59], level set [60], front-
attainable flow rates and drop generation frequencies for a tracking [61], immersed boundary [62] and Lattice–Boltzmann

4
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

[63–65] solutions for two-phase droplet and solid surface other). This can improve mixing rates to millisecond time
flows. Upstream of the nozzle, aqueous and oil microchannel scales [70–72] and facilitate an increase in surface areas
flows are relatively straightforward to resolve [66]. However, between chemically reacting layers [73]. Initial droplet
transient droplet generation, flow, wetting, merging and reagent concentrations can be controlled simply by pre-
splitting increase numerical complexity and increase the range mixing samples, mixing samples at the drop generation inlet,
of length scales that must be resolved for reliable solutions. or using gradient generators connected to multiple droplet
As computing speed improves, numerical models will provide nozzles to create wider experimental parameter spaces [74, 75]
valuable insight into dimensionless scaling parameters beyond (figure 2(c)).
those currently available in the literature. At this time, For aqueous droplets in immiscible oil carrier fluids,
however, there is a need to couple experimental and numerical delivery of reagents and other solutes to and from cells is
results to cover wider parameter spaces. prevented by the low solubility of the aqueous phase in the
Current numerical model implementations are likely not oil phase. However, some lipophilic oils can selectively
feasible when simulating full systems in which the domain dissolve trace amounts of water to concentrate aqueous
sizes are large and complex, or even simple systems where solutes [76], or transport particles across the interface [77],
time and length scales span many orders of magnitude. On but these effects are usually undesirable. A recent study
the other hand, homogeneous two-phase flow correlations noted that while hydrocarbon oils are not compatible with
provide empirical insights into critical design considerations, DNA transfection of encapsulated cells in droplets due to
such as pressure drops, without computationally expensive reduced oxygen transport and reactivity with transfection
numerical solutions. Moreover, while two-phase flow models reagents at the fluid interface, fluorocarbon oils were utilized
have existed since the mid-20th century [67], most have to successfully transfect encapsulated cells [78]. Another
been tailored to macroscale pipe flow applications for vapour- study controllably lowered the fluorocarbon oil oxygen
liquid flows. More recently, microchannel droplet experiments concentration that reportedly caused encapsulated blood cell
and models have been developed which depend on the fluid sickling [79]. Despite adequate gas exchange for certain
properties for the continuous and dispersed phases, volume applications, aqueous droplet microfluidics still suffers from
fractions of the two fluids and two-phase flow regimes [68, 69]. the inability to continuously perfuse fresh media to cell
cultures to replenish nutrients and wash away toxic cell by-
2.3. Chemical species transport: mixing and isolation in products, often limiting encapsulated cell experiment time
droplets scales [18, 80]. However, off-chip cell culture periods of
up to several days are possible using gas-permeable storage
Due to small dimensions in microfluidic channels, Reynolds
materials with larger 600–700 picoliter (pL) droplets [15, 81].
numbers (Rec ) are typically small, on the order of 1 to 100 or
less. For reference, the channel Reynolds number is given by
3. Cell encapsulation methods
ρU Dh
Rec = , (3)
µ High-throughput single-cell droplet microfluidics benefits
from encapsulation methods which efficiently utilize cells
where ρ, U , Dh and µ represent the fluid density, mean
at the highest droplet generation rates. When cell-laden
velocity, channel hydraulic diameter and dynamic viscosity,
respectively. A low Reynolds number also implies a relatively suspensions serve as the aqueous fluid during droplet
low Peclet number (Pe), which compares the advection generation, cells become encapsulated in droplets. For
transport to the diffusion transport as randomly dispersed cell suspensions such as those used in
FACS machines [21], the number of cells in any given
LU drop is based on Poisson statistics, which govern random
Pe = , (4)
D encapsulation. Thus, the fraction of droplets containing k cells
is given by
where L is a characteristic length (such as the hydraulic
λk exp(−λ)
diameter Dh ) and D is a mass diffusion coefficient. Low Dk = , (5)
Pe represents diffusion-dominated transport, while high Pe k!
represents advection dominated mixing. While Rec and Pe where λ is the average number of cells per drop. For
may be large enough in high-throughput applications to leave random single-cell encapsulation, the cell suspension is
the viscous Stokes’ flow regime (i.e. where Rec , P e  1), it often highly diluted to avoid droplets containing multiple
is still below turbulent flow regimes better suited to mixing. cells. A large number of droplets thus remain empty,
However, rapid mixing is essential for applications where leading to wasted reagents and cell throughput decreases
reactions, measurement and actuation must take place on due to additional required droplet sorting or interrogation
millisecond (ms) or faster time scales, and where interrogation of empty droplets [82]. Highlighting the ubiquitous need
times in high-throughput devices are limited. In lieu of for efficient cell encapsulation, a 2009 review of bio-
diffusive mixing, microfluidic channel geometries such as electrospraying (deposition of cell-containing droplets onto
serpentines (figures 2(a) and (b)) are often added to induce surfaces to create ‘living residues’), states that the electrospray
chaotic advection (secondary flows within channels and method suffers from the inability to control the number of cells
droplets which cause folding of layers of fluid onto each per droplet [83].

5
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

Figure 3. (a) Passive droplet sorting following cell-triggered encapsulation. Large droplets represent droplets containing cells, while small
droplets are empty. The devices have been engineered to passively sort droplets containing cells to the upper flow branch. The white arrow
follows a cell-containing droplet to the incorrect branch [84]. Copyright 2008, National Academy of Sciences, USA. (b) Gel particle
encapsulation. Water is added in the first junction to space the particles prior to encapsulation, and oil is added in the second junction to
form drops and encapsulate the particles [85]. (c) Ordered encapsulation (top) where circled droplets represent single-particle encapsulation.
Controlled encapsulation of single HL60 cells (bottom) is also demonstrated. Scale bars represent 100 µm [86]. (d) Controlled
encapsulation of single and multiple particles. Scale bars represent 27 µm [87]. Parts (a) and (d) are reproduced with permission
from [84, 87], respectively. Parts (b) and (c) are reproduced by permission of the Royal Society of Chemistry, DOI: 10.1039/b909386a and
DOI: 10.1039/B805456H, respectively.

3.1. Controlling high-throughput cell encapsulation limitation for random encapsulation, another study utilized
densely packed gel particles in aqueous flows upstream of
To control cell encapsulation, a number of approaches may
the flow-focusing droplet nozzle to encapsulate single gel
be utilized. In one study, self-sorting of cell-containing
particles at 1.5 kHz rates with 98% efficiency [85] (figure 3(b)).
droplets from empty droplets provided singly encapsulated
cells at a throughput of 160 cells s−1 following a 1 kHz droplet However, expansion of the method to cells would require prior
generation rate [84] (figure 3(a)). Here, the empty droplets encapsulation of cells in gel particles to alleviate fluid stresses
were sorted [84] by size. Alternatively, empty droplets can associated with flow-based close packing.
be interrogated as if they contained cells, and their data can Additional studies used long, narrow aqueous flow
be considered later. To overcome the Poisson statistical channels that led to hydrodynamically ordered cell trains of

6
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

equal streamwise spacing [86]. As a result, cells arrived at the


drop generation nozzle at the same frequency as drops were
formed (figure 3(c)). The ordering of cells depends both on
sufficiently high flow rates and particle size in relation to the
channel size. The particle Reynolds number is given by
 a 2
Rep = Rec , (6)
H
where Rec is the channel Reynolds number, a is the cell
diameter and H is a characteristic channel dimension, often the
hydraulic diameter Dh . Single-cell ordered encapsulation of
human HL60 cells has been demonstrated with encapsulation
efficiencies twice those of Poisson statistics at rates of 14.9 kHz
[86], the same order of magnitude as current FACS machines
[21]. The method has also been demonstrated for two-particle
encapsulation (figure 3(d)), useful in cell-signalling studies,
for example. This study also reported simple design guidelines
for encapsulating controllable numbers (3, 4, . . . , N) of cells
per droplet [87]. Note that higher flow rates are generally
required for ordered encapsulation, which may be problematic
when considering the dripping to jetting transitions in droplet
formation. Higher flow rates also increase pressures and shear
forces on cells, though only for fractions of a second.

3.2. From water droplets to cell-encapsulating gel particles


In addition to aqueous droplet formation, agarose and
other polymer hydrogels have been employed to encapsulate
and immobilize cells [82, 88–92] or individual molecules
[89, 93] within monodisperse microspheres. In these
applications, particle composition is carefully selected to
control microcapsule mechanical, diffusive and degradation
properties. Typically, drop generation encapsulates the cells
within the precursors using typical flow focusing or other
Figure 4. Droplet microfluidic fabrication of yeast-cell-laden
junction-type nozzles. Note that the viscosity and interfacial radical-free hyperbranched polyglycerol-polyethylene glycol
tension may change due to the addition of gel precursors, (hPG-PEG) microgel particles. (a) Two sequential cross-junctions
which could greatly affect the drop generation dynamics serve to form monodisperse, micrometer-sized precursor droplets
and limit throughput when compared with encapsulation in which consist of hPG, PEG and cells. (b) Yeast-cell-laden
aqueous cell culture media [94]. In most cases, gel droplets microgel particles formed by gelation of the droplets.
(c) Core-shell-structured microgel with cells concentrated in the
are exposed to thermal or UV curing methods which must centre of the microgel. Reprinted with permission from [95].
not harm encapsulated cells. Often, the resulting hydrogel Copyright 2012, American Chemical Society.
microcapsules may also be re-suspended in a continuous
phase of aqueous media so that longer-term cell studies are mixing and diffusion between the two droplet components
possible, although some of the benefits of droplet confinement to create particles with two distinct halves. Paramagnetic
are eliminated in a re-suspended environment. To highlight Janus particles, fabricated using one magnetic nanoparticle
the ability of hydrogels to sustain longer-term experiments, laden inlet stream and one non-magnetic inlet stream, provide
agarose particles have been utilized in antibiotic resistance not only the ability to move and sort cell-embedded hydrogel
studies of E. coli cells [91] and in a seven day culture of droplets in a magnetic field, but the capability to orient particles
C. militaris cells [92]. An additional study encapsulated into large arrays for parallel manipulations [98].
and cultivated yeast cells in polyethyleneglycol (PEG) and Instead of simple cell immobilization in a gel matrix,
hyperbranched polyglycerol (hPG) without the use of cell- more complex core–shell emulsions can also be utilized to trap
harming polymerization initiators [95] (figure 4). cells inside porous capsules with tunable transport properties.
Interestingly, multiple aqueous hydrogel precursor inlets In addition, double emulsions (water/oil/water) have been
upstream of the drop generation nozzle may be used to form utilized to trap cells within a polymer (whose precursor is
so-called ‘Janus’ particles [96–99] (figure 5). When multiple soluble in the oil phase) [100–102] or other biomaterial (such
precursor streams merge immediately upstream of the droplet as a lipid [103]) membrane. More recently, porous shell
generation nozzle, the fluids are not well-mixed in the droplet. capsules were fabricated using a polymer-gold nanoparticle
Rapid curing following droplet formation therefore limits composite with versatile functional groups at up to 300 Hz

7
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

Figure 5. Parallel scale-up of Janus particle production with a 16-channel module. (a) Schematic of the microchannels on a chip. The ‘b’
and ‘w’ labels specify the inlet positions for the black and white monomers respectively. The aqueous phase is infused from the inner 16
inlets, arranged circularly with an outlet in the centre. (b) Schematic of the layered internal device structure (side view). (c) Top view of the
formation of biphasic droplets in the module. (d) Magnified view of the co-flow geometries. (e) Magnified view of the outlet port in the
centre of the chip. Reprinted with permission from [96].

rates with a demonstrated ability to encapsulate bacterial electric and magnetic fields and any changes which may
cells [104]. Note that these thin membrane and core–shell alter a cell’s phenotype. While serial methods which
methods highlight one critical role of droplet microfluidics interrogate and actuate individual droplets one at a time
in synthetic biology [105, 106] for both single cells and have proven indispensable, they require parallelization of
sub-cellular components. Additional materials and methods sensors and actuators to increase throughput further. For
for core–shell microcapsules have been reviewed in depth higher throughputs, goals include dynamic control, parallel
elsewhere [107–109]. (and ideally simultaneous) actuation and sensing and droplet-
specific tagging to track individual droplets or unique droplet
4. High-throughput droplet sensing and actuation characteristics, all with low rates of error, failure, and
methods interference with natural cellular processes.
While others have reviewed single-cell investigation
Parallel (multiplexed) droplet generation (figure 5) can techniques [110, 111] and microfluidic pumps [112, 113]
increase droplet production tremendously, but providing and valves [2] in depth, we focus this section on droplet
reliable downstream measurement and control of so many microfluidic methods, both serial and parallel, and their
droplets has been much more challenging. To match the potential for throughputs that compete technically and
high-throughput droplet generation capabilities (as shown economically with existing technologies. It is not always
in figure 3) to downstream droplet processing capabilities, straightforward to distinctly separate actuation and sensing
high-throughput actuation and measurement interfaces, data methods, as the two preferably work in conjunction to achieve
processing and analysis and real-time feedback control are maximum throughputs. However, to address sensing, we must
essential. first address how droplets are actuated to provide conditions
For droplet microfluidics at any throughput, actuation and and appropriate time scales for reliable droplet sensing. The
sensing methods should be minimally invasive to preserve the methods highlighted here in no way represent an exhaustive
viability and differentiation of encapsulated cells. Invasive list of techniques in single-cell droplet microfluidics; rather,
methods are those which introduce pressures, shear stresses, we attempt to highlight a few recent advances toward

8
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

high-throughput applications. Additionally, we discuss referenced laser heating studies, stationary droplet samples are
methods that have been demonstrated for actuation and cycled between approximately 60 ◦ C and 95 ◦ C while reporting
manipulation of suspended cells, since these principles may laser powers on the order of 30–70 mW. The laser is defocused
be expanded for use with droplets in the future. to apply a uniform intensity across the droplets with diameters
on the order of 300–400 µm. While electric fields and lasers
4.1. Extracellular environment control provide tools for coalescence, cell lysis and thermal control, it
is important to quantify induced stresses to avoid unintentional
While microfluidic droplets provide unique microreactor cell death.
conditions for test samples, often droplet environments must
be modified by introducing new aqueous fluids and reagents,
4.2. Droplet trapping
extracting fluid and cells from droplets, re-suspending cells
in a bulk aqueous environment, or modifying individual Measurement of microfluidic droplet characteristics requires,
droplet temperatures. Passive droplet splitting provides a for example, optical, magnetic, chemical, or thermal
mechanism to extract multiple samples of the extracellular interrogation of droplets which may be moving at a relatively
fluid environment. Coalescence mechanisms provide means high speed (up to hundreds of mm s−1 ) through finite
for introducing controlled volumes of reagents and media to microscopic fields of view. Therefore, matching the sensor
cell-containing droplets. interrogation time to the droplet lifetime in the field of view is a
Splitting and coalescence have been well-reviewed critical parameter when balancing the need to accurately assess
elsewhere [30, 31]. Briefly, passive splitting is often droplet contents versus the desire to maximize throughput. The
accomplished using a bifurcating T-junction [114, 115] or sensor time scale places an upper limit on the velocity at which
Y-junction [116]. In both T- and Y-junctions, droplet breakup droplets may be actuated for a single measurement in time if
dynamics have shown dependence on capillary number Ca and actuation follows sensing, one drop at a time.
droplet size [114, 117]. Symmetric splitting junctions [114] With cellular processes occurring on time scales from
(equal-sized outlet channels each with equal flow) provide sub-millisecond to hours and days, multiple measurements
equal-sized daughter droplets while asymmetric junctions may be required to obtain transient droplet profiles. Despite
[118] provide one larger droplet and one smaller droplet. It is the requirement to stop the flow, which limits the maximum
important to note that passive splitting methods do not control throughput, static droplet and cell traps provide some of the
which daughter droplet contains the encapsulated cell or cells. more established methods for detecting droplet properties
For passive coalescence, altered channel geometries can with maximum resolution over a wide range of time scales.
induce coalescence in low-stability emulsions simply by U-shaped flow-induced traps provide a method to ‘catch’
bringing a droplet into direct contact with another droplet droplets in a flow field while a continuous oil flow assists in
or bulk fluid interface [119–121]. Alternatively, patterned thermal and chemical transport (depending on oil solubility
biphilic surfaces provide spatial control over droplet wetting and interfacial properties) and in generating secondary mixing
on a small hydrophilic area of the channel wall. When flows within droplets [130, 131]. U-shaped traps may also
enough droplets collect and mix on the wetted surface, the be designed to contain multiple droplets [132, 133]. While
drag force overcomes the droplet capillary force, and the mechanical droplet traps have the benefit of containing cells
combined droplet detaches [122]. Droplets may be actively indefinitely for experiments with longer time scales, sub-
coalesced using electric fields (on the order of 50–200 V in millisecond kinetics were obtained using only droplet contact
some studies [123, 124]) oriented parallel to the two-phase traps [133]. As opposed to droplet-merging reactions, droplet
flow. Additionally, focused lasers can locally heat fluid contact provided a controlled reaction interface where the
interfaces at the point of droplet contact to induce interfacial temporal resolution was not limited by millisecond droplet
tension gradients and Marangoni flows in each droplet resulting mixing times. Three-dimensional packing schemes have also
in coalescence [125]. been utilized to passively arrange up to around one million
When using electric fields and focused lasers for droplet droplets in a wide-field array. As a result, simultaneous
control, care must be taken to avoid damage to encapsulated imaging of multiple droplet planes is possible [116, 134].
cells if cells are to remain viable for later interrogation. One Several trapping mechanisms also allow for quasi-
study characterized droplet-encapsulated cell electroporation continuous flow where droplets are stopped only temporarily.
for both gene delivery and cell lysis using droplet throughputs One two-dimensional array, the ‘dropspot’, passively captures
on the order of 40–250 Hz and applied voltages on the order droplets in continuous flow. This works because the interfacial
of 5–10 V [126]. While electroporation voltage thresholds are tension forces favour droplet locations within traps as spheres
based on many factors (e.g. fluid electrical properties, droplet rather than in the narrow regions between traps. Notably, these
sizes, droplet velocities), these values suggest precluding devices can also release droplets by increasing the pressure
droplet electrocoalescence techniques in some encapsulated differential across the channel following droplet interrogation
cell applications. Laser-induced coalescence applications [135, 136]. More integrated microfluidic designs use series
may be limited when extended cell viability is required. of complex valves such that media and reagents can be added
Rapid photolysis using focused lasers has been demonstrated to any one of hundreds of addressable chambers [137–139].
for enzymatic analysis of droplet-encapsulated cells [127]. While these chambers have been demonstrated with trapped
Heating using laser absorption has been exploited to reduce cells, droplets may also be trapped and subsequently released
droplet PCR cycle times to around 10 s [128, 129]. In the following interrogation.

9
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

4.3. Droplet sorting


4.3.1. Mechanical and hydrodynamic sorting. For
continuous flows, mechanical and hydrodynamic methods can
also provide passive sorting of cells and droplets. For example,
pinched flow fractionation (PFF) utilizes a narrow channel
followed by a sudden expansion to a wide channel to separate
particles based on size [140]. In addition, deterministic lateral
displacement (DLD) utilizes pillar arrays to sort particles based
on size [141]. Recently DLD has also been applied to sort
cell-containing droplets [142]. Passive hydrodynamic effects
in rectangular channels also provide a sorting mechanism.
At sufficiently high flow rates, objects such as particles,
cells and droplets tend to passively equilibrate to discrete
cross-sectional ‘focusing’ positions in both cylindrical and
rectangular channels [143–145] (recall figures 3(c) and (d)).
Combining this straight-channel effect with spiral [146, 147]
and asymmetrical [148, 149] channels introduces secondary Figure 6. Sorting droplets based on droplet fluorescence. When an
Dean flows, forcing larger objects to move toward the inside ac electric field was applied across the electrodes, the droplets were
channel wall while smaller objects stay further from the wall. deflected into the positive arm. In the absence of an electric field,
A number of discrete channel outlets provide separation based the droplets flowed into the negative arm owing to the lower
hydraulic resistance (inset). The length of the scale bar is
on size, although current devices typically require a semi- 100 µm [170]. Reproduced by permission of the Royal Society of
empirical calibration of flow rates to optimize the separation Chemistry, DOI: 10.1039/B902504A.
efficiency.
being exploited to deflect droplets into specific sorting paths.
4.3.2. Optical sorting. Optical methods include the use Acoustic waves provide an attractive method for manipulation
of lasers to control cells and particles [150]. The optical of label-free particles and droplets because forces depend
tweezer uses a single laser to trap a dielectric particle in three
only on droplet size, shape and density [164]. One study
dimensions [151]. For aqueous droplets, optical tweezers are
utilized acoustic forces to perform both size-based separation
not feasible when the refractive index of oil is greater than that
of polystyrene particles and density based separation of red
of the aqueous phase (∼1.33 for pure water), but optical vortex
blood cells, platelets and leukocytes [165]. Additional studies
traps have been utilized to facilitate trapping in this case [152].
demonstrate cell [166] and droplet [167] sorting at kHz rates.
Optical switches can also deflect droplets and cells for sorting
and have achieved actuation rates in the 4 ms range [153]. More
recently, optical pulling of dielectrics has also been discovered, 4.3.5. Electric sorting. In a similar manner to SAWs,
although achievable forces are relatively weak [154]. electric fields can also deflect and sort droplets. In commercial
FACS machines, droplets are electrostatically charged, and
4.3.3. Magnetic sorting. Magnetic activated cell droplet sorting is achieved by switching electric fields [21],
sorting (MACS) involves tagging cells and molecules with a phenomenon that has also been utilized to deflect droplets
functionalized magnetic nanoparticles or simply suspending between outlets [168]. Dielectrophoresis (reviewed elsewhere
magnetic nanoparticles in droplets. When magnetic particles [169]) has also been used to deflect droplets in fluorescence-
are uniformly suspended in all droplets, continuous flow activated droplet sorting (FADS) at rates up to 2 kHz [170]
actuation must be based on a triggered change in magnetic (figure 6). Dielectrophoretic methods using marker-specific
field. Controlling the magnetic nanoparticle concentration labelled cells (where labelled cells exhibited dielectrophoretic
also provides variable deflection (with force proportional to deflection and unlabelled cells did not) have achieved rare cell
concentration) of droplets in flow [155]. However, magnetic sorting at rates of 10 kHz [171].
nanoparticles bound to specific proteins [156] and cells [157–
159] prior to encapsulation can provide both sensing and 4.4. Droplet labelling and indexing
sorting actuation without direct control of the magnetic field
[160]. Studies have reported functional binding to yeast [158] For HTS, processing millions or more droplets on short time
and bacteria [157] cells without loss in cell viability for scales entails very short interrogation periods for each droplet.
selective magnetic manipulation. Magnetic droplet sorting For biological processes occurring on longer time scales
has been demonstrated at relatively low rates of around (minutes, hours or days), tracking individual droplets in static
10 droplets s−1 [155] and 30 droplets s−1 [161], while cell arrays for the entire process is not feasible while still achieving
sorting has been demonstrated using flow velocities on the a high throughput. Ideally, one could interrogate a unique
order of 0.1–1 mm s−1 [162]. droplet, temporarily store it, and recall the same droplet later
at discrete time intervals to measure its transient quantities.
4.3.4. Acoustic sorting. Surface acoustic waves (SAWs), Studies have demonstrated barcode identification of
often created using piezoelectric actuators [163], are also unique non-spherical particles by utilizing UV curing of

10
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

polymer precursors with variable optical mask patterns in encapsulated in droplets, based on enzymatic activity, at a rate
both continuous [172] and stop flow lithography [173]. As of 300 cells s−1 [170].
an alternative strategy for droplet identification, suspending One study combined confocal fluorescence microscopy
quantum dots [174–176] in the extracellular fluid may with fluorescence resonance energy transfer (FRET), which
provide an additional tool which is easily adaptable for uses fluorescent stimulation of one molecule to induce
spherical particle and liquid droplet identification. Briefly, fluorescent emission from an adjacent molecule, to detect
fluorescent quantum dots are suspended in droplets at N millisecond droplet reaction kinetics [181]. Additional
discrete concentrations using microfluidic gradient generation studies have used confocal fluorescence microscopy to detect
combined with standard drop generation techniques. When single DNA molecules in static droplets [131], cell protein
combined with a discrete range of M fluorescent colours, expression in moving droplets [16] and single molecule
(N M − 1) spectral codes are possible [176]. For example, a detection with droplet flow velocities up to 1 mm s−1 [182].
combination of 30 discrete intensity levels with five colours Additional droplet techniques include fluorescent polarization,
leads to over 24 million unique codes, although mapping demonstrated for protein detection [183], and fluorescence
each individual droplet to a unique code is not a trivial in situ hybridization (FISH), demonstrated for rare cell
challenge. Magnetic nanoparticle concentration gradients identification, although the latter method is lethal to cells [6].
could also assist as an additional marker, as droplet and particle
deflections are proportional to the magnetic nanoparticle 4.5.2. Droplet sensing by mass spectrometry. Mass
concentration [155]. spectrometry (MS) uses a label-free system which ionizes
sample compounds to generate charged molecule fragments
4.5. Droplet sensing methods for detection. The ionization process is invasive and deadly to
cells, so post-measurement cell culture is not possible. While
Prior to initiating many of the previously mentioned actuation
MS has been demonstrated using continuous flow microfluidic
and sorting mechanisms, sensing is required to decide on the
devices, the continuous oil phase in droplet microfluidics
appropriate actions. Sensors provide a measure of optical,
interferes with MS techniques such that droplets must be
electromagnetic, or chemical output that must be quickly
coalesced into continuous flow systems for analysis [27].
received, interpreted and converted to an actuation response.
Although droplet-encapsulated cells remain the focus of this Electrospray ionization MS (ESI-MS) has been developed to
review, here we also include several methods which have been re-introduce droplet streams into new aqueous streams for
developed for droplet-encapsulated molecular studies. These aerosol droplet generation and MS analysis at encapsulated
methods are relevant when detecting small concentrations of droplet rates on the order of 0.1 Hz [184, 185]. This is
molecules and proteins which may be expressed or absorbed relatively slow for high-throughput applications, and high
by encapsulated cells. dilution negates some of the benefits of droplet confinement.

4.5.1. Fluorescence droplet sensing. Though some cellular 4.5.3. Droplet sensing by Raman spectroscopy. Raman
proteins and other biomolecules exhibit auto-fluorescence, spectroscopy utilizes non-invasive, label-free interrogation of
they often must be tagged using targeted binding of fluorescent single cells and droplets using a spectrum of Raman bands
dyes or particles. Several techniques are highlighted here, but of molecular vibration initiated by an incident laser to serve
additional methods are reviewed in depth elsewhere [111, 177]. as fingerprint of a cell. Notably, unique bands exist for
To optimize throughput using fluorescent methods, one different sub-cellular components including proteins and DNA
must carefully weigh the importance of image resolution and [186]. Traditional Raman signal acquisition times are on
brightness with the ability to interrogate a larger number the order of minutes, as only a tiny fraction of photons
of droplets in parallel. Wide-field fluorescence utilizes a undergoes Raman scattering [187, 188]. This leads to low
fluorescence microscope, fluorescence-inducing light source throughput in Raman-activated cell sorting (RACS) platforms
and high resolution camera. Due to the wider fields of [189]. Interrogation times may be reduced, however, by
view, multiple droplets may be interrogated in parallel (and increasing the laser power [187, 189], or by using enhanced
in dynamic and static parallel channels [178] and arrays) for Raman spectroscopy techniques. Surface-enhanced Raman
maximizing throughput, although the increase in throughput scattering (SERS) detects molecules attached to noble metals
comes at the expense of decreased resolution [27]. Serpentine [187, 190] such as gold. SERS was used to investigate the
channels have been employed to move samples across the internal contents of gold-nanoparticle laden shells fabricated
field of view at discrete time intervals for fluorescent imaging, using microfluidics [104] and to reduce the interrogation
increasing total on-chip time for transient experiments [179]. time of E. coli cells to 1 s [191]. More recently, surface-
For smaller regions of interest, focused lasers are enhanced resonance Raman scattering (SERRS) systems have
used to induce fluorescence. Laser-induced fluorescence been demonstrated which detect multiple droplet analytes in
has made it possible to detect molecules on the 10−12 M real time at a rate of 4 droplets s−1 [192] and interrogate
scale [180]. Precise fluorescence activation is more moving droplets at sub-ms time scales at an average rate of
complicated in moving droplet flows but has been successfully 13.3 droplets s−1 [193].
implemented. In an integrated fluorescence-activated droplet
sorter (FADS), laser-induced fluorescence was utilized to 4.5.4. Electric droplet sensing. Several studies use insulated
dielectrophoretically sort continuously flowing E. coli cells electrodes placed within the top and bottom channel surfaces to

11
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

measure capacitance as droplets pass between the electrodes, a number of which have been addressed in recent reviews.
since the droplet changes the overall dielectric constant One review [26] highlighted how the absence of dilution in
across the channel, at time scales on the order of 10 ms droplets allows for faster accumulation of biomarkers reducing
[194, 195]. Aqueous droplet flows in silicone oil induce a clinical diagnostic times for detecting antibiotic responses of
change in the measured voltage or capacitance across the bacteria in human blood plasma [211]. This review also
electrodes. This serves as a droplet detector and droplet emphasized how volumetric confinement in droplets allowed
velocity measurement device when multiple electrode pairs are for enzymatic amplification of cell-surface protein biomarkers
included [195]. This simple method using only voltage signals [212]. Confinement also allowed for the detection of rare
provides a valuable alternative to optical droplet detection mammalian cells via cell lysis within droplets followed by
and velocity measurement where channel materials must be PCR amplification of DNA [213]. Several reviews [26, 214]
transparent. The measured signal is also proportional to note how confinement reduces the number of cells required
droplet size and was calibrated to quantify droplet sizes to mimic quorum sensing environments (high cell density)
for pure water, providing discrete volumetric measurements for cell-growth assays [18, 215]. Cell confinement has also
without using optical imaging. Depending on how droplet allowed for the detection of rare slow-growing cells since
composition affects its dielectric properties, capacitive sensors fast-growing cells cannot dominate a mixed population, as
could be exploited to measure droplet composition (including is the case in a bulk sample [216]. Given that a number of
detecting which droplets contain cells and which do not) and single-cell droplet reviews have been recently published, here
transient droplet reactions. Similarly, electrodes were utilized we focus on just a few notable studies based on their high-
to quantify droplet flows using impedance measurements throughput potentials. These studies have not only allowed
[196–198], with one study reporting measurement times on biological questions to be answered in new ways but also
the order of 100 ms [196]. represent significant progress in high-throughput single-cell
droplet microfluidic applications.
4.5.5. Magnetic droplet sensing. As previously mentioned, Based on the discussions from previous sections, it
functionalized magnetic nanoparticles can be tailored to bind is apparent that high-throughput applications of droplet
to specific cells and molecules for their use as both sensor and microfluidics have been limited by the ability to sense droplet
actuator in droplet microfluidics [160]. Additionally, studies contents and sort droplets at the kHz rates at which droplets
have used magnetic nanoparticles to interrogate droplets using can be generated. Significant efforts are now being directed
nuclear magnetic resonance (NMR) imaging coils wrapped to apply on-chip dielectrophoretic sorting of droplets, based
around channels, although throughput was relatively low on fluorescence interrogation, to biological applications that
because of the need to stop the flow for 16 s for sufficient require a high throughput. These include screening cells for
droplet sensing time [199, 200]. Additional studies have used enzyme activity [170], screening drug libraries for cytotoxicity
microfluidic NMR hand-held devices (without droplets) to [15] (figure 7) and screening engineered proteins for directed
detect bacteria in unprocessed biological samples [201, 202] evolution [217]. This represents the logical extension of
and for cancer cell detection [203]. A more detailed review of fluorescence-activated cell sorting to a drop-based system, and
magnetic biosensors is available elsewhere [204]. the throughputs have now reached about 2000 droplets per
second, a significant fraction of the best rates achieved for
4.5.6. Particles and droplets as sensors. In some cases, commercial FACS machines. In contrast to FACS, however,
suspended droplets and hydrogels themselves can function as droplets enable sorting according to accumulated changes
sensors. Stimuli-responsive hydrogels can be engineered to in the extracellular media that are induced by cell-specific
change size based on temperature, pH, droplet composition processes, and this can lead to time-integrated measurements
and electric fields [205–207]. DNA may also be used as a that are not limited to intracellular processes. This is an
reversible cross-linker to modulate the hydrogel rheological impressive capability and will clearly have application to many
and mechanical properties and to provide for controlled critical problems in biotechnology and biomedicine.
interactions between DNA and other molecules and cells [208]. However, nutrient depletion by encapsulated cells sets an
In another study, oxygen-permeable PDMS microparticles important time limit on experiments since even single cells will
were embedded with oxygen-sensitive dye for an optical eventually run out of nutrients and accumulate toxic levels
readout of oxygen concentration [209]. Liquid droplets of waste products in their drop. Although it is inherently
have also been used to monitor reactions if those reactions impossible to replenish media in droplets without sacrificing
induce changes in osmotic pressure. As a result, small some degree of chemical isolation, it is possible to exchange
but measurable volumetric water transfer between droplets the extracellular fluids while still preventing entry of biological
provided quantifiable reaction metrics [210]. contaminants, e.g. bacteria. In one type of approach, gel beads
are formed from emulsion drops, with or without cells inside,
5. Expansion to high-throughput applications and they are then re-suspended in aqueous media to allow for
fluid exchange without cell–cell contact. Since this barrier may
In previous sections, we have addressed droplet mechanics and only be necessary at the oil–water interface, it could be better
methods for measuring some of the most relevant properties in some cases to create a polymer shell with a defined pore
of cell-laden droplets. Here, we attempt to place these size in order to limit the effects of gel precursors on events in
methods in the context of specific biomedical applications, the drop interior (c.f. a subsequent section on capsule density).

12
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

Washing in aqueous media then facilitates a direct coupling


of encapsulated cells to standard FACS [218, 219] (figure 8).
Similarly, w/o/w emulsions can be used directly in FACS
machines yet retain all the benefits of encapsulation [220, 221].
Moreover, though drop-based FACS is appropriate for
many applications, one can aim for even higher throughputs,
and in particular, for passive droplet sorting mechanisms that
select droplets according to specific properties of the cells
inside them. To see why passive sorting can be desirable,
consider that although commercial FACS machines can sort
many thousands of cells per second with multi-colour probes,
this instrument is expensive, takes up significant lab space,
and requires a trained technician to run properly. On the
other hand, MACS can isolate rare cells from a large mixed
sample by specific adhesion of coated magnetic beads to cell
membrane proteins. Although less specific than multi-colour
FACS, the key is that separation is done in parallel as a single
batch operation with an inexpensive magnet.
Furthermore, there are numerous cell separation
approaches which do not require active control of individual
cells. These include affinity-based capture [222], size-based
separation by DLD [141] or inertial focusing [144], and
density-gradient centrifugation [223], among many others that
rely on multiple physics modes, as reviewed elsewhere [224].
The key to each of these is that sensing and actuation are
intrinsically linked so that control per se is not required on Figure 7. Drop-based platforms for toxicity screening of single
a cell-by-cell basis. This greatly simplifies operation even at cells. In addition to integrating cell encapsulation, pairwise droplet
merging and reinjection of encapsulated cells, at combined rates of
ultra-high rates through simpler parallelization. Because many 100 cells per second, this study used eight concentrations of
of the same principles for cell separation might in theory be fluorescent dye to encode each droplet by its drug concentration.
adapted to droplet separation, one promising area of future Copyright 2009, National Academy of Sciences, USA. Reproduced
research would be to find robust means to precisely change the with permission from [15].
sortable properties of droplets in response to the phenotype
exhibited by cells inside each drop. Droplet size, density change in volume was seen for dead cells or those in drops
and viscoelasticity are the obvious physical properties which, without nutrients. The appeal of their approach for high
if modified by a cell, could lead to passive separation of throughput cell-based encapsulation is that droplet sorting
the associated droplets. At first glance, these would seem could be done in parallel based on size alone without the need
to require quite large efforts by cells and do not suggest to actively interrogate and sort each droplet individually. This
specificity in sorting. Nevertheless, in highlighting three recent allows enrichment of cells that continue to grow over several
approaches, it becomes clear that each avenue is possible for hours in drops from inactive cells or empty droplets, potentially
cell-based assays. yielding methods to isolate pathological cell clones based on
their resistance to therapeutics added to the media. In fact, in a
5.1. Droplet size prior study, DLD was used to separate yeast-containing drops
at 12 000 drops per second due to the osmotic shrinkage of
Inert fluorocarbon oils are notable for their impermeability cell-containing droplets [142].
to aqueous and lipophilic molecules and high gas exchange,
making them highly desirable for cell-based assays. However, 5.2. Capsule density
this chemical segregation is not absolute, and recent work
[210, 225] has taken advantage of the tiny but non-zero Interestingly, one recent study [104] reported a simple
mobility of water in FC-40 to turn monodisperse droplets into approach to create porous capsules through self-assembly of a
osmotic sensors for chemical reactions (figure 9(a)). Notably, supramolecular coating as a thin, rigid, film along the FC-40
the authors observed that water migrated through the oil over oil and water interface (figure 9(b)). Specifically, the authors
the course of several hours in response to solute depletion mixed a host molecule with two guest molecules just upstream
by cellular metabolic activity, moving water from yeast- of a T-junction for drop formation and observed that a ternary
containing droplets to nearby empty droplets in a hexagonal complex formed at the droplet boundary which solidified on
close-packed droplet array. This was shown to result from partial dehydration of the drop. After cell encapsulation, the
osmotic pressure differences created by growing cells. Yeast difference in density between the capsule and the surrounding
cells ultimately exhausted their nutrients such that drop volume aqueous media is due entirely to the presence of cells plus
stabilized at about half its initial value after 33 h, where no a thin shell, so capsules could be fractionated according to

13
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

Figure 8. Coupling emulsion drops to standard FACS for high throughput single-cell gene expression. Just upstream of a droplet nozzle,
cells are combined with RT-PCR and cell lysis buffer within a 2% agarose solution. After thermocycling and gelation, agar beads are
re-suspended into aqueous buffer so that they can be used in existing fluorescence-activated cell sorters. Copyright 2012, American
Chemical Society. Reproduced with permission from [218].

the number of cells in a capsule and their densities, enabling Looking to the future, it will be important to increase
sorting by centrifugation. the specificity and flexibility of passive droplet separation
methods. One interesting approach could be to self-assemble
5.3. Droplet viscoelasticity lipid rafts along the oil–water interface, among a larger
coating of stabilizing surfactants, as a platform for receptor–
A third recent report [226] described a step along the ligand interactions. For instance, receptor domains on the
way to creating artificial cells from microfluidic droplets. aqueous side of the droplet surface could be made to change
Specifically, the authors encapsulated Xenopus egg extracts conformation on the oil-phase side after specific interaction
and observed the self-organization of microtubules and actin with ligands secreted by the cell. If these changes led
filaments in droplets. Because these events are under to adhesion of the lipids to corresponding motifs presented
complex biological control, it is possible in theory to on channel walls, affinity-based capture of droplets could
switch between formation and degradation through regulatory become possible. In fact, one recent study grafted lipid
controls. However, it is not clear if these intracellular structures patches onto soybean oil droplets in an aqueous continuous
could be coupled to the activities of encapsulated cells through phase to generate receptor–ligand-like interactions between
specific cell secretions. Nevertheless, if this were possible, the droplet surface and a T-cell line [227]. Alternatively,
it would allow cells which secrete particular biomolecules through the further development of artificial cells in droplets,
to modify the mechanical properties of their encapsulating one can envision the use of combinatorial interactions among
droplet into a form that is highly resistant to internal flow droplet pairs across an artificial lipid bilayer [228] that
(filament growth), or one that behaves normally (degradation), includes specific membrane proteins which cleave the drop–
allowing passive separations based on differential droplet drop connections on exposure to a particular cell secretion.
migration according to viscoelastic properties (figure 9(c)). In Or, droplets could become sticky only when cells secrete
addition, it should be possible to encapsulate cells in specific the target molecules. Either way, separation could then be
extracellular matrix materials to select for cells that are able to accomplished by differential centrifugation. The key benefit
degrade their matrix. in these approaches would be the high degree of specificity

14
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

Figure 9. Strategies for control over droplet physical properties by encapsulated cells. (a) Solute depletion by yeast cell metabolism leads
to osmotic shrinkage of cell-containing drops over 30 h of culture [210], yielding size-based droplet sorting capability [142]. (b) Thin-shell
porous capsules were formed by partial dehydration of droplets in the presence of host-guest chemicals [104]. After resuspension in water,
capsule density is determined by encapsulated cellular materials, enabling separation by density-gradient centrifugation. In addition to
encapsulation of cells in drops filled by a biodegradable extracellular matrix, (c) droplet viscoelastic properties could be modulated by
shifting the balance between growth and disassembly of actin filaments (top) or microtubules (bottom) [226]. Parts (a) and (c) reproduced
by permission of the Royal Chemistry Society, DOI: 10.1039/C2LC20971C and DOI: 10.1039/C0LC00046A, respectively. Part (b)
reprinted with permission from AAAS.

afforded by receptor–ligand interactions, and the passive mechanics, chemistry, physics, biology, computer science,
manner of sorting. materials science, economics and personalized medicine to
bring the great potential of droplet microfluidics to full fruition.
6. Conclusion

In the past ten years, droplet microfluidic platforms have References


provided a new way to carry out single-cell and biomolecular
experiments for drug discovery, ‘omics’ studies and rare cell [1] Beebe D J, Mensing G A and Walker G M 2002 Physics and
detection. Downstream measurement and actuation of droplets applications of microfluidics in biology Annu. Rev. Biomed.
Eng. 4 261–86
provides a major challenge to reaching this high-throughput [2] Melin J and Quake S R 2007 Microfluidic large-scale
potential. Automated systems must acquire and comprehend integration: the evolution of design rules for biological
massive amounts of data to make decisions in the tiny fraction automation Annu. Rev. Biophys. Biomol. Struct. 36 213–31
of a second that a droplet resides within a small region of [3] Mark D, Haeberle S, Roth G, von Stetten F and Zengerle R
interest. When biological processes take place on longer 2010 Microfluidic lab-on-a-chip platforms: requirements,
characteristics and applications Chem. Soc. Rev. 39 1153–82
time scales, the system must either slow droplet speeds at
[4] Duffy D C, McDonald J C, Schueller O J A and
the expense of throughput, or employ methods which can Whitesides G M 1998 Rapid prototyping of microfluidic
interrogate, store, and reacquire a unique droplet over time. systems in poly(dimethylsiloxane) Anal. Chem.
Indexed droplet tracking has been recently explored using 70 4974–84
quantum dots, but much room for improvement remains. [5] Vincent M E, Liu W, Haney E B and Ismagilov R F 2010
Microfluidic stochastic confinement enhances analysis of
Highlighting the challenges above should not overshadow
rare cells by isolating cells and creating high density
the many recent successes in droplet microfluidics. Droplet- environments for control of diffusible signals Chem. Soc.
based PCR methods have demonstrated gains for protein Rev. 39 974–84
engineering and directed evolution studies while using smaller [6] Liu W S, Kim H J, Lucchetta E M, Du W B and Ismagilov R F
volumes of costly reagents. Microfluidic lab on chip devices 2009 Isolation, incubation, and parallel functional testing
and identification by FISH of rare microbial single-copy
such as the ‘FADS’ system, utilizing fluorescence sensing and
cells from multi-species mixtures using the combination of
dielectrophoretic actuation, have made significant progress chemistrode and stochastic confinement Lab Chip
toward high speed automated sorting, reaching 2000 droplets 9 2153–62
per second using an on-chip device and microscope. In [7] Lombardi D and Dittrich P S 2010 Advances in microfluidics
recent years, data acquisition times for single-cell Raman for drug discovery Expert Opin. Drug Discovery 5 1081–94
spectroscopy have been reduced from minutes to sub-second [8] Gong Z, Zhao H, Zhang T, Nie F, Pathak P, Cui K, Wang Z,
Wong S and Que L 2011 Drug effects analysis on cells
durations. Continuing on the path to commercial successes using a high throughput microfluidic chip Biomed.
will take a truly interdisciplinary understanding of fluid Microdevices 13 215–9

15
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

[9] Fair R B 2007 Digital microfluidics: is a true lab-on-a-chip [33] Xu J and Attinger D 2008 Drop on demand in a microfluidic
possible? Microfluidics Nanofluidics 3 245–81 chip J. Micromech. Microeng. 18 065020
[10] Choi K, Kim J Y, Ahn J H, Choi J M, Im M and Choi Y K [34] Kim H, Luo D W, Link D, Weitz D A, Marquez M and
2012 Integration of field effect transistor-based biosensors Cheng Z D 2007 Controlled production of emulsion drops
with a digital microfluidic device for a lab-on-a-chip using an electric field in a flow-focusing microfluidic device
application Lab Chip 12 1533–9 Appl. Phys. Lett. 91 133106
[11] Schaerli Y and Hollfelder F 2009 The potential of microfluidic [35] Anna S L, Bontoux N and Stone H A 2003 Formation of
water-in-oil droplets in experimental biology Mol. Biosyst. dispersions using ‘flow focusing’ in microchannels Appl.
5 1392–404 Phys. Lett. 82 364–6
[12] Pekin D et al 2011 Quantitative and sensitive detection of rare [36] Utada A, Fernandez-Nieves A, Stone H and Weitz D 2007
mutations using droplet-based microfluidics Lab Chip Dripping to jetting transitions in coflowing liquid streams
11 2156–66 Phys. Rev. Lett. 99 094502
[13] Markey A L, Mohr S and Day P J 2010 High-throughput [37] Nisisako T, Torii T and Higuchi T 2002 Droplet formation in a
droplet PCR Methods 50 277–81 microchannel network Lab Chip 2 24–6
[14] Zhu Z, Jenkins G, Zhang W, Zhang M, Guan Z and Yang C J [38] Chu L Y, Utada A S, Shah R K, Kim J W and Weitz D A 2007
2012 Single-molecule emulsion PCR in microfluidic Controllable monodisperse multiple emulsions Angew.
droplets Anal. Bioanal. Chem. 403 2127–43 Chem. Int. Edn Engl. 46 8970–4
[15] Brouzes E, Medkova M, Savenelli N, Marran D, [39] Okushima S, Nisisako T, Torii T and Higuchi T 2004
Twardowski M, Hutchison J B, Rothberg J M, Link D R, Controlled production of monodisperse double emulsions
Perrimon N and Samuels M L 2009 Droplet microfluidic by two-step droplet breakup in microfluidic devices
technology for single-cell high-throughput screening Proc. Langmuir: ACS J. Surf. Colloids 20 9905–8
Natl Acad. Sci. USA 106 14195–200 [40] Dreyfus R, Tabeling P and Willaime H 2003 Ordered and
[16] Huebner A, Srisa-Art M, Holt D, Abell C, Hollfelder F, disordered patterns in two-phase flows in microchannels
deMello A J and Edel J B 2007 Quantitative detection of Phys. Rev. Lett. 90 144505
protein expression in single cells using droplet microfluidics [41] Becker H and Gartner C 2008 Polymer microfabrication
Chem. Commun. 1218–20 technologies for microfluidic systems Anal. Bioanal. Chem.
[17] Srisa-Art M, Bonzani I C, Williams A, Stevens M M, 390 89–111
deMello A J and Edel J B 2009 Identification of rare [42] Tsao C-W and DeVoe D L 2008 Bonding of thermoplastic
progenitor cells from human periosteal tissue using droplet polymer microfluidics Microfluidics Nanofluidics 6 1–16
microfluidics Analyst 134 2239–45 [43] Roy E, Geissler M, Galas J-C and Veres T 2011 Prototyping of
[18] Koster S et al 2008 Drop-based microfluidic devices for microfluidic systems using a commercial thermoplastic
encapsulation of single cells Lab Chip 8 1110–5 elastomer Microfluidics Nanofluidics 11 235–44
[19] 2012 Search Results: Topic=(‘high throughput’ microfluidic*). [44] Berthier E, Young E W K and Beebe D 2012 Engineers are
(http://apps.webofknowledge.com/: Thomson Reuters) from PDMS-land, biologists are from Polystyrenia Lab
[20] Osborne G W 2011 Recent advances in flow cytometric cell Chip 12 1224–37
sorting Methods Cell Biol. 102 533–56 [45] Bong K W, Xu J J, Kim J H, Chapin S C, Strano M S,
[21] Virgo P F and Gibbs G J 2012 Flow cytometry in clinical Gleason K K and Doyle P S 2012
pathology Ann. Clin. Biochem. 49 17–28 Non-polydimethylsiloxane devices for oxygen-free flow
[22] Eisenstein M 2006 Cell sorting: divide and conquer Nature lithography Nature Commun. 3 805
441 1179–85 [46] Garstecki P, Fuerstman M J, Stone H A and Whitesides G M
[23] Ashcroft R G and Lopez P A 2000 Commercial high speed 2006 Formation of droplets and bubbles in a microfluidic
machines open new opportunities in high throughput flow T-junction-scaling and mechanism of break-up Lab Chip
cytometry (HTFC) J. Immunol. Methods 243 13–24 6 437–46
[24] Huebner A, Sharma S, Srisa-Art M, Hollfelder F, Edel J B and [47] De Menech M, Garstecki P, Jousse F and Stone H A 2008
Demello A J 2008 Microdroplets: a sea of applications? Transition from squeezing to dripping in a microfluidic
Lab Chip 8 1244–54 T-shaped junction J. Fluid Mech. 595 141–6
[25] Casadevall i Solvas X and deMello A 2011 Droplet [48] Pathak M 2011 Numerical simulation of membrane
microfluidics: recent developments and future applications emulsification: effect of flow properties in the transition
Chem. Commun. 47 1936–42 from dripping to jetting J. Membrane Sci. 382 166–76
[26] Zagnoni M and Cooper J M 2011 Droplet microfluidics for [49] Abate A R, Mary P, van Steijn V and Weitz D A 2012
high-throughput analysis of cells and particles Methods Cell Experimental validation of plugging during drop formation
Biol. 102 25–48 in a T-junction Lab Chip 12 1516–21
[27] Theberge A B, Courtois F, Schaerli Y, Fischlechner M, [50] Bibette J, Calderon F L and Poulin P 1999 Emulsions: basic
Abell C, Hollfelder F and Huck W T 2010 Microdroplets in principles Rep. Prog. Phys. 62 969–1033
microfluidics: an evolving platform for discoveries in [51] Baret J C 2012 Surfactants in droplet-based microfluidics Lab
chemistry and biology Angew. Chem. Int. Edn Engl. chip 12 422–33
49 5846–68 [52] Holtze C et al 2008 Biocompatible surfactants for
[28] Christopher G F and Anna S L 2007 Microfluidic methods for water-in-fluorocarbon emulsions Lab Chip 8 1632–9
generating continuous droplet streams J. Phys. D: Appl. [53] Holt D J, Payne R J, Chow W Y and Abell C 2010
Phys. 40 R319–36 Fluorosurfactants for microdroplets: interfacial tension
[29] Teh S Y, Lin R, Hung L H and Lee A P 2008 Droplet analysis J. Colloid Interface Sci. 350 205–11
microfluidics Lab Chip 8 198–220 [54] Holt D J, Payne R J and Abell C 2010 Synthesis of novel
[30] Baroud C N, Gallaire F and Dangla R 2010 Dynamics of fluorous surfactants for microdroplet stabilisation in
microfluidic droplets Lab Chip 10 2032–45 fluorous oil streams J. Fluorine Chem. 131 398–407
[31] Seemann R, Brinkmann M, Pfohl T and Herminghaus S [55] Yan N X, Gray M R and Masliyah J H 2001 On water-in-oil
2012 Droplet based microfluidics Rep. Prog. Phys. emulsions stabilized by fine solids Colloid Surf.
75 016601 A—Physicochem. Eng. Asp. 193 97–107
[32] Hardt S and Hahn T 2012 Microfluidics with aqueous [56] Simovic S and Prestidge C A 2004 Nanoparticles of varying
two-phase systems Lab Chip 12 434–42 hydrophobicity at the emulsion droplet–water interface:

16
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

adsorption and coalescence stability Langmuir: ACS J. Surf. rapid oxygen exchange in microfluidic drops Lab Chip
Colloids 20 8357–65 10 2505–12
[57] Sohel Murshed S M, Tan S-H and Nguyen N-T 2008 [80] Lindstrom S and Andersson-Svahn H 2010 Overview of
Temperature dependence of interfacial properties and single-cell analyses: microdevices and applications Lab
viscosity of nanofluids for droplet-based microfluidics Chip 10 3363–72
J. Phys. D: Appl. Phys. 41 085502 [81] Clausell-Tormos J et al 2008 Droplet-based microfluidic
[58] Bresme F and Oettel M 2007 Nanoparticles at fluid interfaces platforms for the encapsulation and screening of
J. Phys.: Condens. Matter 19 413101 mammalian cells and multicellular organisms Chem. Biol.
[59] Anderson D M, McFadden G B and Wheeler A A 1998 15 427–37
Diffuse-interface methods in fluid mechanics Annu. Rev. [82] Velasco D, Tumarkin E and Kumacheva E 2012 Microfluidic
Fluid Mech. 30 139–65 encapsulation of cells in polymer microgels Small
[60] Sethian J A and Smereka P 2003 Level set methods for fluid 8 1633–42
interfaces Annu. Rev. Fluid Mech. 35 341–72 [83] Hong J, deMello A J and Jayasinghe S N 2010
[61] Tryggvason G, Bunner B, Esmaeeli A, Juric D, Al-Rawahi N, Bio-electrospraying and droplet-based microfluidics:
Tauber W, Han J, Nas S and Jan Y J 2001 A front-tracking control of cell numbers within living residues Biomed.
method for the computations of multiphase flow J. Comput. Mater. 5 21001
Phys. 169 708–59 [84] Chabert M and Viovy J L 2008 Microfluidic high-throughput
[62] Mittal R and Iaccarino G 2005 Immersed boundary methods encapsulation and hydrodynamic self-sorting of single cells
Annu. Rev. Fluid Mech. 37 239–61 Proc. Natl Acad. Sci. USA 105 3191–6
[63] Liu H H and Zhang Y H 2009 Droplet formation in a T-shaped [85] Abate A R, Chen C H, Agresti J J and Weitz D A 2009 Beating
microfluidic junction J. Appl. Phys. 106 034906 Poisson encapsulation statistics using close-packed ordering
[64] Chun B and Ladd A J C 2006 Inertial migration of neutrally Lab Chip 9 2628–31
buoyant particles in a square duct: an investigation of [86] Edd J F, Di Carlo D, Humphry K J, Koster S, Irimia D,
multiple equilibrium positions Phys. Fluids 18 031704 Weitz D A and Toner M 2008 Controlled encapsulation of
[65] Chen S and Doolen G D 1998 Lattice Boltzmann method for single-cells into monodisperse picolitre drops Lab Chip
fluid flows Annu. Rev. Fluid Mech. 30 329–64 8 1262–4
[66] Steinke M and Kandlikar S 2006 Single-phase liquid [87] Lagus T P and Edd J F 2012 High throughput single-cell and
friction factors in microchannels Int. J. Therm. Sci. multiple-cell micro-encapsulation J. Visualized Exp.
45 1073–83 64 4096
[67] Lockhart R W and Martinelli R C 1949 Proposed correlation [88] Luo D, Pullela S R, Marquez M and Cheng Z 2007 Cell
of data for isothermal two-phase, two-component flow in encapsules with tunable transport and mechanical properties
pipes Chem. Eng. Prog. 45 39–45 Biomicrofluidics 1 34102
[68] Labrot V, Schindler M, Guillot P, Colin A and Joanicot M [89] Um E, Lee D-S, Pyo H-B and Park J-K 2008 Continuous
2009 Extracting the hydrodynamic resistance of droplets generation of hydrogel beads and encapsulation of
from their behavior in microchannel networks biological materials using a microfluidic droplet-merging
Biomicrofluidics 3 12804 channel Microfluidics Nanofluidics 5 541–9
[69] Choi C and Kim M 2011 Flow pattern based correlations of [90] Leonard A, Dandoy P, Danloy E, Leroux G, Meunier C F,
two-phase pressure drop in rectangular microchannels Int. Rooke J C and Su B L 2011 Whole-cell based hybrid
J. Heat Fluid Flow 32 1199–207 materials for green energy production, environmental
[70] Atencia J and Beebe D J 2005 Controlled microfluidic remediation and smart cell-therapy Chem. Soc. Rev.
interfaces Nature 437 648–55 40 860–85
[71] Capretto L, Cheng W, Hill M and Zhang X 2011 Micromixing [91] Eun Y J, Utada A S, Copeland M F, Takeuchi S and
within microfluidic devices Top. Curr. Chem. 304 27–68 Weibel D B 2011 Encapsulating bacteria in agarose
[72] Bringer M R, Gerdts C J, Song H, Tice J D and Ismagilov R F microparticles using microfluidics for high-throughput cell
2004 Microfluidic systems for chemical kinetics that rely on analysis and isolation ACS Chem. Biol. 6 260–6
chaotic mixing in droplets Phil. Trans. R. Soc A [92] Lin Y S, Yang C H, Lu K, Huang K S and Zheng Y Z 2011
362 1087–104 Synthesis of agar microparticles using
[73] Song H, Chen D L and Ismagilov R F 2006 Reactions in temperature-controlled microfluidic devices for Cordyceps
droplets in microflulidic channels Angew. Chem. Int. Edn militaris cultivation Electrophoresis 32 3157–63
Engl. 45 7336–56 [93] Leng X, Zhang W, Wang C, Cui L and Yang C J 2010 Agarose
[74] Yeh C-H, Chen Y-C and Lin Y-C 2011 Generation of droplets droplet microfluidics for highly parallel and efficient single
with different concentrations using gradient-microfluidic molecule emulsion PCR Lab Chip 10 2841–3
droplet generator Microfluidics Nanofluidics 11 245–53 [94] Berthier J, Le Vot S, Tiquet P, David N, Lauro D,
[75] Cai L F, Zhu Y, Du G S and Fang Q 2012 Droplet-based Benhamou P Y and Rivera F 2010 Highly viscous fluids in
microfluidic flow injection system with large-scale pressure actuated flow focusing devices Sensors Actuators
concentration gradient by a single nanoliter-scale injection A 158 140–8
for enzyme inhibition assay Anal. Chem. 84 446–52 [95] Rossow T, Heyman J A, Ehrlicher A J, Langhoff A,
[76] Bajpayee A, Edd J F, Chang A and Toner M 2010 Weitz D A, Haag R and Seiffert S 2012 Controlled
Concentration of glycerol in aqueous microdroplets by synthesis of cell-laden microgels by radical-free gelation in
selective removal of water Anal. Chem. 82 1288–91 droplet microfluidics J. Am. Chem. Soc. 134 4983–9
[77] Wong E H M, Rondeau E, Schuetz P and Cooper-White J 2009 [96] Nisisako T, Torii T, Takahashi T and Takizawa Y 2006
A microfluidic-based method for the transfer of biopolymer Synthesis of monodisperse bicolored janus particles with
particles from an oil phase to an aqueous phase Lab Chip electrical anisotropy using a microfluidic co-flow system
9 2582–90 Adv. Mater. 18 1152–6
[78] Chen F, Zhan Y, Geng T, Lian H, Xu P and Lu C 2011 [97] Walther A and Müller A H E 2008 Janus particles Soft Matter
Chemical transfection of cells in picoliter aqueous droplets 4 663
in fluorocarbon oil Anal. Chem. 83 8816–20 [98] Yuet K P, Hwang D K, Haghgooie R and Doyle P S 2010
[79] Abbyad P, Tharaux P L, Martin J L, Baroud C N and Multifunctional superparamagnetic Janus particles
Alexandrou A 2010 Sickling of red blood cells through Langmuir: ACS J. Surf. Colloids 26 4281–7

17
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

[99] Nisisako T and Hatsuzawa T 2009 A microfluidic [120] Niu X, Gulati S, Edel J B and deMello A J 2008
cross-flowing emulsion generator for producing biphasic Pillar-induced droplet merging in microfluidic circuits Lab
droplets and anisotropically shaped polymer particles Chip 8 1837–41
Microfluidics Nanofluidics 9 427–37 [121] Niu X Z, Gielen F, Edel J B and deMello A J 2011 A
[100] Kim J W, Utada A S, Fernandez-Nieves A, Hu Z and microdroplet dilutor for high-throughput screening Nature
Weitz D A 2007 Fabrication of monodisperse gel shells and Chem. 3 437–42
functional microgels in microfluidic devices Angew. Chem. [122] Fidalgo L M, Abell C and Huck W T S 2007
Int. Edn Engl. 46 1819–22 Surface-induced droplet fusion in microfluidic devices Lab
[101] Seiffert S, Romanowsky M B and Weitz D A 2010 Janus Chip 7 984–6
microgels produced from functional precursor polymers [123] Zagnoni M, Le Lain G and Cooper J M 2010
Langmuir: ACS J. Surf. Colloids 26 14842–7 Electrocoalescence mechanisms of microdroplets using
[102] Seiffert S and Weitz D A 2010 Controlled fabrication of localized electric fields in microfluidic channels Langmuir:
polymer microgels by polymer-analogous gelation in ACS J. Surf. Colloids 26 14443–9
droplet microfluidics Soft Matter 6 3184 [124] Niu X Z, Gielen F, deMello A J and Edel J B 2009
[103] Teh S Y, Khnouf R, Fan H and Lee A P 2011 Stable, Electro-coalescence of digitally controlled droplets Anal.
biocompatible lipid vesicle generation by solvent Chem. 81 7321–5
extraction-based droplet microfluidics Biomicrofluidics [125] Baroud C N, de Saint Vincent M R and Delville J P 2007 An
5 44113-4411312 optical toolbox for total control of droplet microfluidics Lab
[104] Zhang J, Coulston R J, Jones S T, Geng J, Scherman O A and Chip 7 1029–33
Abell C 2012 One-step fabrication of supramolecular [126] Zhan Y H, Wang J, Bao N and Lu C 2009 Electroporation
microcapsules from microfluidic droplets Science of cells in microfluidic droplets Anal. Chem. 81 2027–31
335 690–4 [127] He M Y, Edgar J S, Jeffries G D M, Lorenz R M, Shelby J P
[105] Gulati S, Rouilly V, Niu X, Chappell J, Kitney R I, Edel J B, and Chiu D T 2005 Selective encapsulation of single cells
Freemont P S and deMello A J 2009 Opportunities for and subcellular organelles into picoliter- and
microfluidic technologies in synthetic biology J. R. Soc. femtoliter-volume droplets Anal. Chem. 77 1539–44
Interface 6 (Suppl 4) S493–506 [128] Kim H, Vishniakou S and Faris G W 2009 Petri dish PCR:
[106] Vinuselvi P, Park S, Kim M, Park J M, Kim T and Lee S K laser-heated reactions in nanoliter droplet arrays Lab Chip
2011 Microfluidic technologies for synthetic biology Int. J. 9 1230–5
Mol. Sci. 12 3576–93 [129] Kim H, Dixit S, Green C J and Faris G W 2009 Nanodroplet
[107] van Dongen S F M, de Hoog H P M, Peters R J R W, real-time PCR system with laser assisted heating Opt.
Nallani M, Nolte R J M and van Hest J C M 2009 Biohybrid Express 17 218–27
polymer capsules Chem. Rev. 109 6212–74 [130] Di Carlo D, Wu L Y and Lee L P 2006 Dynamic single cell
[108] Bysell H, Mansson R, Hansson P and Malmsten M 2011 culture array Lab Chip 6 1445–9
Microgels and microcapsules in peptide and protein drug [131] Srisa-Art M, deMello A J and Edel J B 2010
delivery Adv. Drug Delivery Rev. 63 1172–85 High-efficiency single-molecule detection within
[109] Rabanel J M, Banquy X, Zouaoui H, Mokhtar M and trapped aqueous microdroplets J. Phys. Chem. B
Hildgen P 2009 progress technology in microencapsulation 114 15766–72
methods for cell therapy Biotechnol. Prog. 25 946–63 [132] Bai Y P, He X M, Liu D S, Patil S N, Bratton D, Huebner A,
[110] Yin H and Marshall D 2012 Microfluidics for single cell Hollfelder F, Abell C and Huck W T S 2010 A double
analysis Curr. Opin. Biotechnol. 23 110–9 droplet trap system for studying mass transport across a
[111] Fritzsch F S, Dusny C, Frick O and Schmid A 2012 droplet–droplet interface Lab Chip 10 1281–5
Single-cell analysis in biotechnology, systems biology, [133] Huebner A M, Abell C, Huck W T, Baroud C N and
and biocatalysis Annu. Rev. Chem. Biomol. Eng. Hollfelder F 2011 Monitoring a reaction at
3 129–55 submillisecond resolution in picoliter volumes Anal. Chem.
[112] Iverson B D and Garimella S V 2008 Recent advances in 83 1462–8
microscale pumping technologies: a review and evaluation [134] Hatch A C, Fisher J S, Tovar A R, Hsieh A T, Lin R,
Microfluidics Nanofluidics 5 145–74 Pentoney S L, Yang D L and Lee A P 2011 1-Million
[113] Nisar A, Afzulpurkar N, Mahaisavariya B and Tuantranont A droplet array with wide-field fluorescence imaging for
2008 MEMS-based micropumps in drug delivery and digital PCR Lab Chip 11 3838–45
biomedical applications Sensors Actuators B 130 917–42 [135] Schmitz C H, Rowat A C, Koster S and Weitz D A 2009
[114] Link D R, Anna S L, Weitz D A and Stone H A 2004 Dropspots: a picoliter array in a microfluidic device Lab
Geometrically mediated breakup of drops in microfluidic Chip 9 44–9
devices Phys. Rev. Lett. 92 054503 [136] Edd J F, Humphry K J, Irimia D, Weitz D A and Toner M
[115] Leshansky A M and Pismen L M 2009 Breakup of drops 2009 Nucleation and solidification in static arrays of
in a microfluidic T junction Phys. Fluids monodisperse drops Lab Chip 9 1859–65
21 023303 [137] Taylor R J, Falconnet D, Niemisto A, Ramsey S A, Prinz S,
[116] Hatch A C, Fisher J S, Pentoney S L, Yang D L and Lee A P Shmulevich I, Galitski T and Hansen C L 2009 Dynamic
2011 Tunable 3D droplet self-assembly for analysis of MAPK signaling using a high-throughput
ultra-high-density digital micro-reactor arrays Lab Chip microfluidic single-cell imaging platform Proc. Natl Acad.
11 2509–17 Sci. USA 106 3758–63
[117] Carlson A, Do-Quang M and Amberg G 2010 Droplet [138] Falconnet D, Niemisto A, Taylor R J, Ricicova M, Galitski T,
dynamics in a bifurcating channel Int. J. Multiphase Flow Shmulevich I and Hansen C L 2011 High-throughput
36 397–405 tracking of single yeast cells in a microfluidic imaging
[118] Menetrier-Deremble L and Tabeling P 2006 Droplet matrix Lab Chip 11 466–73
breakup in microfluidic junctions of arbitrary angles Phys. [139] Gomez-Sjoberg R, Leyrat A A, Pirone D M, Chen C S and
Rev. E 74 035303 Quake S R 2007 Versatile, fully automated, microfluidic
[119] Lai A, Bremond N and Stone H A 2009 Separation-driven cell culture system Anal. Chem. 79 8557–63
coalescence of droplets: an analytical criterion for the [140] Yamada M, Nakashima M and Seki M 2004 Pinched flow
approach to contact J. Fluid Mech. 632 97–107 fractionation: Continuous size separation of particles

18
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

utilizing a laminar flow profile in a pinched microchannel [162] Liu C X, Stakenborg T, Peeters S and Lagae L 2009 Cell
Anal. Chem. 76 5465–71 manipulation with magnetic particles toward microfluidic
[141] Huang L R, Cox E C, Austin R H and Sturm J C 2004 cytometry J. Appl. Phys. 105 102014
Continuous particle separation through deterministic lateral [163] Petersson F, Nilsson A, Holm C, Jonsson H and Laurell T
displacement Science 304 987–90 2005 Continuous separation of lipid particles from
[142] Joensson H N, Uhlen M and Svahn H A 2011 Droplet size erythrocytes by means of laminar flow and acoustic
based separation by deterministic lateral standing wave forces Lab Chip 5 20–2
displacement-separating droplets by cell-induced shrinking [164] Bruus H 2011 Acoustofluidics 1: Governing equations in
Lab Chip 11 1305–10 microfluidics Lab Chip 11 3742–51
[143] Segré G and Silberberg A 1962 Behaviour of macroscopic [165] Petersson F, Aberg L, Sward-Nilsson A M and Laurell T
rigid spheres in Poiseuille flow: II. Experimental results and 2007 Free flow acoustophoresis: Microfluidic-based
interpretation J. Fluid Mech. 14 136 mode of particle and cell separation Anal. Chem.
[144] Di Carlo D 2009 Inertial microfluidics Lab Chip 9 3038–46 79 5117–23
[145] Di Carlo D, Edd J, Humphry K, Stone H and Toner M 2009 [166] Franke T, Braunmuller S, Schmid L, Wixforth A and
Particle segregation and dynamics in confined flows Phys. Weitz D A 2010 Surface acoustic wave actuated cell sorting
Rev. Lett. 102 094503 (SAWACS) Lab Chip 10 789–94
[146] Kuntaegowdanahalli S S, Bhagat A A, Kumar G and [167] Franke T, Abate A R, Weitz D A and Wixforth A 2009
Papautsky I 2009 Inertial microfluidics for continuous Surface acoustic wave (SAW) directed droplet flow in
particle separation in spiral microchannels Lab Chip microfluidics for PDMS devices Lab Chip 9 2625–7
9 2973–80 [168] Link D R, Grasland-Mongrain E, Duri A, Sarrazin F,
[147] Russom A, Gupta A K, Nagrath S, Di Carlo D, Edd J F and Cheng Z, Cristobal G, Marquez M and Weitz D A 2006
Toner M 2009 Differential inertial focusing of particles in Electric control of droplets in microfluidic devices Angew.
curved low-aspect-ratio microchannels New J. Phys. Chem. Int. Edn Engl. 45 2556–60
11 75025 [169] Pethig R 2010 Review Article-Dielectrophoresis: status of
[148] Di Carlo D, Edd J F, Irimia D, Tompkins R G and Toner M the theory, technology, and applications Biomicrofluidics
2008 Equilibrium separation and filtration of particles using 4 022811
differential inertial focusing Anal. Chem. 80 2204–11 [170] Baret J C et al 2009 Fluorescence-activated droplet sorting
[149] Oakey J, Applegate R W, Arellano E, Di Carlo D, (FADS): efficient microfluidic cell sorting based on
Graves S W and Toner M 2010 Particle focusing in staged enzymatic activity Lab Chip 9 1850–8
inertial microfluidic devices for flow cytometry Anal. Chem. [171] Hu X Y, Bessette P H, Qian J R, Meinhart C D,
82 3862–7 Daugherty P S and Soh H T 2005 Marker-specific sorting of
[150] Jonas A and Zemanek P 2008 Light at work: The use of rare cells using dielectrophoresis Proc. Natl Acad Sci. USA
optical forces for particle manipulation, sorting, and 102 15757–61
analysis Electrophoresis 29 4813–51 [172] Pregibon D C, Toner M and Doyle P S 2007 Multifunctional
[151] Ashkin A, Dziedzic J M, Bjorkholm J E and Chu S 1986 encoded particles for high-throughput biomolecule analysis
Observation of a single-beam gradient force optical trap for Science 315 1393–6
dielectric particles Opt. Lett. 11 288–90 [173] Bong K W, Chapin S C and Doyle P S 2010 Magnetic
[152] Lorenz R M, Edgar J S, Jeffries G D M and Chiu D T 2006 barcoded hydrogel microparticles for multiplexed detection
Microfluidic and optical systems for the on-demand Langmuir: ACS J. Surf. Colloids 26 8008–14
generation and manipulation of single femtoliter-volume [174] Ji X-H, Cheng W, Guo F, Liu W, Guo S-S, He Z-K and
aqueous droplets Anal. Chem. 78 6433–9 Zhao X-Z 2011 On-demand preparation of quantum
[153] Wang M M et al 2005 Microfluidic sorting of mammalian dot-encoded microparticles using a droplet microfluidic
cells by optical force switching Nature Biotechnol. system Lab Chip 11 2561
23 83–7 [175] Ji X-H, Zhang N-G, Cheng W, Guo F, Liu W, Guo S-S,
[154] Chen J, Ng J, Lin Z F and Chan C T 2011 Optical pulling He Z-K and Zhao X-Z 2011 Integrated parallel microfluidic
force Nature Photon. 5 531–4 device for simultaneous preparation of multiplex
[155] Zhang K, Liang Q, Ma S, Mu X, Hu P, Wang Y and Luo G optical-encoded microbeads with distinct quantum dot
2009 On-chip manipulation of continuous picoliter-volume barcodes J. Mater. Chem. 21 13380
superparamagnetic droplets using a magnetic force Lab [176] Zhao Y, Shum H C, Chen H, Adams L L, Gu Z and Weitz D A
Chip 9 2992–9 2011 Microfluidic generation of multifunctional quantum
[156] Lombardi D and Dittrich P S 2010 Droplet microfluidics with dot barcode particles J. Am. Chem. Soc. 133 8790–3
magnetic beads: a new tool to investigate drug–protein [177] Goldner L S, Jofre A M and Tang J Y 2010 Methods in
interactions Anal. Bioanal. Chem. 399 347–52 Enzymology, vol 472: Single Molecule Tools, A:
[157] Zhang D Y, Fakhrullin R F, Ozmen M, Wang H, Wang J, Fluorescence Based Approaches ed N G Walter (San Diego,
Paunov V N, Li G H and Huang W E 2011 CA: Elsevier Academic Press Inc) pp 61–88
Functionalization of whole-cell bacterial reporters with [178] Damean N, Olguin L F, Hollfelder F, Abell C and Huck W T
magnetic nanoparticles Microb. Biotechnol. 4 89–97 S 2009 Simultaneous measurement of reactions in
[158] Fakhrullin R F, Garcia-Alonso J and Paunov V N 2010 A microdroplets filled by concentration gradients Lab Chip
direct technique for preparation of magnetically 9 1707–13
functionalised living yeast cells Soft Matter 6 391–7 [179] Song H and Ismagilov R F 2003 Millisecond kinetics on a
[159] Kekarainen T, Mannelin S, Laine J and Jaatinen T 2006 microfluidic chip using nanoliters of reagents J. Am. Chem.
Optimization of immunomagnetic separation for cord Soc. 125 14613–9
blood-derived hematopoietic stem cells BMC Cell Biol. 7 30 [180] Ng K C, Heredia K H and Kliewer D 2012 Note: A method
[160] Pankhurst Q A, Connolly J, Jones S K and Dobson J 2003 to isolate and detect a large number of single molecules
Applications of magnetic nanoparticles in biomedicine by microdroplet fluorescence spectroscopy Rev. Sci.
J. Phys. D: Appl. Phys. 36 R167–81 Instrum. 83 036107
[161] Lombardi D and Dittrich P S 2011 Droplet microfluidics with [181] Srisa-Art M, Dyson E C, Demello A J and Edel J B 2008
magnetic beads: a new tool to investigate drug–protein Monitoring of real-time streptavidin-biotin binding kinetics
interactions Anal. Bioanal. Chem. 399 347–52 using droplet microfluidics Anal. Chem. 80 7063–7

19
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

[182] Dittrich P S, Jahnz M and Schwille P 2005 A new embedded [201] Lee H, Sun E, Ham D and Weissleder R 2008 Chip-NMR
process for compartmentalized cell-free protein expression biosensor for detection and molecular analysis of cells
and on-line detection in microfluidic devices Nature Med. 14 869–74
Chembiochem: Eur. J. Chem. Biol. 6 811–4 [202] Lee H, Yoon T J and Weissleder R 2009 Ultrasensitive
[183] Joensson H N, Zhang C, Uhlen M and Andersson-Svahn H detection of bacteria using core–shell nanoparticles and an
2012 A homogeneous assay for protein analysis in droplets NMR-filter system Angew. Chem. Int. Edn Engl.
by fluorescence polarization Electrophoresis 33 436–9 48 5657–60
[184] Fidalgo L M, Whyte G, Bratton D, Kaminski C F, Abell C [203] Haun J B, Castro C M, Wang R, Peterson V M,
and Huck W T S 2008 From microdroplets to microfluidics: Marinelli B S, Lee H and Weissleder R 2011 Micro-NMR
Selective emulsion separation in microfluidic devices for rapid molecular analysis of human tumor samples Sci.
Angew. Chem. Int. Edn Engl. 47 2042–5 Transl. Med. 3 71ra16
[185] Zhu Y and Fang Q 2010 Integrated droplet analysis system [204] Haun J B, Yoon T J, Lee H and Weissleder R 2010 Magnetic
with electrospray ionization-mass spectrometry using a nanoparticle biosensors Wiley Interdiscip. Rev. Nanomed.
hydrophilic tongue-based droplet extraction interface Anal. Nanobitechnol. 2 291–304
Chem. 82 8361–6 [205] Prabaharan M and Mano J F 2006 Stimuli-responsive
[186] Huang W E, Griffiths R I, Thompson I P, Bailey M J and hydrogels based on polysaccharides incorporated with
Whiteley A S 2004 Raman microscopic analysis of single thermo-responsive polymers as novel biomaterials
microbial cells Anal. Chem. 76 4452–8 Macromol. Biosci. 6 991–1008
[187] Li M, Xu J, Romero-Gonzalez M, Banwart S A and [206] Madsen J and Armes S P 2012 (Meth)acrylic
Huang W E 2012 Single cell Raman spectroscopy for stimulus-responsive block copolymer hydrogels Soft Matter
cell sorting and imaging Curr. Opin. Biotechnol. 8 592–605
23 56–63 [207] Salehpour S and Dube M A 2012 Application properties of
[188] Huang W E, Li M Q, Jarvis R M, Goodacre R and stimuli-responsive polyglycerol hydrogels J. Macromol.
Banwart S A 2010 Advances in Applied Microbiology Sci. A—Pure Appl. Chem. 49 103–10
vol 70, ed A I Laskin et al (San Diego, CA: Elsevier [208] Liu J W 2011 Oligonucleotide-functionalized hydrogels as
Academic Press Inc) pp 153–86 stimuli responsive materials and biosensors Soft Matter
[189] Lau A Y, Lee L P and Chan J W 2008 An integrated 7 6757–67
optofluidic platform for Raman-activated cell sorting Lab [209] Jiang K, Thomas P C, Forry S P, DeVoe D L and
Chip 8 1116–20 Raghavan S R 2012 Microfluidic synthesis of monodisperse
[190] Lee S E and Lee L P 2010 Biomolecular plasmonics for PDMS microbeads as discrete oxygen sensors Soft Matter
quantitative biology and nanomedicine Curr. Opin. 8 923
Biotechnol. 21 489–97 [210] Hofmann T W, Hanselmann S, Janiesch J W, Rademacher A
[191] Walter A, Marz A, Schumacher W, Rosch P and Popp J 2011 and Bohm C H 2012 Applying microdroplets as sensors for
Towards a fast, high specific and reliable discrimination of label-free detection of chemical reactions Lab Chip
bacteria on strain level by means of SERS in a microfluidic 12 916–22
device Lab Chip 11 1013–21 [211] Boedicker J Q, Li L, Kline T R and Ismagilov R F 2008
[192] Syme C D, Martino C, Yusvana R, Sirimuthu N M and Detecting bacteria and determining their susceptibility to
Cooper J M 2012 Quantitative characterization of individual antibiotics by stochastic confinement in nanoliter
microdroplets using surface-enhanced resonance Raman droplets using plug-based microfluidics Lab Chip
scattering spectroscopy Anal. Chem. 84 1491–5 8 1265–72
[193] Cecchini M P, Hong J, Lim C, Choo J, Albrecht T, [212] Joensson H N, Samuels M L, Brouzes E R, Medkova M,
Demello A J and Edel J B 2011 Ultrafast surface enhanced Uhlen M, Link D R and Andersson-Svahn H 2009
resonance Raman scattering detection in droplet-based Detection and analysis of low-abundance cell-surface
microfluidic systems Anal. Chem. 83 3076–81 biomarkers using enzymatic amplification in
[194] Ernst A, Streule W, Schmitt N, Zengerle R and Koltay P 2009 microfluidic droplets Angew. Chem. Int. Edn Engl.
A capacitive sensor for non-contact nanoliter droplet 48 2518–21
detection Sensors Actuators A 153 57–63 [213] Kumaresan P, Yang C J, Cronier S A, Blazej R G and
[195] Elbuken C, Glawdel T, Chan D and Ren C L 2011 Detection Mathies R A 2008 High-throughput single copy DNA
of microdroplet size and speed using capacitive sensors amplification and cell analysis in engineered nanoliter
Sensors Actuators A 171 55–62 droplets Anal. Chem. 80 3522–9
[196] Cahill B P, Land R, Nacke T, Min M and Beckmann D 2011 [214] Guo M T, Rotem A, Heyman J A and Weitz D A 2012
Contactless sensing of the conductivity of aqueous droplets Droplet microfluidics for high-throughput biological assays
in segmented flow Sensors Actuators B 159 286–93 Lab Chip 12 2146–55
[197] Moiseeva E V, Fletcher A A and Harnett C K 2011 Thin-film [215] Boedicker J Q, Vincent M E and Ismagilov R F 2009
electrode based droplet detection for microfluidic systems Microfluidic confinement of single cells of bacteria in small
Sensors Actuators B 155 408–14 volumes initiates high-density behavior of quorum sensing
[198] Sadeghi S, Ding H, Shah G J, Chen S, Keng P Y, Kim C-J C and growth and reveals its variability Angew. Chem. Int.
and van Dam R M 2012 On Chip droplet characterization: Edn Engl. 48 5908–11
a practical, high-sensitivity measurement of droplet [216] Lecault V et al 2011 High-throughput analysis of single
impedance in digital microfluidics Anal. Chem. hematopoietic stem cell proliferation in microfluidic cell
84 1915–23 culture arrays Nature Methods 8 581–6
[199] Kautz R A, Goetzinger W K and Karger B L 2005 [217] Fallah-Araghi A, Baret J C, Ryckelynck M and Griffiths A D
High-throughput microcoil NMR of compound libraries 2012 A completely in vitro ultrahigh-throughput
using zero-dispersion segmented flow analysis J. Comb. droplet-based microfluidic screening system for protein
Chem. 7 14–20 engineering and directed evolution Lab Chip 12 882–91
[200] Lin Y Q, Schiavo S, Orjala J, Vouros P and Kautz R 2008 [218] Zhang H, Jenkins G, Zou Y, Zhu Z and Yang C J 2012
Microscale LC-MS-NMR platform applied to the Massively parallel single-molecule and single-cell emulsion
identification of active cyanobacterial metabolites Anal. reverse transcription polymerase chain reaction using
Chem. 80 8045–54 agarose droplet microfluidics Anal. Chem. 84 3599–606

20
J. Phys. D: Appl. Phys. 46 (2013) 114005 T P Lagus and J F Edd

[219] Zhang W Y, Zhang W, Liu Z, Li C, Zhu Z and Yang C J 2012 [224] Gossett D R, Weaver W M, Mach A J, Hur S C, Tse H T K,
Highly parallel single-molecule amplification approach Lee W, Amini H and Di Carlo D 2010 Label-free cell
based on agarose droplet polymerase chain reaction for separation and sorting in microfluidic systems Anal.
efficient and cost-effective aptamer selection Anal. Chem. Bioanal. Chem. 397 3249–67
84 350–5 [225] Boitard L, Cottinet D, Kleinschmitt C, Bremond N, Baudry J,
[220] Mastrobattista E, Taly V, Chanudet E, Treacy P, Kelly B T Yvert G and Bibette J 2012 Monitoring single-cell
and Griffiths A D 2005 High-throughput screening of bioenergetics via the coarsening of emulsion droplets Proc.
enzyme libraries: In vitro evolution of a beta-galactosidase Natl Acad. Sci USA 109 7181–6
by fluorescence-activated sorting of double emulsions [226] Jimenez A M, Roche M, Pinot M, Panizza P, Courbin L and
Chem. Biol. 12 1291–300 Gueroui Z 2011 Towards high throughput production of
[221] Bernath K, Hai M T, Mastrobattista E, Griffiths A D, artificial egg oocytes using microfluidics Lab Chip
Magdassi S and Tawfik D S 2004 In vitro 11 429–34
compartmentalization by double emulsions: sorting and [227] Bourouina N, Husson J, Hivroz C and Henry N 2012
gene enrichment by fluorescence activated cell sorting Anal. Biomimetic droplets for artificial engagement of living cell
Biochem. 325 151–7 surface receptors: the specific case of the T-Cell Langmuir:
[222] Nagrath S et al 2007 Isolation of rare circulating tumour cells ACS J. Surf. Colloids 28 6106–13
in cancer patients by microchip technology Nature [228] Zagnoni M 2012 Miniaturised technologies for the
450 1235–9 development of artificial lipid bilayer systems Lab Chip
[223] Boyum A 1984 Separation of lymphocytes, granulocytes, and 12 1026–39
monocytes from human-blood using iodinated density
gradient media Method Enzymol. 108 88–102

21

You might also like