SlideShare a Scribd company logo
1 of 10
Download to read offline
Microfluidic single cell analysis: from promise to practice
Ve´ ronique Lecault1,2,3,5
, Adam K White1,5
, Anupam Singhal1,3,5
and
Carl L Hansen1,4
Methods for single-cell analysis are critical to revealing cell-to-
cell variability in biological systems, especially in cases where
relevant minority cell populations can be obscured by
population-averaged measurements. However, to date single
cell studies have been limited by the cost and throughput
required to examine large numbers of cells and the difficulties
associated with analyzing small amounts of starting material.
Microfluidic approaches are well suited to resolving these
issues by providing increased senstitivity, economy of scale,
and automation. After many years of development microfluidic
systems are now finding traction in a variety of single-cell
analytics including gene expression measurements, protein
analysis, signaling response, and growth dynamics. With newly
developed tools now being applied in fields ranging from
human haplotyping and drug discovery to stem cell and cancer
research, the long-heralded promise of microfluidic single cell
analysis is now finally being realized.
Addresses
1
Centre for High-Throughput Biology, University of British Columbia,
2185 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada
2
Michael Smith Laboratories, University of British Columbia, 2125 East
Mall, Vancouver, British Columbia, V6T 1Z4, Canada
3
Department of Chemical and Biological Engineering, University of
British Columbia, 2360 East Mall, Vancouver, British Columbia, V6T 1Z3,
Canada
4
Department of Physics and Astronomy, University of British Columbia,
6224 Agricultural Road, East Mall, Vancouver, British Columbia, V6T
1Z1, Canada
Corresponding author: Hansen,
Carl L (chansen@physics.ubc.ca, chansen@phas.ubc.ca)
5
Authors contributed equally to this work.
Current Opinion in Chemical Biology 2012, 16:381–390
This review comes from a themed issue on Analytical techniques
Edited by Shana O Kelley and Petra S Dittrich
For a complete overview see the Issue and the Editorial
Available online 21st April 2012
1367-5931/$ – see front matter, # 2012 Elsevier Ltd. All rights
reserved.
http://dx.doi.org/10.1016/j.cbpa.2012.03.022
Introduction
Cells are the fundamental units of biological processes.
However, the vast majority of our understanding of bio-
chemistry and genetics has been gleaned through the bulk
analysis of large populations of cells, which are required to
obtain sufficient starting material for conventional analysis
methods. Interpretation of such data often implies the tacit
assumption that each cell in the population is similar. More
often than notthisisfalse.Cellular heterogeneityisinfacta
generalfeatureofbiologicalsystemsandhasbeenobserved
across all levels of life, from single bacterial cells to human
tissues. Even cells with identical functions generally
respondasynchronously, making precise studies of kinetics
and dynamics of cell populations impossible. Moreover, in
many important fields, minority subpopulations of cells are
often the most relevant. For instance, in microbial geno-
mics it is frequently impossible to selectively isolate or
culture a particular microbial species, thus necessitating
the study of DNA shrapnel derived from a mixture of
organisms. Similarly, in stem cell science even the most
advanced isolation methods can only provide enriched
populations of stem cells. In the best case, murine hema-
topoietic stem cells, functional purities are generally below
50% [1], and in many other systems they are much lower.
Thus, bulk measurements of the molecular signatures of
these important cells are obscured by significant, and often
overwhelming, contamination from other cell types of
unknown state and relative abundance. This scenario is
paralleled in numerous fields of research including cancer,
immunology and developmental biology. Understanding
the extent and importance of cellular heterogeneity is one
of the most vexing problems facing biological research.
The challenge of understanding cellular heterogeneity
has been a major thrust of technological development
over the past decade, resulting in an increasingly powerful
suite of instrumentation, protocols, and methods for ana-
lyzing single cells at the level of DNA sequence, RNA
expression and protein abundance [2–4]. Flow cytometry
and cell sorting have been widely adopted as a corner-
stone of cellular phenotyping and purification, allowing
for high-throughput quantitative analysis of protein
expression and phosphorylation state in single cells [5–
7]. Recent advances in coupling this technology with
metal labeling have now extended the analysis of fixed
cells to sensitive multiplexing of dozens of targets per run
[8]. At the same time, increasingly rapid and sensitive
imaging instrumentation now allows high-throughput
single cell measurements of proteins and nucleic acids
with single molecule resolution [9], and the precise
tracking of cellular growth and responses over extended
periods [10–13]. Sensitive PCR-based protocols for
measuring large panels of mRNA from single cells are
now well established and have been coupled to FACS
isolation and robotic assay assembly [14,15
,16–19]. At
the same time, the rise of high-throughput sequencing
Available online at www.sciencedirect.com
www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
instrumentation [20] has now made single-cell genomics a
practical proposition [21,22] and increasingly focus has
turned to the development of appropriate protocols for
high-fidelity amplification of DNA and RNA from single
cells [23–25]. Sequencing approaches have further been
coupled with virus-based molecular barcoding strategies
to dissect heterogeneity in cellular processes in vivo [26].
Despite this suite of available approaches we are still only
beginning to face the measurement challenges of cellular
heterogeneity. There is much work to be done, both in
enabling new modes of analysis, and in improving the
speed, throughput, and economy of those that exist.
Although each measurement application carries its own
specific requirements, the recurring challenge in single-
cell analysis is to simultaneously achieve the sensitivity,
precision, throughput, and economy needed to detect and
study complex subpopulations of cells. Microfluidic for-
mats are ideally suited to addressing these problems by
providing reduced reagent costs, high effective template
concentrations in small volumes, scalability, ease of auto-
mation, improved cell handling, and multi-step integration
(Table 1). Over a decade of interdisciplinary research and
development, spanning fields of engineering, molecular
biology, imaging, cell biology, and chemistry, has now
brought microfluidic single cell analysis to a turning point,
with technologies increasingly being adopted in biological
andbiomedicalresearch[27,28].Herewereviewhowthese
approaches are pushing new avenues of research and high-
light the next generation of advances that seem poised to
transition from the bread-board to the bench-top.
Microfluidic approaches to high-throughput
single-cell RT-qPCR
Reverse transcription quantitative PCR (RT-qPCR) pro-
vides both excellent specificity and sensitivity, making it
well suited to gene expression measurements in single
cells. New preamplification strategies [29] have enabled
RT-qPCR measurements, which were typically per-
formed on a modest numbers of target genes [30,31],
to be multiplexed for the analysis of many dozens of
transcripts from a single cell [32,33]. However, appli-
cation of this approach to the analysis of large numbers
of single cells creates a ‘tyranny of numbers’ with serious
challenges in both throughput and cost. A commercially
available valve-based microfluidic qPCR system, the
Dynamic ArrayTM
(Fluidigm), provides a low-volume
(nanoliter) and high-throughput (thousands of PCR reac-
tions per device) solution to this detection problem and
has become increasingly popular for large-scale single cell
studies. For instance, Guo et al. used microfluidic qPCR
arrays to measure the expression dynamics of 48 genes
over 500 individual cells harvested during the first four
days of mouse embryo development [17]. Correlation of
expression measurements with surface markers revealed
that co-expression of lineage-specific transcription factors
occurred at the 32-cell stage of development, but that
three distinct transcriptional programs emerged at the 64-
cell stage. Scalable single cell expression measurements
have also been used to provide new insights into cellular
heterogeneity that exists within rare populations isolated
from different developmental stages and across hierar-
chies of differentiation, both at the level of transcript and
miRNA expression [18,19,34,35]. Of particular interest
has been the implication of cellular heterogeneity within
cancer, both in terms of disease progression and optimal
treatment strategies [36,37]. Dalerba et al. identified dis-
tinct single cell gene expression signatures that are pre-
dictive of patient survival and clinical outcomes in colon
cancer patients [15
]. Diehn et al. used single-cell
analysis to identify a radioresistant subset of breast tumor
cells with increased capacity for reactive oxygen species
scavenging [16].
382 Analytical techniques
Table 1
Advantages of microfluidics for single cell analysis.
Application Challenges with traditional methods Advantages of microfluidics Examples
Single-cell RT-qPCR Limited abundance of starting template Concentration enhancement in small volumes [38
,40]
Cost and throughput required for analysis
of large numbers of cells and target genes
Parallelization, automation, and economy of scale [17,38
]
Single-cell genomics Amplification bias and sensitivity Improved reaction bias and sensitivity in nL volumes
and reduced contaminant DNA
[47]
Isolating individual cells Integrated microfluidic cell sorting and processing [48]
Single-cell measurements
of intracellular proteins
Movement of living cells Confinement of live cells in microfluidic structures [50
]
Low amount of signal Integrated single-cell handling allows lysate analysis [54,55
]
Single-cell measurements
of secreted proteins
Small amounts of secreted products from
single cells
Concentration enhancement in small volumes [57,58]
Difficult to co-localize multiple cells in
defined chemical environments
Ease of confinement in droplets or microchambers [62]
Signaling studies Mostly limited to static conditions Easy temporal stimulation [67,70
,71
]
Inability to rapidly exchange conditions
on suspension cells
Laminar flow and proper design enables cell
sequestration
[68,70
]
Live cell imaging Difficulties of tracking cells through
multiple frames
Confinement of clones facilitates cell tracking [76]
Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
The examples above show how microfluidics can provide
scalability, reduced reagent consumption, and throughput
to enable large single cell studies that would otherwise be
impractical or prohibitively expensive. However, these
analyses still require off-chip cell handling and processing
steps to generate products needed for qPCR. The
inclusion of microfluidic cell handling and processing
thus offers important avenues to improved throughput
and cost, while also improving precision and sensitivity
through small-volume confinement. To this end White
et al. recently described a fully integrated microfluidic
RT-qPCR device that implements all steps of cell trap-
ping, lysis, cDNA synthesis, and qPCR analysis (Figure 1)
at a throughput of 300 cells per run [38
]. This system
achieves improved precision and sensitivity over large-
volume RT-qPCR analysis and offers a general solution
for cell-handling automation and integration that may be
adapted to a variety of amplification and analysis protocols
[39], including high-throughput microfluidic qPCR, high-
density digital PCR [40,41], sequencing, and microarray
analysis.
Single cell genomes
In addition to gene expression analysis, microfluidic
approaches are finding increasing applications in studying
diversity and variations in single cell genomes, with
applications spanning cancer biology to environmental
microbiology. While new high-throughput sequencing
instrumentation has made exome and whole-genome
shotgun sequencing standard practice in cancer research,
attention has now turned towards dissecting clonal
heterogeneity. In one approach the sequencing of ampli-
fied single nuclei was used to infer the clonal evolution of
a breast tumor by measuring single cell copy number
variations [24]. Scalable and low-cost implementations of
single cell/nuclei isolation and amplification are likely to
expand the power and pervasiveness of such analyses. In
this line, Fan et al. recently reported a haplotyping
approach on the basis of using a microfluidic device to
amplify and genotype individual chromosomes isolated
from a single cell [42
]. The isolation and lysis of a single
metaphase cell was used to distribute chromosomes
across an array of 20 nL reaction chambers, followed by
phi29 polymerase multiple displacement amplification
[43], recovery of the isolated amplified products, and
analysis on SNP arrays to establish haplotypes [42].
Beyond the utility in phasing genomes, more scalable
implementations of this technology may soon allow for
the study of heterogeneity in chromosome partitioning
and translocation events in cancer.
Single cell genome analysis is particularly compelling in
the study of microbial organisms and communities. The
vast majority of microorganisms on the planet have yet to
be isolated in culture, necessitating metagenomic strat-
egies that attempt to infer the identity and relative
abundance of constituent members by sequencing and
analysis of mixed pools of DNA. Microfluidic digital PCR
in valve-based devices has been used to help untangle
this genomic information by co-amplification of specific
genes in isolated individual bacteria cells. This has been
applied to establish co-existence of functional genes
within a single organism and to reveal virus–host relation-
ships in complex environments [44,45]. In a similar single
cell genotyping application, microfluidic droplet-based
systems for digital PCR have been used for the detection
Microfluidic single cell analysis Lecault et al. 383
Figure 1
i
ii
iv
iii
(a) (b) (c)
Current Opinion in Chemical Biology
A microfluidic device for high-throughput single-cell RT-qPCR. (a) Fluorescence image of entire device showing 300 reactions after 40 cycles of PCR.
(b) Enlarged view of individual reactors from (a) with dyes highlighting fluid paths (blue) and valves (red). Each array unit consists of (i) a reagent inlet, (ii)
a 0.6 nL cell capture region with integrated cell traps, (iii) a 10 nL RT chamber and (iv) a 50 nL PCR chamber. Scale bar: 400 mm. (c) Optical micrograph
of single cells trapped in the cell capture chambers (indicated by black arrows). Scale bar, 400 mm.
Adapted from [38
].
www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
of pathogenic E. coli within a high background of non-
pathogenic cells, achieving a detection limit of 1/105
[46].
The coupling of new low-bias whole genome amplification
strategies [43] with sequencing provides a powerful
approach to global single cell genome analysis and has
been implemented in conventional tubes using single cell
isolation by FACS or micropipette [23]. Microfluidic sys-
tems offer an integrated ‘front-end’ solution to bacterial
imaging, isolation, and processing, and have been shown to
improve MDA performance by providing reduced ampli-
fication bias in nanoliter volume reactions and suppression
of contaminating DNA [23]. Although the physical separ-
ation of single cells remains an important obstacle for many
complex samples, this approach has been applied to obtain
highly enriched metagenomes for T7 microbes from the
human mouth [47], and has been further integrated with
optical tweezer manipulation to obtain a novel genome for
an ammonia-oxidizing archaeon [48]. Leung et al. have
recently developed a programmable droplet-based format
for single microbe isolation and multiparameter single-cell
analysis [49]. This system provides improvements in-
cluding facilitated single cell handling, flexible protocol
development, and significantly higher throughput, and was
used to dissect microbial diversity in environmental
samples by whole genome amplification and sequencing
of single cells and cell aggregates.
Microfluidic analysis of proteins in single cells
Beyond genomics applications, the scalability and small-
volume advantages of microfluidic methods have increas-
ingly found applications in the measurement of intra-
cellular and secreted proteins from single cells. Taniguchi
et al. used single molecule imaging in a parallel micro-
fluidic format to measure intracellular protein expression,
localization and abundance in a library of over 1018
Escherichia coli fluorescent protein fusion strains [50
]
(Figure 2a–c). Protein abundances were found to vary
from 0.1 to 104
molecules per cell, spanning a dynamic
range of over 5 orders of magnitude. At low copy numbers
(10), differences in protein expression were attributed
to intrinsic noise (e.g. stochastic binding of transcription
factors to DNA promoter sites), whereas variations in
higher expressed proteins, including almost all essential
proteins, was generally governed by extrinsic fluctuations
in cellular metabolites, ribosomes, and polymerases.
Interestingly, for any given gene, a single cell’s protein
and mRNA abundance were found to be uncorrelated,
likely reflecting the relatively rapid degradation of
mRNA compared to the long lifetimes of proteins in
the cell. In contrast, Cheong and colleagues exploited
the parallelization of microfluidic culture to reconstruct
signaling kinetics using endpoint staining of fixed cells at
different time points after stimulation, thereby enabling
the detection of multiple proteins on each cell [51]. With
384 Analytical techniques
Figure 2
Stage scan
PDMS
Bacteria
Coverslip
Objective lens
Strain n-1
Strain n+1
Strain n
25 μm
10 μm
Hybridoma
cellAntibody
capture beads
10 μm
150 μm
(a)
Side view
(b) (c)
(e)
Adk
Probability
NormalizedFluorescence
Protein copy number
Time (min)
(d)
0.1
0.08
0.06
0.04
0.02
0
1.2
1
0.8
0.6
0.4
0.2
0 20 40 60 80 100 120
0 500 1000 1500 2000
a = 6.8
b = 99
Current Opinion in Chemical Biology
Microfluidic analysis of single cell intracellular (a)–(c) and extracellular (d) and (e) protein expression. (a) Microfluidic device for the parallel molecular
imaging of multiple bacterial strains with single protein resolution. (b) High-resolution fluorescence imaging to quantify protein abundance in single E.
coli cells using genetically fused fluorescent reporter strains. (c) Distribution of intracellular Adk protein in single E. coli cells. (d) A microfluidic
fluorescence bead assay to measure antibody–antigen binding kinetics from antibodies secreted by a single cell. (e) Binding kinetics of a mouse mAb
with hen egg lysozyme measured from a single bead.(a)–(c) Adapted from [50
] with permission. (d) and (e) adapted from [59].
Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
this method, it was shown that JNK signaling exhibited a
binary switch-like response after anisomycin stimulation
in HeLa cells [52]. Microfluidic-based immunocyto-
chemistry has also been used to assess intratumoral
and intertumoral heterogeneity in brain tumor samples,
showing that cluster analysis of four intracellular sig-
naling proteins could predict tumor progression and
survival outcomes [53]. Single-cell protein measure-
ments have also been performed by integrating micro-
fluidic single-cell handling (i.e. sorting, lysis, labeling)
with electrophoretic separation [54] or micro-patterned
antibody capture arrays [55
] In the latter approach,
protein expression variability in a glioblastoma tumor
cell line was measured using a quantitative fluorescent
sandwich assay on multiple target proteins per cell [55
].
This single-cell barcode chip (SCBC) approach is scal-
able and has already been demonstrated at a throughput
of tens of cells per chip with duplicate measurements of
nine proteins.
The concentration enhancement and rapid diffusive mix-
ing afforded by subnanoliter microfluidic chambers has
enabled the single-cell analysis of secreted effector
proteins from immune cells (e.g. B cells, T cells, and
macrophage). Ma et al. applied the SCBC to simul-
taneously measure multiple cytokines (e.g. IL-10,
TNF-b, IFN-g) from human macrophages and cytotoxic
T lymphocytes (CTLs) obtained from both healthy
donors and a metastatic melanoma patient [56]. Although
not strictly microfluidic, open arrays of microfabricated
chambers have also been used to screen and select B cells
secreting antigen-specific antibodies from both immu-
nized humans and mice [57,58]. This approach can be
improved by using microfluidics to automate cell hand-
ling and fluid-exchange, allowing antibodies from single
cells to be directly screened by measuring antibody-
antigen binding kinetics and specificities to different
target antigens [59] (Figure 2d,e).
An alternative approach uses microfluidic devices for the
high-throughput analysis of secreted proteins from single
cells by encapsulating them in emulsions consisting of
subnanoliter aqueous droplets in oil [60,61]. Tumarkin
and colleagues used a variant of this approach to study
cellular paracrine signaling by co-encapsulating cells in
microfluidic-generated agarose beads, and demonstrating
that cell survival could be modulated by the ratio of
MBA2 IL-3 secreting cells to M07e factor-dependent
cells [62]. Microfluidic droplet generation is also being
developed for drug screening applications by measuring
viability of encapsulated single cells exposed to different
chemical compositions [63
].
Single cell growth and response phenotypes
In addition to facilitating multiplexed genetic and protein
analysis of single cells, microfluidic devices provide
numerous advantages to the study of live cells such as
precise spatio-temporal control of medium conditions,
parallelization, and cell confinement for improved ima-
ging. These capabilities are particularly well suited to
measuring the kinetics and cellular heterogeneity of cell
signaling. Because of the ease of genetic manipulation
and cell culture, microfluidic approaches have been used
extensively to study the response of archetypical protein
signaling networks in yeast, most notably the pheromone
and HOG mitogen activated protein kinase (MAPK)
networks. The combination of microfluidic flow control,
precise cell immobilization, fluorescent protein reporters,
and image processing has been used to access several
experimental regimes that are difficult or impossible to
implement in bulk: the study of cell signaling under
stable and precisely defined spatial gradients [64]; the
frequency analysis of signaling response under oscillating
stimulation conditions [65,66]; and the high-throughput
analysis of network response under combined chemical
and genetic perturbations [67,68]. Importantly, exper-
iments that track individual cells through time are critical
to understanding mechanisms of response heterogeneity,
including effects of cell cycle, cell age, and familial
relationships. For instance, Falconnet et al. found that
nonuniform gene expression and phenotypic responses
observed at intermediate concentrations of pheromone
were not random, but rather correlated with genealogical
relationships (Figure 3a), illustrating how nongenetic
heritable traits may influence the critical threshold for
cellular decision making [68].
In a similar vein, microfluidic approaches are becoming
increasingly important as an in vitro method for studying
complex and highly dynamic interactions that govern
growth and differentiation of mammalian cells in vivo.
These approaches have used both surface patterning of
microwells to more precisely dissect and mimic factors
present in the in vivo microenvironment [11,69], and
automated fluid control to examine temporally varying
medium conditions [70
,71
]. Tay and colleagues
exploited the later approach to demonstrate that single
3T3 mouse fibroblast cells respond to tumor necrosis
factor alpha (TNF-a) in a digital manner; that is, lower
numbers of cells responded to lower doses of TNF-a
even though the amplitude of transcription factor nuclear
factor (NF-kB) remained high [71
]. Pulse stimulation
studies showed that prior exposure to TNF-a played a
role in subsequent cellular responses, indicating that this
pathway is not a purely stochastic system. In addition to
deterministic control of applied factors, Molidena and
colleagues reported the use of varying microfluidic flows
to modulate the effect of endogenously secreted factors,
showing that endocrine signaling is an important deter-
minant of differentiation and cellular heterogeneity in
murine embryonic stem cells [72
].
Nonadherent cell types, including hematopoietic cells
and industrially important suspension-adapted cell lines,
Microfluidic single cell analysis Lecault et al. 385
www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
present unique challenges for microfluidic analysis due to
the need for immobilization during medium exchange.
Lecault et al. reported a cell culture device optimized for
suspension cell types that solves this problem by using
gravity to drop cells into the bottom of high aspect ratio
chambers (Figure 3b,c), thereby sequestering them from
flow forces while maintaining the ability to exchange
medium conditions using diffusion [70
]. In addition, this
device featured a thin cell-culture layer that was overlaid
with a large medium reservoir, essentially blocking dehy-
dration effects and allowing for robust growth and the
selective recovery of viable cells following culture. The
ability to perform immunohistochemistry on live cells
without disturbing their spatial locations was used to
correlate heterogeneous clonal growth of preleukemic
cells with varying differentiation state. In a separate
study, exposure of rare primary hematopoietic stem cells
to temporally varying Steel factor (SF) concentrations
revealed that this growth factor is critical for cell survival
during a short window as cells exit quiescence, but does
not directly influence cell growth kinetics.
Single cell growth analysis
Microfluidic systems greatly facilitate long-term time-
lapse imaging studies of clonal growth and death kinetics
under different medium conditions. This is perhaps one
of the simplest and most underexploited assays of cellular
heterogeneity. For instance, the early division kinetics of
primary murine HSCs transduced with NUP98-HOXA10
homeodomain (NA10hd), a fusion gene known to stimu-
late stem cell expansion in vitro, was investigated using a
microfluidic cell culture array [73]. Clonal analysis of
infected and control populations has confirmed that
NA10hd did not affect cell cycling times, but that the
growth advantage of overexpressing cells was due to an
increase in self-renewal. In a different study, Albrecht
et al. used the expression of H2B-EGFP to facilitate
tracking of mitotic events in murine embryonic stem
cells and reported an apparent synchronization of division
events across the microfluidic device [74].
In addition to quantitative measures of clonal growth and
death,genealogicalanalysis,allowingfor thereconstruction
386 Analytical techniques
Figure 3
MAPK pathway GFP response320 min α-factor stimulation40 min α-factor stimulation
(a)
(b) (c)
Current Opinion in Chemical Biology
Microfluidic cell culture devices for live analysis of clonal heterogeneity. (a) Example of heterogeneous MAPK responses to alpha-factor pheromone
being passed on from mother to daughter yeast cells. (b) Microfluidic chambers with high-aspect ratio for the culture of mammalian suspension cells.
(c) Microfluidic cell culture array and image analysis show intraclonal heterogeneity in hematopoietic progenitor NUP98-HOXD13 cells. Green labels
represent viable cells, red labels represent dead cells and blue lines highlight cell contours. Scale bar, 100 mm.
(a) Adapted from [68]. Reproduced by permission of the Royal Society of Chemistry (RSC). (b) Reproduced from [70
]. (c) Adapted from [70
].
Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
of the ‘family tree’ in each clone, provides a rich phe-
notype for understanding cellular decision-making and
retrospectively assessing the heterogeneity of the starting
cell populations. Scherf and colleagues combined genea-
logical tracing and surface patterning in a microfluidic
device to show that the symmetry of human stem and
progenitor cell divisions could be altered by exposure to
exogenous factors [75]. The generation of division pedi-
grees from a large number of single cells is assisted by
microfluidic designs that confine clones in a limited space.
In one example, microfluidic chambers were used to grow
yeast colonies in linear geometries in order to facilitate
the assignment of mother-daughter relationships, showing
that observed bursts of heat shock protein expression are
synchronized between a cell and its immediate progeny
[76]. As a high-precision alternative to optical analysis of
cellular growth, the Manalis group have developed an
innovative approach that directly measures the mass of
single cells using microfabricated cantilevers with inte-
grated flow channels. This technique was used to show
that heavier cells grew faster than lighter cells [77], and that
cellular density could be used to distinguish subpopu-
lations of blood cells [78]. Although this approach has
not yet been adapted to high-throughput formats, it pro-
vides an excellent illustration of how microfluidic
approaches can enable previously inaccessible, and often
even unexpected, measurement tools for understanding
cellular heterogeneity.
Future impact of microfluidic technologies
Although microfluidic analysis methods have long been
a focus of technology development, it is only recently
that these tools are impacting biological and biomedical
research. These are auspicious times. The pace of de-
velopment and adoption is accelerating rapidly and the
next few years will see many important technological
developments and applications. Microfluidic cell proces-
sing will emerge as a cornerstone of single cell genomics,
enabling routine and scalable measurements of tran-
scription and DNA sequence with improved perform-
ance and economy. Although global proteomic analysis
of single cells is still far away, requiring major advances
in detection sensitivity and instrumentation, measure-
ments of specific protein panels using either antibody
capture or alternative specific reagents will continue to
develop and should enable new avenues of inquiry
ranging from immune response to cancer biology. Sim-
ilarly, the profiling of metabolites in single cells con-
stitutes an important area for continued advancement
[79]. Another exciting prospect for dissecting complex
cell populations is the integration of live-cell imaging
and genealogical analysis with transcriptional and
protein expression measurements, and the correlation
of these with functional assays. While these microfluidic
tools will be driven in the near term by interdisciplinary
research teams, broad adoption and long-term impact
will require increasing commercialization activities.
New developments in programmable microfluidic
devices will also be important in providing a general
fluid-handling platform that may be adapted to niche or
user-specific applications [49,80,81]. Given the rapid
pace of development, the enormous impact of single
cell approaches and the inherent advantages of micro-
fluidic analysis, it would seem that answers to many of
the biggest questions in single cell variability may very
well come on a tiny chip.
Acknowledgements
The authors would like to thank Marketa Ricikova for providing the original
images for Figure 3a. Funding support has been provided by grants from the
Natural Sciences and Engineering Research Council (NSERC), Genome
BC, Genome Canada, Western Diversification, the Terry Fox Foundation,
and the Canadian Institute for Health Research (CIHR). The authors also
thank the Michael Smith Foundation for Health Research (VL, AS, AKW,
CLH), NSERC (VL, AS, AKW), and CIHR (CLH) for salary support.
References and recommended reading
Papers of particular interest, published within the period of review,
have been highlighted as:
 of special interest
 of outstanding interest
1. Kent DG, Copley MR, Benz C, Wohrer S, Dykstra BJ, Ma E,
Cheyne J, Zhao YJ, Bowie MB, Zhao Y et al.: Prospective
isolation and molecular characterization of hematopoietic
stem cells with durable self-renewal potential. Blood 2009,
113:6342-6350.
2. Wu M, Singh AK: Single-cell protein analysis. Curr Opin
Biotechnol 2012, 23:83-88.
3. Wang D, Bodovitz S: Single cell analysis: the new frontier in
‘omics’. Trends Biotechnol 2010, 28:281-290.
4. Kalisky T, Blainey P, Quake SR: Genomic analysis at the single-
cell level. Annu Rev Genet 2011, 45:431-445.
5. Tarnok A, Ulrich H, Bocsi J: Phenotypes of stem cells from
diverse origin. Cytometry A 2010, 77A:6-10.
6. Tracy BP, Gaida SM, Papoutsakis ET: Flow cytometry for
bacteria: enabling metabolic engineering, synthetic biology
and the elucidation of complex phenotypes. Curr Opin
Biotechnol 2010, 21:85-99.
7. Krutzik PO, Irish JM, Nolan GP, Perez OD: Analysis of protein
phosphorylation and cellular signaling events by flow
cytometry: techniques and clinical applications. Clin Immunol
2004, 110:206-221.
8. Bendall SC, Simonds EF, Qiu P, Amir E-aD, Krutzik PO, Finck R,
Bruggner RV, Melamed R, Trejo A, Ornatsky OI et al.: Single-cell
mass cytometry of differential immune and drug responses
across a human hematopoietic continuum. Science 2011,
332:687-696.
9. Raj A, van den Bogaard P, Rifkin SA, van Oudenaarden A, Tyagi S:
Imaging individual mRNA molecules using multiple singly
labeled probes. Nat Methods 2008, 5:877-879.
10. Rieger MA, Hoppe PS, Smejkal BM, Eitelhuber AC, Schroeder T:
Hematopoietic cytokines can instruct lineage choice. Science
2009, 325:217-218.
11. Lutolf MP, Doyonnas R, Havenstrite K, Koleckar K, Blau HM:
Perturbation of single hematopoietic stem cell fates in
artificial niches. Integr Biol 2009, 1:59-69.
12. Dykstra B, Ramunas J, Kent D, McCaffrey L, Szumsky E, Kelly L,
Farn K, Blaylock A, Eaves C, Jervis E: High-resolution video
monitoring of hematopoietic stem cells cultured in single-cell
arrays identifies new features of self-renewal. Proc Natl Acad
Sci U S A 2006, 103:8185-8190.
Microfluidic single cell analysis Lecault et al. 387
www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
13. Cheong R, Rhee A, Wang CJ, Nemenman I, Levchenko A:
Information transduction capacity of noisy biochemical
signaling networks. Science 2011, 334:354-358.
14. Chen CF, Ridzon DA, Broomer AJ, Zhou ZH, Lee DH, Nguyen JT,
Barbisin M, Xu NL, Mahuvakar VR, Andersen MR et al.: Real-time
quantification of microRNAs by stem-loop RT-PCR. Nucl Acids
Res 2005, 33:e179.
15.

Dalerba P, Kalisky T, Sahoo D, Rajendran PS, Rothenberg ME,
Leyrat AA, Sim S, Okamoto J, Johnston DM, Qian DL et al.: Single-
cell dissection of transcriptional heterogeneity in human colon
tumors. Nat Biotechnol 2011, 29:1120-1127.
This study used high-throughput microfluidic qPCR arrays to dissect
human colon cancer tissue by single-cell gene expression analysis, and
showed that gene expression signatures were predictive of clinical
outcomes.
16. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D,
Lam JS, Ailles LE, Wong M et al.: Association of reactive oxygen
species levels and radioresistance in cancer stem cells. Nature
2009, 458:780-783.
17. Guo G, Huss M, Tong GQ, Wang C, Sun LL, Clarke ND, Robson P:
Resolution of cell fate decisions revealed by single-cell gene
expression analysis from zygote to blastocyst. Dev Cell 2010,
18:675-685.
18. Spike BT, Engle DD, Lin JC, Cheung SK, La J, Wahl GM: A
mammary stem cell population identified and characterized in
late embryogenesis reveals similarities to human breast
cancer. Cell Stem Cell 2012, 10:183-197.
19. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee-
Messer C, Dolmetsch RE, Tsien RW, Crabtree GR: MicroRNA-
mediated conversion of human fibroblasts to neurons. Nature
2011, 476:228-231.
20. Metzker ML: Applications of next-generation sequencing:
sequencing technologies – the next generation. Nat Rev Genet
2010, 11:31-46.
21. Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, Wang X,
Bodeau J, Tuch BB, Siddiqui A et al.: mRNA-Seq whole-
transcriptome analysis of a single cell. Nat Methods 2009,
6:377-382.
22. Tang F, Lao K, Surani MA: Development and applications of
single-cell transcriptome analysis. Nat Methods 2011,
8:S6-S11.
23. Lasken RS: Single-cell genomic sequencing using multiple
displacement amplification. Curr Opin Microbiol 2007,
10:510-516.
24. Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J,
Cook K, Stepansky A, Levy D, Esposito D et al.: Tumour evolution
inferred by single-cell sequencing. Nature 2011, 472:90-94.
25. Tang F, Barbacioru C, Nordman E, Li B, Xu N, Bashkirov VI, Lao K,
Surani MA: RNA-Seq analysis to capture the transcriptome
landscape of a single cell. Nat Protocols 2010, 5:516-535.
26. Lu R, Neff NF, Quake SR, Weissman IL: Tracking single
hematopoietic stem cells in vivo using high-throughput
sequencing in conjunction with viral genetic barcoding. Nat
Biotechnol 2011, 29:928-933.
27. Yin H, Marshall D: Microfluidics for single cell analysis. Curr
Opin Biotechnol 2012, 23:110-119.
28. Zare RN, Kim S: Microfluidic platforms for single-cell analysis.
Annu Rev Biomed Eng 2010, 12:187-201.
29. Mestdagh P, Feys T, Bernard N, Guenther S, Chen C, Speleman F,
Vandesompele J: High-throughput stem-loop RT-qPCR miRNA
expression profiling using minute amounts of input RNA. Nucl
Acids Res 2008, 36:e143.
30. Bengtsson M, Hemberg M, Rorsman P, Stahlberg A:
Quantification of mRNA in single cells and modelling of RT-
qPCR induced noise. BMC Mol Biol 2008, 9:63.
31. Bengtsson M, Stahlberg A, Rorsman P, Kubista M: Gene
expression profiling in single cells from the pancreatic islets of
Langerhans reveals lognormal distribution of mRNA levels.
Genome Res 2005, 15:1388-1392.
32. Tang F, Hajkova P, Barton SC, Lao K, Surani MA: MicroRNA
expression profiling of single whole embryonic stem cells.
Nucl Acids Res 2006, 34:e9.
33. Diercks A, Kostner H, Ozinsky A: Resolving cell population
heterogeneity: real-time PCR for simultaneous multiplexed
gene detection in multiple single-cell samples. PLoS ONE
2009, 4:e6326.
34. Narsinh KH, Sun N, Sanchez-Freire V, Lee AS, Almeida P, Hu S,
Jan T, Wilson KD, Leong D, Rosenberg J et al.: Single cell
transcriptional profiling reveals heterogeneity of human
induced pluripotent stem cells. J Clin Investig 2011,
121:1217-1221.
35. Petriv O, Kuchenbauer F, Delaney A, Lecault V, White A, Kent D,
Marmolejo L, Heuser M, Berg T, Copley M et al.: Comprehensive
microRNA expression profiling of the hematopoietic
hierarchy. Proc Natl Acad Sci U S A 2010, 107:15443-15448.
36. Flatz L, Roychoudhuri R, Honda M, Filali-Mouhim A, Goulet J-P,
Kettaf N, Lin M, Roederer M, Haddad EK, Sekaly RP et al.: Single-
cell gene-expression profiling reveals qualitatively distinct
CD8 T cells elicited by different gene-based vaccines. Proc
Natl Acad Sci U S A 2011, 108:5724-5729.
37. Kikushige Y, Ishikawa F, Miyamoto T, Shima T, Urata S,
Yoshimoto G, Mori Y, Iino T, Yamauchi T, Eto T et al.: Self-
renewing hematopoietic stem cell is the primary target in
pathogenesis of human chronic lymphocytic leukemia. Cancer
Cell 2011, 20:246-259.
38.

White AK, VanInsberghe M, Petriv OI, Hamidi M, Sikorski D,
Marra MA, Piret J, Aparicio S, Hansen CL: High-throughput
microfluidic single-cell RT-qPCR. Proc Natl Acad Sci U S A
2011, 108:13999-14004.
The authors addressed the bottleneck between cell handling and high-
density qPCR arrays by developing a fully integrated microfluidic RT-
qPCR device that implements all steps of cell trapping, lysis, cDNA
synthesis, and qPCR analysis at a throughput of 300 cells per run.
39. Bontoux N, Dauphinot L, Vitalis T, Studer V, Chen Y, Rossier J,
Potier MC: Integrating whole transcriptome assays on a lab-
on-a-chip for single cell gene profiling. Lab Chip 2008,
8:443-450.
40. Warren L, Bryder D, Weissman IL, Quake SR: Transcription
factor profiling in individual hematopoietic progenitors by
digital RT-PCR. Proc Natl Acad Sci U S A 2006,
103:17807-17812.
41. Heyries KA, Tropini C, VanInsberghe M, Doolin C, Petriv OI,
Singhal A, Leung K, Hughesman CB, Hansen CL: Megapixel
digital PCR. Nat Methods 2011, 8:649-651.
42.

Fan HC, Wang J, Potanina A, Quake SR: Whole-genome
molecular haplotyping of single cells. Nat Biotechnol 2011,
29:51-57.
The authors developed of a microfluidic device capable of amplifying and
genotyping individual chromosomes from a single cell. This technique is
scalable, and may be used in conjunction with SNP arrays and sequen-
cing for applications in personal genomics such as haplotyping.
43. Dean FB, Hosono S, Fang LH, Wu XH, Faruqi AF, Bray-Ward P,
Sun ZY, Zong QL, Du YF, Du J et al.: Comprehensive human
genome amplification using multiple displacement
amplification. Proc Natl Acad Sci U S A 2002, 99:5261-5266.
44. Ottesen EA, Hong JW, Quake SR, Leadbetter JR: Microfluidic
digital PCR enables multigene analysis of individual
environmental bacteria. Science 2006, 314:1464-1467.
45. Tadmor AD, Ottesen EA, Leadbetter JR, Phillips R: Probing
individual environmental bacteria for viruses by using
microfluidic digital PCR. Science 2011, 333:58-62.
46. Zeng Y, Novak R, Shuga J, Smith MT, Mathies RA: High-
performance single cell genetic analysis using microfluidic
emulsion generator arrays. Anal Chem 2010, 82:3183-3190.
47. Marcy Y, Ishoey T, Lasken RS, Stockwell TB, Walenz BP,
Halpern AL, Beeson KY, Goldberg SMD, Quake SR: Nanoliter
reactors improve multiple displacement amplification of
genomes from single cells. PLoS Genet 2007,
3:1702-1708.
388 Analytical techniques
Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
48. Blainey PC, Mosier AC, Potanina A, Francis CA, Quake SR:
Genome of a low-salinity ammonia-oxidizing archaeon
determined by single-cell and metagenomic analysis. PLoS
ONE 2011, 6:e16626.
49. Leung K, Zahn H, Leaver T, Konwar KM, Hanson NW, Page´ AP, Lo
C-C, Chain PS, Hallam S, Hansen CL: A programmable droplet-
based microfluidic device applied to multiparameter analysis
of single microbes and microbial communities. Proc Natl Acad
Sci U S A 2012, http://dx.doi.org/10.1073/pnas.1106752109,
in press.
50.

Taniguchi Y, Choi P, Li G, Chen H, Babu M, Hearn J, Emili A, Xie X:
Quantifying E-coli proteome and transcriptome with single-
molecule sensitivity in single cells. Science 2010, 329:533-538.
The authors used a microfluidic device to perform parallel measurements
of single-cell protein and mRNA expression with single-molecule sensi-
tivity across over 1000 different strains of Escherichia coli.
51. Cheong R, Wang CJ, Levchenko A: High content cell screening
in a microfluidic device. Mol Cell Proteomics 2009,
8:433-442.
52. Fosbrink M, Aye-Han NN, Cheong R, Levchenko A, Zhang J:
Visualization of JNK activity dynamics with a genetically
encoded fluorescent biosensor. Proc Natl Acad Sci U S A 2010,
107:5459-5464.
53. Sun J, Masterman-Smith MD, Graham NA, Jiao J, Mottahedeh J,
Laks DR, Ohashi M, DeJesus J, Kamei K-i, Lee K-B et al.: A
microfluidic platform for systems pathology: multiparameter
single-cell signaling measurements of clinical brain tumor
specimens. Cancer Res 2010, 70:6128-6138.
54. Huang B, Wu H, Bhaya D, Grossman A, Granier S, Kobilka BK,
Zare RN: Counting low-copy number proteins in a single cell.
Science 2007, 315:81-84.
55.

Shi Q, Qin L, Wei W, Geng F, Fan R, Shin YS, Guo D, Hood L,
Mischel PS, Heath JR: Single-cell proteomic chip for profiling
intracellular signaling pathways in single tumor cells. Proc Natl
Acad Sci U S A 2011, 109:419-424.
The authors describe a microfluidic ‘Single Cell Barcode Chip’ (SCBC) for
multiplexed profiling of over 10 different cytokines and effector proteins
secreted by single macrophage and T cells, and demonstrate how this
tool may be useful for clinical monitoring of the immune system.
56. Ma C, Fan R, Ahmad H, Shi Q, Comin-Anduix B, Chodon T,
Koya RC, Liu C-C, Kwong GA, Radu CG et al.: A clinical
microchip for evaluation of single immune cells reveals high
functional heterogeneity in phenotypically similar T cells. Nat
Med 2011, 17:738-743.
57. Story CM, Papa E, Hu C-CA, Ronan JL, Herlihy K, Ploegh HL,
Love JC: Profiling antibody responses by multiparametric
analysis of primary B cells. Proc Natl Acad Sci U S A 2008,
105:17902-17907.
58. Jin A, Ozawa T, Tajiri K, Obata T, Kondo S, Kinoshita K,
Kadowaki S, Takahashi K, Sugiyama T, Kishi H et al.: A rapid and
efficient single-cell manipulation method for screening
antigen-specific antibody-secreting cells from human
peripheral blood. Nat Med 2009, 15:1088-1092.
59. Singhal A, Haynes CA, Hansen CL: Microfluidic measurement of
antibody–antigen binding kinetics from low-abundance
samples and single cells. Anal Chem 2010, 82:8671-8679.
60. Koester S, Angile FE, Duan H, Agresti JJ, Wintner A, Schmitz C,
Rowat AC, Merten CA, Pisignano D, Griffiths AD et al.: Drop-
based microfluidic devices for encapsulation of single cells.
Lab Chip 2008, 8:1110-1115.
61. Konry T, Dominguez-Villar M, Baecher-Allan C, Hafler DA,
Yarmush ML: Droplet-based microfluidic platforms for single T
cell secretion analysis of IL-10 cytokine. Biosens Bioelectron
2011, 26:2707-2710.
62. Tumarkin E, Tzadu L, Csaszar E, Seo M, Zhang H, Lee A,
Peerani R, Purpura K, Zandstra PW, Kumacheva E: High-
throughput combinatorial cell co-culture using microfluidics.
Integr Biol 2011, 3:653-662.
63.

Brouzes E, Medkova M, Savenelli N, Marran D, Twardowski M,
Hutchison JB, Rothberg JM, Link DR, Perrimon N, Samuels ML:
Droplet microfluidic technology for single-cell high-
throughput screening. Proc Natl Acad Sci U S A 2009,
106:14195-14200.
The authors present a droplet-based microfluidic technology for high-
throughput encapsulation of single mammalian cells and cytotoxicity
screening against a library of drugs.
64. Paliwal S, Iglesias PA, Campbell K, Hilioti Z, Groisman A,
Levchenko A: MAPK-mediated bimodal gene expression and
adaptive gradient sensing in yeast. Nature 2007,
446:46-51.
65. Bennett MR, Pang WL, Ostroff NA, Baumgartner BL, Nayak S,
Tsimring LS, Hasty J: Metabolic gene regulation in a
dynamically changing environment. Nature 2008,
454:1119-1122.
66. Hersen P, McClean MN, Mahadevan L, Ramanathan S: Signal
processing by the HOG MAP kinase pathway. Proc Natl Acad
Sci U S A 2008, 105:7165-7170.
67. Taylor RJ, Falconnet D, Niemisto A, Ramsey SA, Prinz S,
Shmulevich I, Galitski T, Hansen CL: Dynamic analysis of MAPK
signaling using a high-throughput microfluidic single-cell
imaging platform. Proc Natl Acad Sci U S A 2009,
106:3758-3763.
68. Falconnet D, Niemisto A, Taylor RJ, Ricicova M, Galitski T,
Shmulevich I, Hansen CL: High-throughput tracking of single
yeast cells in a microfluidic imaging matrix. Lab Chip 2011,
11:466-473.
69. Gobaa S, Hoehnel S, Roccio M, Negro A, Kobel S, Lutolf MP:
Artificial niche microarrays for probing single stem cell fate in
high throughput. Nat Methods 2011, 8:949-955.
70.

Lecault V, VanInsberghe M, Sekulovic S, Knapp DJHF, Wohrer S,
Bowden W, Viel F, McLaughlin T, Jarandehei A, Miller M et al.:
High-throughput analysis of single hematopoietic stem cell
proliferation in microfluidic cell culture arrays. Nat Methods
2011, 8:581-586.
This article demonstrates robust long-term microfluidic culture of pri-
mary hematopoietic stem and progenitor single cells with the integration
of an iso-osmotic bath to block evaporation. The design features an
array of high aspect ratio chambers and allows for medium exchange,
temporal stimulation, time-lapse imaging, live immunostaining and
clone recovery.
71.

Tay S, Hughey JJ, Lee TK, Lipniacki T, Quake SR, Covert MW:
Single-cell NF-kappa B dynamics reveal digital activation and
analogue information processing. Nature 2010, 466:267-271.
This study combines fluorescent gene expression with dynamic micro-
fluidic cell culture to measure NF-kB activity in response to temporal
stimulation with TNF-a in single mammalian cells.
72.

Moledina F, Clarke G, Oskooei A, Onishi K, Gunther A,
Zandstra PW: Predictive microfluidic control of regulatory
ligand trajectories in individual pluripotent cells. Proc Natl
Acad Sci U S A 2012, 109:3264-3269.
The authors study the effects of autocrine and paracrine signaling on cell
fate in murine embryonic stem cells by precisely modulating laminar flow
in microfluidic channels.
73. Sekulovic S, Gasparetto M, Lecault V, Hoesli CA, Kent DG,
Rosten P, Wan A, Brookes C, Hansen CL, Piret JM et al.:
Ontogeny stage-independent and high-level clonal expansion
in vitro of mouse hematopoietic stem cells stimulated by an
engineered NUP98-HOX fusion transcription factor. Blood
2011, 118:4366-4376.
74. Albrecht DR, Underhill GH, Resnikoff J, Mendelson A, Bhatia SN,
Shah JV: Microfluidics-integrated time-lapse imaging for
analysis of cellular dynamics. Integr Biol 2010,
2:278-287.
75. Scherf N, Franke K, Glauche I, Kurth I, Bornhauser M, Werner C,
Pompe T, Roeder I: On the symmetry of siblings: automated
single-cell tracking to quantify the behavior of hematopoietic
stem cells in a biomimetic setup. Exp Hematol 2012,
40:119-130.
76. Rowat AC, Bird JC, Agresti JJ, Rando OJ, Weitz DA: Tracking
lineages of single cells in lines using a microfluidic device.
Proc Natl Acad Sci U S A 2009, 106:18149-18154.
77. Godin M, Delgado FF, Son SM, Grover WH, Bryan AK, Tzur A,
Jorgensen P, Payer K, Grossman AD, Kirschner MW et al.: Using
Microfluidic single cell analysis Lecault et al. 389
www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
buoyant mass to measure the growth of single cells. Nat
Methods 2010, 7:387-390.
78. Grover WH, Bryan AK, Diez-Silva M, Suresh S, Higgins JM,
Manalis SR: Measuring single-cell density. Proc Natl Acad Sci U
S A 2011, 108:10992-10996.
79. Dada OO, Essaka DC, Hindsgaul O, Palcic MM, Prendergast J,
Schnaar RL, Dovichi NJ: Nine orders of magnitude dynamic
range: picomolar to millimolar concentration measurement in
capillary electrophoresis with laser induced fluorescence
detection employing cascaded avalanche photodiode photon
counters. Anal Chem 2011, 83:2748-2753.
80. Fidalgo LM, Maerkl SJ: A software-programmable microfluidic
device for automated biology. Lab Chip 2011, 11:1612-1619.
81. Srigunapalan S, Eydelnant IA, Simmons CA, Wheeler AR: A digital
microfluidic platform for primary cell culture and analysis. Lab
Chip 2012, 12:369-375.
390 Analytical techniques
Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com

More Related Content

What's hot

Pluripotent stem cells An in vitro model for nanotoxicity
Pluripotent stem cells An in vitro model for nanotoxicityPluripotent stem cells An in vitro model for nanotoxicity
Pluripotent stem cells An in vitro model for nanotoxicityDr. Harish Handral
 
Updates in Gastric Cancer
Updates in Gastric CancerUpdates in Gastric Cancer
Updates in Gastric CancerSamieh Asadian
 
NGS in cancer treatment
NGS in cancer treatmentNGS in cancer treatment
NGS in cancer treatmentNur Suhaida
 
Genes and Tissue Culture Technology Assignment (G6)
Genes and Tissue Culture Technology Assignment (G6)Genes and Tissue Culture Technology Assignment (G6)
Genes and Tissue Culture Technology Assignment (G6)Rohini Krishnan
 
SCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationSCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationJayden On
 
Alexey_Ball_CV_v2
Alexey_Ball_CV_v2Alexey_Ball_CV_v2
Alexey_Ball_CV_v2Lex Ball
 
An Overview on Gene Expression Analysis
An Overview on Gene Expression AnalysisAn Overview on Gene Expression Analysis
An Overview on Gene Expression AnalysisIOSR Journals
 
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsUmbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsComprehensiveBiologi
 
2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generation2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generationNatalia Muñoz Roa
 
Teresa Coque Hospital Universitario Ramón y Cajal.
Teresa Coque  Hospital Universitario Ramón y Cajal. Teresa Coque  Hospital Universitario Ramón y Cajal.
Teresa Coque Hospital Universitario Ramón y Cajal. Fundación Ramón Areces
 
Umbilical cord wj_greatest___msc.vangsness2015_(1)
Umbilical cord wj_greatest___msc.vangsness2015_(1)Umbilical cord wj_greatest___msc.vangsness2015_(1)
Umbilical cord wj_greatest___msc.vangsness2015_(1)ComprehensiveBiologi
 
APPLICATION OF NEXT GENERATION SEQUENCING (NGS) IN CANCER TREATMENT
APPLICATION OF  NEXT GENERATION SEQUENCING (NGS)  IN CANCER TREATMENTAPPLICATION OF  NEXT GENERATION SEQUENCING (NGS)  IN CANCER TREATMENT
APPLICATION OF NEXT GENERATION SEQUENCING (NGS) IN CANCER TREATMENTDinie Fariz
 
Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)ComprehensiveBiologi
 
SCT60103 - Chemotherapy
SCT60103 - Chemotherapy SCT60103 - Chemotherapy
SCT60103 - Chemotherapy KaiXuan Lim
 
Compartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) posterCompartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) posterJackie Lau
 
Discoidin receptors a proteomic portret
Discoidin receptors a proteomic portretDiscoidin receptors a proteomic portret
Discoidin receptors a proteomic portretMaciej Luczynski
 

What's hot (19)

Pluripotent stem cells An in vitro model for nanotoxicity
Pluripotent stem cells An in vitro model for nanotoxicityPluripotent stem cells An in vitro model for nanotoxicity
Pluripotent stem cells An in vitro model for nanotoxicity
 
Updates in Gastric Cancer
Updates in Gastric CancerUpdates in Gastric Cancer
Updates in Gastric Cancer
 
NGS in cancer treatment
NGS in cancer treatmentNGS in cancer treatment
NGS in cancer treatment
 
FRD Grant Paper
FRD Grant PaperFRD Grant Paper
FRD Grant Paper
 
Genes and Tissue Culture Technology Assignment (G6)
Genes and Tissue Culture Technology Assignment (G6)Genes and Tissue Culture Technology Assignment (G6)
Genes and Tissue Culture Technology Assignment (G6)
 
SCT60103 Group 2 Presentation
SCT60103 Group 2 PresentationSCT60103 Group 2 Presentation
SCT60103 Group 2 Presentation
 
Alexey_Ball_CV_v2
Alexey_Ball_CV_v2Alexey_Ball_CV_v2
Alexey_Ball_CV_v2
 
An Overview on Gene Expression Analysis
An Overview on Gene Expression AnalysisAn Overview on Gene Expression Analysis
An Overview on Gene Expression Analysis
 
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applicationsUmbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
Umbilical cord-derived-whartons-jelly-for-regenerative-medicine-applications
 
2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generation2000 weinberg. hallsmarcks of cancer new generation
2000 weinberg. hallsmarcks of cancer new generation
 
Teresa Coque Hospital Universitario Ramón y Cajal.
Teresa Coque  Hospital Universitario Ramón y Cajal. Teresa Coque  Hospital Universitario Ramón y Cajal.
Teresa Coque Hospital Universitario Ramón y Cajal.
 
Umbilical cord wj_greatest___msc.vangsness2015_(1)
Umbilical cord wj_greatest___msc.vangsness2015_(1)Umbilical cord wj_greatest___msc.vangsness2015_(1)
Umbilical cord wj_greatest___msc.vangsness2015_(1)
 
International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
 
APPLICATION OF NEXT GENERATION SEQUENCING (NGS) IN CANCER TREATMENT
APPLICATION OF  NEXT GENERATION SEQUENCING (NGS)  IN CANCER TREATMENTAPPLICATION OF  NEXT GENERATION SEQUENCING (NGS)  IN CANCER TREATMENT
APPLICATION OF NEXT GENERATION SEQUENCING (NGS) IN CANCER TREATMENT
 
Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)
 
SCT60103 - Chemotherapy
SCT60103 - Chemotherapy SCT60103 - Chemotherapy
SCT60103 - Chemotherapy
 
Compartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) posterCompartment specific micro rna expression profiles (poster) poster
Compartment specific micro rna expression profiles (poster) poster
 
Discoidin receptors a proteomic portret
Discoidin receptors a proteomic portretDiscoidin receptors a proteomic portret
Discoidin receptors a proteomic portret
 
International Journal of Stem Cells & Research
International Journal of Stem Cells & ResearchInternational Journal of Stem Cells & Research
International Journal of Stem Cells & Research
 

Similar to Lecaut et al 2012

A statistical framework for multiparameter analysis at the single cell level
A statistical framework for multiparameter analysis at the single cell levelA statistical framework for multiparameter analysis at the single cell level
A statistical framework for multiparameter analysis at the single cell levelShashaanka Ashili
 
Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...
Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...
Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...The Lifesciences Magazine
 
Advances-in-cancer-research.pdf
Advances-in-cancer-research.pdfAdvances-in-cancer-research.pdf
Advances-in-cancer-research.pdfssuser54a9d9
 
Cimetta et al., 2013
Cimetta et al., 2013Cimetta et al., 2013
Cimetta et al., 2013Fran Flores
 
Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...Shashaanka Ashili
 
Array_nmeth.3507
Array_nmeth.3507Array_nmeth.3507
Array_nmeth.3507Aana Hahn
 
2008-0471S1-4
2008-0471S1-42008-0471S1-4
2008-0471S1-4pharmdude
 
Soergel oa week-2014-lightning
Soergel oa week-2014-lightningSoergel oa week-2014-lightning
Soergel oa week-2014-lightningDavid Soergel
 
Optimizing Transfection of Umbilical Cord Mesenchymal Stem Cells Utilizing
Optimizing Transfection of Umbilical Cord Mesenchymal Stem Cells UtilizingOptimizing Transfection of Umbilical Cord Mesenchymal Stem Cells Utilizing
Optimizing Transfection of Umbilical Cord Mesenchymal Stem Cells UtilizingMohadese Hashem Boroojerdi
 
A physical sciences network characterization of non-tumorigenic and metastati...
A physical sciences network characterization of non-tumorigenic and metastati...A physical sciences network characterization of non-tumorigenic and metastati...
A physical sciences network characterization of non-tumorigenic and metastati...Shashaanka Ashili
 
2D CAT Based Modeling of Tumour Growth and Drug Transport
2D CAT Based Modeling of Tumour Growth and Drug Transport2D CAT Based Modeling of Tumour Growth and Drug Transport
2D CAT Based Modeling of Tumour Growth and Drug TransportEditor IJMTER
 
Advances and Problems in Preclinical Models for Childhood Cancer
Advances and Problems in Preclinical Models for Childhood CancerAdvances and Problems in Preclinical Models for Childhood Cancer
Advances and Problems in Preclinical Models for Childhood Cancerijtsrd
 
Biotechnology And Chemical Weapons Control
Biotechnology And Chemical Weapons ControlBiotechnology And Chemical Weapons Control
Biotechnology And Chemical Weapons Controlguest971b1073
 
Ellison MolBioSys b905602e published (2)
Ellison MolBioSys b905602e published (2)Ellison MolBioSys b905602e published (2)
Ellison MolBioSys b905602e published (2)Dr David Ellison
 

Similar to Lecaut et al 2012 (20)

A statistical framework for multiparameter analysis at the single cell level
A statistical framework for multiparameter analysis at the single cell levelA statistical framework for multiparameter analysis at the single cell level
A statistical framework for multiparameter analysis at the single cell level
 
Assay Development in Cell Culture.pdf
Assay Development in Cell Culture.pdfAssay Development in Cell Culture.pdf
Assay Development in Cell Culture.pdf
 
Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...
Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...
Flow Cytometry: Guide to Understanding Applications and Benefits | The Lifesc...
 
Advances-in-cancer-research.pdf
Advances-in-cancer-research.pdfAdvances-in-cancer-research.pdf
Advances-in-cancer-research.pdf
 
nature19764
nature19764nature19764
nature19764
 
JALANov2000
JALANov2000JALANov2000
JALANov2000
 
Cimetta et al., 2013
Cimetta et al., 2013Cimetta et al., 2013
Cimetta et al., 2013
 
Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...
 
cancer
cancercancer
cancer
 
Array_nmeth.3507
Array_nmeth.3507Array_nmeth.3507
Array_nmeth.3507
 
2008-0471S1-4
2008-0471S1-42008-0471S1-4
2008-0471S1-4
 
Gene therapy
Gene therapyGene therapy
Gene therapy
 
Soergel oa week-2014-lightning
Soergel oa week-2014-lightningSoergel oa week-2014-lightning
Soergel oa week-2014-lightning
 
Optimizing Transfection of Umbilical Cord Mesenchymal Stem Cells Utilizing
Optimizing Transfection of Umbilical Cord Mesenchymal Stem Cells UtilizingOptimizing Transfection of Umbilical Cord Mesenchymal Stem Cells Utilizing
Optimizing Transfection of Umbilical Cord Mesenchymal Stem Cells Utilizing
 
multiomics-ebook.pdf
multiomics-ebook.pdfmultiomics-ebook.pdf
multiomics-ebook.pdf
 
A physical sciences network characterization of non-tumorigenic and metastati...
A physical sciences network characterization of non-tumorigenic and metastati...A physical sciences network characterization of non-tumorigenic and metastati...
A physical sciences network characterization of non-tumorigenic and metastati...
 
2D CAT Based Modeling of Tumour Growth and Drug Transport
2D CAT Based Modeling of Tumour Growth and Drug Transport2D CAT Based Modeling of Tumour Growth and Drug Transport
2D CAT Based Modeling of Tumour Growth and Drug Transport
 
Advances and Problems in Preclinical Models for Childhood Cancer
Advances and Problems in Preclinical Models for Childhood CancerAdvances and Problems in Preclinical Models for Childhood Cancer
Advances and Problems in Preclinical Models for Childhood Cancer
 
Biotechnology And Chemical Weapons Control
Biotechnology And Chemical Weapons ControlBiotechnology And Chemical Weapons Control
Biotechnology And Chemical Weapons Control
 
Ellison MolBioSys b905602e published (2)
Ellison MolBioSys b905602e published (2)Ellison MolBioSys b905602e published (2)
Ellison MolBioSys b905602e published (2)
 

More from Fran Flores

Friedlander et al 2012
Friedlander et al 2012Friedlander et al 2012
Friedlander et al 2012Fran Flores
 
Toh et al. 2014
Toh et al. 2014 Toh et al. 2014
Toh et al. 2014 Fran Flores
 
Balagaddé et al 2005
 Balagaddé et al 2005 Balagaddé et al 2005
Balagaddé et al 2005Fran Flores
 
Cimetta et al 2010
Cimetta et al 2010Cimetta et al 2010
Cimetta et al 2010Fran Flores
 
Adriana San Miguel and Hang Lu (2013)
Adriana San Miguel and Hang Lu (2013)Adriana San Miguel and Hang Lu (2013)
Adriana San Miguel and Hang Lu (2013)Fran Flores
 
Atencia, Cooksey and .Locascio 2012
Atencia, Cooksey and .Locascio 2012Atencia, Cooksey and .Locascio 2012
Atencia, Cooksey and .Locascio 2012Fran Flores
 

More from Fran Flores (10)

Sackmann 2014
Sackmann 2014Sackmann 2014
Sackmann 2014
 
Friedlander et al 2012
Friedlander et al 2012Friedlander et al 2012
Friedlander et al 2012
 
Toh et al. 2014
Toh et al. 2014 Toh et al. 2014
Toh et al. 2014
 
Whitesides 2006
Whitesides 2006Whitesides 2006
Whitesides 2006
 
Duffy 1998
Duffy 1998Duffy 1998
Duffy 1998
 
Balagaddé et al 2005
 Balagaddé et al 2005 Balagaddé et al 2005
Balagaddé et al 2005
 
Cimetta et al 2010
Cimetta et al 2010Cimetta et al 2010
Cimetta et al 2010
 
Adriana San Miguel and Hang Lu (2013)
Adriana San Miguel and Hang Lu (2013)Adriana San Miguel and Hang Lu (2013)
Adriana San Miguel and Hang Lu (2013)
 
Atencia, Cooksey and .Locascio 2012
Atencia, Cooksey and .Locascio 2012Atencia, Cooksey and .Locascio 2012
Atencia, Cooksey and .Locascio 2012
 
Adler 1975
Adler 1975Adler 1975
Adler 1975
 

Recently uploaded

Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024hassan khalil
 
HARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICS
HARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICSHARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICS
HARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICSRajkumarAkumalla
 
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur High Profile
 
OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...
OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...
OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...Soham Mondal
 
Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝
Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝
Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝soniya singh
 
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur EscortsCall Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur High Profile
 
chaitra-1.pptx fake news detection using machine learning
chaitra-1.pptx  fake news detection using machine learningchaitra-1.pptx  fake news detection using machine learning
chaitra-1.pptx fake news detection using machine learningmisbanausheenparvam
 
APPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICS
APPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICSAPPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICS
APPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICSKurinjimalarL3
 
GDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentationGDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentationGDSCAESB
 
Call Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile serviceCall Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile servicerehmti665
 
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINEMANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINESIVASHANKAR N
 
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptxthe ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptxhumanexperienceaaa
 
VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130
VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130
VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130Suhani Kapoor
 
VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130
VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130
VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130Suhani Kapoor
 
MANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLS
MANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLSMANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLS
MANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLSSIVASHANKAR N
 
What are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptxWhat are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptxwendy cai
 
Call Girls in Nagpur Suman Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Suman Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur Suman Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Suman Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur High Profile
 
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escortsranjana rawat
 

Recently uploaded (20)

Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024Architect Hassan Khalil Portfolio for 2024
Architect Hassan Khalil Portfolio for 2024
 
HARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICS
HARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICSHARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICS
HARDNESS, FRACTURE TOUGHNESS AND STRENGTH OF CERAMICS
 
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Meera Call 7001035870 Meet With Nagpur Escorts
 
OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...
OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...
OSVC_Meta-Data based Simulation Automation to overcome Verification Challenge...
 
Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝
Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝
Model Call Girl in Narela Delhi reach out to us at 🔝8264348440🔝
 
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur EscortsCall Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
Call Girls Service Nagpur Tanvi Call 7001035870 Meet With Nagpur Escorts
 
chaitra-1.pptx fake news detection using machine learning
chaitra-1.pptx  fake news detection using machine learningchaitra-1.pptx  fake news detection using machine learning
chaitra-1.pptx fake news detection using machine learning
 
APPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICS
APPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICSAPPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICS
APPLICATIONS-AC/DC DRIVES-OPERATING CHARACTERISTICS
 
GDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentationGDSC ASEB Gen AI study jams presentation
GDSC ASEB Gen AI study jams presentation
 
Call Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile serviceCall Girls Delhi {Jodhpur} 9711199012 high profile service
Call Girls Delhi {Jodhpur} 9711199012 high profile service
 
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINEMANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
MANUFACTURING PROCESS-II UNIT-2 LATHE MACHINE
 
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptxthe ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
the ladakh protest in leh ladakh 2024 sonam wangchuk.pptx
 
VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130
VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130
VIP Call Girls Service Kondapur Hyderabad Call +91-8250192130
 
VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130
VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130
VIP Call Girls Service Hitech City Hyderabad Call +91-8250192130
 
Roadmap to Membership of RICS - Pathways and Routes
Roadmap to Membership of RICS - Pathways and RoutesRoadmap to Membership of RICS - Pathways and Routes
Roadmap to Membership of RICS - Pathways and Routes
 
MANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLS
MANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLSMANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLS
MANUFACTURING PROCESS-II UNIT-5 NC MACHINE TOOLS
 
DJARUM4D - SLOT GACOR ONLINE | SLOT DEMO ONLINE
DJARUM4D - SLOT GACOR ONLINE | SLOT DEMO ONLINEDJARUM4D - SLOT GACOR ONLINE | SLOT DEMO ONLINE
DJARUM4D - SLOT GACOR ONLINE | SLOT DEMO ONLINE
 
What are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptxWhat are the advantages and disadvantages of membrane structures.pptx
What are the advantages and disadvantages of membrane structures.pptx
 
Call Girls in Nagpur Suman Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Suman Call 7001035870 Meet With Nagpur EscortsCall Girls in Nagpur Suman Call 7001035870 Meet With Nagpur Escorts
Call Girls in Nagpur Suman Call 7001035870 Meet With Nagpur Escorts
 
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur EscortsHigh Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
High Profile Call Girls Nagpur Isha Call 7001035870 Meet With Nagpur Escorts
 

Lecaut et al 2012

  • 1. Microfluidic single cell analysis: from promise to practice Ve´ ronique Lecault1,2,3,5 , Adam K White1,5 , Anupam Singhal1,3,5 and Carl L Hansen1,4 Methods for single-cell analysis are critical to revealing cell-to- cell variability in biological systems, especially in cases where relevant minority cell populations can be obscured by population-averaged measurements. However, to date single cell studies have been limited by the cost and throughput required to examine large numbers of cells and the difficulties associated with analyzing small amounts of starting material. Microfluidic approaches are well suited to resolving these issues by providing increased senstitivity, economy of scale, and automation. After many years of development microfluidic systems are now finding traction in a variety of single-cell analytics including gene expression measurements, protein analysis, signaling response, and growth dynamics. With newly developed tools now being applied in fields ranging from human haplotyping and drug discovery to stem cell and cancer research, the long-heralded promise of microfluidic single cell analysis is now finally being realized. Addresses 1 Centre for High-Throughput Biology, University of British Columbia, 2185 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada 2 Michael Smith Laboratories, University of British Columbia, 2125 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada 3 Department of Chemical and Biological Engineering, University of British Columbia, 2360 East Mall, Vancouver, British Columbia, V6T 1Z3, Canada 4 Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, East Mall, Vancouver, British Columbia, V6T 1Z1, Canada Corresponding author: Hansen, Carl L (chansen@physics.ubc.ca, chansen@phas.ubc.ca) 5 Authors contributed equally to this work. Current Opinion in Chemical Biology 2012, 16:381–390 This review comes from a themed issue on Analytical techniques Edited by Shana O Kelley and Petra S Dittrich For a complete overview see the Issue and the Editorial Available online 21st April 2012 1367-5931/$ – see front matter, # 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.cbpa.2012.03.022 Introduction Cells are the fundamental units of biological processes. However, the vast majority of our understanding of bio- chemistry and genetics has been gleaned through the bulk analysis of large populations of cells, which are required to obtain sufficient starting material for conventional analysis methods. Interpretation of such data often implies the tacit assumption that each cell in the population is similar. More often than notthisisfalse.Cellular heterogeneityisinfacta generalfeatureofbiologicalsystemsandhasbeenobserved across all levels of life, from single bacterial cells to human tissues. Even cells with identical functions generally respondasynchronously, making precise studies of kinetics and dynamics of cell populations impossible. Moreover, in many important fields, minority subpopulations of cells are often the most relevant. For instance, in microbial geno- mics it is frequently impossible to selectively isolate or culture a particular microbial species, thus necessitating the study of DNA shrapnel derived from a mixture of organisms. Similarly, in stem cell science even the most advanced isolation methods can only provide enriched populations of stem cells. In the best case, murine hema- topoietic stem cells, functional purities are generally below 50% [1], and in many other systems they are much lower. Thus, bulk measurements of the molecular signatures of these important cells are obscured by significant, and often overwhelming, contamination from other cell types of unknown state and relative abundance. This scenario is paralleled in numerous fields of research including cancer, immunology and developmental biology. Understanding the extent and importance of cellular heterogeneity is one of the most vexing problems facing biological research. The challenge of understanding cellular heterogeneity has been a major thrust of technological development over the past decade, resulting in an increasingly powerful suite of instrumentation, protocols, and methods for ana- lyzing single cells at the level of DNA sequence, RNA expression and protein abundance [2–4]. Flow cytometry and cell sorting have been widely adopted as a corner- stone of cellular phenotyping and purification, allowing for high-throughput quantitative analysis of protein expression and phosphorylation state in single cells [5– 7]. Recent advances in coupling this technology with metal labeling have now extended the analysis of fixed cells to sensitive multiplexing of dozens of targets per run [8]. At the same time, increasingly rapid and sensitive imaging instrumentation now allows high-throughput single cell measurements of proteins and nucleic acids with single molecule resolution [9], and the precise tracking of cellular growth and responses over extended periods [10–13]. Sensitive PCR-based protocols for measuring large panels of mRNA from single cells are now well established and have been coupled to FACS isolation and robotic assay assembly [14,15 ,16–19]. At the same time, the rise of high-throughput sequencing Available online at www.sciencedirect.com www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
  • 2. instrumentation [20] has now made single-cell genomics a practical proposition [21,22] and increasingly focus has turned to the development of appropriate protocols for high-fidelity amplification of DNA and RNA from single cells [23–25]. Sequencing approaches have further been coupled with virus-based molecular barcoding strategies to dissect heterogeneity in cellular processes in vivo [26]. Despite this suite of available approaches we are still only beginning to face the measurement challenges of cellular heterogeneity. There is much work to be done, both in enabling new modes of analysis, and in improving the speed, throughput, and economy of those that exist. Although each measurement application carries its own specific requirements, the recurring challenge in single- cell analysis is to simultaneously achieve the sensitivity, precision, throughput, and economy needed to detect and study complex subpopulations of cells. Microfluidic for- mats are ideally suited to addressing these problems by providing reduced reagent costs, high effective template concentrations in small volumes, scalability, ease of auto- mation, improved cell handling, and multi-step integration (Table 1). Over a decade of interdisciplinary research and development, spanning fields of engineering, molecular biology, imaging, cell biology, and chemistry, has now brought microfluidic single cell analysis to a turning point, with technologies increasingly being adopted in biological andbiomedicalresearch[27,28].Herewereviewhowthese approaches are pushing new avenues of research and high- light the next generation of advances that seem poised to transition from the bread-board to the bench-top. Microfluidic approaches to high-throughput single-cell RT-qPCR Reverse transcription quantitative PCR (RT-qPCR) pro- vides both excellent specificity and sensitivity, making it well suited to gene expression measurements in single cells. New preamplification strategies [29] have enabled RT-qPCR measurements, which were typically per- formed on a modest numbers of target genes [30,31], to be multiplexed for the analysis of many dozens of transcripts from a single cell [32,33]. However, appli- cation of this approach to the analysis of large numbers of single cells creates a ‘tyranny of numbers’ with serious challenges in both throughput and cost. A commercially available valve-based microfluidic qPCR system, the Dynamic ArrayTM (Fluidigm), provides a low-volume (nanoliter) and high-throughput (thousands of PCR reac- tions per device) solution to this detection problem and has become increasingly popular for large-scale single cell studies. For instance, Guo et al. used microfluidic qPCR arrays to measure the expression dynamics of 48 genes over 500 individual cells harvested during the first four days of mouse embryo development [17]. Correlation of expression measurements with surface markers revealed that co-expression of lineage-specific transcription factors occurred at the 32-cell stage of development, but that three distinct transcriptional programs emerged at the 64- cell stage. Scalable single cell expression measurements have also been used to provide new insights into cellular heterogeneity that exists within rare populations isolated from different developmental stages and across hierar- chies of differentiation, both at the level of transcript and miRNA expression [18,19,34,35]. Of particular interest has been the implication of cellular heterogeneity within cancer, both in terms of disease progression and optimal treatment strategies [36,37]. Dalerba et al. identified dis- tinct single cell gene expression signatures that are pre- dictive of patient survival and clinical outcomes in colon cancer patients [15 ]. Diehn et al. used single-cell analysis to identify a radioresistant subset of breast tumor cells with increased capacity for reactive oxygen species scavenging [16]. 382 Analytical techniques Table 1 Advantages of microfluidics for single cell analysis. Application Challenges with traditional methods Advantages of microfluidics Examples Single-cell RT-qPCR Limited abundance of starting template Concentration enhancement in small volumes [38 ,40] Cost and throughput required for analysis of large numbers of cells and target genes Parallelization, automation, and economy of scale [17,38 ] Single-cell genomics Amplification bias and sensitivity Improved reaction bias and sensitivity in nL volumes and reduced contaminant DNA [47] Isolating individual cells Integrated microfluidic cell sorting and processing [48] Single-cell measurements of intracellular proteins Movement of living cells Confinement of live cells in microfluidic structures [50 ] Low amount of signal Integrated single-cell handling allows lysate analysis [54,55 ] Single-cell measurements of secreted proteins Small amounts of secreted products from single cells Concentration enhancement in small volumes [57,58] Difficult to co-localize multiple cells in defined chemical environments Ease of confinement in droplets or microchambers [62] Signaling studies Mostly limited to static conditions Easy temporal stimulation [67,70 ,71 ] Inability to rapidly exchange conditions on suspension cells Laminar flow and proper design enables cell sequestration [68,70 ] Live cell imaging Difficulties of tracking cells through multiple frames Confinement of clones facilitates cell tracking [76] Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
  • 3. The examples above show how microfluidics can provide scalability, reduced reagent consumption, and throughput to enable large single cell studies that would otherwise be impractical or prohibitively expensive. However, these analyses still require off-chip cell handling and processing steps to generate products needed for qPCR. The inclusion of microfluidic cell handling and processing thus offers important avenues to improved throughput and cost, while also improving precision and sensitivity through small-volume confinement. To this end White et al. recently described a fully integrated microfluidic RT-qPCR device that implements all steps of cell trap- ping, lysis, cDNA synthesis, and qPCR analysis (Figure 1) at a throughput of 300 cells per run [38 ]. This system achieves improved precision and sensitivity over large- volume RT-qPCR analysis and offers a general solution for cell-handling automation and integration that may be adapted to a variety of amplification and analysis protocols [39], including high-throughput microfluidic qPCR, high- density digital PCR [40,41], sequencing, and microarray analysis. Single cell genomes In addition to gene expression analysis, microfluidic approaches are finding increasing applications in studying diversity and variations in single cell genomes, with applications spanning cancer biology to environmental microbiology. While new high-throughput sequencing instrumentation has made exome and whole-genome shotgun sequencing standard practice in cancer research, attention has now turned towards dissecting clonal heterogeneity. In one approach the sequencing of ampli- fied single nuclei was used to infer the clonal evolution of a breast tumor by measuring single cell copy number variations [24]. Scalable and low-cost implementations of single cell/nuclei isolation and amplification are likely to expand the power and pervasiveness of such analyses. In this line, Fan et al. recently reported a haplotyping approach on the basis of using a microfluidic device to amplify and genotype individual chromosomes isolated from a single cell [42 ]. The isolation and lysis of a single metaphase cell was used to distribute chromosomes across an array of 20 nL reaction chambers, followed by phi29 polymerase multiple displacement amplification [43], recovery of the isolated amplified products, and analysis on SNP arrays to establish haplotypes [42]. Beyond the utility in phasing genomes, more scalable implementations of this technology may soon allow for the study of heterogeneity in chromosome partitioning and translocation events in cancer. Single cell genome analysis is particularly compelling in the study of microbial organisms and communities. The vast majority of microorganisms on the planet have yet to be isolated in culture, necessitating metagenomic strat- egies that attempt to infer the identity and relative abundance of constituent members by sequencing and analysis of mixed pools of DNA. Microfluidic digital PCR in valve-based devices has been used to help untangle this genomic information by co-amplification of specific genes in isolated individual bacteria cells. This has been applied to establish co-existence of functional genes within a single organism and to reveal virus–host relation- ships in complex environments [44,45]. In a similar single cell genotyping application, microfluidic droplet-based systems for digital PCR have been used for the detection Microfluidic single cell analysis Lecault et al. 383 Figure 1 i ii iv iii (a) (b) (c) Current Opinion in Chemical Biology A microfluidic device for high-throughput single-cell RT-qPCR. (a) Fluorescence image of entire device showing 300 reactions after 40 cycles of PCR. (b) Enlarged view of individual reactors from (a) with dyes highlighting fluid paths (blue) and valves (red). Each array unit consists of (i) a reagent inlet, (ii) a 0.6 nL cell capture region with integrated cell traps, (iii) a 10 nL RT chamber and (iv) a 50 nL PCR chamber. Scale bar: 400 mm. (c) Optical micrograph of single cells trapped in the cell capture chambers (indicated by black arrows). Scale bar, 400 mm. Adapted from [38 ]. www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
  • 4. of pathogenic E. coli within a high background of non- pathogenic cells, achieving a detection limit of 1/105 [46]. The coupling of new low-bias whole genome amplification strategies [43] with sequencing provides a powerful approach to global single cell genome analysis and has been implemented in conventional tubes using single cell isolation by FACS or micropipette [23]. Microfluidic sys- tems offer an integrated ‘front-end’ solution to bacterial imaging, isolation, and processing, and have been shown to improve MDA performance by providing reduced ampli- fication bias in nanoliter volume reactions and suppression of contaminating DNA [23]. Although the physical separ- ation of single cells remains an important obstacle for many complex samples, this approach has been applied to obtain highly enriched metagenomes for T7 microbes from the human mouth [47], and has been further integrated with optical tweezer manipulation to obtain a novel genome for an ammonia-oxidizing archaeon [48]. Leung et al. have recently developed a programmable droplet-based format for single microbe isolation and multiparameter single-cell analysis [49]. This system provides improvements in- cluding facilitated single cell handling, flexible protocol development, and significantly higher throughput, and was used to dissect microbial diversity in environmental samples by whole genome amplification and sequencing of single cells and cell aggregates. Microfluidic analysis of proteins in single cells Beyond genomics applications, the scalability and small- volume advantages of microfluidic methods have increas- ingly found applications in the measurement of intra- cellular and secreted proteins from single cells. Taniguchi et al. used single molecule imaging in a parallel micro- fluidic format to measure intracellular protein expression, localization and abundance in a library of over 1018 Escherichia coli fluorescent protein fusion strains [50 ] (Figure 2a–c). Protein abundances were found to vary from 0.1 to 104 molecules per cell, spanning a dynamic range of over 5 orders of magnitude. At low copy numbers (10), differences in protein expression were attributed to intrinsic noise (e.g. stochastic binding of transcription factors to DNA promoter sites), whereas variations in higher expressed proteins, including almost all essential proteins, was generally governed by extrinsic fluctuations in cellular metabolites, ribosomes, and polymerases. Interestingly, for any given gene, a single cell’s protein and mRNA abundance were found to be uncorrelated, likely reflecting the relatively rapid degradation of mRNA compared to the long lifetimes of proteins in the cell. In contrast, Cheong and colleagues exploited the parallelization of microfluidic culture to reconstruct signaling kinetics using endpoint staining of fixed cells at different time points after stimulation, thereby enabling the detection of multiple proteins on each cell [51]. With 384 Analytical techniques Figure 2 Stage scan PDMS Bacteria Coverslip Objective lens Strain n-1 Strain n+1 Strain n 25 μm 10 μm Hybridoma cellAntibody capture beads 10 μm 150 μm (a) Side view (b) (c) (e) Adk Probability NormalizedFluorescence Protein copy number Time (min) (d) 0.1 0.08 0.06 0.04 0.02 0 1.2 1 0.8 0.6 0.4 0.2 0 20 40 60 80 100 120 0 500 1000 1500 2000 a = 6.8 b = 99 Current Opinion in Chemical Biology Microfluidic analysis of single cell intracellular (a)–(c) and extracellular (d) and (e) protein expression. (a) Microfluidic device for the parallel molecular imaging of multiple bacterial strains with single protein resolution. (b) High-resolution fluorescence imaging to quantify protein abundance in single E. coli cells using genetically fused fluorescent reporter strains. (c) Distribution of intracellular Adk protein in single E. coli cells. (d) A microfluidic fluorescence bead assay to measure antibody–antigen binding kinetics from antibodies secreted by a single cell. (e) Binding kinetics of a mouse mAb with hen egg lysozyme measured from a single bead.(a)–(c) Adapted from [50 ] with permission. (d) and (e) adapted from [59]. Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
  • 5. this method, it was shown that JNK signaling exhibited a binary switch-like response after anisomycin stimulation in HeLa cells [52]. Microfluidic-based immunocyto- chemistry has also been used to assess intratumoral and intertumoral heterogeneity in brain tumor samples, showing that cluster analysis of four intracellular sig- naling proteins could predict tumor progression and survival outcomes [53]. Single-cell protein measure- ments have also been performed by integrating micro- fluidic single-cell handling (i.e. sorting, lysis, labeling) with electrophoretic separation [54] or micro-patterned antibody capture arrays [55 ] In the latter approach, protein expression variability in a glioblastoma tumor cell line was measured using a quantitative fluorescent sandwich assay on multiple target proteins per cell [55 ]. This single-cell barcode chip (SCBC) approach is scal- able and has already been demonstrated at a throughput of tens of cells per chip with duplicate measurements of nine proteins. The concentration enhancement and rapid diffusive mix- ing afforded by subnanoliter microfluidic chambers has enabled the single-cell analysis of secreted effector proteins from immune cells (e.g. B cells, T cells, and macrophage). Ma et al. applied the SCBC to simul- taneously measure multiple cytokines (e.g. IL-10, TNF-b, IFN-g) from human macrophages and cytotoxic T lymphocytes (CTLs) obtained from both healthy donors and a metastatic melanoma patient [56]. Although not strictly microfluidic, open arrays of microfabricated chambers have also been used to screen and select B cells secreting antigen-specific antibodies from both immu- nized humans and mice [57,58]. This approach can be improved by using microfluidics to automate cell hand- ling and fluid-exchange, allowing antibodies from single cells to be directly screened by measuring antibody- antigen binding kinetics and specificities to different target antigens [59] (Figure 2d,e). An alternative approach uses microfluidic devices for the high-throughput analysis of secreted proteins from single cells by encapsulating them in emulsions consisting of subnanoliter aqueous droplets in oil [60,61]. Tumarkin and colleagues used a variant of this approach to study cellular paracrine signaling by co-encapsulating cells in microfluidic-generated agarose beads, and demonstrating that cell survival could be modulated by the ratio of MBA2 IL-3 secreting cells to M07e factor-dependent cells [62]. Microfluidic droplet generation is also being developed for drug screening applications by measuring viability of encapsulated single cells exposed to different chemical compositions [63 ]. Single cell growth and response phenotypes In addition to facilitating multiplexed genetic and protein analysis of single cells, microfluidic devices provide numerous advantages to the study of live cells such as precise spatio-temporal control of medium conditions, parallelization, and cell confinement for improved ima- ging. These capabilities are particularly well suited to measuring the kinetics and cellular heterogeneity of cell signaling. Because of the ease of genetic manipulation and cell culture, microfluidic approaches have been used extensively to study the response of archetypical protein signaling networks in yeast, most notably the pheromone and HOG mitogen activated protein kinase (MAPK) networks. The combination of microfluidic flow control, precise cell immobilization, fluorescent protein reporters, and image processing has been used to access several experimental regimes that are difficult or impossible to implement in bulk: the study of cell signaling under stable and precisely defined spatial gradients [64]; the frequency analysis of signaling response under oscillating stimulation conditions [65,66]; and the high-throughput analysis of network response under combined chemical and genetic perturbations [67,68]. Importantly, exper- iments that track individual cells through time are critical to understanding mechanisms of response heterogeneity, including effects of cell cycle, cell age, and familial relationships. For instance, Falconnet et al. found that nonuniform gene expression and phenotypic responses observed at intermediate concentrations of pheromone were not random, but rather correlated with genealogical relationships (Figure 3a), illustrating how nongenetic heritable traits may influence the critical threshold for cellular decision making [68]. In a similar vein, microfluidic approaches are becoming increasingly important as an in vitro method for studying complex and highly dynamic interactions that govern growth and differentiation of mammalian cells in vivo. These approaches have used both surface patterning of microwells to more precisely dissect and mimic factors present in the in vivo microenvironment [11,69], and automated fluid control to examine temporally varying medium conditions [70 ,71 ]. Tay and colleagues exploited the later approach to demonstrate that single 3T3 mouse fibroblast cells respond to tumor necrosis factor alpha (TNF-a) in a digital manner; that is, lower numbers of cells responded to lower doses of TNF-a even though the amplitude of transcription factor nuclear factor (NF-kB) remained high [71 ]. Pulse stimulation studies showed that prior exposure to TNF-a played a role in subsequent cellular responses, indicating that this pathway is not a purely stochastic system. In addition to deterministic control of applied factors, Molidena and colleagues reported the use of varying microfluidic flows to modulate the effect of endogenously secreted factors, showing that endocrine signaling is an important deter- minant of differentiation and cellular heterogeneity in murine embryonic stem cells [72 ]. Nonadherent cell types, including hematopoietic cells and industrially important suspension-adapted cell lines, Microfluidic single cell analysis Lecault et al. 385 www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
  • 6. present unique challenges for microfluidic analysis due to the need for immobilization during medium exchange. Lecault et al. reported a cell culture device optimized for suspension cell types that solves this problem by using gravity to drop cells into the bottom of high aspect ratio chambers (Figure 3b,c), thereby sequestering them from flow forces while maintaining the ability to exchange medium conditions using diffusion [70 ]. In addition, this device featured a thin cell-culture layer that was overlaid with a large medium reservoir, essentially blocking dehy- dration effects and allowing for robust growth and the selective recovery of viable cells following culture. The ability to perform immunohistochemistry on live cells without disturbing their spatial locations was used to correlate heterogeneous clonal growth of preleukemic cells with varying differentiation state. In a separate study, exposure of rare primary hematopoietic stem cells to temporally varying Steel factor (SF) concentrations revealed that this growth factor is critical for cell survival during a short window as cells exit quiescence, but does not directly influence cell growth kinetics. Single cell growth analysis Microfluidic systems greatly facilitate long-term time- lapse imaging studies of clonal growth and death kinetics under different medium conditions. This is perhaps one of the simplest and most underexploited assays of cellular heterogeneity. For instance, the early division kinetics of primary murine HSCs transduced with NUP98-HOXA10 homeodomain (NA10hd), a fusion gene known to stimu- late stem cell expansion in vitro, was investigated using a microfluidic cell culture array [73]. Clonal analysis of infected and control populations has confirmed that NA10hd did not affect cell cycling times, but that the growth advantage of overexpressing cells was due to an increase in self-renewal. In a different study, Albrecht et al. used the expression of H2B-EGFP to facilitate tracking of mitotic events in murine embryonic stem cells and reported an apparent synchronization of division events across the microfluidic device [74]. In addition to quantitative measures of clonal growth and death,genealogicalanalysis,allowingfor thereconstruction 386 Analytical techniques Figure 3 MAPK pathway GFP response320 min α-factor stimulation40 min α-factor stimulation (a) (b) (c) Current Opinion in Chemical Biology Microfluidic cell culture devices for live analysis of clonal heterogeneity. (a) Example of heterogeneous MAPK responses to alpha-factor pheromone being passed on from mother to daughter yeast cells. (b) Microfluidic chambers with high-aspect ratio for the culture of mammalian suspension cells. (c) Microfluidic cell culture array and image analysis show intraclonal heterogeneity in hematopoietic progenitor NUP98-HOXD13 cells. Green labels represent viable cells, red labels represent dead cells and blue lines highlight cell contours. Scale bar, 100 mm. (a) Adapted from [68]. Reproduced by permission of the Royal Society of Chemistry (RSC). (b) Reproduced from [70 ]. (c) Adapted from [70 ]. Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
  • 7. of the ‘family tree’ in each clone, provides a rich phe- notype for understanding cellular decision-making and retrospectively assessing the heterogeneity of the starting cell populations. Scherf and colleagues combined genea- logical tracing and surface patterning in a microfluidic device to show that the symmetry of human stem and progenitor cell divisions could be altered by exposure to exogenous factors [75]. The generation of division pedi- grees from a large number of single cells is assisted by microfluidic designs that confine clones in a limited space. In one example, microfluidic chambers were used to grow yeast colonies in linear geometries in order to facilitate the assignment of mother-daughter relationships, showing that observed bursts of heat shock protein expression are synchronized between a cell and its immediate progeny [76]. As a high-precision alternative to optical analysis of cellular growth, the Manalis group have developed an innovative approach that directly measures the mass of single cells using microfabricated cantilevers with inte- grated flow channels. This technique was used to show that heavier cells grew faster than lighter cells [77], and that cellular density could be used to distinguish subpopu- lations of blood cells [78]. Although this approach has not yet been adapted to high-throughput formats, it pro- vides an excellent illustration of how microfluidic approaches can enable previously inaccessible, and often even unexpected, measurement tools for understanding cellular heterogeneity. Future impact of microfluidic technologies Although microfluidic analysis methods have long been a focus of technology development, it is only recently that these tools are impacting biological and biomedical research. These are auspicious times. The pace of de- velopment and adoption is accelerating rapidly and the next few years will see many important technological developments and applications. Microfluidic cell proces- sing will emerge as a cornerstone of single cell genomics, enabling routine and scalable measurements of tran- scription and DNA sequence with improved perform- ance and economy. Although global proteomic analysis of single cells is still far away, requiring major advances in detection sensitivity and instrumentation, measure- ments of specific protein panels using either antibody capture or alternative specific reagents will continue to develop and should enable new avenues of inquiry ranging from immune response to cancer biology. Sim- ilarly, the profiling of metabolites in single cells con- stitutes an important area for continued advancement [79]. Another exciting prospect for dissecting complex cell populations is the integration of live-cell imaging and genealogical analysis with transcriptional and protein expression measurements, and the correlation of these with functional assays. While these microfluidic tools will be driven in the near term by interdisciplinary research teams, broad adoption and long-term impact will require increasing commercialization activities. New developments in programmable microfluidic devices will also be important in providing a general fluid-handling platform that may be adapted to niche or user-specific applications [49,80,81]. Given the rapid pace of development, the enormous impact of single cell approaches and the inherent advantages of micro- fluidic analysis, it would seem that answers to many of the biggest questions in single cell variability may very well come on a tiny chip. Acknowledgements The authors would like to thank Marketa Ricikova for providing the original images for Figure 3a. Funding support has been provided by grants from the Natural Sciences and Engineering Research Council (NSERC), Genome BC, Genome Canada, Western Diversification, the Terry Fox Foundation, and the Canadian Institute for Health Research (CIHR). The authors also thank the Michael Smith Foundation for Health Research (VL, AS, AKW, CLH), NSERC (VL, AS, AKW), and CIHR (CLH) for salary support. References and recommended reading Papers of particular interest, published within the period of review, have been highlighted as: of special interest of outstanding interest 1. Kent DG, Copley MR, Benz C, Wohrer S, Dykstra BJ, Ma E, Cheyne J, Zhao YJ, Bowie MB, Zhao Y et al.: Prospective isolation and molecular characterization of hematopoietic stem cells with durable self-renewal potential. Blood 2009, 113:6342-6350. 2. Wu M, Singh AK: Single-cell protein analysis. Curr Opin Biotechnol 2012, 23:83-88. 3. Wang D, Bodovitz S: Single cell analysis: the new frontier in ‘omics’. Trends Biotechnol 2010, 28:281-290. 4. Kalisky T, Blainey P, Quake SR: Genomic analysis at the single- cell level. Annu Rev Genet 2011, 45:431-445. 5. Tarnok A, Ulrich H, Bocsi J: Phenotypes of stem cells from diverse origin. Cytometry A 2010, 77A:6-10. 6. Tracy BP, Gaida SM, Papoutsakis ET: Flow cytometry for bacteria: enabling metabolic engineering, synthetic biology and the elucidation of complex phenotypes. Curr Opin Biotechnol 2010, 21:85-99. 7. Krutzik PO, Irish JM, Nolan GP, Perez OD: Analysis of protein phosphorylation and cellular signaling events by flow cytometry: techniques and clinical applications. Clin Immunol 2004, 110:206-221. 8. Bendall SC, Simonds EF, Qiu P, Amir E-aD, Krutzik PO, Finck R, Bruggner RV, Melamed R, Trejo A, Ornatsky OI et al.: Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 2011, 332:687-696. 9. Raj A, van den Bogaard P, Rifkin SA, van Oudenaarden A, Tyagi S: Imaging individual mRNA molecules using multiple singly labeled probes. Nat Methods 2008, 5:877-879. 10. Rieger MA, Hoppe PS, Smejkal BM, Eitelhuber AC, Schroeder T: Hematopoietic cytokines can instruct lineage choice. Science 2009, 325:217-218. 11. Lutolf MP, Doyonnas R, Havenstrite K, Koleckar K, Blau HM: Perturbation of single hematopoietic stem cell fates in artificial niches. Integr Biol 2009, 1:59-69. 12. Dykstra B, Ramunas J, Kent D, McCaffrey L, Szumsky E, Kelly L, Farn K, Blaylock A, Eaves C, Jervis E: High-resolution video monitoring of hematopoietic stem cells cultured in single-cell arrays identifies new features of self-renewal. Proc Natl Acad Sci U S A 2006, 103:8185-8190. Microfluidic single cell analysis Lecault et al. 387 www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
  • 8. 13. Cheong R, Rhee A, Wang CJ, Nemenman I, Levchenko A: Information transduction capacity of noisy biochemical signaling networks. Science 2011, 334:354-358. 14. Chen CF, Ridzon DA, Broomer AJ, Zhou ZH, Lee DH, Nguyen JT, Barbisin M, Xu NL, Mahuvakar VR, Andersen MR et al.: Real-time quantification of microRNAs by stem-loop RT-PCR. Nucl Acids Res 2005, 33:e179. 15. Dalerba P, Kalisky T, Sahoo D, Rajendran PS, Rothenberg ME, Leyrat AA, Sim S, Okamoto J, Johnston DM, Qian DL et al.: Single- cell dissection of transcriptional heterogeneity in human colon tumors. Nat Biotechnol 2011, 29:1120-1127. This study used high-throughput microfluidic qPCR arrays to dissect human colon cancer tissue by single-cell gene expression analysis, and showed that gene expression signatures were predictive of clinical outcomes. 16. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, Qian D, Lam JS, Ailles LE, Wong M et al.: Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458:780-783. 17. Guo G, Huss M, Tong GQ, Wang C, Sun LL, Clarke ND, Robson P: Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell 2010, 18:675-685. 18. Spike BT, Engle DD, Lin JC, Cheung SK, La J, Wahl GM: A mammary stem cell population identified and characterized in late embryogenesis reveals similarities to human breast cancer. Cell Stem Cell 2012, 10:183-197. 19. Yoo AS, Sun AX, Li L, Shcheglovitov A, Portmann T, Li Y, Lee- Messer C, Dolmetsch RE, Tsien RW, Crabtree GR: MicroRNA- mediated conversion of human fibroblasts to neurons. Nature 2011, 476:228-231. 20. Metzker ML: Applications of next-generation sequencing: sequencing technologies – the next generation. Nat Rev Genet 2010, 11:31-46. 21. Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, Wang X, Bodeau J, Tuch BB, Siddiqui A et al.: mRNA-Seq whole- transcriptome analysis of a single cell. Nat Methods 2009, 6:377-382. 22. Tang F, Lao K, Surani MA: Development and applications of single-cell transcriptome analysis. Nat Methods 2011, 8:S6-S11. 23. Lasken RS: Single-cell genomic sequencing using multiple displacement amplification. Curr Opin Microbiol 2007, 10:510-516. 24. Navin N, Kendall J, Troge J, Andrews P, Rodgers L, McIndoo J, Cook K, Stepansky A, Levy D, Esposito D et al.: Tumour evolution inferred by single-cell sequencing. Nature 2011, 472:90-94. 25. Tang F, Barbacioru C, Nordman E, Li B, Xu N, Bashkirov VI, Lao K, Surani MA: RNA-Seq analysis to capture the transcriptome landscape of a single cell. Nat Protocols 2010, 5:516-535. 26. Lu R, Neff NF, Quake SR, Weissman IL: Tracking single hematopoietic stem cells in vivo using high-throughput sequencing in conjunction with viral genetic barcoding. Nat Biotechnol 2011, 29:928-933. 27. Yin H, Marshall D: Microfluidics for single cell analysis. Curr Opin Biotechnol 2012, 23:110-119. 28. Zare RN, Kim S: Microfluidic platforms for single-cell analysis. Annu Rev Biomed Eng 2010, 12:187-201. 29. Mestdagh P, Feys T, Bernard N, Guenther S, Chen C, Speleman F, Vandesompele J: High-throughput stem-loop RT-qPCR miRNA expression profiling using minute amounts of input RNA. Nucl Acids Res 2008, 36:e143. 30. Bengtsson M, Hemberg M, Rorsman P, Stahlberg A: Quantification of mRNA in single cells and modelling of RT- qPCR induced noise. BMC Mol Biol 2008, 9:63. 31. Bengtsson M, Stahlberg A, Rorsman P, Kubista M: Gene expression profiling in single cells from the pancreatic islets of Langerhans reveals lognormal distribution of mRNA levels. Genome Res 2005, 15:1388-1392. 32. Tang F, Hajkova P, Barton SC, Lao K, Surani MA: MicroRNA expression profiling of single whole embryonic stem cells. Nucl Acids Res 2006, 34:e9. 33. Diercks A, Kostner H, Ozinsky A: Resolving cell population heterogeneity: real-time PCR for simultaneous multiplexed gene detection in multiple single-cell samples. PLoS ONE 2009, 4:e6326. 34. Narsinh KH, Sun N, Sanchez-Freire V, Lee AS, Almeida P, Hu S, Jan T, Wilson KD, Leong D, Rosenberg J et al.: Single cell transcriptional profiling reveals heterogeneity of human induced pluripotent stem cells. J Clin Investig 2011, 121:1217-1221. 35. Petriv O, Kuchenbauer F, Delaney A, Lecault V, White A, Kent D, Marmolejo L, Heuser M, Berg T, Copley M et al.: Comprehensive microRNA expression profiling of the hematopoietic hierarchy. Proc Natl Acad Sci U S A 2010, 107:15443-15448. 36. Flatz L, Roychoudhuri R, Honda M, Filali-Mouhim A, Goulet J-P, Kettaf N, Lin M, Roederer M, Haddad EK, Sekaly RP et al.: Single- cell gene-expression profiling reveals qualitatively distinct CD8 T cells elicited by different gene-based vaccines. Proc Natl Acad Sci U S A 2011, 108:5724-5729. 37. Kikushige Y, Ishikawa F, Miyamoto T, Shima T, Urata S, Yoshimoto G, Mori Y, Iino T, Yamauchi T, Eto T et al.: Self- renewing hematopoietic stem cell is the primary target in pathogenesis of human chronic lymphocytic leukemia. Cancer Cell 2011, 20:246-259. 38. White AK, VanInsberghe M, Petriv OI, Hamidi M, Sikorski D, Marra MA, Piret J, Aparicio S, Hansen CL: High-throughput microfluidic single-cell RT-qPCR. Proc Natl Acad Sci U S A 2011, 108:13999-14004. The authors addressed the bottleneck between cell handling and high- density qPCR arrays by developing a fully integrated microfluidic RT- qPCR device that implements all steps of cell trapping, lysis, cDNA synthesis, and qPCR analysis at a throughput of 300 cells per run. 39. Bontoux N, Dauphinot L, Vitalis T, Studer V, Chen Y, Rossier J, Potier MC: Integrating whole transcriptome assays on a lab- on-a-chip for single cell gene profiling. Lab Chip 2008, 8:443-450. 40. Warren L, Bryder D, Weissman IL, Quake SR: Transcription factor profiling in individual hematopoietic progenitors by digital RT-PCR. Proc Natl Acad Sci U S A 2006, 103:17807-17812. 41. Heyries KA, Tropini C, VanInsberghe M, Doolin C, Petriv OI, Singhal A, Leung K, Hughesman CB, Hansen CL: Megapixel digital PCR. Nat Methods 2011, 8:649-651. 42. Fan HC, Wang J, Potanina A, Quake SR: Whole-genome molecular haplotyping of single cells. Nat Biotechnol 2011, 29:51-57. The authors developed of a microfluidic device capable of amplifying and genotyping individual chromosomes from a single cell. This technique is scalable, and may be used in conjunction with SNP arrays and sequen- cing for applications in personal genomics such as haplotyping. 43. Dean FB, Hosono S, Fang LH, Wu XH, Faruqi AF, Bray-Ward P, Sun ZY, Zong QL, Du YF, Du J et al.: Comprehensive human genome amplification using multiple displacement amplification. Proc Natl Acad Sci U S A 2002, 99:5261-5266. 44. Ottesen EA, Hong JW, Quake SR, Leadbetter JR: Microfluidic digital PCR enables multigene analysis of individual environmental bacteria. Science 2006, 314:1464-1467. 45. Tadmor AD, Ottesen EA, Leadbetter JR, Phillips R: Probing individual environmental bacteria for viruses by using microfluidic digital PCR. Science 2011, 333:58-62. 46. Zeng Y, Novak R, Shuga J, Smith MT, Mathies RA: High- performance single cell genetic analysis using microfluidic emulsion generator arrays. Anal Chem 2010, 82:3183-3190. 47. Marcy Y, Ishoey T, Lasken RS, Stockwell TB, Walenz BP, Halpern AL, Beeson KY, Goldberg SMD, Quake SR: Nanoliter reactors improve multiple displacement amplification of genomes from single cells. PLoS Genet 2007, 3:1702-1708. 388 Analytical techniques Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com
  • 9. 48. Blainey PC, Mosier AC, Potanina A, Francis CA, Quake SR: Genome of a low-salinity ammonia-oxidizing archaeon determined by single-cell and metagenomic analysis. PLoS ONE 2011, 6:e16626. 49. Leung K, Zahn H, Leaver T, Konwar KM, Hanson NW, Page´ AP, Lo C-C, Chain PS, Hallam S, Hansen CL: A programmable droplet- based microfluidic device applied to multiparameter analysis of single microbes and microbial communities. Proc Natl Acad Sci U S A 2012, http://dx.doi.org/10.1073/pnas.1106752109, in press. 50. Taniguchi Y, Choi P, Li G, Chen H, Babu M, Hearn J, Emili A, Xie X: Quantifying E-coli proteome and transcriptome with single- molecule sensitivity in single cells. Science 2010, 329:533-538. The authors used a microfluidic device to perform parallel measurements of single-cell protein and mRNA expression with single-molecule sensi- tivity across over 1000 different strains of Escherichia coli. 51. Cheong R, Wang CJ, Levchenko A: High content cell screening in a microfluidic device. Mol Cell Proteomics 2009, 8:433-442. 52. Fosbrink M, Aye-Han NN, Cheong R, Levchenko A, Zhang J: Visualization of JNK activity dynamics with a genetically encoded fluorescent biosensor. Proc Natl Acad Sci U S A 2010, 107:5459-5464. 53. Sun J, Masterman-Smith MD, Graham NA, Jiao J, Mottahedeh J, Laks DR, Ohashi M, DeJesus J, Kamei K-i, Lee K-B et al.: A microfluidic platform for systems pathology: multiparameter single-cell signaling measurements of clinical brain tumor specimens. Cancer Res 2010, 70:6128-6138. 54. Huang B, Wu H, Bhaya D, Grossman A, Granier S, Kobilka BK, Zare RN: Counting low-copy number proteins in a single cell. Science 2007, 315:81-84. 55. Shi Q, Qin L, Wei W, Geng F, Fan R, Shin YS, Guo D, Hood L, Mischel PS, Heath JR: Single-cell proteomic chip for profiling intracellular signaling pathways in single tumor cells. Proc Natl Acad Sci U S A 2011, 109:419-424. The authors describe a microfluidic ‘Single Cell Barcode Chip’ (SCBC) for multiplexed profiling of over 10 different cytokines and effector proteins secreted by single macrophage and T cells, and demonstrate how this tool may be useful for clinical monitoring of the immune system. 56. Ma C, Fan R, Ahmad H, Shi Q, Comin-Anduix B, Chodon T, Koya RC, Liu C-C, Kwong GA, Radu CG et al.: A clinical microchip for evaluation of single immune cells reveals high functional heterogeneity in phenotypically similar T cells. Nat Med 2011, 17:738-743. 57. Story CM, Papa E, Hu C-CA, Ronan JL, Herlihy K, Ploegh HL, Love JC: Profiling antibody responses by multiparametric analysis of primary B cells. Proc Natl Acad Sci U S A 2008, 105:17902-17907. 58. Jin A, Ozawa T, Tajiri K, Obata T, Kondo S, Kinoshita K, Kadowaki S, Takahashi K, Sugiyama T, Kishi H et al.: A rapid and efficient single-cell manipulation method for screening antigen-specific antibody-secreting cells from human peripheral blood. Nat Med 2009, 15:1088-1092. 59. Singhal A, Haynes CA, Hansen CL: Microfluidic measurement of antibody–antigen binding kinetics from low-abundance samples and single cells. Anal Chem 2010, 82:8671-8679. 60. Koester S, Angile FE, Duan H, Agresti JJ, Wintner A, Schmitz C, Rowat AC, Merten CA, Pisignano D, Griffiths AD et al.: Drop- based microfluidic devices for encapsulation of single cells. Lab Chip 2008, 8:1110-1115. 61. Konry T, Dominguez-Villar M, Baecher-Allan C, Hafler DA, Yarmush ML: Droplet-based microfluidic platforms for single T cell secretion analysis of IL-10 cytokine. Biosens Bioelectron 2011, 26:2707-2710. 62. Tumarkin E, Tzadu L, Csaszar E, Seo M, Zhang H, Lee A, Peerani R, Purpura K, Zandstra PW, Kumacheva E: High- throughput combinatorial cell co-culture using microfluidics. Integr Biol 2011, 3:653-662. 63. Brouzes E, Medkova M, Savenelli N, Marran D, Twardowski M, Hutchison JB, Rothberg JM, Link DR, Perrimon N, Samuels ML: Droplet microfluidic technology for single-cell high- throughput screening. Proc Natl Acad Sci U S A 2009, 106:14195-14200. The authors present a droplet-based microfluidic technology for high- throughput encapsulation of single mammalian cells and cytotoxicity screening against a library of drugs. 64. Paliwal S, Iglesias PA, Campbell K, Hilioti Z, Groisman A, Levchenko A: MAPK-mediated bimodal gene expression and adaptive gradient sensing in yeast. Nature 2007, 446:46-51. 65. Bennett MR, Pang WL, Ostroff NA, Baumgartner BL, Nayak S, Tsimring LS, Hasty J: Metabolic gene regulation in a dynamically changing environment. Nature 2008, 454:1119-1122. 66. Hersen P, McClean MN, Mahadevan L, Ramanathan S: Signal processing by the HOG MAP kinase pathway. Proc Natl Acad Sci U S A 2008, 105:7165-7170. 67. Taylor RJ, Falconnet D, Niemisto A, Ramsey SA, Prinz S, Shmulevich I, Galitski T, Hansen CL: Dynamic analysis of MAPK signaling using a high-throughput microfluidic single-cell imaging platform. Proc Natl Acad Sci U S A 2009, 106:3758-3763. 68. Falconnet D, Niemisto A, Taylor RJ, Ricicova M, Galitski T, Shmulevich I, Hansen CL: High-throughput tracking of single yeast cells in a microfluidic imaging matrix. Lab Chip 2011, 11:466-473. 69. Gobaa S, Hoehnel S, Roccio M, Negro A, Kobel S, Lutolf MP: Artificial niche microarrays for probing single stem cell fate in high throughput. Nat Methods 2011, 8:949-955. 70. Lecault V, VanInsberghe M, Sekulovic S, Knapp DJHF, Wohrer S, Bowden W, Viel F, McLaughlin T, Jarandehei A, Miller M et al.: High-throughput analysis of single hematopoietic stem cell proliferation in microfluidic cell culture arrays. Nat Methods 2011, 8:581-586. This article demonstrates robust long-term microfluidic culture of pri- mary hematopoietic stem and progenitor single cells with the integration of an iso-osmotic bath to block evaporation. The design features an array of high aspect ratio chambers and allows for medium exchange, temporal stimulation, time-lapse imaging, live immunostaining and clone recovery. 71. Tay S, Hughey JJ, Lee TK, Lipniacki T, Quake SR, Covert MW: Single-cell NF-kappa B dynamics reveal digital activation and analogue information processing. Nature 2010, 466:267-271. This study combines fluorescent gene expression with dynamic micro- fluidic cell culture to measure NF-kB activity in response to temporal stimulation with TNF-a in single mammalian cells. 72. Moledina F, Clarke G, Oskooei A, Onishi K, Gunther A, Zandstra PW: Predictive microfluidic control of regulatory ligand trajectories in individual pluripotent cells. Proc Natl Acad Sci U S A 2012, 109:3264-3269. The authors study the effects of autocrine and paracrine signaling on cell fate in murine embryonic stem cells by precisely modulating laminar flow in microfluidic channels. 73. Sekulovic S, Gasparetto M, Lecault V, Hoesli CA, Kent DG, Rosten P, Wan A, Brookes C, Hansen CL, Piret JM et al.: Ontogeny stage-independent and high-level clonal expansion in vitro of mouse hematopoietic stem cells stimulated by an engineered NUP98-HOX fusion transcription factor. Blood 2011, 118:4366-4376. 74. Albrecht DR, Underhill GH, Resnikoff J, Mendelson A, Bhatia SN, Shah JV: Microfluidics-integrated time-lapse imaging for analysis of cellular dynamics. Integr Biol 2010, 2:278-287. 75. Scherf N, Franke K, Glauche I, Kurth I, Bornhauser M, Werner C, Pompe T, Roeder I: On the symmetry of siblings: automated single-cell tracking to quantify the behavior of hematopoietic stem cells in a biomimetic setup. Exp Hematol 2012, 40:119-130. 76. Rowat AC, Bird JC, Agresti JJ, Rando OJ, Weitz DA: Tracking lineages of single cells in lines using a microfluidic device. Proc Natl Acad Sci U S A 2009, 106:18149-18154. 77. Godin M, Delgado FF, Son SM, Grover WH, Bryan AK, Tzur A, Jorgensen P, Payer K, Grossman AD, Kirschner MW et al.: Using Microfluidic single cell analysis Lecault et al. 389 www.sciencedirect.com Current Opinion in Chemical Biology 2012, 16:381–390
  • 10. buoyant mass to measure the growth of single cells. Nat Methods 2010, 7:387-390. 78. Grover WH, Bryan AK, Diez-Silva M, Suresh S, Higgins JM, Manalis SR: Measuring single-cell density. Proc Natl Acad Sci U S A 2011, 108:10992-10996. 79. Dada OO, Essaka DC, Hindsgaul O, Palcic MM, Prendergast J, Schnaar RL, Dovichi NJ: Nine orders of magnitude dynamic range: picomolar to millimolar concentration measurement in capillary electrophoresis with laser induced fluorescence detection employing cascaded avalanche photodiode photon counters. Anal Chem 2011, 83:2748-2753. 80. Fidalgo LM, Maerkl SJ: A software-programmable microfluidic device for automated biology. Lab Chip 2011, 11:1612-1619. 81. Srigunapalan S, Eydelnant IA, Simmons CA, Wheeler AR: A digital microfluidic platform for primary cell culture and analysis. Lab Chip 2012, 12:369-375. 390 Analytical techniques Current Opinion in Chemical Biology 2012, 16:381–390 www.sciencedirect.com