Sei sulla pagina 1di 56

Accepted Manuscript

Title: Biochemical analysis on microfluidic chips

Author: Jing Wu, Ziyi He, Qiushui Chen, Jin-Ming Lin

PII: S0165-9936(15)30145-X
DOI: http://dx.doi.org/doi: 10.1016/j.trac.2016.03.013
Reference: TRAC 14704

To appear in: Trends in Analytical Chemistry

Please cite this article as: Jing Wu, Ziyi He, Qiushui Chen, Jin-Ming Lin, Biochemical analysis
on microfluidic chips, Trends in Analytical Chemistry (2016), http://dx.doi.org/doi:
10.1016/j.trac.2016.03.013.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service
to our customers we are providing this early version of the manuscript. The manuscript will
undergo copyediting, typesetting, and review of the resulting proof before it is published in its
final form. Please note that during the production process errors may be discovered which could
affect the content, and all legal disclaimers that apply to the journal pertain.
Biochemical Analysis on Microfluidic Chips
Jing Wua, Ziyi Heb, Qiushui Chenb, Jin-Ming Linb*
a
School of Science, China University of Geosciences (Beijing), Beijing 100083,
China
b
Department of Chemistry, Beijing Key Laboratory of Microanalytical Methods and
Instrumentation, Tsinghua University, Beijing 100084, China

Highlights
 Biochemical analysis plays a crucial role in understanding mechanism of life
activities and giving biological insights into life process.
 Microfluidic chip shows great promise in biochemical analysis field.
 Recent advances in integrated technologies on microfluidic chip for
biochemical analysis have been reviewed.
 New paradigms of biochemical analysis on microfluidic platforms have been
summarized.

Abstract
Biochemical analysis is crucial in understanding mechanism of life activities and
giving biological insights into life process. With inherent merits in flexible design,
microscale of operation, good incorporation with other techniques for manipulation
and detection, microfluidic chip has introduced new paradigms in biochemical
analysis field. Considering the explosive development of microfluidics over the past
decades, in this review, we summarized recent advances in biochemical analysis on
microfluidic platforms. Highlight was put on the integrated technologies such as
optical, electrical, acoustic and magnetic techniques. Focus also was given on relative
applications in biomimetics, drug screening, biomolecular detection, single cell and
stem cell analysis.
Keywords: biochemical analysis; microfluidic chip; optical detector; electronic
methods; acoustic wave; magnetic operation; chip-MS platform; biomimetics; drug

Page 1 of 55
discovery; biomolecular analysis; single cell; stem cell.

1. Introduction
In native cellular microenvironment, cells are subject to complex biochemical cues
that vary in temporal and spatial scales [1]. These cues can be divided into soluble and
insoluble signaling molecules, including chemokines [2], gradients of cytokines [3],
growth factor secretion from neighboring cells [4], and even biophysical interactions
with the extracellular matrix (ECM) [5]. The autocrine and paracrine signals are
sensed by cells in different ways and regulate cell physiology temporally and spatially.
Meanwhile, cells exert some factors to alter their surrounding microenvironment [6,
7]. Probing the biochemical processes could govern cell behavior and show great
current and potential impact on the bio-analytical chemistry researches. However,
most in vitro cell-based biochemical experiments are performed in two-dimensional
(2D) manner that cells are cultured onto plastic surfaces and treated by coating them
with various solutions [8, 9]. The standard 2D conditions poorly mimic the cellular
microenvironment and are absent of three-dimensional (3D) cues.
Fortunately, microsystems bring unprecedented opportunities for giving analytical
insights into cells and tissues [1, 10-12]. Microfluidic chips enable cells reside in a
more physiologically relevant context and present cells with biochemical cues in a
controllable and reproducible way [13-15]. 3D cell culture on microfluidic chips
makes it possible to retain native tissue-specific functions of cells and recapitulate
tissue-tissue interfaces, spatiotemporal chemical gradients [16, 17]. Various artificial
organ systems have been particularly constructed on microfluidic platforms to
reconstitute the critical organ functions [18, 19]. By integrating with biochemical
analysis techniques, microdevices show great promise for basic biomedical and
pharmaceutical researches. Over the past decade, the microfluidic chips have been
recognized as ideal platforms for biochemical assays which have been widely
expanded to tissue engineering, drug screening, biomolecular detection, single cell
and stem cell analysis (Fig. 1).

Page 2 of 55
Fig. 1
In this Review, we describe methods and techniques of biochemical analysis on a
micrometer scale and discuss the application of these microtechnologies in biological
sample analysis. We further present a critical survey of recent advances and future
trends of biochemical analysis in microsystems. Emphasis is placed on works aimed
at gaining biological insights, as well as on efforts to realize advanced biochemical
analysis on chips.

2. Biochemical analysis techniques on chips


Almost every conventional analytical tool in biochemistry labs has an equivalent
microfabricated counterpart on chips. With tremendous advances, microfluidics could
integrate technologies in various fields to perform rapid and reproducible analysis on
small sample volumes as well as eliminate the labor-intensive and potentially
error-prone laboratory manipulations. Here, we highlight several kinds of analytical
methods applied in microfluidic chips in recent years.
2.1 Optical detector
Due to the ubiquitous optical instrumentation in laboratories, quantitative detections
of samples by optical techniques become common in microsystems. Conventional
optical detection technologies, such as absorbance, fluorescence, infrared (IR) and
surface plasmon resonance (SPR), have all been applied in microfluidic chips. Single
mouse hybridoma cells, a fluorescent probe and single beads coated with anti-mouse
IgG antibodies were co-compartmentalized in droplets to generate a distinguishable
fluorescence signal enabling intracellular, cell-surface or secreted protein screening
(Fig. 2) [20]. The production rate of fluorescent proteins in bacteria was monitored on
microfluidic chip revealing a long period of constant protein production in stationary
phase [21]. Microscopic imaging of whole aging process of budding yeast was
successfully conducted on a microfluidic dissection platform integrated with arrays of
soft elastomer-based micropads [22]. Spatially resolved IR spectroscopy as an
analytical method possessing characteristic of label-free and nondestructive detection
can provide spatiotemporal information on functional groups of biomolecules through

Page 3 of 55
observing their characteristic vibrational modes.
Fig. 2
With the help of spatially resolved IR spectroscopy, Holman et al. [23] monitored the
spatiotemporal chemical changes in living PC12 cells and found that the cells at the
boundary and center of the colony tended to produce more glycogen and glycoprotein
on an open-channel membrane device. The device composed of a gold-coated porous
membrane between a feeding channel and a viewing chamber to allow long-term
continuous IR measurement of live, adherent mammalian cells. On-chip protein
microarray was fabricated by in vitro transcription and translation to be implemented
for SPR imaging in both clinical and research applications [24]. Cytosolic glucose
levels in yeast were analyzed using genetically encoded Förster resonance energy
transfer (FRET) sensors. Glucose-induced conformational change in the bacterial
periplasmic glucose/galactose binding protein MglB was detected by monitoring
FRET signals between two fluorescent protein variants [25]. Optical method was even
used to pattern different types of cells at single-cell patterning precision. The
propagating light signals of the patterned cells could be detected in real-time giving
promise for the detection of transduction signals among patterned cells [26].
All the above conventional detection units which also called “off-chip” strategies are
separated from the chip platform. While optofluidic is an “on-chip” mode in which
the optics are integrated with or fabricated together with the fluidic functional units
and the optical property of the integrated optics can be tuned by fluidic technology.
All the relevant units, including light sources, filters, mirrors, waveguides, lenses and
detectors, are integrated on the chip. Kim et al. [27] conducted a single-molecule
optofluidic analysis on a microfluidic mixing device which integrated valves and
pumps to accurately accomplish titration of biomolecules with picoliter resolution. An
optical fiber-based, on-chip detection unit with a droplet-based microfluidic unit was
integrated onto a single-layer, optofluidic device for real-time, high-throughput,
quantitative analysis of droplet contents. A detection throughput of 2000 droplets per
second, a detection limit of 20 nM and an excellent reproducibility were achieved on
the device [28]. A random gold nanoisland substrate was used for laser-controlled
4

Page 4 of 55
optofluidic manipulation based on plasmonic heating. Microfluidic flow guiding,
valving and mixing were firstly realized within the same functional substrate [29].
The more functional units are integrated on the chip, the degree of miniaturization of
the microdevice is higher. With the advance of manufacturing technologies, full
integration of optical elements onto microfluidic chip is becoming feasible.
2.2 Electronic methods
Electrical analysis of biological samples on microfluidic platforms shows prominent
advantages that rich expertise on recording and processing of electronic signals,
readily available technologies for miniaturization and integration, and possibility for
label-free and non-invasive detection [30]. By giving information on cell membrane
capacitance and resistance and cytoplasm conductivity and permittivity, impedance
spectroscopy has been developed as a robust tool for characterization of biological
cells. Its application has been expanded to areas of drug screening [31-34], cellular
biology [35], neuroscience [36-38] and disease diagnosis [39, 40]. Automated
quantification of ovarian cancer cells from whole blood was reported on an
electrochemical Lab-on-a-Disc platform by detecting impedance [41]. The
combination of peptides with electrodes opens a robust, evolutionary avenue in
studies of the interactions between bacteria and antimicrobial peptides (AMPs).
Mannoor et al. [42] reported selective and sensitive detection of infectious agents via
impedance spectroscopy based on hybridization of the AMP magainin I with
functionalized microcapacitive electrode arrays. These devices with electronic
read-out may work as portable pathogen detectors (Fig. 3). Flow impedance
measurement of cells on microfluidic devices provides several advantages over
conventional techniques, such as high sensitivity, saving reagent and sample,
integration of reference measurement electrodes, the use of sheath flow or
dielectrophoretic forces for cell centering and the possibility of online implementation
of cell sorting [43].
Fig. 3
Dielectrophoresis (DEP)-based methods also serve as powerful strategies for
continuous-flow cell sorting on chip. Renaud et al. [44-46] developed
5

Page 5 of 55
equilibrium-based continuous cell sorters based on DEP. Applying opposite negative
DEP force fields from electrodes defines a position of equilibrium for the dielectric
particles placed in these fields. In addition to the equilibrium-based sorting device,
some novel cell sorting microdevices based on DEP have been reported such as
Raman-activated cell sorting [47], label-free stem cell sorting [48, 49], DEP viable
cell sorting [50, 51] and electrodeless DEP [52, 53]. Single cells were captured and
repulsed by DEP forces generated by a bipolar electrode. Both faradaic ion
enrichment and depletion zones generated by bipolar electrodes can be exploited to
shape and extend the electric field gradients that are responsible for DEP force. DEP
technologies for manipulating biological cells also show many distinct merits over
other cell-handling techniques including label-free selectivity, cost-saving device
components, and amenability to single-cell and array-based applications [54].
Currently, many novel analytical approaches based on electronic detection at high
speed have been developed on chips. Western blotting assay was conducted in a single
glass microchannel under purely electronic control. Western blotting on microfluidic
platform is an effort trying to reach the long-sought bioanalytical goal in the life
sciences that developing rapid and quantitative analysis methods [55]. Protein
biomarkers in complex biological samples were detected by multiplexed
immunoarrays on microfluidic devices [56-58]. Electrodes even were printed on
paper-based microfluidic device to achieve some biological and chemical applications
such as cancer cell detection [59], enzyme-linked immunosorbent assay (ELISA) [60].
All of them are the results of the technological advances in microfluidics,
microelectronics and electrophysiology.
2.3 Acoustic wave
The introduction of surface acoustic wave (SAW) technology onto lab-on-a-chip
platforms has opened a new frontier in microfluidics. Numerous advantages are
provided by SAW microfluidics: compact and inexpensive devices and accessories,
fast fluid actuation, contact-free particle manipulation, and compatibility with other
microfluidic components [61]. According to the property of acoustic wave, SAW
microfluidics are divided into two types: travelling SAW (TSAW) and standing SAW
6

Page 6 of 55
(SSAW). Ultrafast particle and cell concentration are essential to the success of
subsequent analytical procedures and the development of miniaturized biological and
chemical sensors. TSAW devices were used to excite a MHz-order acoustic wave that
propagated into a microlitre droplet to facilitate spatial partitioning between two
different sized suspended microparticles [62]. A novel microfluidic cell sorter
operating in continuous flow at high sorting rates was successfully used to direct
HaCaT cells, fibroblasts from mice and MV3 melanoma cells by acoustic streaming
excited by TSAW [63]. TSAW also allows fast drop formation and real time control of
drop size at quick response times over a wide range [64]. A sophisticated fluid
manipulation tool based on TSAW was demonstrated to carry out the complex
sequence of fluidic manipulations to detect the rodent malaria parasite in blood [65].
Acoustic fields were used to produce the required rotational vortices to mechanically
lyse both the red blood cells and the parasitic cells presented in a drop of blood (Fig.
4a).
Fig. 4
In contrast to TSAW-based devices harnessing the acoustic streaming, SSAW-based
devices use the primary acoustic radiation forces acting on particles via the
surrounding fluid. On-chip functional blocks for sample preprocessing are
indispensable elements for fully portable micrototal analysis systems. Pretreatment of
whole-blood samples [66] and cell washing [67] were successfully conducted on the
SSAW-based microfluidic device. Similar to optical tweezers, “acoustic tweezers”
based on SSAW was provided to trap and manipulate single cells and entire organisms
in a single-layer microfluidic chip (Fig. 4b). The acoustic tweezers utilize the wide
resonance band of chirped interdigital transducers to achieve real-time control of a
SSAW field which enables flexible manipulation of most known microparticles. The
tweezers give promise as powerful tools for many disciplines of science and
engineering [68]. Supported lipid bilayers (SLBs) are crucial in cellular biology
because they regulate the intracellular and intercellular movement of ions, proteins
and other molecules [69]. As a result, SLBs work as a critically important tool in
studying the physical properties of biological membranes and cells. SSAW were used
7

Page 7 of 55
to generate lateral standing waves leading to a lateral modulation in lipid
concentration. This pattern formation was demonstrated to be reversible and brought
no effect on the integrity of the lipid bilayer [70]. Arrays of molecules, such as DNA
[71] and proteins [72], were bounded to SLBs to enable on-chip screening and
binding assays. Titled-angle SSAW (ta-SSAW)-based microfluidic device was capable
of high-throughput separation of circulating tumor cells (CTCs) from peripheral blood
samples obtained from cancer patients (Fig. 4c). In the fields of cancer research,
diagnostics, drug efficacy assessment, and therapeutics, this method will become an
invaluable supplement tool owning to its simple design, label-free automated
operation and capability to isolate rare CTCs in a viable state [73]. Ta-SSAW also was
introduced to microfluidic channel for improving the efficiency and sensitivity of
acoustic separation techniques. Human breast cancer cells were effectively separated
from non-malignant leukocytes, while preserving the integrity [74]. An organized cell
co-culture platform was constructed by sequentially patterning different types of cells
in the SSAW field. Cell migration dynamics of epithelial cancer cells was real-time
monitored on this platform when they were co-cultured with endothelial cells [75].
SAW-based microfluidics has come a long way in the past decade. Its applications
have been spanned the full spectrum of lab functions.
2.4 Magnetic operation
Utilization of magnetic particles has a combination of advantages that competing
techniques lack: a large surface-to-volume ratio, convenient bio-functionalization,
manipulation by magnetic fields, simplified extraction and buffer replacement steps.
Manipulation of magnetic particles by magnetic fields is compatible with system
miniaturization on account of the following reasons: (1) the tendency of explorations
toward miniaturization has a solid basis that availability of magnetic particles and
corresponding assay reagents; (2) the magnetic fields are strong close to the field
generators; (3) magnetic field gradients are large in the vicinity of structures with high
curvature; (4) only short distances need to be travelled in miniaturized devices [76].
Magnetic particles have been applied in mixing fluids, selective capturing,
concentrating, transferring, labeling specific analytes, and probing biophysical
8

Page 8 of 55
properties of analytes.
Rapid mixing of fluids or solutions is a necessary step in most point-of-care testing
devices and has been a topic of everlasting interest in the microfluidic field. Magnetic
field was used to assemble an artificial biomimetic cilia as long chains of spherical
superparamagnetic particles and to actuate the cilia in a simple nonreciprocal manner
resulting in a fluid flow [77]. A flow system consisting of a water-based ferrofluid and
a mixture of DI water and glycerol was mixed by the interaction between a uniform
magnetic field and a magnetic fluid in a microfluidic chamber [78]. The controlled
rotation and torque was generated by uniaxial magnetic microactuators formed by two
bound superparamagnetic particles in a fluid. This demonstration opened a range of
possibilities in lab-on-a-chip applications, such as the actuation of single molecules,
fluid mixing in microfluidic chambers and novel cluster-based assays [79].
High surface-to-volume ratio and many bio-functionalization options are beneficial
for magnetic particles to capture, concentrate, transfer and label analyte in biological
samples. A novel microfluidic platform using “phase-transfer magnetophoresis”
enabled continuous DNA extraction from bacterial Escherichia coli which was
cultured with superparamagnetic bead suspension, lysis and binding buffers (Fig. 5a).
These DNA carrying beads were subsequently transported across the interfaces
between co-flowing laminar streams of sample mixture, washing and elution buffer
actuating by applying a time-varying magnetic field generated by a rotating
permanent magnet [80]. Magneto-capillary valve technology also was reported to be a
flexible approach for integrating purification and enrichment of nucleic acids and
proteins [81]. Transportation of magnetic beads between multiple microfluidic
chambers was even realized to be automated on a centrifugal microfluidic cartridge
“LabDisk” [82]. A concept of multidimensional magnetic and optical barcoding of
droplets based on a magnetiofluidic platform was presented. The platform comprised
multiple functional blocks: encoding area, encoded droplet pool and magnetic
decoding area (Fig. 5b). The platform paved the way for the development of novel
non-optical encoding schemes for multiplexed droplet-based biological assays [83].
Fig. 5
9

Page 9 of 55
After the capture, concentration, transferring and labeling of target analytes, further
processing is accurate and specific detection. Concentration of avidin added to the
solution could be quantitatively determinated by monitoring changes in the amplitude
of the optical transmittance of the solution which was caused by self-assembled
chains of functionalized superparamagnetic beads rotating in the solution (Fig. 5c).
This protocol offered a rapid, highly sensitive, inexpensive and homogeneous means
of detecting biorecognition processes [84]. Magnetic particle-scanning for on-chip
ultrasensitive immunodetection also was described. A magnetic particle carrying a
low number of antigens was transported by hydrodynamic forces and was subjected to
a pattern of substrate-bound small magnetic particles functionalized with antibodies
[85]. An electromagnetically actuated platform was demonstrated to perform nucleic
acid extraction from whole blood followed by real-time polymerase chain reaction
(PCR) detection of KRAS oncogene [86]. Isolation and detection of CTCs from blood
samples was reported using a system that integrated a microchip with
immunomagnetics, high-throughput fluidics and size-based filtration [87]. Concerning
the magnetic particles, it is foreseen that particle-based assays will benefit from the
ongoing optimization of particles regarding their surface bio-functionalization,
surface smoothness and their size and magnetization uniformity [88].
2.5 Chip-MS platform
Mass spectrometry (MS) is an important analytical tool which can provide both
qualitative and quantitative information about a specific analyte and identify its
chemical structure. Gas chromatography (GC) [89, 90], liquid chromatography (LC)
[91, 92] and capillary electrophoresis (CE) [93-95] have been coupled with MS and
studied for a long time. All of the technologies have made great contributions in
separation and detection of molecules. However, MS-based technology with higher
throughput, more rapid and convenient analysis, and lower sample consumption is
still in great demand to meet the new challenges in biochemical analysis. In 1997,
Karger [96] and Ramsey [97] provided the concept of microchip-MS for modern
biological analysis. The new method is superior to other detection methods coupling
with chips in giving information for characterization of chemical structure [98]. Since
10

Page 10 of 55
then, a number of microfluidic chip-MS studies have been reported.
A series of analytical methods relative to microchip-MS have been developed in our
group, which show their potential as an integrated platform for cell-based biochemical
analysis. C30 beads were used to imitate cells to be introduced into a hole on a simple
microchip. The combination of microfluidic device with electrospray ionization
quadrupole time-of-flight mass spectrometer (ESI-Q-TOF-MS) was successfully
detected a series of herbicides on a single C30 bead [99]. Then, the ESI-Q-TOF-MS
coupled microchip platform was employed to monitor carnosine-protection process
against A42-induced glutamate released from PC12 cells [100]. We detected
multiple cell metabolites on the microchip coupled with ESI-Q-TOF-MS. The
metabolism of vitamin E in human lung epithelial A549 cells was successfully studied
by online microchip-MS platform with high sensitivity and short analysis time. By
integrating solid-phase extraction (SPE) microcolumns, cell culture chambers with
microchips, all the steps of cell culture, metabolism generation, sample pretreatment
and detection were conducted on the chip-MS platform [101]. Multiple gradient
generator also was integrated onto the chip-MS platform. The absorption of
methotrexate and its effects on HepG2 and Caco-2 cells were investigated on the
combination system [102]. Curcumin was used as a model drug to detect drug
permeability in Caco-2 monolayers on a membrane-based microfluidic device
coupled with MS [103]. In order to improve the stability, sensitivity and repeatability
of quantitative analysis on the chip-MS system, we developed a stable isotope
labeling assisted microfluidic chip ESI-MS (SIL-chip-ESI-MS) (Fig. 6). A
dual-isotopic labeling was presented for effective qualitative analysis of multiplex
metabolites. Despite complex biological matrixes, three coeluting pairs of
isotopomers could be easily recognized and identified by SIL-chip-ESI-MS [104]. We
even extended the application of chip-MS platform to study cell-to-cell
communication. The inhibition on growth hormone secretion from GH3 cells by
dopamine released from PC12 cells was investigated and demonstrated [105]. A
“Surface Tension Plug” was presented on a microchip to control the communication
between the 293 and the L-02 cells. Signaling molecules epinephrine and glucose
11

Page 11 of 55
were successfully detected using ESI-Q-TOF-MS [106]. An automatic and online
microfluidic chip-MS system was established to quantify noncovalent protein-protein
interactions [107].
Fig. 6
Recently, novel chip-MS platforms have been developed by researchers. An integrated
microfluidic CE-ESI device was demonstrated to separate intact monoclonal antibody
charge variants with online MS identification [108]. An electrochemical chip was
coupled online to MS or LC-MS to generate phase I and phase II drug metabolites and
to demonstrate protein modification by reactive metabolites. This new screening
method gave the potential of this electrochemical chip as a complementary tool for a
variety of drug metabolism studies in the early stages of drug discovery [109]. A
multi-layer microfluidic device was developed for characterization of drug
metabolism in human liver microsomes by coupled with ESI-Q-TOF-MS [110]. A
method combining microfluidics and a miniature MS was applied to quantify abuse
drugs in urine. Cocaine, benzoylecgonine and codeine were quantified from four
samples in less than 15 min using the new method [111]. A Swan-shaped probe
coupled with ESI-MS was designed and applied for high-throughput and
nanoliter-scale analysis of biological samples in both a microfluidic droplet array and
a multiwall plate. The Swan probe had two sections: a U-shaped section with a
micrometer-sized hole for sampling and a tapered tip for sample electrospray
ionization. 256 droplets were analyzed within 90 min with a peak height relative
standard deviation (RSD) of 12.6% [112]. Efforts also were put on miniaturization of
microchip-based MS using SU-8 as a material [113]. All this demonstrate that the
microfluidic chip-MS platform is developing to be a potential useful tool for
biochemical researches [114, 115].

3. Applications
By integrating various technologies from other fields, microfluidic device as a
powerful analytical technique is increasingly being used in various biochemical
researches, such as biomimetics, drug screening, biomolecular analysis and cell

12

Page 12 of 55
analysis. Some recent examples of these applications have been reviewed here.
3.1 Biomimetics
Cell culture on microscale platforms better mimics the in vivo cellular
microenvironment than conventional, whereas macroscale systems enabling
physiologically realistic in vitro tissue models to be constructed. Biomimetic
hydrogels are the preferred material for microfluidic chip fabrication. Recently, the
effects of microenvironmental parameters on cell fate were demonstrated in 3D model
by incorporating multiple cell types in microgels which having defined and tunable
mechanical and biochemical properties [116]. CTCs were encapsulated on-chip in a
biomimetic hydrogel matrix and their clonal 3D spheroid growth potential was
assayed by microscopy over one week. The possibility to clonally expand a subset of
captured CTCs in a near-physiological in vitro model powerfully exploit CTC-chip
application and ultimately should improve prediction of treatment responses and
disease progression [117]. Tethered protein gradients were patterned on the surface of
soft synthetic hydrogels in any user-defined shape to mimic the biophysical
characteristics of the natural extracellular milieu and validated to have a certain
impact on single-cell migration [118-120]. Probably, in vitro human skin models are
the most developed and understood engineered constructs due to the clinic and
cosmetics industry requirements [121]. By using a microfluidics-based in vitro skin
wound healing model, it was revealed that the bottom side of the bacterial cellulose
film could better promote the migration of cells to facilitate wound healing through
MTT assay [122]. The microenvironment of bone marrow contains a complex set of
cellular, chemical, structural and physical cues to maintain the viability and function
of the hematopoietic system. In vitro hematopoiesis models always fail to demonstrate
the complex microenvironment and functions of living bone marrow. A method for
fabricating “bone marrow-on-a-chip” allowed in vitro culturing of live marrow with a
functional hematopoietic niche. Hematopoietic stem and progenitor cells could be
retained in the engineering bone marrow for at least 1 week [123]. We developed a
microfluidic 3D culture device to imitate the diffusion process between blood vessels
and tissues. On this microfluidic device, QD cytotoxicity was evaluated by
13

Page 13 of 55
determining cell apoptosis, intracellular reactive oxygen species and glutathione with
specific fluorescence probes. The degree of QD cytotoxicity was demonstrated to be
relative to the diffusion distance [124].
Microfluidic devices also serve as novel tools for recapitulating complex organ
functions to improve the biochemical analysis on chips. A biomimetic microsystem of
human lung was fabricated by reconstituting the critical functional alveolar- capillary
interface (Fig. 7a). Complex integrated organ-level responses to bacteria and
inflammatory cytokines were reproduced on this bioinspired microdevice [19]. In
human body, liver is a central nexus integrating metabolic and immunologic
homeostasis. Also, it is the direct or indirect target of most molecular therapeutics
[125]. We integrated bioreactors containing poly(ethylene) glycol hydrogel
encapsulated human liver microsomes onto the microfluidic device to imitate drug
metabolism in human liver and its cytotoxicity on cells. The products of
5’-diphosphate-glucuronosyltransferase (UGT) metabolism of acetaminophen (AP)
were injected into the cell culture chamber for cytotoxicity assay and simultaneously
detected online with ESI-Q-TOF-MS [126]. Alzheimer’s disease (AD) classified as
one of the most costly brain disease imposes an enormous burden on society. A 3D
brain-on-a-chip with an interstitial level of flow was developed as an in vitro model of
AD to meet the growing need for understanding of etiology and faster development of
treatment strategies (Fig. 7b) [127]. The microvasculature is an extensive organ that
supports metabolic activity and mediates the interaction between blood and tissues.
Engineering living microvascular networks in 3D tissue scaffolds developed by Zhang
et al. provided a platform for studying complex vascular phenomena (Fig. 7c). The
angiogenic activities and nonthrombotic nature of the vascular endothelium and its
transition to a prothrombotic state during an inflammatory response were
demonstrated [128]. Metastasis of cancer cells from a primary tumor to secondary loci
is the main cause for cancer-related mortality. A microfluidic 3D in vitro model was
constructed to analyze human breast cancer cell extravasation into bone- and
muscle-mimicking microenvironments through a microvascular network
concentrically wrapped with mural cells and unveil the underlying mechanisms of
14

Page 14 of 55
cancer metastases [129]. In vivo-like oriented neural networks between superior
cervical ganglion (SCG) neurons and their effector smooth muscle cells (SMC) were
modeled in a microfluidic device which allowed amperometric detection of individual
neurotransmitter release events inside functional SCG-SMC synapse with carbon fiber
nanoelectrodes as well as recording of postsynaptic potential using glass nanopipette
electrodes [130]. Further advances in the field of organ-on-a-chip platforms can
revolutionize some biochemical analysis process such as drug screening, genetic and
protein analysis. The final goal is to develop a body-on-a-chip platform for systemic
biochemistry analysis.
Fig. 7
3.2 Drug discovery
Bringing a new drug to market is a complex, lengthy and expensive process. An
adequate cell-based assay to efficiently screen and validate potential drug candidates
is crucial in the initial stage of drug discovery [131]. Microfluidic platforms provide
several unique advantages for drug screening. Physical and chemical properties of
drug carriers can be easily and effectively modified by tuning the flow rate and
geometries of microchannels on microfluidic devices. Batches of carriers can be
fabricated with minimal effort and with little variation. Tissue or organ models can be
mimicked in microfluidic systems to be used as in vitro drug screening tools [132].
A robust microfluidic approach increased the number of data points in dose-response
analysis to approximately 10 000 per compound (Fig. 8a). The increased number of
data points resulted in highly precise and reproducible IC50 values [133]. A
microfluidic device featuring an electrokinetic size and mobility trap was formed to
overcome the bottleneck that extraction of target analytes from biological samples.
Ampicillin levels in blood were analyzed within 5 min and a linear response over the
range of 2.5-20 g mL-1 on this device [134]. Concentration gradients play essential
roles in drug screening because they can affect various cell behaviors. A novel
gradient generator was developed to achieve long range and linear chemical gradients
with a dynamic control function. This gradient generator overcome the disadvantages
of the “Christmas tree” design that lacking dynamic control of gradient profile and
15

Page 15 of 55
limited length of the gradient. The effect of deoxycycline on the viability of PC-9 lung
cancer cells was tested on this device [135]. Anti-angiogenesis drugs suppress tumor
growth by disrupting oxygen and nutrient supplied from blood to the tumor. Kim et al.
[136] described a method for screening and quantifying the vascular endothelial
growth factor (VEGF)-induced chemotactic response on human umbilical vein
endothelial cells (HUVEC) cultured with different concentrations of bortezomib, a
selective 26S proteasome inhibitor. A 3D microfluidic cell array which reconstructed a
3D tumor microenvironment with cancer cells and microvascular endothelial cells
was used to predict anticancer drug responses in human tumors [137]. Cell gradient
also is a key factor in cytotoxicity assay. We developed a microfluidic cell density
gradient generator composed of eight parallel channels which were surrounded by 1-8
microwells. Series of cell density gradients were successfully generated on this chip
and QD cytotoxicity exhibited obvious cell density-dependence [138].
Fig. 8
The development of microengineered models of the functional units of human organs
or tissues could improve the effectiveness of preclinical predictions of human drug
responses [139-141]. Shuler et al. [142] designed and developed a microfluidic
platform that allowed for long-term maintenance of full thickness human skin
equivalents which were comprised of both the epidermal and dermal compartments.
The toxic effects of doxorubicin on skin cells and structure was examined on this
platform. Our group developed an in vitro liver model to imitate and detect prodrug
metabolism (Fig. 8b). Capecitabine (CAP) was selected as a model compound
because it needed to be metabolized into active intermediate in the liver and then
transformed into final effective drug in tumor cells. This process was realized and
confirmed within this in vitro liver model. The intermediate product was successfully
detected by MS and the anti-tumor effect of the active metabolite was observed
through cell viability assays of MCF-7 cells [143]. Hepatic organoids could be also
formed from co-cultures of HepG2 and NIH-3T3 cells embedded in hydrogel matrices
on a digital microfluidic device. The organoids exhibited better mimics of in vivo liver
tissue than comparable 2D cell culture systems. The effects of a dilution series of
16

Page 16 of 55
acetaminophen on apoptosis and necrosis were analyzed on this device [144]. A
higher throughput “heart on a chip” which recapitulated the laminar architecture of
the heart ventricle was presented. The microdevice was employed to test the positive
inotropic effect of isoproterenol on cardiac contractility at dosages ranging from 1 nM
to 100 M [145]. An in vitro microfluidic absorption, distribution, metabolism and
excretion (ADME) profiling system was established to study systemic absorption of
drugs in the small intestine, metabolism by the liver as well as excretion by the kidney.
Dose systemic toxicity testing of drug candidates had been lasted for over 28 days in
the microsystem [146]. Achievements in the organ-on-a-chip field exploit exciting
new avenues for drug discovery and development. The tremendous potential that
organ-on-a-chip technology holds will bring a broad impact on drug discovery and
give a surge in its future progress.
3.3 Biomolecular analysis
3.3.1 Genetic analysis
So far, microsystem for DNA and RNA measurement is one of the most developed
analytical microfluidic field. PCR and other techniques for sample amplification are
currently powerful and popular technique for sensitive genetic analysis. Due to the
high surface-to-volume ratio inside microchannels or microscale reactors, microscale
PCR has been realized and demonstrated to be rapid. A digital PCR using aqueous
droplets in oil as microreaction vessels for individual DNA molecules known as
droplet digital PCR was conducted on a microfluidic chip [147]. Single-cell PCR
gene-expression analysis was implemented on microfluidic chip to dissect the cellular
composition of primary human normal colon and colon cancer epithelia. It was
showed that different gene-expression programs linked to multilineage differentiation
were strongly associated with patient survival [148]. We also established a method
based on PCR coupled with microchip electrophoresis to analyze BCR-ABL fusion
gene [149] and mini-short tandem repeat loci [150]. A crucial step in the genetic assay
process is the isolation of the nucleic acids. Purification of the nucleic acids from raw
samples could enhance the device sensitivity and pave the way for downstream
processes [151]. An array of surface-adhering droplets was reported to facilitate the
17

Page 17 of 55
transportation of magnetic purification beads for parallel mRNA extraction through
individual buffer solutions without solid structures [152]. However, the device in Fig.
9a executing all steps of single-cell processing, including cell capture, cell lysis,
reverse transcription and quantitative PCR made it possible to skip nucleic acid
isolation [153].
Fig. 9
Hybridization arrays, real-time probes and electrophoretic sizing for analysis are
always contained on microchips. A microfluidic chip with 3 × 3 arrayed
electrochemical sensors for the analysis of DNA hybridization events was presented.
The electrochemical signal given by the microfluidic chip was validated and
demonstrated to be repeatable [154]. A label-free DNA sensing platform was
developed with low-voltage electrolyte-gated transistors. A floating-gate electrode
functionalized with ssDNA whose potential was determined by both capacitive
coupling with a primary, addressable gate electrode and the presence of adsorbed
molecules. When DNA was hybridized at the floating gate, it offset the primary gate
voltage. The offset was related to the number density of dsDNA molecules [155]. A
temperature jump (T-jump) setup enabled heating and cooling of aqueous solutions by
~ 50 ℃ on a 1-s time scale. The setup was comprised by a thermally conductive
sapphire substrate with light-absorptive nano-coating, a microfluidic device and a
rapidly switched moderate-power IR laser with the laser beam focused on the
nano-coating. The setup could be used to probe folding and unfolding dynamics of
DNA hairpins after direct and inverse temperature jumps [156]. Chip-base analysis of
mRNA levels in enriched tumor exosomes obtained from blood even could monitor
the level change during treatment. A microfluidic platform termed immune-magnetic
exosome RNA (iMER) analysis was presented in Fig. 9b which integrated
immunomagnetic selection, RNA collection and real-time PCR into the format of a
single chip [157]. Zero-mode waveguides (ZMWs) are attractive technology for
single-molecule imaging. Conical lens-based dark-field fluorescence microscopy was
combined with a ZMW/microfluidic chip and revealed weak DNA-protein and
protein-protein interactions found with T4 replisomal proteins [158]. “DNA
18

Page 18 of 55
dumbbells”— single molecules of DNA with a microscopic bead attached at each end
were generated on a microfluidic device. This design facilitated the examination of
the role of 3D DNA conformation and dynamics in protein-DNA interactions [159].
The advantage of microfluidic technology that high-throughput features more
prominently in genetic analysis. A million-membered metagenomic library was
screened in microfluidic picolitre droplet compartments [160]. Ohler et. al [161]
developed a high-throughput microfluidic device in which 64 Arabidopsis thaliana
seedlings could be grown and their roots were imaged by confocal microscopy over
several days without manual intervention. They tracked hundreds of roots to capture
detailed expression patterns of 12 transgenic reporter lines under different conditions.
The integration of microfluidic selection with high-throughput DNA sequencing
technology was described for rapid and efficient discovery of nucleic acid aptamers.
The copy number and enrichment-fold of more than 10 million individual sequences
were tracked by this method through multiple selection rounds [162]. Huang et. al
[163] prepared 94 libraries consisting of single mouse embryonic cells and technical
replicates of extracted RNA through implementing microfluidic technology in
single-cell RNA sequencing. They also quantified variation between and within
different types of mouse embryonic cells. The advantages of microfluidic approaches
that enhanced measurement precision, detection sensitivity and high throughput were
throughly validated in the single-cell transcriptome analysis. Integration of multiple
instruments has been a major trend in fabricating microfludic chips for biochemical
analysis. An efficient microfluidic single-cell analysis chip in combination with
high-resolution time-lapse microscopy was developed to firstly determine the yeast
replicative lifespan in a high-throughput manner. This chip gave a promise for the
usefulness of yeast replicative lifespan assays as a broad genetic screening platform
for research on aging [164].

3.3.2 Protein analysis


The separation of required protein analytes from raw biosamples is of critical
importance in modern proteomics. An on-chip all SiNW filtering, selective separation,
19

Page 19 of 55
desalting, and preconcentration platform for target proteins analysis was developed.
The SiNW forest with ultralarge binding surface area was modified with antibody to
separate required protein, followed by the release of these proteins in a controlled
liquid media. The released proteins were detected by supersensitive SiNW-based
field-effect transistor arrays fabricated on the same chip [165]. A robust
chemomechanical sorting system also was designed to catch and release target
biomolecules from a solution mixture. Target-specific, reversible binding sites
attached to microscopic fins were the main part of the system. The microscopic fins
were embedded in a responsive hydrogel that moved the cargo between two
chemically distinct environments. Thrombin was separated as an example on this
platform by synchronizing the pH-dependent binding strength of a thrombin-specific
aptamer with volume changes of the pH-responsive hydrogel in a biphasic
microfluidic regime (Fig. 10) [166]. The evolution of enzyme catalyst was performed
on a microfluidic device by caged in gel-shell beads. These hydrogel beads were
surrounded with a polyelectrolyte shell that enclosed an enzyme, its encoding DNA
and the fluorescent reaction product [167]. The activity of millions of glycosidase
sequence variants was mapped by Abate et. al through combining droplet microfluidic
screening with next-generation DNA sequencing. Microfluidic-based deep mutational
scanning provided a comprehensive and unbiased view to dissect enzyme function
[168]. Otten et al. [169] described a microfluidic platform for on-chip expression,
covalent surface attachment and measurement of single-molecule protein mechanical
properties. A dockerin tag on each protein molecule allowed thousands of pulling
cycles performed by a single cohesion-modified cantilever. We also reported some
microchips for protein analysis. An integrated microfluidic device was developed for
real-time analysis of secreted protein VEGF165 by aptamer-functionalized
microchannels. Besides that, investigation of various oxygen concentrations and
distances effect on cell migration was conducted on this platform [170]. An
aptamer-modified microchip for ultrasensitive detection of thrombin using rolling
circle amplification and G-quadruplex DNAzyme was developed in our lab [171]. We
also assayed multiplex proteins based on tunable aptamer by microchip
20

Page 20 of 55
electrophoresis. Different lengths of aptamers modulated the electrophoretic mobility
of proteins allowing the protein to be effectively separated on the microchip. Under
optimum conditions, good linearities of quantitative assays of three kinds of proteins
(PDGF-BB, VEGF165 and thrombin) were achieved with a dynamic range of
5.00-150.0 nM [172]. ELISA which measures protein abundance using bulky
fluorescent scanners is the current gold standard method for protein biomarker
detection. ELISA is widely used in biochemical analysis with high specificity and
sensitivity but lacks of portability. Carrying out ELISA on microfluidic platform can
eliminate this disadvantage. A digital microfluidic platform present by Javanmard et.
al making use of simple electronics was translated into a portable handheld device and
could be adapted to assay different types of proteomic biomarkers [173]. Micro
open-sandwich ELISA allowed rapid and noncompetitive detection of a small
biomarker peptide [174]. A novel microfluidic ELISA microplate with standard
SBS-configured 96-well microplate architecture was introduced as the next generation
assay platform for unparalleled assay performances [175]. An integrated microfluidic
solid-phase ELISA platform was developed for rapid and ultrasensitive detection of
proteins with a wide dynamic range. Two key novelties existed in this design that an
integrated microwell-patterned assay chamber which could be pneumatically actuated
to reduce the volume of chemifluorescent reaction and monolithic integration of
on-chip pumps to allow programmable fluid delivery and effective mixing [176].
ELISA was even performed in a 96-microzone plate fabricated in paper [177].
Interaction between biomacromolecules plays a significant role in revealing the
underlying mechanisms of cellular activities. A unique continuous-flow laminar mixer
based on microfluidic dual-hydrodynamic focusing was investigated to characterize
the kinetics of DNA-protein interactions. The interaction kinetics of G-quadruplex
and single-stranded DNA binding protein was successfully interrogated by this device
[178]. Biomolecular assay on microfluidic chip can give fundamental biology
investigations to decipher mechanisms of some in vivo biochemical processes.
Fig. 10
3.4 Cell analysis
21

Page 21 of 55
3.4.1 Single cells
The capability of integrated microfluidics to accurately manipulate, handle and
analyze very small volumes has opened up new opportunities for analysis of
intracellular constituent at single-cell level. Compared to conventional biological
assays that statistically analyze the average response from a large population of cells,
single-cell assay can tell the differences between individual cells allowing more
precise understanding of single-cell behavior. The challenging in studying single cells
is requiring hundreds or thousands of isolated single cells. Numerous
microfluidic-based techniques such as microwell arrays [179, 180], DEP [181, 182],
microscale physical trap arrays [183] and microdroplets [184, 185] have been
developed and successfully utilized to capture single cells. Using fluid dynamics
simulations in combination with particle image velocimetry to systematically
optimize trap architectures, Kobel et al. [186] enhanced single cell trapping efficiency
up to 97%. A U-shaped hydrodynamic trap was incorporated into a two-layer
microfluidic device platform for single cell capture, culture and clonal expansion.
Both adherent and non-adherent cells were trapped with high efficiency and in a high
throughput manner [187]. An optical micro-tweezers was integrated into a hybrid
polymethylmethacrylate (PMMA)-glass microfluidic circuit to trap a single cell and
excite its Raman and fluorescence response, thus making monitoring the single cell
reaction to different stimuli to be possible [188]. Single-cell encapsulation rate was
improved by Liu et. al through integrating droplet generation with
fluorescence-activated droplet sorting which isolated droplets containing one cell
[189]. Our group fabricated rounded bottom microwell arrays in PDMS by moulding
a monolayer of ordered polystyrene (PS) microspheres. PS microspheres were
self-assembled on a glass slide and partially melted at 240 ℃ to be used as master to
generate microwell arrays. Single cells can be retained in the microwells with high
efficiency [190]. We also designed a novel microfluidic platform integrated with
cell-recognizable aptamer-encoded microwells to isolate single tumor cells with
satisfied single-cell occupancy and unique bioselectivity. The single-cell capture
efficiency was significantly enhanced from 0.5% to 88.2% through the introduction of
22

Page 22 of 55
the aptamer into the microwells with optimized size [191]. After isolation,
microfluidic approaches are found to be well suited to resolve a variety of single-cell
analytics including signal response, growth dynamics, gene expression measurements
and protein analysis. A chemical signal generator based on hydrodynamic gating
permitted resolving temporal dynamics of single cells [192]. A microfluidic
hydrodynamic trapping system was designed with arrays of bypass structures for
trapping individual cells and developed as a digital single-cell assay system to
monitor cellular dynamics under drug treatment over time [193]. A high-throughput
single-cell multidrug resistance analysis device was developed to examine both the
chemo-sensitizing effect and the cytotoxity of the potential chemo-sensitizing agents
[194]. Five clinically established chemotherapeutic reagents were conveniently
assayed on this system. Large-scale, highly parallel single-cell transcriptomics was
realized in tiny droplets in which single cells were captured along with sets of
barcoded primer beads. Transcriptionally distinct cell populations in mouse retinal
tissue were revealed by this analytical method [195] . Similar method also was used to
enable single-cell transcriptomics of a large number of cells in a heterogeneous
population. Population structure, gene expression relationships, and the heterogeneous
onset of differentiation in mouse embryonic stem cells were uncovered depending on
this development [196]. Single-cell genomic damage which was generally identified
with a “comet assay” was demonstrated to be analyzed in an ultrahigh-throughput
approach with an agarose-based microfluidic chip. Up to 10 000 individual cells were
simultaneously detected with better reproducibility [197]. Picoliter-sized droplets
were present for compartmentalized whole genome amplification to minimize
amplification bias and contaminants showing potential as an efficient tool for
effective amplification of low-input DNA for single-cell genomics [198]. Voldman et
al. [199] presented a microfluidic device to trap and properly pair thousands of cells.
The device was compatible with both chemical and electrical fusion protocols.
Recently, they further applied this device to investigate immune response by pairing
lymphocytes (Fig. 11a). Using this platform, they characterized early activation
dynamics of CD8 T cells and investigated the extent of heterogeneity in T-cell
23

Page 23 of 55
activation and the correlation of multiple readouts [200]. A microfluidic migration
platform to simultaneously analyze four qualitative migration patterns:
chemoattraction, -repulsion, -kinesis and -inhibition was designed and validated using
single-cell quantitative matrics of direction, speed, persistence and fraction of cells
responding (Fig. 11b) [201]. We designed a microfluidic system consisted of a PDMS
microwell array for single-cell arrangement. Depending on this platform, we found
that QD cytotoxicity was dependent on cellular uptake in various cell cycle phases
because the accumulation and dilution of QDs happened in single cell cycles. The
rank of QD cytotoxicity in different cell cycle phases was G2/M > S > G0/G1 [202].
A single-cell analysis pipeline for studies of signaling dynamics was described.
Microfluidic cell culture, precise stimulation of cells with signaling molecules or
drugs, live-cell microscopy, computerized cell tracking, on-chip staining of key
proteins and subsequent retrieval of cells for high-throughput gene expression
analysis were incorporated into this pipeline. This pipeline represented a major step
forward in dynamic single-cell analysis because it enhanced experimental precision
and throughput by orders of magnitude and introduced much-desired new capabilities
in cell and fluid handling [203]. A microchip was designed to quantify the levels of a
dozen cytoplasmic and membrane proteins from single cells. Single-cell analysis
uniquely revealed single-cell heterogeneity and different types and strengths of
protein-protein interactions. This platform helped to comprehensively understand
altered signal transduction networks in tumor cells and gave insight into the effect of
targeted therapies on protein signaling networks [204]. A microscope slide supporting
a 30-m-thick photoactive polyacrylamide gel was developed to conduct ~103
concurrent single-cell western blots in ~4 h. This method was applied to monitor
single-cell differentiation of rat neural stem cells and responses to mitogen
stimulation [205]. Liu et. al [206] proposed a novel single-cell chemical proteomics
strategy to profile low-abundance membrane proteins in single cells. They further
applied this method to investigate single primary cells and observed significant
heterogeneity among individual mouse cerebellar granule neurons. A complete cycle
of single cell analysis was achieved on a lab-on-a-chip that combines
24

Page 24 of 55
micro/nano-fabricated features with a Convex Lens-Induced Confinement device. All
the steps of cell trapping, cell isolation, lysis, protein digestion, genomic DNA
extraction and on-chip genomic DNA linearization were included [207]. A novel
microfluidic single-cell culture method was present by Buckanovich et. al to directly
observe the differentiation capacity of four heterogeneous ovarian cancer cell
populations. The data suggested a hierarchical differentiation pattern in which bone
morphologenetic protein 2 acted as a feedback mechanism promoting ovarian cancer
stem-like cell expansion and suppressing progenitor proliferation [208]. Microfluidics
will work as a necessary single-cell analytical technique in biotechnological process
and strain characterization.
Fig. 11
3.4.2 Stem cells
Stem cells are one kind of cells that capable of potentially differentiating into specific
tissue types and continuously self-renewaling through replication. Thus, they can
offer a consistent supply of physiologically relevant cells from validated
pathogen-free sources that differentiate into mature somatic cells [209]. Stem cell
analysis is an emerging field of great significance in healing damaged tissues and
replacing non-functional organs. The combination of microfabrication-based
technologies with stem cell analysis shows advantages in spatially and temporally
controlling cell growth and stimuli by combining surfaces that mimic complex
biochemistry and geometry properties of the ECM to maintain a homogeneous culture
of undifferentiated cells and direct stem cell differentiation reliably. Occhetta et al.
[210] designed and validated a microfluidic platform to culture 3D micromasses of
human bone marrow-derived mesenchymal stromal cells under continuous flow
perfusion. Defined concentrations of morphogens were delivered to specific culture
units yielding more uniform cell response throughout the 3D structures of perfused
micromasses and a 34-fold higher percentage of proliferating cells at day 7. A
microfluidic platform containing thousands of nanoliter-scale chambers suitable for
live-cell imaging studies was used for single hematopoietic stem cell proliferation
analysis (Fig. 11c). The automated medium exchange of this system made it possible
25

Page 25 of 55
to define when Steel factor stimulation was first required by adult hematopoietic stem
cells (HSCs) in vitro as the point of exit from quiescence [211]. A large array of 2D
no-flow chambers was designed to allow the monitoring of single HSCs for several
days. Live human HSCs could be stained with fluorescent primary antibodies to
reveal their stage in the hematopoiesis differentiation pathway. Human HSCs’ growth
rate, polarization and migration were correlated to their differentiation stage on this
chip [212]. We proposed a novel PDMS porous membrane-assembled 3D microdevice
for feeder-separated co-culture of mouse embryonic stem (mES) cells (Fig. 11d). Pure
mES cells of good pluripotency were separated from mouse embryonic fibroblasts
with easy and high efficiency [213].
Niches are complex microenvironments that regulate adult stem cell fate in vivo by
molecular interactions [214] and are critical to maintain stem cells in their multipotent
and often quiescent state. Upon activation, inches influence the decision of single
cells to self-renew or to undergo differentiation into the various lineages of a tissue.
Microfluidics is ideally suitable to give well-defined chemical and physical stimuli to
stem cells, so artificial niches are suitable constructed on microfluidic platform to
investigate biological behavior of stem cells. Lutolf’s group have made a great
contribution in this field. They applied microfabrication and micropatterning
technologies into dissecting the complex molecular interplay of stem cells and their
niches [215]. Hydrogel engineering was combined with droplet microfluidic
technology to form cell-type-specific artificial extracellular matrix in high-throughput.
The robust expansion or differentiation of stem cells was stimulated via display of
specific niche components on the microcarrier surface [216]. Artificial niche
microarrays in which individual microwells was functionalized with combinations of
proteins were reported to test the effect of cell-cell interactions on adipogenic
differentiation of adherent human mesenchymal stem cells (MSCs) and the effect of
substrate stiffness on osteogenic MSC differentiation. Artificial niches supporting
extensive self-renewal of nonadherent mouse neural stem cells also were identified
[217]. Single-cell analysis in microwell arrays revealed a highly heterogeneous
division behavior of HSCs [218]. Recently, droplet-based microfluidics has grown to
26

Page 26 of 55
be a promising strategy to build in vitro stem cell niches in a miniaturized and highly
precise fashion for stem cell biology [219]. A microfluidic gradient generator was
used to study brain-derived neurotrophic factor (BDNF)-enhanced neural stem cell
(NSC) chemotaxis which was critical for NSC migration during brain development
and neural tissue regeneration [220]. Osteogenic differentiated factors were
encapsulated in poly(lactic-co-glycolic acid) (PLGA) microspheres which were
patterned into grooves by Teflon chips. These factors induced the osteogenic
differentiation of adipose-derived stem cells. The results demonstrated that a
combination of chemical and topographical cues was more effective to direct the
osteogenic commitment of stem cells than either was alone [221]. Cell-secreted
factors were demonstrated to be necessary to maintain self-renewal of mES cells on a
microfluidic perfusion device [222]. Another major advantage of microfluidic
technology is integrating electrical, optical, magnetic and acoustic sensors into a
single device to give observation on stem cells. Kang et al. [223] presented a
microfluidic device that coupled on-chip culture of adherent cells and transfection by
localized electroporation. Differentiation of NSCs and transfection of postmitotic
neurons with a green fluorescent protein plasmid were demonstrated on this device. A
radio frequency identification-based sensor platform was designed by integrating
sensitive elements onto glass substrate comprised of two comb-shaped interdigitated
gold electrodes covering an area of 1.8 mm × 2 mm. The platform was adopted to
characterize cultivation and differentiation of human bone marrow-derived
multipotent stem cells over periods of up to several days and weeks [224]. Osteogenic
differentiation of MSCs was evaluated on a SPR-based system. A high correlation
between the duration of osteogenic induction and the difference in refractive angle
shift with coefficient was demonstrated [225]. In the light of an increasing demand of
stem cells for cytotoxicity assay, disease modeling, drug screening and cell-based
therapies, there is an increasing necessity for advanced in vitro stem cell analysis
systems. Advances in biomaterials and microtechnology for stem cell analysis even
make it possible to offer mechanistic insight into organogenesis and unveil powerful
new models for drug discovery, as well as strategies for tissue regeneration in the
27

Page 27 of 55
clinic [226-228].

4. Conclusions and outlook


Owning to the inherent advances in device design, fabrication and flexible integration
with other microscale techniques, microfluidics has experienced an explosive growth
during the past decades and become a major analytical technology with enlarging
application fields. As a technique enabling precise manipulation of fluid at the
micrometer scale, microfluidics has paved a revolutionary way for time-saving and
cost-saving detection in sophisticated biochemical analysis. Not only traditional
biochemical operations were miniaturized onto the microfluidic device, but also new
platforms for generating unconventional strategies to overcome the challenges in
biochemical analysis were provided.
For improving microfluidic applications in biochemical analysis, there are still some
challenges have to be addressed: (1) Highly integrated microdevices coupled with
large and multifunctional instruments such as MS, nuclear magnetic resonance
spectrometer have to be designed. Although we have explored in the field of chip
coupled with MS, fully-integrated chip-MS platforms need to be further developed.
Besides that, how to miniaturize these large instruments to make them adjustable to be
coupled with chip still demand prompt development. (2) Automated manipulation of
microfluidic chips desire more convenient sample pretreatment and high-throughput
analysis. This promotion will advance microfluidics to the upper level in biochemical
analysis. (3) Real-time analysis of multiple targets of in vivo biochemical process in
situ is a crucial area in microfluidics. With the development of online microfluidic
chip-based biochemical analysis, it is hoped that portable and inexpensive
microfluidic devices will play a great role in point-of-care diagnosis and drug
discovery in the future.

Acknowledgements
This work was supported by the National Natural Science Foundation of China (No.
21405143, 21435002, 21227006) and the Fundamental Research Funds for the
Central Universities (No. 2-9-2014-023).
28

Page 28 of 55
Corresponding Author
*E-mail: jmlin@mail.tsinghua.edu.cn.

References:
[1] J. El-Ali, P.K. Sorger, K.F. Jensen, Cells on chips, Nature 442 (2006) 403-411.
[2] M.M. Rosenkilde, T.W. Schwartz, The chemokine system -- a major regulator of
angiogenesis in health and disease, APMIS 112 (2004) 481-495.
[3] L. Liu, B.D. Ratner, E.H. Sage, S. Jiang, Endothelial cell migration on surface-density
gradients of fibronectin, VEGF, or both proteins, Langmuir 23 (2007) 11168-11173.
[4] A.S. Belloni, G. Albertin, M.L. Forneris, G.G. Nussdorfer, Proadrenomedullin-derived
peptides as autocrine-paracrine regulators of cell growth, Histol. Histopathol. 16 (2001) 1263-1274.
[5] F. Rosso, A. Giordano, M. Barbarisi, A. Barbarisi, From cell-ECM interactions to tissue
engineering, J. Cell Physiol. 199 (2004) 174-180.
[6] C. Jamora, E. Fuchs, Intercellular adhesion, signalling and the cytoskeleton, Nat. Cell Biol. 4
(2002) E101-E108.
[7] B. Geiger, A. Bershadsky, R. Pankov, K.M. Yamada, Transmembrane extracellular
matrix-cytoskeleton crosstalk, Nat. Rev. Mol. Cell Bio. 2 (2001) 793-805.
[8] Y. Li, Y. Zhou, H. Wang, S. Perrett, Y. Zhao, Z. Tang, G. Nie, Chirality of glutathione
surface coating affects the cytotoxicity of quantum dots, Angew. Chem. Int. Edit. 50 (2011)
5860-5864.
[9] A. Hoshino, K. Fujioka, T. Oku, M. Suga, Y.F. Sasaki, T. Ohta, M. Yasuhara, K. Suzuki, K.
Yamamoto, Physicochemical properties and cellular toxicity of nanocrystal quantum dots depend on
their surface modification, Nano Lett 4 (2004) 2163-2169.
[10] A. Arora, G. Simone, G.B. Salieb-Beugelaar, J.T. Kim, A. Manz, Latest developments in
micro total analysis systems, Anal. Chem. 82 (2010) 4830-4847.
[11] G.B. Salieb-Beugelaar, G. Simone, A. Arora, A. Philippi, A. Manz, Latest developments in
microfluidic cell biology and analysis systems, Anal. Chem. 82 (2010) 4848-4864.
[12] D. Mark, S. Haeberle, G. Roth, F. von Stetten, R. Zengerle, Microfluidic lab-on-a-chip
platforms: Requirements, characteristics and applications, Chem. Soc. Rev. 39 (2010) 1153-1182.
[13] I. Meyvantsson, D.J. Beebe, Cell culture models in microfluidic systems, Annu. Rev. Anal.
Chem. 1 (2008) 423-449.
[14] A. Khademhosseini, R. Langer, J. Borenstein, J.P. Vacanti, Microscale technologies for
tissue engineering and biology, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 2480-2487.
[15] D. Gao, H. Liu, Y. Jiang, J.-M. Lin, Recent developments in microfluidic devices for in vitro
cell culture for cell-biology research, TrAC, Trends Anal. Chem. 35 (2012) 150-164.
[16] M.C. Cushing, K.S. Anseth, Hydrogel cell cultures, Science 316 (2007) 1133-1134.
[17] D. Huh, G.A. Hamilton, D.E. Ingber, From 3D cell culture to organs-on-chips, Trends Cell
Biol. 21 (2011) 745-754.
[18] Y. Imura, K. Sato, E. Yoshimura, Micro total bioassay system for ingested substances:
Assessment of intestinal absorption, hepatic metabolism, and bioactivity, Anal. Chem. 82 (2010)
9983-9988.
[19] D. Huh, B.D. Matthews, A. Mammoto, M. Montoya-Zavala, H.Y. Hsin, D.E. Ingber,
29

Page 29 of 55
Reconstituting organ-level lung functions on a chip, Science 328 (2010) 1662-1668.
[20] L. Mazutis, J. Gilbert, W.L. Ung, D.A. Weitz, A.D. Griffiths, J.A. Heyman, Single-cell
analysis and sorting using droplet-based microfluidics, Nat. Protoc. 8 (2013) 870-891.
[21] O. Gefen, O. Fridman, I. Ronin, N.Q. Balaban, Direct observation of single stationary-phase
bacteria reveals a surprisingly long period of constant protein production activity, Proc. Natl. Acad. Sci.
U. S. A. 111 (2014) 556-561.
[22] S.S. Lee, I. Avalos Vizcarra, D.H.E.W. Huberts, L.P. Lee, M. Heinemann, Whole lifespan
microscopic observation of budding yeast aging through a microfluidic dissection platform, Proc. Natl.
Acad. Sci. U. S. A. 109 (2012) 4916-4920.
[23] K. Loutherback, L. Chen, H.N. Holman, Open-channel microfluidic membrane device for
long-term FT-IR spectromicroscopy of live adherent cells, Anal. Chem. 87 (2015) 4601-4606.
[24] T.H. Seefeld, A.R. Halpern, R.M. Corn, On-Chip synthesis of protein microarrays from
DNA microarrays via coupled in vitro transcription and translation for surface plasmon resonance
imaging biosensor applications, J. Am. Chem. Soc. 134 (2012) 12358-12361.
[25] C. Bermejo, F. Haerizadeh, H. Takanaga, D. Chermak, W.B. Frommer, Optical sensors for
measuring dynamic changes of cytosolic metabolite levels in yeast, Nat. Protoc. 6 (2011) 1806-1817.
[26] H. Xin, Y. Li, B. Li, Controllable patterning of different cells via optical assembly of 1D
periodic cell structures, Adv. Funct. Mater. 25 (2015) 2816-2823.
[27] S. Kim, A.M. Streets, R.R. Lin, S.R. Quake, S. Weiss, D.S. Majumdar, High-throughput
single-molecule optofluidic analysis, Nat. Methods 8 (2011) 242-245.
[28] F. Guo, M.I. Lapsley, A.A. Nawaz, Y. Zhao, S.S. Lin, Y. Chen, S. Yang, X. Zhao, T.J.
Huang, A droplet-based, optofluidic device for high-throughput, quantitative bioanalysis, Anal. Chem.
84 (2012) 10745-10749.
[29] J. Chen, Z. Kang, G. Wang, J.F.C. Loo, S.K. Kong, H. Ho, Optofluidic guiding, valving,
switching and mixing based on plasmonic heating in a random gold nanoisland substrate, Lab Chip 15
(2015) 2504-2512.
[30] A. Valero, T. Braschler, P. Renaud, A unified approach to dielectric single cell analysis:
Impedance and dielectrophoretic force spectroscopy, Lab Chip 10 (2010) 2216-2225.
[31] T.B. Tran, S. Cho, J. Min, Hydrogel-based diffusion chip with Electric Cell-substrate
Impedance Sensing (ECIS) integration for cell viability assay and drug toxicity screening, Biosens.
Bioelectron. 50 (2013) 453-459.
[32] Y. Xu, Y. Lv, L. Wang, W. Xing, J. Cheng, A microfluidic device with passive air-bubble
valves for real-time measurement of dose-dependent drug cytotoxicity through impedance sensing,
Biosens. Bioelectron. 32 (2012) 300-304.
[33] K. Zor, A. Heiskanen, C. Caviglia, M. Vergani, E. Landini, F. Shah, M. Carminati, A.
Martinez-Serrano, T. Ramos Moreno, M. Kokaia, D. Benayahu, Z. Keresztes, D. Papkovsky, U.
Wollenberger, W.E. Svendsen, M. Dimaki, G. Ferrari, R. Raiteri, M. Sampietro, M. Dufva, J. Emneus,
A compact multifunctional microfluidic platform for exploring cellular dynamics in real-time using
electrochemical detection, RSC Adv. 4 (2014) 63761-63771.
[34] R. Meissner, B. Eker, H. Kasi, A. Bertsch, P. Renaud, Distinguishing drug-induced minor
morphological changes from major cellular damage via label-free impedimetric toxicity screening, Lab
Chip 11 (2011) 2352-2361.
[35] A.C. Sabuncu, J. Zhuang, J.F. Kolb, A. Beskok, Microfluidic impedance spectroscopy as a
tool for quantitative biology and biotechnology, Biomicrofluidics 6 (2012) 34103.

30

Page 30 of 55
[36] B.J. Dworak, B.C. Wheeler, Novel MEA platform with PDMS microtunnels enables the
detection of action potential propagation from isolated axons in culture, Lab Chip 9 (2009) 404-410.
[37] A. Hai, J. Shappir, M.E. Spira, In-cell recordings by extracellular microelectrodes, Nat.
Methods 7 (2010) 200-202.
[38] G. Cellot, E. Cilia, S. Cipollone, V. Rancic, A. Sucapane, S. Giordani, L. Gambazzi, H.
Markram, M. Grandolfo, D. Scaini, F. Gelain, L. Casalis, M. Prato, M. Giugliano, L. Ballerini, Carbon
nanotubes might improve neuronal performance by favouring electrical shortcuts, Nat. Nanotechnol. 4
(2009) 126-133.
[39] S.Y. Ng, J. Reboud, K.Y.P. Wang, K.C. Tang, L. Zhang, P. Wong, K.T. Moe, W. Shim, Y.
Chen, Label-free impedance detection of low levels of circulating endothelial progenitor cells for
point-of-care diagnosis, Biosens. Bioelectron. 25 (2010) 1095-1101.
[40] E. Du, S. Ha, M. Diez-Silva, M. Dao, S. Suresh, A.P. Chandrakasan, Electric impedance
microflow cytometry for characterization of cell disease states, Lab Chip 13 (2013) 3903-3909.
[41] C.E. Nwankire, A. Venkatanarayanan, T. Glennon, T.E. Keyes, R.J. Forster, J. Ducrée,
Label-free impedance detection of cancer cells from whole blood on an integrated centrifugal
microfluidic platform, Biosens. Bioelectron. 68 (2015) 382-389.
[42] M.S. Mannoor, S. Zhang, A.J. Link, M.C. McAlpine, Electrical detection of pathogenic
bacteria via immobilized antimicrobial peptides, Proc. Natl. Acad. Sci. U. S. A. 107 (2010)
19207-19212.
[43] S. Gawad, K. Cheung, U. Seger, A. Bertsch, P. Renaud, Dielectric spectroscopy in a
micromachined flow cytometer: Theoretical and practical considerations, Lab Chip 4 (2004) 241-251.
[44] T. Braschler, N. Demierre, E. Nascimento, T. Silva, A.G. Oliva, P. Renaud, Continuous
separation of cells by balanced dielectrophoretic forces at multiple frequencies, Lab Chip 8 (2008)
280-286.
[45] N. Demierre, T. Braschler, P. Linderholm, U. Seger, H. van Lintel, P. Renaud,
Characterization and optimization of liquid electrodes for lateral dielectrophoresis, Lab Chip 7 (2007)
355-365.
[46] N. Demierre, T. Braschler, R. Muller, P. Renaud, Focusing and continuous separation of
cells in a microfluidic device using lateral dielectrophoresis, Sens. Actuator B-Chem. 132 (2008)
388-396.
[47] P. Zhang, L. Ren, X. Zhang, Y. Shan, Y. Wang, Y. Ji, H. Yin, W.E. Huang, J. Xu, B. Ma,
Raman-Activated Cell Sorting Based on Dielectrophoretic Single-Cell Trap and Release, Anal. Chem.
87 (2015) 2282-2289.
[48] M.G. Simon, Y. Li, J. Arulmoli, L.P. McDonnell, A. Akil, J.L. Nourse, A.P. Lee, L.A.
Flanagan, Increasing label-free stem cell sorting capacity to reach transplantation-scale throughput,
Biomicrofluidics 8 (2014) 64106.
[49] H. Song, J.M. Rosano, Y. Wang, C.J. Garson, B. Prabhakarpandian, K. Pant, G.J. Klarmann,
A. Perantoni, L.M. Alvarez, E. Lai, Continuous-flow sorting of stem cells and differentiation products
based on dielectrophoresis, Lab Chip 15 (2015) 1320-1328.
[50] G. Lee, H. Wu, P. Yang, J.D. Mai, Optically induced dielectropheresis sorting with
automated medium exchange in an integrated optofluidic device resulting in higher cell viability, Lab
Chip 14 (2014) 2837-2843.
[51] Z. Wei, X. Li, D. Zhao, H. Yan, Z. Hu, Z. Liang, Z. Li, Flow-through cell electroporation
microchip integrating dielectrophoretic viable cell sorting, Anal. Chem. 86 (2014) 10215-10222.

31

Page 31 of 55
[52] J. DuBose, X. Lu, S. Patel, S. Qian, S.W. Joo, X. Xuan, Microfluidic electrical sorting of
particles based on shape in a spiral microchannel, Biomicrofluidics 8 (2014) 14101.
[53] J. Zhu, X. Xuan, Curvature-induced dielectrophoresis for continuous separation of particles
by charge in spiral microchannels, Biomicrofluidics 5 (2011) 24111.
[54] R.K. Anand, E.S. Johnson, D.T. Chiu, Negative dielectrophoretic capture and repulsion of
single cells at a bipolar electrode: The impact of faradaic ion enrichment and depletion, J. Am. Chem.
Soc. 137 (2015) 776-783.
[55] A.J. Hughes, A.E. Herr, Microfluidic western blotting, Proc. Natl. Acad. Sci. U. S. A. 109
(2012) 21450-21455.
[56] P. Batalla, A. Martin, M. Angel Lopez, M. Cristina Gonzalez, A. Escarpa, Enzyme-based
microfluidic chip coupled to graphene electrodes for the detection of D-Amino acid
enantiomer-biomarkers, Anal. Chem. 87 (2015) 5074-5078.
[57] K. Kadimisetty, S. Malla, N.P. Sardesai, A.A. Joshi, R.C. Faria, N.H. Lee, J.F. Rusling,
Automated multiplexed ECL immunoarrays for cancer biomarker proteins, Anal. Chem. 87 (2015)
4472-4478.
[58] R. Vaidyanathan, L.M. van Leeuwen, S. Rauf, M.J.A. Shiddiky, M. Trau, A multiplexed
device based on tunable nanoshearing for specific detection of multiple protein biomarkers in serum,
Sci. Rep. 5 (2015) 9756.
[59] L. Wu, C. Ma, L. Ge, Q. Kong, M. Yan, S. Ge, J. Yu, Paper-based
electrochemiluminescence origami cyto-device for multiple cancer cells detection using porous AuPd
alloy as catalytically promoted nanolabels, Biosens. Bioelectron. 63 (2015) 450-457.
[60] A.C. Glavan, D.C. Christodouleas, B. Mosadegh, H.D. Yu, B.S. Smith, J. Lessing, M.
Teresa Fernandez-Abedul, G.M. Whitesides, Folding analytical devices for electrochemical ELISA in
hydrophobic R-H paper, Anal. Chem. 86 (2014) 11999-12007.
[61] X. Ding, P. Li, S.S. Lin, Z.S. Stratton, N. Nama, F. Guo, D. Slotcavage, X. Mao, J. Shi, F.
Costanzo, T.J. Huang, Surface acoustic wave microfluidics, Lab Chip 13 (2013) 3626-3649.
[62] P.R. Rogers, J.R. Friend, L.Y. Yeo, Exploitation of surface acoustic waves to drive
size-dependent microparticle concentration within a droplet, Lab Chip 10 (2010) 2979-2985.
[63] T. Franke, S. Braunmuller, L. Schmid, A. Wixforth, D.A. Weitz, Surface acoustic wave
actuated cell sorting (SAWACS), Lab Chip 10 (2010) 789-794.
[64] L. Schmid, T. Franke, SAW-controlled drop size for flow focusing, Lab Chip 13 (2013)
1691-1694.
[65] J. Reboud, Y. Bourquin, R. Wilson, G.S. Pall, M. Jiwaji, A.R. Pitt, A. Graham, A.P. Waters,
J.M. Cooper, Shaping acoustic fields as a toolset for microfluidic manipulations in diagnostic
technologies, Proc. Natl. Acad. Sci. U. S. A. 109 (2012) 15162-15167.
[66] M. Travagliati, R.J. Shilton, M. Pagliazzi, I. Tonazzini, F. Beltram, M. Cecchini,
Acoustofluidics and whole-blood Manipulation in Surface Acoustic Wave Counterflow Devices, Anal.
Chem. 86 (2014) 10633-10638.
[67] S. Li, X. Ding, Z. Mao, Y. Chen, N. Nama, F. Guo, P. Li, L. Wang, C.E. Cameron, T.J.
Huang, Standing surface acoustic wave (SSAW)-based cell washing, Lab Chip 15 (2015) 331-338.
[68] X. Ding, S.S. Lin, B. Kiraly, H. Yue, S. Li, I. Chiang, J. Shi, S.J. Benkovic, T.J. Huang,
On-chip manipulation of single microparticles, cells, and organisms using surface acoustic waves, Proc.
Natl. Acad. Sci. U. S. A. 109 (2012) 11105-11109.
[69] R.P. Richter, R. Bérat, A.R. Brisson, Formation of solid-supported Lipid Bilayers: An

32

Page 32 of 55
Integrated View, Langmuir 22 (2006) 3497-3505.
[70] M. Hennig, J. Neumann, A. Wixforth, J.O. Radler, M.F. Schneider, Dynamic patterns in a
supported lipid bilayer driven by standing surface acoustic waves, Lab Chip 9 (2009) 3050-3053.
[71] M. Hennig, M. Wolff, J. Neumann, A. Wixforth, M.F. Schneider, J.O. Rädler, DNA
concentration modulation on supported lipid bilayers switched by surface acoustic waves, Langmuir 27
(2011) 14721-14725.
[72] J. Neumann, M. Hennig, A. Wixforth, S. Manus, J.O. Rädler, M.F. Schneider, Transport,
separation, and accumulation of proteins on supported lipid bilayers, Nano Lett 10 (2010) 2903-2908.
[73] P. Li, Z. Mao, Z. Peng, L. Zhou, Y. Chen, P. Huang, C.I. Truica, J.J. Drabick, W.S. El-Deiry,
M. Dao, S. Suresh, T.J. Huang, Acoustic separation of circulating tumor cells, Proc. Natl. Acad. Sci. U.
S. A. 112 (2015) 4970-4975.
[74] X. Ding, Z. Peng, S.S. Lin, M. Geri, S. Li, P. Li, Y. Chen, M. Dao, S. Suresh, T.J. Huang,
Cell separation using tilted-angle standing surface acoustic waves, Proc. Natl. Acad. Sci. U. S. A. 111
(2014) 12992-12997.
[75] S. Li, F. Guo, Y. Chen, X. Ding, P. Li, L. Wang, C.E. Cameron, T.J. Huang, Standing
surface acoustic wave based cell coculture, Anal. Chem. 86 (2014) 9853-9859.
[76] A. van Reenen, A.M. de Jong, J.M.J. den Toonder, M.W.J. Prins, Integrated lab-on-chip
biosensing systems based on magnetic particle actuation - a comprehensive review, Lab Chip 14 (2014)
1966-1986.
[77] M. Vilfan, A. Potocnik, B. Kavcic, N. Osterman, I. Poberaj, A. Vilfan, D. Babic,
Self-assembled artificial cilia, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 1844-1847.
[78] G. Zhu, N. Nguyen, Rapid magnetofluidic mixing in a uniform magnetic field, Lab Chip 12
(2012) 4772-4780.
[79] A. Ranzoni, X.J.A. Janssen, M. Ovsyanko, L.J. van IJzendoorn, M.W.J. Prins, Magnetically
controlled rotation and torque of uniaxial microactuators for lab-on-a-chip applications, Lab Chip 10
(2010) 179-188.
[80] M. Karle, J. Miwa, G. Czilwik, V. Auwarter, G. Roth, R. Zengerle, F. von Stetten,
Continuous microfluidic DNA extraction using phase-transfer magnetophoresis, Lab Chip 10 (2010)
3284-3290.
[81] R.C. den Dulk, K.A. Schmidt, G. Sabatte, S. Liebana, M.W.J. Prins, Magneto-capillary valve
for integrated purification and enrichment of nucleic acids and proteins, Lab Chip 13 (2013) 106-118.
[82] O. Strohmeier, A. Emperle, G. Roth, D. Mark, R. Zengerle, F. von Stetten, Centrifugal
gas-phase transition magnetophoresis (GTM) - a generic method for automation of magnetic bead
based assays on the centrifugal microfluidic platform and application to DNA purification, Lab Chip 13
(2013) 146-155.
[83] G. Lin, D. Makarov, M. Medina-Sanchez, M. Guix, L. Baraban, G. Cuniberti, O.G. Schmidt,
Magnetofluidic platform for multidimensional magnetic and optical barcoding of droplets, Lab Chip 15
(2015) 216-224.
[84] S.Y. Park, H. Handa, A. Sandhu, Magneto-optical biosensing platform based on light
scattering from self-assembled chains of functionalized rotating magnetic beads, Nano Lett 10 (2010)
446-451.
[85] M. Cornaglia, R. Trouillon, H.C. Tekin, T. Lehnert, M.A.M. Gijs, Magnetic
particle-scanning for ultrasensitive immunodetection On-Chip, Anal. Chem. 86 (2014) 8213-8223.
[86] C. Chiou, D. Jin Shin, Y. Zhang, T. Wang, Topography-assisted electromagnetic platform

33

Page 33 of 55
for blood-to-PCR in a droplet, Biosens. Bioelectron. 50 (2013) 91-99.
[87] C. Chang, W. Huang, S.I. Jalal, B. Chan, A. Mahmood, S. Shahda, B.H. O'Neil, D.E. Matei,
C.A. Savran, Circulating tumor cell detection using a parallel flow micro-aperture chip system, Lab
Chip 15 (2015) 1677-1688.
[88] A. van Reenen, Y. Gao, A.H. Bos, A.M. de Jong, M.A. Hulsen, J.M.J. den Toonder, M.W.J.
Prins, Accurate quantification of magnetic particle properties by intra-pair magnetophoresis for
nanobiotechnology, Appl. Phys. Lett. 103 (2013) 43704.
[89] F. Hernández, T. Portolés, E. Pitarch, F.J. López, Gas chromatography coupled to
high-resolution time-of-flight mass spectrometry to analyze trace-level organic compounds in the
environment, food safety and toxicology, TrAC, Trends Anal. Chem. 30 (2011) 388-400.
[90] G. Wei, E.Y. Zeng, Gas chromatography-mass spectrometry and high-performance liquid
chromatography-tandem mass spectrometry in quantifying fatty acids, TrAC, Trends Anal. Chem. 30
(2011) 1429-1436.
[91] T. Cajka, O. Fiehn, Comprehensive analysis of lipids in biological systems by liquid
chromatography-mass spectrometry, TrAC, Trends Anal. Chem. 61 (2014) 192-206.
[92] P.D. Rainville, G. Theodoridis, R.S. Plumb, I.D. Wilson, Advances in liquid
chromatography coupled to mass spectrometry for metabolic phenotyping, TrAC, Trends Anal. Chem.
61 (2014) 181-191.
[93] A. Hirayama, M. Wakayama, T. Soga, Metabolome analysis based on capillary
electrophoresis-mass spectrometry, TrAC, Trends Anal. Chem. 61 (2014) 215-222.
[94] C. Seger, S. Sturm, H. Stuppner, Mass spectrometry and NMR spectroscopy: Modern
high-end detectors for high resolution separation techniques - state of the art in natural product
HPLC-MS, HPLC-NMR, and CE-MS hyphenations, Nat. Prod. Rep. 30 (2013) 970-987.
[95] A.R. Timerbaev, Capillary electrophoresis coupled to mass spectrometry for biospeciation
analysis: Critical evaluation, TrAC, Trends Anal. Chem. 28 (2009) 416-425.
[96] Q.F. Xue, F. Foret, Y.M. Dunayevskiy, P.M. Zavracky, N.E. McGruer, B.L. Karger,
Multichannel microchip electrospray mass spectrometry, Anal. Chem. 69 (1997) 426-430.
[97] R.S. Ramsey, J.M. Ramsey, Generating electrospray from microchip devices using
electroosmotic pumping, Anal. Chem. 69 (1997) 1174-1178.
[98] X. Feng, B. Liu, J. Li, X. Liu, Advances in coupling microfluidic chips to mass spectrometry,
Mass Spectrom. Rev. 34 (2015) 535-557.
[99] H. Wei, H. Li, J.-M. Lin, Analysis of herbicides on a single C30 bead via a microfluidic
device combined with electrospray ionization quadrupole time-of-flight mass spectrometer, J.
Chromatogr. A 1216 (2009) 9134-9142.
[100] H. Wei, H. Li, D. Gao, J.-M. Lin, Multi-channel microfluidic devices combined with
electrospray ionization quadrupole time-of-flight mass spectrometry applied to the monitoring of
glutamate release from neuronal cells, Analyst 135 (2010) 2043-2050.
[101] D. Gao, H. Wei, G.S. Guo, J.-M. Lin, Microfluidic cell culture and metabolism detection
with electrospray ionization quadrupole time-of-flight mass spectrometer, Anal. Chem. 82 (2010)
5679-5685.
[102] D. Gao, H. Li, N. Wang, J.-M. Lin, Evaluation of the absorption of methotrexate on cells and
its cytotoxicity assay by using an integrated microfluidic device coupled to a mass spectrometer, Anal.
Chem. 84 (2012) 9230-9237.
[103] D. Gao, H. Liu, J.-M. Lin, Y. Wang, Y. Jiang, Characterization of drug permeability in

34

Page 34 of 55
Caco-2 monolayers by mass spectrometry on a membrane-based microfluidic device, Lab Chip 13
(2013) 978-985.
[104] Q. Chen, J. Wu, Y. Zhang, J.-M. Lin, Qualitative and quantitative analysis of tumor cell
metabolism via stable isotope labeling assisted microfluidic chip electrospray ionization mass
spectrometry, Anal. Chem. 84 (2012) 1695-1701.
[105] H. Wei, H. Li, S. Mao, J.-M. Lin, Cell signaling analysis by mass spectrometry under
coculture conditions on an integrated microfluidic device, Anal. Chem. 83 (2011) 9306-9313.
[106] S. Mao, J. Zhang, H. Li, J.-M. Lin, Strategy for signaling molecule detection by using an
integrated microfluidic device coupled with mass spectrometry to study Cell-to-Cell communication,
Anal. Chem. 85 (2013) 868-876.
[107] W. Liu, Q. Chen, X. Lin, J.-M. Lin, Online multi-channel microfluidic chip-mass
spectrometry and its application for quantifying noncovalent protein-protein interactions, Analyst 140
(2015) 1551-1554.
[108] E.A. Redman, N.G. Batz, J.S. Mellors, J.M. Ramsey, Integrated microfluidic capillary
electrophoresis-electrospray ionization devices with online MS detection for the separation and
characterization of intact monoclonal antibody variants, Anal. Chem. 87 (2015) 2264-2272.
[109] F.T.G. van den Brink, L. Büter, M. Odijk, W. Olthuis, U. Karst, A. van den Berg, Mass
spectrometric detection of short-lived drug metabolites generated in an electrochemical microfluidic
chip, Anal. Chem. 87 (2015) 1527-1535.
[110] Q. Wu, D. Gao, J. Wei, F. Jin, W. Xie, Y. Jiang, H. Liu, Development of a novel multi-layer
microfluidic device towards characterization of drug metabolism and cytotoxicity for drug screening,
Chem. Commun. 50 (2014) 2762-2764.
[111] A.E. Kirby, N.M. Lafrenière, B. Seale, P.I. Hendricks, R.G. Cooks, A.R. Wheeler, Analysis
on the go: Quantitation of drugs of abuse in dried urine with digital microfluidics and miniature mass
spectrometry, Anal. Chem. 86 (2014) 6121-6129.
[112] D. Jin, Y. Zhu, Q. Fang, Swan probe: A nanoliter-scale and high-throughput sampling
interface for coupling electrospray ionization mass spectrometry with microfluidic droplet array and
multiwell plate, Anal. Chem. 86 (2014) 10796-10803.
[113] S. Arscott, SU-8 as a material for lab-on-a-chip-based mass spectrometry, Lab Chip 14
(2014) 3668-3689.
[114] D. Gao, H. Liu, Y. Jiang, J.-M. Lin, Recent advances in microfluidics combined with mass
spectrometry: Technologies and applications, Lab Chip 13 (2013) 3309-3322.
[115] X. He, Q. Chen, Y. Zhang, J.-M. Lin, Recent advances in microchip-mass spectrometry for
biological analysis, TrAC, Trends Anal. Chem. 53 (2014) 84-97.
[116] S. Allazetta, L. Kolb, S. Zerbib, J. Bardy, M.P. Lutolf, Cell-instructive microgels with
Tailor-made physicochemical properties, Small 11 (2015) 5647-5656.
[117] C.A. Bichsel, S. Gobaa, S. Kobel, C. Secondini, G.N. Thalmann, M.G. Cecchini, M.P.
Lutolf, Diagnostic microchip to assay 3D colony-growth potential of captured circulating tumor cells,
Lab Chip 12 (2012) 2313-2316.
[118] S. Allazetta, S. Cosson, M.P. Lutolf, Programmable microfluidic patterning of protein
gradients on hydrogels, Chem. Commun. 47 (2011) 191-193.
[119] S. Cosson, S. Allazetta, M.P. Lutolf, Patterning of cell-instructive hydrogels by
hydrodynamic flow focusing, Lab Chip 13 (2013) 2099-2105.
[120] S. Cosson, S.A. Kobel, M.P. Lutolf, Capturing complex protein gradients on biomimetic

35

Page 35 of 55
hydrogels for cell-based assays, Adv. Funct. Mater. 19 (2009) 3411-3419.
[121] S.H. Mathes, H. Ruffner, U. Graf-Hausner, The use of skin models in drug development,
Adv. Drug Deliver. Rev. 69–70 (2014) 81-102.
[122] Y. Li, S. Wang, R. Huang, Z. Huang, B. Hu, W. Zheng, G. Yang, X. Jiang, Evaluation of the
effect of the structure of bacterial cellulose on full thickness skin wound repair on a microfluidic chip,
Biomacromolecules 16 (2015) 780-789.
[123] Y. Torisawa, C.S. Spina, T. Mammoto, A. Mammoto, J.C. Weaver, T. Tat, J.J. Collins, D.E.
Ingber, Bone marrow-on-a-chip replicates hematopoietic niche physiology in vitro, Nat. Methods 11
(2014) 663-669.
[124] J. Wu, Q. Chen, W. Liu, Y. Zhang, J.-M. Lin, Cytotoxicity of quantum dots assay on a
microfluidic 3D-culture device based on modeling diffusion process between blood vessels and tissues,
Lab Chip 12 (2012) 3474-3480.
[125] M.R. Ebrahimkhani, J.A.S. Neiman, M.S.B. Raredon, D.J. Hughes, L.G. Griffith, Bioreactor
technologies to support liver function in vitro, Adv. Drug Deliver. Rev. 69–70 (2014) 132-157.
[126] S. Mao, D. Gao, W. Liu, H. Wei, J.-M. Lin, Imitation of drug metabolism in human liver and
cytotoxicity assay using a microfluidic device coupled to mass spectrometric detection, Lab Chip 12
(2012) 219-226.
[127] J. Park, B.K. Lee, G.S. Jeong, J.K. Hyun, C.J. Lee, S. Lee, Three-dimensional
brain-on-a-chip with an interstitial level of flow and its application as an in vitro model of Alzheimer's
disease, Lab Chip 15 (2015) 141-150.
[128] Y. Zheng, J. Chen, M. Craven, N.W. Choi, S. Totorica, A. Diaz-Santana, P. Kermani, B.
Hempstead, C. Fischbach-Teschl, J.A. Lopez, A.D. Stroock, In vitro microvessels for the study of
angiogenesis and thrombosis, Proc. Natl. Acad. Sci. U. S. A. 109 (2012) 9342-9347.
[129] J.S. Jeon, S. Bersini, M. Gilardi, G. Dubini, J.L. Charest, M. Moretti, R.D. Kamm, Human
3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation, Proc. Natl.
Acad. Sci. U. S. A. 112 (2015) 214-219.
[130] Y. Li, S. Zhang, X. Wang, X. Zhang, A.I. Oleinick, I. Svir, C. Amatore, W. Huang,
Real-time monitoring of discrete synaptic release events and excitatory potentials within
self-reconstructed neuromuscular junctions, Angew. Chem. Int. Edit. 54 (2015) 9313-9318.
[131] M. Wu, S. Huang, G. Lee, Microfluidic cell culture systems for drug research, Lab Chip 10
(2010) 939-956.
[132] J.H. Tsui, W. Lee, S.H. Pun, J. Kim, D. Kim, Microfluidics-assisted in vitro drug screening
and carrier production, Adv. Drug Deliver. Rev. 65 (2013) 1575-1588.
[133] O.J. Miller, A. El Harrak, T. Mangeat, J. Baret, L. Frenz, B. El Debs, E. Mayot, M.L.
Samuels, E.K. Rooney, P. Dieu, M. Galvan, D.R. Link, A.D. Griffiths, High-resolution dose-response
screening using droplet-based microfluidics, Proc. Natl. Acad. Sci. U. S. A. 109 (2012) 378-383.
[134] A.I. Shallan, R.M. Guijt, M.C. Breadmore, Electrokinetic size and mobility traps for on-site
therapeutic drug monitoring, Angew. Chem. Int. Edit. 54 (2015) 7359-7362.
[135] H. Wang, C. Chen, Z. Xiang, M. Wang, C. Lee, A convection-driven long-range linear
gradient generator with dynamic control, Lab Chip 15 (2015) 1445-1450.
[136] C. Kim, J. Kasuya, J. Jeon, S. Chung, R.D. Kamm, A quantitative microfluidic angiogenesis
screen for studying anti-angiogenic therapeutic drugs, Lab Chip 15 (2015) 301-310.
[137] Z. Dereli-Korkut, H.D. Akaydin, A.H.R. Ahmed, X. Jiang, S. Wang, Three dimensional
microfluidic cell arrays for ex vivo drug screening with mimicked vascular flow, Anal. Chem. 86 (2014)

36

Page 36 of 55
2997-3004.
[138] J. Wu, Q. Chen, W. Liu, J.-M. Lin, A simple and versatile microfluidic cell density gradient
generator for quantum dot cytotoxicity assay, Lab Chip 13 (2013) 1948-1954.
[139] E.W. Esch, A. Bahinski, D. Huh, Organs-on-chips at the frontiers of drug discovery, Nat.
Rev. Drug Discov. 14 (2015) 248-260.
[140] P. Neuži, S. Giselbrecht, K. Länge, T.J. Huang, A. Manz, Revisiting lab-on-a-chip
technology for drug discovery, Nat. Rev. Drug Discov. 11 (2012) 620-632.
[141] C.Y. Chan, P. Huang, F. Guo, X. Ding, V. Kapur, J.D. Mai, P.K. Yuen, T.J. Huang,
Accelerating drug discovery via organs-on-chips, Lab Chip 13 (2013) 4697-4710.
[142] H.E. Abaci, K. Gledhill, Z. Guo, A.M. Christiano, M.L. Shuler, Pumpless microfluidic
platform for drug testing on human skin equivalents, Lab Chip 15 (2015) 882-888.
[143] J. Zhang, J. Wu, H. Li, Q. Chen, J.-M. Lin, An in vitro liver model on microfluidic device
for analysis of capecitabine metabolite using mass spectrometer as detector, Biosens. Bioelectron. 68
(2015) 322-328.
[144] S.H. Au, M.D. Chamberlain, S. Mahesh, M.V. Sefton, A.R. Wheeler, Hepatic organoids for
microfluidic drug screening, Lab Chip 14 (2014) 3290-3299.
[145] A. Agarwal, J.A. Goss, A. Cho, M.L. McCain, K.K. Parker, Microfluidic heart on a chip for
higher throughput pharmacological studies, Lab Chip 13 (2013) 3599-3608.
[146] I. Maschmeyer, A.K. Lorenz, K. Schimek, T. Hasenberg, A.P. Ramme, J. Hubner, M.
Lindner, C. Drewell, S. Bauer, A. Thomas, N.S. Sambo, F. Sonntag, R. Lauster, U. Marx, A
four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney
equivalents, Lab Chip 15 (2015) 2688-2699.
[147] H. Tanaka, S. Yamamoto, A. Nakamura, Y. Nakashoji, N. Okura, N. Nakamoto, K.
Tsukagoshi, M. Hashimoto, Hands-Off preparation of monodisperse emulsion droplets using a
poly(dimethylsiloxane) microfluidic chip for droplet digital PCR, Anal. Chem. 87 (2015) 4134-4143.
[148] P. Dalerba, T. Kalisky, D. Sahoo, P.S. Rajendran, M.E. Rothenberg, A.A. Leyrat, S. Sim, J.
Okamoto, D.M. Johnston, D. Qian, M. Zabala, J. Bueno, N.F. Neff, J. Wang, A.A. Shelton, B. Visser,
S. Hisamori, Y. Shimono, M. van de Wetering, H. Clevers, M.F. Clarke, S.R. Quake, Single-cell
dissection of transcriptional heterogeneity in human colon tumors, Nat. Biotechnol. 29 (2011)
1120-1127.
[149] X. Lin, J. Wu, W. Liu, H. Li, Z. Wang, J.-M. Lin, Detection of BCR-ABL using one step
reverse transcriptase- polymerase chain reaction and microchip electrophoresis, Methods 64 (2013)
250-254.
[150] X. Lin, J. Wu, H. Li, Z. Wang, J.-M. Lin, Determination of mini-short tandem repeat
(miniSTR) loci by using the combination of polymerase chain reaction (PCR) and microchip
electrophoresis, Talanta 114 (2013) 131-137.
[151] S.J. Reinholt, A.J. Baeumner, Microfluidic isolation of nucleic acids, Angew. Chem. Int.
Edit. 53 (2014) 13988-14001.
[152] X. Shi, C. Chen, W. Gao, S. Chao, D.R. Meldrum, Parallel RNA extraction using magnetic
beads and a droplet array, Lab Chip 15 (2015) 1059-1065.
[153] A.K. White, M. VanInsberghe, O.I. Petriv, M. Hamidi, D. Sikorski, M.A. Marra, J. Piret, S.
Aparicio, C.L. Hansen, High-throughput microfluidic single-cell RT-qPCR, Proc. Natl. Acad. Sci. U. S.
A. 108 (2011) 13999-14004.
[154] H. Ben-Yoav, P.H. Dykstra, W.E. Bentley, R. Ghodssi, A controlled microfluidic

37

Page 37 of 55
electrochemical lab-on-a-chip for label-free diffusion-restricted DNA hybridization analysis, Biosens.
Bioelectron. 64 (2015) 579-585.
[155] S.P. White, K.D. Dorfman, C.D. Frisbie, Label-Free DNA sensing platform with
Low-Voltage Electrolyte-Gated transistors, Anal. Chem. 87 (2015) 1861-1866.
[156] M.E. Polinkovsky, Y. Gambin, P.R. Banerjee, M.J. Erickstad, A. Groisman, A.A. Deniz,
Ultrafast cooling reveals microsecond-scale biomolecular dynamics, Nat. Commun. 5 (2014) 5737.
[157] H. Shao, J. Chung, K. Lee, L. Balaj, C. Min, B.S. Carter, F.H. Hochberg, X.O. Breakefield,
H. Lee, R. Weissleder, Chip-based analysis of exosomal mRNA mediating drug resistance in
glioblastoma, Nat. Commun. 6 (2015) 6999.
[158] Y. Zhao, D. Chen, H. Yue, M.M. Spiering, C. Zhao, S.J. Benkovic, T.J. Huang, Dark-field
illumination on zero-mode waveguide/microfluidic hybrid chip reveals T4 replisomal protein
interactions, Nano Lett 14 (2014) 1952-1960.
[159] A.L. Forget, C.C. Dombrowski, I. Amitani, S.C. Kowalczykowski, Exploring protein-DNA
interactions in 3D using in situ construction, manipulation and visualization of individual DNA
dumbbells with optical traps, microfluidics and fluorescence microscopy, Nat. Protoc. 8 (2013)
525-538.
[160] P. Colin, B. Kintses, F. Gielen, C.M. Miton, G. Fischer, M.F. Mohamed, M. Hyvonen, D.P.
Morgavi, D.B. Janssen, F. Hollfelder, Ultrahigh-throughput discovery of promiscuous enzymes by
picodroplet functional metagenomics, Nat. Commun. 6 (2015) 10008.
[161] W. Busch, B.T. Moore, B. Martsberger, D.L. Mace, R.W. Twigg, J. Jung, I.
Pruteanu-Malinici, S.J. Kennedy, G.K. Fricke, R.L. Clark, U. Ohler, P.N. Benfey, A microfluidic
device and computational platform for high-throughput live imaging of gene expression, Nat. Methods
9 (2012) 1101-1106.
[162] M. Cho, Y. Xiao, J. Nie, R. Stewart, A.T. Csordas, S.S. Oh, J.A. Thomson, H.T. Soh,
Quantitative selection of DNA aptamers through microfluidic selection and high-throughput
sequencing, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 15373-15378.
[163] A.M. Streets, X. Zhang, C. Cao, Y. Pang, X. Wu, L. Xiong, L. Yang, Y. Fu, L. Zhao, F.
Tang, Y. Huang, Microfluidic single-cell whole-transcriptome sequencing, Proc. Natl. Acad. Sci. U. S.
A. 111 (2014) 7048-7053.
[164] M.C. Jo, W. Liu, L. Gu, W. Dang, L. Qin, High-throughput analysis of yeast replicative
aging using a microfluidic system, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) 9364-9369.
[165] V. Krivitsky, L. Hsiung, A. Lichtenstein, B. Brudnik, R. Kantaev, R. Elnathan, A. Pevzner,
A. Khatchtourints, F. Patolsky, Si nanowires forest-based on-chip biomolecular filtering, separation
and preconcentration devices: Nanowires do it all, Nano Lett 12 (2012) 4748-4756.
[166] A. Shastri, L.M. McGregor, Y. Liu, V. Harris, H. Nan, M. Mujica, Y. Vasquez, A.
Bhattacharya, Y. Ma, M. Aizenberg, O. Kuksenok, A.C. Balazs, J. Aizenberg, X. He, An
aptamer-functionalized chemomechanically modulated biomolecule catch-and-release system, Nat.
Chem. 7 (2015) 447-454.
[167] M. Fischlechner, Y. Schaerli, M.F. Mohamed, S. Patil, C. Abell, F. Hollfelder, Evolution of
enzyme catalysts caged in biomimetic gel-shell beads, Nat. Chem. 6 (2014) 791-796.
[168] P.A. Romero, T.M. Tran, A.R. Abate, Dissecting enzyme function with microfluidic-based
deep mutational scanning, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) 7159-7164.
[169] M. Otten, W. Ott, M.A. Jobst, L.F. Milles, T. Verdorfer, D.A. Pippig, M.A. Nash, H.E. Gaub,
From genes to protein mechanics on a chip, Nat. Methods 11 (2014) 1127-1130.

38

Page 38 of 55
[170] X. Lin, Q. Chen, W. Liu, J. Zhang, S. Wang, Z. Lin, J.-M. Lin, Oxygen-induced cell
migration and on-line monitoring biomarkers modulation of cervical cancers on a microfluidic system,
Sci. Rep. 5 (2015) 9643.
[171] X. Lin, Q. Chen, W. Liu, H. Li, J.-M. Lin, A portable microchip for ultrasensitive and
high-throughput assay of thrombin by rolling circle amplification and hemin/G-quadruplex system,
Biosens. Bioelectron. 56 (2014) 71-76.
[172] X. Lin, Q. Chen, W. Liu, L. Yi, H. Li, Z. Wang, J.-M. Lin, Assay of multiplex proteins from
cell metabolism based on tunable aptamer and microchip electrophoresis, Biosens. Bioelectron. 63
(2015) 105-111.
[173] J. Mok, M.N. Mindrinos, R.W. Davis, M. Javanmard, Digital microfluidic assay for protein
detection, Proc. Natl. Acad. Sci. U. S. A. 111 (2014) 2110-2115.
[174] M. Ihara, A. Yoshikawa, Y. Wu, H. Takahashi, K. Mawatari, K. Shimura, K. Sato, T.
Kitamori, H. Ueda, Micro OS-ELISA: Rapid noncompetitive detection of a small biomarker peptide by
open-sandwich enzyme-linked immunosorbent assay (OS-ELISA) integrated into microfluidic device,
Lab Chip 10 (2010) 92-100.
[175] J. Kai, A. Puntambekar, N. Santiago, S.H. Lee, D.W. Sehy, V. Moore, J. Han, C.H. Ahn, A
novel microfluidic microplate as the next generation assay platform for enzyme linked immunoassays
(ELISA), Lab Chip 12 (2012) 4257-4262.
[176] T. Wang, M. Zhang, D.D. Dreher, Y. Zeng, Ultrasensitive microfluidic solid-phase ELISA
using an actuatable microwell-patterned PDMS chip, Lab Chip 13 (2013) 4190-4197.
[177] C. Cheng, A.W. Martinez, J. Gong, C.R. Mace, S.T. Phillips, E. Carrilho, K.A. Mirica, G.M.
Whitesides, Paper-based ELISA, Angew. Chem. Int. Edit. 49 (2010) 4771-4774.
[178] Y. Li, F. Xu, C. Liu, Y. Xu, X. Feng, B. Liu, A novel microfluidic mixer based on
dual-hydrodynamic focusing for interrogating the kinetics of DNA-protein interaction, Analyst 138
(2013) 4475-4482.
[179] C. Han, Q. Zhang, R. Ma, L. Xie, T. Qiu, L. Wang, K. Mitchelson, J. Wang, G. Huang, J.
Qiao, J. Cheng, Integration of single oocyte trapping, in vitrofertilization and embryo culture in a
microwell-structured microfluidic device, Lab Chip 10 (2010) 2848-2854.
[180] M.C. Park, J.Y. Hur, H.S. Cho, S. Park, K.Y. Suh, High-throughput single-cell
quantification using simple microwell-based cell docking and programmable time-course live-cell
imaging, Lab Chip 11 (2011) 79-86.
[181] P. Zhang, L. Ren, X. Zhang, Y. Shan, Y. Wang, Y. Ji, H. Yin, W.E. Huang, J. Xu, B. Ma,
Raman-activated cell sorting based on dielectrophoretic single-cell trap and release, Anal. Chem. 87
(2015) 2282-2289.
[182] N. Chen, C. Chen, M. Chen, L. Jang, M. Wang, Single-cell trapping and impedance
measurement utilizing dielectrophoresis in a parallel-plate microfluidic device, Sens. Actuator B-Chem.
190 (2014) 570-577.
[183] D. Wlodkowic, S. Faley, M. Zagnoni, J.P. Wikswo, J.M. Cooper, Microfluidic single-cell
array cytometry for the analysis of tumor apoptosis, Anal. Chem. 81 (2009) 5517-5523.
[184] S. Koster, F.E. Angile, H. Duan, J.J. Agresti, A. Wintner, C. Schmitz, A.C. Rowat, C.A.
Merten, D. Pisignano, A.D. Griffiths, D.A. Weitz, Drop-based microfluidic devices for encapsulation
of single cells, Lab Chip 8 (2008) 1110-1115.
[185] J. Clausell-Tormos, D. Lieber, J. Baret, A. El-Harrak, O.J. Miller, L. Frenz, J. Blouwolff, K.J.
Humphry, S. Köster, H. Duan, C. Holtze, D.A. Weitz, A.D. Griffiths, C.A. Merten, Droplet-based

39

Page 39 of 55
microfluidic platforms for the encapsulation and screening of mammalian cells and multicellular
organisms, Chem. Biol. 15 (2008) 427-437.
[186] S. Kobel, A. Valero, J. Latt, P. Renaud, M. Lutolf, Optimization of microfluidic single cell
trapping for long-term on-chip culture, Lab Chip 10 (2010) 857-863.
[187] H. Chen, J. Sun, E. Wolvetang, J. Cooper-White, High-throughput, deterministic single cell
trapping and long-term clonal cell culture in microfluidic devices, Lab Chip 15 (2015) 1072-1083.
[188] C. Liberale, G. Cojoc, F. Bragheri, P. Minzioni, G. Perozziello, R. La Rocca, L. Ferrara, V.
Rajamanickam, E. Di Fabrizio, I. Cristiani, Integrated microfluidic device for single-cell trapping and
spectroscopy, Sci. Rep. 3 (2013) 1258.
[189] L. Wu, P. Chen, Y. Dong, X. Feng, B. Liu, Encapsulation of single cells on a microfluidic
device integrating droplet generation with fluorescence-activated droplet sorting, Biomed.
Microdevices 15 (2013) 553-560.
[190] C. Liu, J. Liu, D. Gao, M. Ding, J.-M. Lin, Fabrication of microwell arrays based on
two-dimensional ordered polystyrene microspheres for high-throughput single-cell analysis, Anal.
Chem. 82 (2010) 9418-9424.
[191] Q. Chen, J. Wu, Y. Zhang, Z. Lin, J.-M. Lin, Targeted isolation and analysis of single tumor
cells with aptamer-encoded microwell array on microfluidic device, Lab Chip 12 (2012) 5180-5185.
[192] J. Sun, J. Wang, P. Chen, X. Feng, W. Du, B. Liu, A chemical signal generator for resolving
temporal dynamics of single cells, Anal. Bioanal. Chem. 400 (2011) 2973-2981.
[193] Y. Wang, X. Tang, X. Feng, C. Liu, P. Chen, D. Chen, B. Liu, A microfluidic digital
single-cell assay for the evaluation of anticancer drugs, Anal. Bioanal. Chem. 407 (2015) 1139-1148.
[194] Y. Li, D. Chen, Y. Zhang, C. Liu, P. Chen, Y. Wang, X. Feng, W. Du, B. Liu,
High-throughput single cell multidrug resistance analysis with multifunctional gradients-customizing
microfluidic device, Sens. Actuator B-Chem. 225 (2016) 563-571.
[195] E.Z. Macosko, A. Basu, R. Satija, J. Nemesh, K. Shekhar, M. Goldman, I. Tirosh, A.R.
Bialas, N. Kamitaki, E.M. Martersteck, J.J. Trombetta, D.A. Weitz, J.R. Sanes, A.K. Shalek, A. Regev,
S.A. McCarroll, Highly parallel genome-wide expression profiling of individual cells using nanoliter
droplets, Cell 161 (2015) 1202-1214.
[196] A.M. Klein, L. Mazutis, I. Akartuna, N. Tallapragada, A. Veres, V. Li, L. Peshkin, D.A.
Weitz, M.W. Kirschner, Droplet barcoding for single-cell transcriptomics applied to embryonic stem
cells, Cell 161 (2015) 1187-1201.
[197] Y. Li, X. Feng, W. Du, Y. Li, B. Liu, Ultrahigh-throughput approach for analyzing
single-cell genomic damage with an agarose-based microfluidic comet array, Anal. Chem. 85 (2013)
4066-4073.
[198] Y. Nishikawa, M. Hosokawa, T. Maruyama, K. Yamagishi, T. Mori, H. Takeyama,
Monodisperse picoliter droplets for Low-Bias and Contamination-Free reactions in Single-Cell whole
genome amplification, Plos One 10 (2015) e138733.
[199] A.M. Skelley, O. Kirak, H. Suh, R. Jaenisch, J. Voldman, Microfluidic control of cell pairing
and fusion, Nat. Methods 6 (2009) 147-152.
[200] B. Dura, S.K. Dougan, M. Barisa, M.M. Hoehl, C.T. Lo, H.L. Ploegh, J. Voldman, Profiling
lymphocyte interactions at the single-cell level by microfluidic cell pairing, Nat. Commun. 6 (2015)
5940.
[201] L. Boneschansker, J. Yan, E. Wong, D.M. Briscoe, D. Irimia, Microfluidic platform for the
quantitative analysis of leukocyte migration signatures, Nat. Commun. 5 (2014) 4787.

40

Page 40 of 55
[202] J. Wu, H. Li, Q. Chen, X. Lin, W. Liu, J.-M. Lin, Statistical single-cell analysis of cell
cycle-dependent quantum dot cytotoxicity and cellular uptake using a microfluidic system, RSC Adv. 4
(2014) 24929-24934.
[203] R.A. Kellogg, R. Gómez-Sjöberg, A.A. Leyrat, S. Tay, High-throughput microfluidic
single-cell analysis pipeline for studies of signaling dynamics, Nat. Protoc. 9 (2014) 1713-1726.
[204] Q. Shi, L. Qin, W. Wei, F. Geng, R. Fan, Y.S. Shin, D. Guo, L. Hood, P.S. Mischel, J.R.
Heath, Single-cell proteomic chip for profiling intracellular signaling pathways in single tumor cells,
Proc. Natl. Acad. Sci. U. S. A. 109 (2012) 419-424.
[205] A.J. Hughes, D.P. Spelke, Z. Xu, C. Kang, D.V. Schaffer, A.E. Herr, Single-cell western
blotting, Nat. Methods 11 (2014) 749-755.
[206] F. Xu, H. Zhao, X. Feng, L. Chen, D. Chen, Y. Zhang, F. Nan, J. Liu, B. Liu, Single-cell
chemical proteomics with an activity-based probe: Identification of low-copy membrane proteins on
primary neurons, Angew. Chem. Int. Edit. 53 (2014) 6730-6733.
[207] S. Mahshid, M.J. Ahamed, D. Berard, S. Amin, R. Sladek, S.R. Leslie, W. Reisner,
Development of a platform for single cell genomics using convex lens-induced confinement, Lab Chip
15 (2015) 3013-3020.
[208] Y. Choi, P.N. Ingram, K. Yang, L. Coffman, M. Iyengar, S. Bai, D.G. Thomas, E. Yoon, R.J.
Buckanovich, Identifying an ovarian cancer cell hierarchy regulated by bone morphogenetic protein 2,
Proc Natl Acad Sci U. S. A. 112 (2015) E6882-E6888.
[209] P. Ertl, D. Sticker, V. Charwat, C. Kasper, G. Lepperdinger, Lab-on-a-chip technologies for
stem cell analysis, Trends Biotechnol. 32 (2014) 245-253.
[210] P. Occhetta, M. Centola, B. Tonnarelli, A. Redaelli, I. Martin, M. Rasponi, High-throughput
microfluidic platform for 3D cultures of mesenchymal stem cells, towards engineering developmental
processes, Sci. Rep. 5 (2015) 10288.
[211] V. Lecault, M. VanInsberghe, S. Sekulovic, D.J.H.F. Knapp, S. Wohrer, W. Bowden, F. Viel,
T. McLaughlin, A. Jarandehei, M. Miller, D. Falconnet, A.K. White, D.G. Kent, M.R. Copley, F.
Taghipour, C.J. Eaves, R.K. Humphries, J.M. Piret, C.L. Hansen, High-throughput analysis of single
hematopoietic stem cell proliferation in microfluidic cell culture arrays, Nat. Methods 8 (2011)
581-586.
[212] T. Cambier, T. Honegger, V. Vanneaux, J. Berthier, D. Peyrade, L. Blanchoin, J. Larghero,
M. Thery, Design of a 2D no-flow chamber to monitor hematopoietic stem cells, Lab Chip 15 (2015)
77-85.
[213] Q. Chen, J. Wu, Q. Zhuang, X. Lin, J. Zhang, J.-M. Lin, Microfluidic isolation of highly
pure embryonic stem cells using feeder-separated co-culture system, Sci. Rep. 3 (2013) 2433.
[214] S.J. Morrison, A.C. Spradling, Stem cells and niches: Mechanisms that promote stem cell
maintenance throughout life, Cell 132 (2008) 598-611.
[215] S. Kobel, M.P. Lutolf, High-throughput methods to define complex stem cell niches,
Biotechniques 48 (2010) IX-XXII.
[216] S. Allazetta, T.C. Hausherr, M.P. Lutolf, Microfluidic synthesis of cell-type-specific
artificial extracellular matrix hydrogels, Biomacromolecules 14 (2013) 1122-1131.
[217] S. Gobaa, S. Hoehnel, M. Roccio, A. Negro, S. Kobel, M.P. Lutolf, Artificial niche
microarrays for probing single stem cell fate in high throughput, Nat. Methods 8 (2011) 949-955.
[218] A. Roch, V. Trachsel, M.P. Lutolf, Brief Report: Single-cell analysis reveals cell
division-independent emergence of megakaryocytes from phenotypic hematopoietic stem cells, Stem

41

Page 41 of 55
Cells 33 (2015) 3152-3157.
[219] S. Allazetta, M.P. Lutolf, Stem cell niche engineering through droplet microfluidics, Curr.
Opin. Biotech. 35 (2015) 86-93.
[220] H. Xu, S.C. Heilshorn, Microfluidic investigation of BDNF-enhanced neural stem cell
chemotaxis in CXCL12 gradients, Small 9 (2013) 585-595.
[221] X. Shi, S. Chen, J. Zhou, H. Yu, L. Li, H. Wu, Directing osteogenesis of stem cells with
drug-laden, polymer-microsphere-based micropatterns generated by teflon microfluidic chips, Adv.
Funct. Mater. 22 (2012) 3799-3807.
[222] L.M. Przybyla, J. Voldman, Attenuation of extrinsic signaling reveals the importance of
matrix remodeling on maintenance of embryonic stem cell self-renewal, Proc. Natl. Acad. Sci. U. S. A.
109 (2012) 835-840.
[223] W. Kang, J.P. Giraldo-Vela, S.S.P. Nathamgari, T. McGuire, R.L. McNaughton, J.A.
Kessler, H.D. Espinosa, Microfluidic device for stem cell differentiation and localized electroporation
of postmitotic neurons, Lab Chip 14 (2014) 4486-4495.
[224] S. Reitinger, J. Wissenwasser, W. Kapferer, R. Heer, G. Lepperdinger, Electric impedance
sensing in cell-substrates for rapid and selective multipotential differentiation capacity monitoring of
human mesenchymal stem cells, Biosens. Bioelectron. 34 (2012) 63-69.
[225] Y. Kuo, J.H. Ho, T. Yen, H. Chen, O.K. Lee, Development of a surface plasmon resonance
biosensor for real-time detection of osteogenic differentiation in live mesenchymal stem cells, Plos One
6 (2011) e22382.
[226] A. Ranga, N. Gjorevski, M.P. Lutolf, Drug discovery through stem cell-based organoid
models, Adv. Drug Deliver. Rev. 69–70 (2014) 19-28.
[227] S. Kobel, M.P. Lutolf, Biomaterials meet microfluidics: Building the next generation of
artificial niches, Curr. Opin. Biotech. 22 (2011) 690-697.
[228] N. Gjorevski, A. Ranga, M.P. Lutolf, Bioengineering approaches to guide stem cell-based
organogenesis, Development 141 (2014) 1794-1804.

42

Page 42 of 55
Figure captions:
Fig. 1. Biochemical analysis integrated on microfluidic chip.

Fig. 2. Principle of single mouse hybridoma cell analysis and sorting depending on fluorescent

signals (Reprinted with permission from ref. [20]. Copyright 2013 Nature America, Inc.).

Fig. 3. AMP-based electrical detection of bacteria. (a) Schematic of AMPs immobilized on an

interdigitated microelectrode array. (b) Magnified image of theAMP magainin I in helical form,

modified with a terminal cysteine residue, and with clearly defined hydrophobic and hydrophilic

faces. (c) Detection of bacteriais achieved via binding of target cells to the immobilized AMPs. (d)

Image of the microfluidic chip (Reprinted with permission from ref. [42]. Copyright 2010

National Academy of Sciences.).

Fig. 4. SAW-based microfluidic devices. (a) A TSAW-based microfluidic tool detect the rodent

malaria parasite in blood (Reprinted with permission from ref. [65]. Copyright 2012 National

Academy of Sciences.). (b) Device structure and working mechanism of the acoustic tweezers

(Reprinted with permission from ref. [68].Copyright 2012 National Academy of Sciences.). (c)

Schematic illustration and image of the high-throughput ta-SSAW device for cancer cell

separation (Reprinted with permission from ref. [73]. Copyright 2015 National Academy of

Sciences.).

Fig. 5. Magnetic-based microfluidic technologies. (a) Photograph of the chip that was used for

continuous DNA extraction using phase-transfer magnetophoresis (Reprinted with permission

from ref. [80]. Copyright 2010 The Royal Society of Chemistry.). (b) Conceptual image of a

lab-on-a-chip droplet-based magnetofluidic platform which is composed of several functional

blocks (Reprinted with permission from ref. [83]. Copyright 2015 The Royal Society of

Chemistry.). (c) Schematic illustration of the process of magneto-optical biomolecular detection

by rotating super paramagnetic bead chains in solution (Reprinted with permission from ref. [84].

Copyright 2010 American Chemical Society.).

Fig. 6. Schematic diagram of the SIL-chip-ESI-MS system (Reprinted with permission from ref.

[104]. Copyright 2012 American Chemical Society.).

Fig. 7. Biomimetics on microfluidic chips. (a) Biologically inspired design of a human breathing

lung-on-a-chip microdevice (Reprinted with permission from ref. [19]. Copyright 2010 by the

43

Page 43 of 55
American Association for the Advancement of Science.). (b) Schematic diagram of a 3D

brain-on-a-chip with an interstitial level of flow (Reprinted with permission from ref. [127].

Copyright 2015 The Royal Society of Chemistry.). (c) Schematic cross-sectional view of a

microvasculature model (Reprinted with permission from ref. [128]. Copyright 2012 National

Academy of Sciences.).

Fig. 8. Drug discovery on microfluidic devices. (a) The microfluidic screening system (Reprinted

with permission from ref. [133]. Copyright 2012 National Academy of Sciences.). (b) Schematic

of the microfluidic device and cell co-culture process (Reprinted with permission from ref. [143].

Copyright 2015 Elsevier B.V.).

Fig. 9. Genetic analysis on microfluidic chip. (a) Design and operation of the microfluidic device

for single-cell gene expression analysis (Reprinted with permission from ref. [153]. Copyright

2011 National Academy of Sciences.). (b) The immunomagnetic exosomal RNA (iMER) platform

(Reprinted with permission from ref. [157]. Copyright 2015 Macmillan Publishers Limited.).

Fig. 10. Scheme depicting a cross-sectional view of the design of the chemomechanically

modulated biomolecule catch-and-release system (Reprinted with permission from ref. [166].

Copyright 2015 Macmillan Publishers Limited.).

Fig. 11. Cell analysis on microfluidic chips. (a) Microfluidic device for immune cell pairing

(Reprinted with permission from ref. [200]. Copyright 2015 Macmillan Publishers Limited.). (b)

Measuring attraction, repulsion, kinesis and inhibition during leukocyte responses to chemical

gradients (Reprinted with permission from ref. [201]. Copyright 2014 Macmillan Publishers

Limited.). (c) Iso-osmotic perfusion microfluidic cell culture array (Reprinted with permission

from ref. [211]. Copyright 2011 Nature America, Inc.). (d) Schematic of PDMS porous

membrane-assembled microfluidic feeder-separated co-culture system (Reprinted with permission

from ref. [213]. Copyright 2013 Nature Publishing Group, a division of Macmillan Publishers

Limited.).

44

Page 44 of 55
Fig. 1.

45

Page 45 of 55
Fig. 2.

46

Page 46 of 55
Fig. 3.

47

Page 47 of 55
Fig. 4.

48

Page 48 of 55
Fig. 5.

49

Page 49 of 55
Fig. 6.

50

Page 50 of 55
Fig. 7.

51

Page 51 of 55
Fig. 8.

52

Page 52 of 55
Fig. 9.

53

Page 53 of 55
Fig. 10.

54

Page 54 of 55
Fig. 11.

55

Page 55 of 55

Potrebbero piacerti anche