Sensors 15 29848
Sensors 15 29848
Sensors 15 29848
Abstract: Recent advances in biomedical technologies are mostly related to the convergence of
biology with microengineering. For instance, microfluidic devices are now commonly found in
most research centers, clinics and hospitals, contributing to more accurate studies and therapies
as powerful tools for drug delivery, monitoring of specific analytes, and medical diagnostics. Most
remarkably, integration of cellularized constructs within microengineered platforms has enabled the
recapitulation of the physiological and pathological conditions of complex tissues and organs. The
so-called “organ-on-a-chip” technology, which represents a new avenue in the field of advanced
in vitro models, with the potential to revolutionize current approaches to drug screening and
toxicology studies. This review aims to highlight recent advances of microfluidic-based devices
towards a body-on-a-chip concept, exploring their technology and broad applications in the
biomedical field.
1. Introduction
Twenty years after the first definition of tissue engineering (TE) came out [1], tissue engineers
are now facing new challenges concerning the standardization of the production protocols, cost
reduction and up-scaling of these standardized procedures to the clinical setting. Most remarkably,
knowledge deriving from tissue engineering is finding increasing application in the development of
micro-engineered models of human tissues and organs, which are being investigated as potential
alternatives to animal models in elucidating the biological mechanisms underlying morphogenetic
and pathogenetic processes, as well as drug screening platforms [2–4]. In this scenario, tissue
engineering adds the third dimension (3D) to in vitro cell cultures, better mimicking the complexity
of native tissues and giving access to full-human models.
Major advances in this field are related to the integration of tissue engineering with
microelectronics, microfabrication and microfluidics. Electronic devices have been employed as
integrative systems for tissue engineering research. Biosensors, initially dedicated to the detection of
biomolecules such as proteins [5,6], peptides [7,8], enzymes [9,10] and DNA [11,12], are now proposed
in the tissue engineering field as tools to monitor cell behavior on a miniaturized scale, with high
sensitivity and resolution and low associated costs [13–15]. By detecting cellular analytes, electrical
activity, physical and chemical signals transmitted by the cells, biosensors can provide insights into
cellular activities and responses in real time. Therefore microfluidic-based biosensors—also known as
lab-on-a-chip (LOC) and Biological/Biomedical Micro Electro Mechanical Systems (BioMEMS)—are
becoming more and more popular.
Microfluidic-based biosensors consist of devices in which the manipulation and analysis of fluids
occur within micrometer-sized channels [16,17]. Thanks to this miniaturization, the applications
of microfluidic devices are countless. To date, microfluidics has been successfully in monitoring
and controlling diagnostics [18], cell manipulation [19,20] and drug delivery [21]. Furthermore, the
most advanced microfluidic devices not only allow for the monitoring of signals but also actively
respond and adapt to them. Here, we highlight and summarize current cutting-edge research on
microfluidic devices, their application at a 3D level in tissue engineering and recent developments
towards body-on-a-chip concept.
31143
Sensors 2015, 15, 31142–31170
microfluidic cartridges for DNA purification and genotyping, by using standard laboratory
instruments as integrative systems [50].
New microfluidic platforms appear every day as a toolbox for the development of new solutions,
either to solve benchwork issues or to meet biomedical needs. The complexity and characteristics of
the systems obviously depend on the application-specific requirements, varying from very simple
devices to engineered complex platforms.
Paper-based microfluidics systems are the simplest technology for point-of-care diagnostics,
combining the well-known methods of lateral flow tests with paper microfluidic technology, where a
thin sheet of porous material is the substrate for the bioassays, taking advantage of the substrate high
internal surface area, capillary action and absorptive capacity [51]. Notably, the dramatic reduction
of costs brought in by paper-based microfluidics holds promise to bring point-of-care diagnostics to
developing countries [52].
Microfluidics finds an application in the standardization of cell culture protocols and in
the setup of reliable and sensitive bio-sensing assay protocols. Indeed, cells need optimal
physiological conditions (pH, temperature and CO2 ) to ensure their viability and activity, they must
be continuously and uniformly perfused with nutrients and oxygen and precautions are needed to
avoid biofouling effects [53] and side reactions due to non-specific adsorption of biomolecules [54].
Microscale fluid regulators as valves, pumps, mixers and other functional elements allow cell
perfusion with fresh media and assay reagents. Furthermore, automated liquid handling, electronic
control of switches and valves, multiplexing capability and appropriate detectors to monitor cellular
stimuli make a high-throughput screening format feasible. The wide range of different laboratory
activities, which already benefit from advanced systems in microfluidics-dependent cell assays, is
reviewed elsewhere [55,56].
31144
Sensors 2015, 15, 31142–31170
Table 1. Cont.
31145
Sensors 2015, 15, 31142–31170
Table 1. Cont.
31146
Sensors 2015, 15, 31142–31170
31147
Sensors 2015, 15, 31142–31170
31148
Sensors 2015, 15, 31142–31170
Sensors 2015, 15 10
Figure 1. Illustration of the diverse microfluidic devices used to study biological processes
Figure 1. Illustration of the diverse microfluidic devices used to study biological processes
occurring in vascular, respiratory, nervous, digestive and excretory systems. A. Biochip with
occurring in vascular,
subdividing respiratory,
interconnecting nervous,(array
microchannels digestive andthat
of pillars) excretory
decreasesystems.
in size toA. Biochip
mimic cell with
flow and adhesion in microvasculature to study of vaso-occlusive processes. B. Human breathing
subdividing interconnecting microchannels (array of pillars) that decrease in size to mimic
lung-on-a-chip microdevice, a biomimetic microsystem that reconstitutes the alveolar-capillary
cell flow and ofadhesion
interface the lungs.inThe
microvasculature to study ofchambers
device uses compartmentalized vaso-occlusive
to form anprocesses. B. Human
alveolar-capillary
barrierlung-on-a-chip
breathing on a porous membrane and produces
microdevice, cyclic stretching
a biomimetic of such membrane
microsystem by vacuum
that reconstitutes the
actuation. C. Two-compartment microfluidic culture system bridged by microchannels. It allows the
alveolar-capillary interface of the lungs. The device uses compartmentalized chambers to
visualization of cell interactions in co-culture, namely as a model for synaptic connectivity between
form an mixedalveolar-capillary
hippocampal co-culturesbarrier on a microgrooves
in which porous membraneallow bothand axons produces cyclic
and dendrites stretching
to enter and of
form synapses. D. Vertical cross-section representing the on-chip generation of intestinal villi obtained
such membrane by vacuum actuation. C. Two-compartment microfluidic culture system
by villus morphogenesis of Caco-2 cells. The up-scale of this system leads to the production of
bridged by microchannels.
gut-on-a-chip It allows
platforms to study the visualization
pharmacokinetics of cell interactions
and diffusion processes. E. Artificial liverin co-culture,
sinusoid
namely withasa microfluidic
a model for synaptic connectivity
endothelial-like between
barrier for primary hepatocyte mixed
culturehippocampal
to study diffusiveco-cultures
nutrient in
transport in liver-mediated metabolism. This unit consists of a cord of hepatocytes fed by diffusion
whichofmicrogrooves allow both axons and dendrites to enter and form synapses. D. Vertical
nutrients across the narrow microfluidic channels from a convective transport vessel. F. Kidney
cross-section representing the
proximal tubule-on-a-chip. on-chip generation
The microfluidic device consists ofof intestinal villi separated
an apical channel obtainedfrom bya villus
bottom reservoir by a porous membrane upon which primary human proximal
morphogenesis of Caco-2 cells. The up-scale of this system leads to the production of gut- tubule epithelial cells
are cultured in the presence of apical fluid shear stress. This design mimics the dynamically active
on-a-chip platforms
mechanical to study pharmacokinetics
microenvironment and diffusion
of the living kidney proximal tubule and processes. E. Artificial
allows the study of active liver
sinusoidand with
passivea epithelial
microfluidic endothelial-like barrier for primary hepatocyte culture to study
transport.
31150
Sensors 2015, 15, 31142–31170
to the gut-on-a-chip, it was possible to induce Caco-2 cells to spontaneously undergo intestinal
villi morphogenesis [128] (Figure 1D). This model was recently upscaled by the development
of a platform that can be adaptable to produce several functional units of other organs [152].
Further improvements in the recapitulation of the “intestinal epithelium-on-a-chip” consist in
the fabrication of 3D-shaped microporous polymeric membranes mimicking the geometry of the
intestinal villi [129] or by the so called “intestinal epithelium-on-a-chip” being reproduced by using
a novel hydrogel microfabrication technique and showing a superior structural maturation [130].
Their microfluidic device was further used to study the kinetics of diffusion processes in the 3D villi
scaffold. A much higher degree of complexity was reached by Ramadan and collaborators with the
microfluidic platform called NutriChip. With the aim to analyze the passage of nutrients through the
gastrointestinal tract (GIT), they developed a miniaturized GIT including the epithelial and immune
cell components, in which the response of immune cells to pro and anti-inflammatory stimuli was
monitored [130].
The liver also plays an important role in the digestion processes, being responsible for
the filtration of nutrients and digestion products. Furthermore, liver represents a fundamental
key in the metabolism of xenobiotics and thus a number of publications came out in the last
years, describing strategies to recreate liver-specific functions through microengineered models,
with the purpose of studying drug metabolism and ultimately improving drug development
processes [118–120,134,135]. Moreover, numerous publications described the use of microfluidics
for the development of physiologically-relevant hepatocyte cell culture [110,111,115,117] (Figure 1E),
differentiation [122] and co-culture systems [117,132,144,193] as well as the design of platforms
for diagnostic applications [146]. Additionally, the development of microfluidic-based devices to
investigate liver drug metabolism and toxicity [131,133,137,153,154,159,194] are to be considered
fundamental tools to address liver pathologies, better understand molecular toxicity mechanisms
and simulate drug-drug and organ-organ interactions. A comprehensive review about this topic is
given by van Midwoud and colleagues [195].
Pancreas also plays an important role in the digestion process, as it is responsible for
producing enzymes and hormones to be secreted into the small intestine. Deficient production of
digestive enzymes and hormones, as well pancreas blockage by tumors and gallstones, leads to
subsequent malfunction of the entire digestive system and further compliances. Trying to solve
these life-threatening conditions, researchers have made use of microfluidics to study and diagnose
pancreatic cancer [125–127,148], culture pancreatic islets [112–114,138], monitor stimulus-secretion
factors [139,140,143] and promote tissue-specific cell differentiation [124].
The perfect example of how microfluidics can be successfully applied to treat pancreatic
dysfunctions comes from the “bionic pancreas” developed for type 1 diabetes, that uses continuous
glucose monitoring along with subcutaneous delivery of both rapid-acting insulin and glucagon to
lower/increase blood glucose levels [196].
Due to the important functions on processing digestion products, water balance and blood
pressure regulation, kidneys are of fundamental importance for whole-body homeostasis. Therefore,
there is a considerable interest to develop strategies to adequately treat the most problematic
conditions affecting kidneys. Among them, chronic kidney disease often results in end-stage renal
failure, requiring renal replacement therapy and eventually transplantation, causing a massive
burden on the healthcare systems. Microfluidic systems, as the one developed by Leonard and
collaborators, appear as innovative tools to improve the outcome of classical approaches [150,151].
For instance, a membraneless dialysis strategy was developed, opening possibilities to create
wearable blood processing devices [150,151]. Other microfluidic systems enable the culture of kidney
cells in tubular structures, mimicking the organ structure and function [109,121,123,160] (Figure 1F).
The potential of application of microfluidic also includes disease modeling and metabolism studies,
giving insights about kidney cell toxicity and renal clearance [141,155,156,160]. Renal excretion and
metabolism are the actual subjects of preclinical safety studies, with the goal of investigating drug
31151
Sensors 2015, 15, 31142–31170
31152
Sensors 2015, 15, 31142–31170
BoC devices consist of microfluidic chips into which several modules can be installed holding
different cell types or engineered human organs [201]. Samples are interconnected in a hierarchic
and physiologically relevant fashion, thus allowing the functional modeling and monitoring of the
circulatory, endocrine, digestive, immune, lymphatic, nervous, respiratory and urinary systems, as
an advanced human in vitro model (Figure 2).
Since BoC models mimic physiological context and key aspects of human metabolism, they
allow for:
‚ high accuracy prediction and comprehensive analyses of novel therapeutic candidates during
preclinical stages, by a closer estimation of efficacy and dose response;
‚ reduction and likely replacement of animals in preclinical drug development, thereby reducing
costs and time to market;
‚ creation of a drug development tool that helps modern medicine rapidly respond to fast-moving
pandemics or chemical warfare/bioterrorism attacks;
‚ study cell signaling by monitoring the metabolites that are consumed, produced, and exchanged
between all tissues at physiologically relevant concentrations in real time;
study
Sensors
‚ 2015,embryology
15 and its signaling pathways by following intercellular signals and/or 15
bioelectrical messages;
‚ conduct
conductexperiments
experimentsthat
thatcannot
cannotbebeperformed
performedinincell
cellculture,
culture,e.g.,
e.g. study
study of
of tissue-tissue
tissue-tissue
interactions that occur as a result of metabolite travelling from one tissue to other distant tissue,
interactions that occur as a result of metabolite travelling from one tissue to other distant tissue,
and through dynamic forces that resemble blood circulation;
and through dynamic forces that resemble blood circulation;
‚ efficient and reliable cell–cell and cell-drug/biomaterial interaction studies, narrowing the gap
between
efficient and reliable cell–cell and cell-drug/biomaterial interaction studies, narrowing the gap
in vivo and in vitro conditions.
between in vivo and in vitro conditions.
Microfluidics can bring more benefits if complemented with sensitive analytical methods (namely
mass spectroscopy and sensors), enabling the31153 metabolic profiling and comprehensive molecular
characterization of the chip-based cell systems. Furthermore, as the chip channels are usually
transparent, it is also possible to monitor cell response and perform cell-tracking through time lapse live-
Sensors 2015, 15, 31142–31170
Microfluidics can bring more benefits if complemented with sensitive analytical methods
(namely mass spectroscopy and sensors), enabling the metabolic profiling and comprehensive
molecular characterization of the chip-based cell systems. Furthermore, as the chip channels are
usually transparent, it is also possible to monitor cell response and perform cell-tracking through
time lapse live-cell imaging.
It is important to recognize that there are two complementary approaches for BoC development.
Bottom-up approaches start from a detailed specification of each organ, and then proceed with
the design of coupled systems (e.g., heart–lung and intestine–liver), adding organs to create more
complex models. Top–down approaches, on the contrary, consider the abstract, system-level
architecture of an organism and then break the system down into the functionality of compositional
organ systems.
Moreover, it is also possible to explore the process of inflammation response by adding cytokines
or living immune cells to the system [202,203]. Also, BoC devices with biopsy samples or cells from
individual patients can be very helpful in the development of individualized medicine to predict how
the patient might react to a certain pharmacological treatment, prior to administration, thus reducing
risks [204].
31154
Sensors 2015, 15, 31142–31170
Organs /
Device / Platform Name Application References
Interactions
Brain, Heart, Lung, ADME profiling and
Physiologically-based
Skin, Adipose, quantification of the amount of
pharmacokinetic [212,213]
Muscle, Liver, Bone drugs in different parts of
(PBPK) model
Marrow, Kidney the body
Evaluating nanoparticle toxicity
Gastrointestinal µCCA [200]
and interactions with tissues
Tract and Liver
Investigate paracetamol intestinal
Gut—parallel tube model [206]
and liver first pass metabolism
Heart, Liver,
HeLiVa Drug testing in human health [205]
Vascular System
and disease
Intestine, Liver,
Four-Organ-Chip ADME profiling and [207]
Skin and Kidney
toxicity testing
Liver, Colorectal 96-well format-based
Testing drug effects at different [209–211]
Tissues microfluidic platform
concentrations in several tissues
Liver, Heart, Lung
ATHENA (“Homo Minutus”) Screening new drugs for potency [214]
and Kidney
and potential side-effects
Pharmacokinetic-pharmacodynamic Testing drug toxicity and
Liver, Tumor and
(PK-PD) model combined with improve insights into the drug’s [212]
Marrow
a µCCA mechanism of action
Lung, Gut PDMS-based organs-on-chip Prediction of clinical responses in [152,215]
humans
31155
Sensors 2015, 15, 31142–31170
collaborators developed a microfluidic size-based sorting platform, with the advantage of capturing
tumor cells without taking into account the expression of specific cell surface markers [234].
With a different aim, Ying and collaborators fabricated a 3D microfluidic chip generating
a concentration gradient of hepatocyte growth factor (HGF) to investigate its impact on Met/
PI3K/AKT activation, glucose regulatory protein expression and paclitaxel-induced A549 cell
apoptosis [235], as to mimic the in vivo secretion of the growth factor by cancer-associated fibroblasts.
Also, in order to modulate chemotaxis and electrotaxis of lung cancer cells, Kao employed
direct-current electric fields in a microfluidic cell culture device obtaining both stable electric field
and concentration gradients [77]. Recently, trying to be closer to a personalized medicine approach,
Ruppen and collaborators demonstrated the possibility to reproduce, at least partly, the barrier
induced by the tumor microenvironment to protect the tumor from drug exposure by testing the
chemosensitivity of patient lung cancer cell spheroids in a perfused microfluidic platform [236].
Phenomena such as tumor extravasation and metastatic site specificity have also been investigated
using 3D microfluidic models [237–239].
Several systems allow for the capture of CTCs and clusters from blood samples for further
detailed analysis of biomarkers by flow cytometry technology and multi-imaging [205,221–223].
Furthermore, the capture of these cells/clusters is suited for the identification of patients with
metastatic cancer and RNA sequencing of cancer cells can elucidate about the presence of cell
mutations [226] and identify the tumor origin.
31156
Sensors 2015, 15, 31142–31170
drug development process significantly, but also to improve the knowledge on tissue-tissue and/or
tissue-biomaterials interactions, diminishing the gap between in vivo and in vitro conditions in tissue
engineering applications and disease progression studies.
5. Concluding Remarks
The potential of microfluidics to fuel research applications and enter routine clinical practice is
indeed impressive. The use of microfluidic platforms as biomedical tools holds the promise to be
further implemented as clinically-relevant devices to be included in the daily healthcare practice,
by anticipating and monitoring the onset of diseases. Due to the interplay among microfluidics,
biosensors and tissue engineering know-how, diagnostics is becoming faster and cheaper, and
biomedical devices are getting more comprehensive and able to restore complex lost functions of
diseased or damaged tissues and organs.
In research, microfluidics is used as a complement to several methodologies and prototypes to
solve bench issues or to improve existing technologies. Several publications describe proof-of-concept
devices as innovative and smart alternatives for biomedical applications, although their FDA
approval, standardization and further manufacturing in large-scale is still far to come. FDA rules
are very restrictive in the direct use of microfluidics for tissue engineering applications; however
this technology can be easily adapted to host 3D microtissues and BoC devices, offering better
predictability of drug effects than conventional 2D test systems. These models enable a deep
understanding of interactions between drugs and their metabolites in various organs with regard to
toxic effects and/or drug efficacy. Despite the majority of the biomedical applications of microfluidics
being in vitro or ex vivo, in the near future the use of microfluidic devices will most likely be preferred
to in vivo studies, and upscaled to be suitable for the diagnostics and clinical scenarios, like in the
capture of circulating tumor cells and clusters.
Microfluidic technology is thus deemed to have a huge impact on science and medicine practice
due to its rapid progress, to its tunability and scalability, which leads to outline a trajectory of
tremendous innovation with countless potential. From simple to complex systems, microfluidics will
be evolving, being part of breakthrough and futurist ideas, thus playing a role in the improvement of
organ-on-a chip studies to body-on-a-chip approaches.
Acknowledgments: The present work was supported by the European Regional Development Fund-Project
FNUSA-ICRC (No. CZ.1.05/1.1.00/02.0123). A.C.P.Águas is a F.C.T. doctoral fellow (SFRH/BD/88958/2012).
This work was supported by Fundação para a Ciência e a Tecnologia (FCT), Portugal (UID/BIM/04773/2013).
A.R. is supported by the Internal Research Grant Program, Università Campus Bio-Medico di Roma. We
acknowledge Victorio Pozo Devoto, Rui Borges dos Santos and Carina Silva proof reading and scientific
discussion and Dr. Jorge Oliver de la Cruz for support in Adobe Illustrator software.
Conflicts of Interest: The authors declare no conflict of interest.
List of Abbreviations
2D—two-dimensional
3D—three-dimensional
3MD—Three-dimensional microfluidic microanalytical micro-organ device
ADME—absorption, distribution, metabolism, and excretion
BBB—blood-brain barrier
BioMEMS—biological/biomedical micro electro mechanical systems
BOC—body-on-a-chip
COPD—chronic obstructive pulmonary disease
CTCs—circulating tumor cells
ECM—extracellular matrix
GEDI—geometrically enhanced differential immunocapture
GIT—gastrointestinal tract
31157
Sensors 2015, 15, 31142–31170
References
1. Langer, R.; Vacanti, J.P. Tissue Engineering. Science 1993, 260, 920–926. [CrossRef] [PubMed]
2. Caplin, J.D.; Granados, N.G.; James, M.R.; Montazami, R.; Hashemi, N. Microfluidic organ-on-a-chip
technology for advancement of drug development and toxicology. Adv. Healthc. Mater. 2015, 4, 1426–1450.
[CrossRef] [PubMed]
3. Esch, E.W.; Bahinski, A.; Huh, D. Organs-on-chips at the frontiers of drug discovery. Nat. Rev. Drug Discov.
2015, 14, 248–260. [CrossRef] [PubMed]
4. Skommer, J.; Wlodkowic, D. Successes and future outlook for microfluidics-based cardiovascular drug
discovery. Exp. Opin. Drug Discov. 2015, 10, 231–244. [CrossRef] [PubMed]
5. Esteves-Villanueva, J.O.; Trzeciakiewicz, H.; Martic, S. A protein-based electrochemical biosensor for
detection of tau protein, a neurodegenerative disease biomarker. Analyst 2014, 139, 2823–2831. [CrossRef]
[PubMed]
6. Vistas, C.R.; Soares, S.S.; Rodrigues, R.M.M.; Chu, V.; Conde, J.P.; Ferreira, G.N.M. An amorphous silicon
photodiode microfluidic chip to detect nanomolar quantities of HIV-1 virion infectivity factor. Analyst 2014,
139, 3709–3713. [CrossRef] [PubMed]
7. Zhang, M.; Yin, B.-C.; Wang, X.-F.; Ye, B.-C. Interaction of peptides with graphene oxide and its application
for real-time monitoring of protease activity. Chem. Commun. 2011, 47, 2399–2401. [CrossRef] [PubMed]
8. Das, G.; Chirumamilla, M.; Toma, A.; Gopalakrishnan, A.; Zaccaria, R.P.; Alabastri, A.; Leoncini, M.;
Di Fabrizio, E. Plasmon based biosensor for distinguishing different peptides mutation states. Sci. Rep.
2013, 3, 1–6. [CrossRef] [PubMed]
9. Xu, J.J.; Chen, H.Y. Amperometric glucose sensor based on coimmobilization of glucose oxidase and
poly(p-phenylenediamine) at a platinum microdisk electrode. Anal. Biochem. 2000, 280, 221–226.
10. Mishra, G.K.; Sharma, A.; Deshpande, K.; Bhand, S. Flow injection analysis biosensor for urea analysis in
urine using enzyme thermistor. Appl. Biochem. Biotechnol. 2014, 174, 998–1009. [CrossRef] [PubMed]
11. Javanmard, M.; Davis, R.W. A microfluidic platform for electrical detection of DNA hybridization.
Sens. Actuators B Chem. 2011, 154, 22–27. [CrossRef] [PubMed]
12. De-Carvalho, J.; Rodrigues, R.M.M.; Tomé, B.; Henriques, S.F.; Mira, N.P.; Sá-Correia, I.; Ferreira, G.N.M.
Conformational and mechanical changes of DNA upon transcription factor binding detected by a QCM
and transmission line model. Analyst 2014, 139, 1847–1855. [CrossRef] [PubMed]
13. Nguyen, T.A.; Yin, T.-I.; Reyes, D.; Urban, G.A. A microfluidic chip with integrated electrical cell-impedance
sensing for monitoring single cancer cell migration in three-dimensional matrixes. Anal. Chem. 2013, 85,
11068–11076. [CrossRef] [PubMed]
31158
Sensors 2015, 15, 31142–31170
14. Weltin, A.; Slotwinski, K.; Kieninger, J.; Moser, I.; Jobst, G.; Wego, M.; Ehret, R.; Urban, G.A. Cell culture
monitoring for drug screening and cancer research: A transparent, microfluidic, multi-sensor microsystem.
Lab Chip 2014, 14, 138–146. [CrossRef] [PubMed]
15. Tian, B.; Liu, J.; Dvir, T.; Jin, L.; Tsui, J.H.; Qing, Q.; Suo, Z.; Langer, R.; Kohane, D.S.; Lieber, C.M.
Macroporous nanowire nanoelectronic scaffolds for synthetic tissues. Nat. Mater. 2012, 11, 986–994.
[CrossRef] [PubMed]
16. Manz, A.; Harrison, D.J.; Verpoorte, E.M.J.; Fettinger, J.C.; Paulus, A.; Lüdi, H.; Widmer, H.M.
Planar chips technology for miniaturization and integration of separation techniques into monitoring
systems—Capillary electrophoresis on a chip. J. Chromatogr. A 1992, 593, 253–258. [CrossRef]
17. Whitesides, G.M. The origins and the future of microfluidics. Nature 2006, 442, 368–373. [CrossRef]
[PubMed]
18. Madadi, H.; Casals-Terré, J.; Mohammadi, M. Self-driven filter-based blood plasma separator microfluidic
chip for point-of-care testing. Biofabrication 2015, 7, 025007:1–025007:11. [CrossRef] [PubMed]
19. Lee, L.M.; Liu, A.P. The application of micropipette aspiration in molecular mechanics of single cells.
J. Nanotechnol. Eng. Med. 2014, 5, 040801:1–040801:6. [CrossRef] [PubMed]
20. Giobbe, G.G.; Michielin, F.; Luni, C.; Giulitti, S.; Martewicz, S.; Dupont, S.; Floreani, A.; Elvassore, N.
Functional differentiation of human pluripotent stem cells on a chip. Nat. Methods 2015, 12, 637–640.
[CrossRef] [PubMed]
21. Vasiliauskas, R.; Liu, D.; Cito, S.; Zhang, H.; Shahbazi, M.-A.; Sikanen, T.; Mazutis, L.; Santos, H.A. Simple
microfluidic approach to fabricate monodisperse hollow microparticles for multidrug delivery. ACS Appl.
Mater. Interfaces 2015, 7, 14822–14832. [CrossRef] [PubMed]
22. Pullagurla, S.R.; Witek, M.A.; Jackson, J.M.; Lindell, M.A.M.; Hupert, M.L.; Nesterova, I.V.; Baird, A.E.;
Soper, S.A. Parallel affinity-based isolation of leukocyte subsets using microfluidics: Application for stroke
diagnosis. Anal. Chem. 2014, 86, 4058–4065. [CrossRef] [PubMed]
23. Ho, S.S.Y.; Chua, C.; Gole, L.; Biswas, A.; Koay, E.; Choolani, M. Same-day prenatal diagnosis of common
chromosomal aneuploidies using microfluidics-fluorescence in situ hybridization. Prenat. Diagn. 2012, 32,
321–328. [CrossRef] [PubMed]
24. De la Rica, R.; Stevens, M.M. Plasmonic ELISA for the detection of analytes at ultralow concentrations with
the naked eye. Nat. Protoc. 2013, 8, 1759–1764. [CrossRef] [PubMed]
25. Stern, E.; Vacic, A.; Rajan, N.K.; Criscione, J.M.; Park, J.; Ilic, B.R.; Mooney, D.J.; Reed, M.A.; Fahmy, T.M.
Label-free biomarker detection from whole blood. Nat. Nanotechnol. 2010, 5, 138–142. [CrossRef] [PubMed]
26. Andreou, C.; Hoonejani, M.R.; Barmi, M.R.; Moskovits, M.; Meinhart, C.D. Rapid detection of drugs
of abuse in saliva using surface enhanced raman spectroscopy and microfluidics. ACS Nano 2013, 7,
7157–7164. [CrossRef] [PubMed]
27. Zhu, K.Y.; Leung, K.W.; Ting, A.K.L.; Wong, Z.C.F.; Ng, W.Y.Y.; Choi, R.C.Y.; Dong, T.T.X.; Wang, T.;
Lau, D.T.W.; Tsim, K.W.K. Microfluidic chip based nano liquid chromatography coupled to tandem mass
spectrometry for the determination of abused drugs and metabolites in human hair. Anal. Bioanal. Chem.
2012, 402, 2805–2815. [CrossRef] [PubMed]
28. Swensen, J.S.; Xiao, Y.; Ferguson, B.S.; Lubin, A.A.; Lai, R.Y.; Heeger, A.J.; Plaxco, K.W.; Soh, H.T.
Continuous, real-time monitoring of cocaine in undiluted blood serum via a microfluidic, electrochemical
aptamer-based sensor. J. Am. Chem. Soc. 2009, 131, 4262–4266. [CrossRef] [PubMed]
29. Bell, S.C.; Hanes, R.D. A microfluidic device for presumptive testing of controlled substances. J. Forensic
Sci. 2007, 52, 884–888. [CrossRef] [PubMed]
30. Wang, J.; Sun, J.; Song, Y.; Xu, Y.; Pan, X.; Sun, Y.; Li, D. A label-free microfluidic biosensor for activity
detection of single microalgae cells based on chlorophyll fluorescence. Sensors 2013, 13, 16075–16089.
[CrossRef] [PubMed]
31. Buffi, N.; Merulla, D.; Beutier, J.; Barbaud, F.; Beggah, S.; van Linte, H.; Renaud, P.; van der Meer, J.R.
Miniaturized bacterial biosensor system for arsenic detection holds great promise for making integrated
measurement device. Bioeng. Bugs 2011, 2, 296–298. [CrossRef] [PubMed]
32. Duford, D.A.; Xi, Y.; Salin, E.D. Enzyme inhibition-based determination of pesticide residues in vegetable
and soil in centrifugal microfluidic devices. Anal. Chem. 2013, 85, 7834–7841. [CrossRef] [PubMed]
33. Foudeh, A.M.; Brassard, D.; Tabrizian, M.; Veres, T. Rapid and multiplex detection of Legionella’s RNA
using digital microfluidics. Lab Chip 2015, 15, 1609–1618. [CrossRef] [PubMed]
31159
Sensors 2015, 15, 31142–31170
34. Charles, P.T.; Adams, A.A.; Deschamps, J.R.; Veitch, S.; Hanson, A.; Kusterbeck, A.W. Detection of
explosives in a dynamic marine environment using a moored TNT immunosensor. Sensors 2014, 14,
4074–4085. [CrossRef] [PubMed]
35. Tan, H.Y.; Loke, W.K.; Tan, Y.T.; Nguyen, N.-T. A lab-on-a-chip for detection of nerve agent sarin in blood.
Lab Chip 2008, 8, 885–891. [CrossRef] [PubMed]
36. De Santis, R.; Ciammaruconi, A.; Faggioni, G.; Fillo, S.; Gentile, B.; Di Giannatale, E.; Ancora, M.; Lista, F.
High throughput MLVA-16 typing for Brucella based on the microfluidics technology. BMC Microbiol. 2011,
11, 1–9. [CrossRef] [PubMed]
37. Dulay, S.B.; Gransee, R.; Julich, S.; Tomaso, H.; O’Sullivan, C.K. Automated microfluidically controlled
electrochemical biosensor for the rapid and highly sensitive detection of Francisella tularensis.
Biosens. Bioelectron. 2014, 59, 342–349. [CrossRef] [PubMed]
38. Matatagui, D.; Fontecha, J.L.; Fernández, M.J.; Gràcia, I.; Cané, C.; Santos, J.P.; Horrillo, M.C. Love-wave
sensors combined with microfluidics for fast detection of biological warfare agents. Sensors 2014, 14,
12658–12669. [CrossRef] [PubMed]
39. Shapiro, M.S.; Haswell, S.J.; Lye, G.J.; Bracewell, D.G. Microfluidic chromatography for early stage
evaluation of biopharmaceutical binding and separation conditions. Sep. Sci. Technol. 2011, 46, 185–194.
[CrossRef]
40. Chen, D.L.; Ismagilov, R.F. Microfluidic cartridges preloaded with nanoliter plugs of reagents: An
alternative to 96-well plates for screening. Curr. Opin. Chem. Biol. 2006, 10, 226–231. [CrossRef] [PubMed]
41. Zhang, C.; Xing, D. Miniaturized PCR chips for nucleic acid amplification and analysis: Latest advances
and future trends. Nucleic Acids Res. 2007, 35, 4223–4237. [CrossRef] [PubMed]
42. Cao, Q.; Mahalanabis, M.; Chang, J.; Carey, B.; Hsieh, C.; Stanley, A.; Odell, C.A.; Mitchell, P.; Feldman, J.;
Pollock, N.R.; et al. Microfluidic chip for molecular amplification of influenza a RNA in human respiratory
specimens. PLoS ONE 2012, 7, e33176. [CrossRef] [PubMed]
43. Schell, W.A.; Benton, J.L.; Smith, P.B.; Poore, M.; Rouse, J.L.; Boles, D.J.; Johnson, M.D.; Alexander, B.D.;
Pamula, V.K.; Eckhardt, A.E.; et al. Evaluation of a digital microfluidic real-time PCR platform to detect
DNA of Candida albicans in blood. Eur. J. Clin. Microbiol. Infect. Dis. 2012, 31, 2237–2245. [CrossRef]
[PubMed]
44. Ishii, S.; Segawa, T.; Okabe, S. Simultaneous quantification of multiple food- and waterborne pathogens by
use of microfluidic quantitative PCR. Appl. Environ. Microbiol. 2013, 79, 2891–2898. [CrossRef] [PubMed]
45. Spurgeon, S.L.; Jones, R.C.; Ramakrishnan, R. High throughput gene expression measurement with real
time PCR in a microfluidic dynamic array. PLoS ONE 2008, 3, e1662. [CrossRef] [PubMed]
46. Chen, Y.; Zhong, J.F. Microfluidic devices for high-throughput gene expression profiling of single
hESC-derived neural stem cells. Methods Mol. Biol. 2008, 438, 293–303. [PubMed]
47. May-Panloup, P.; Ferre-L’Hotellier, V.; Moriniere, C.; Marcaillou, C.; Lemerle, S.; Malinge, M.-C.;
Coutolleau, A.; Lucas, N.; Reynier, P.; Descamps, P.; et al. Molecular characterization of corona radiata cells
from patients with diminished ovarian reserve using microarray and microfluidic-based gene expression
profiling. Hum. Reprod. 2012, 27, 829–843. [CrossRef] [PubMed]
48. Shaw, K.J.; Hughes, E.M.; Dyer, C.E.; Greenman, J.; Haswell, S.J. Integrated RNA extraction and RT-PCR
for semi-quantitative gene expression studies on a microfluidic device. Lab. Investig. 2013, 93, 961–966.
[CrossRef] [PubMed]
49. Mellors, J.S.; Gorbounov, V.; Ramsey, R.S.; Ramsey, J.M. Fully integrated glass microfluidic device for
performing high-efficiency capillary electrophoresis and electrospray ionization mass spectrometry. Anal.
Chem. 2008, 80, 6881–6887. [CrossRef] [PubMed]
50. Focke, M.; Mark, D.; Stumpf, F.; Müller, M.; Roth, G.; Zengerle, R.; von Stetten, F. Microfluidic cartridges
for DNA purification and genotyping processed in standard laboratory instruments. In Proceedings of
the SPIE, Smart Sensors, Actuators, and MEMS V, Prague, Czech Republic, 18–20 April 2011; Schmid, U.,
Sánchez-Rojas, J.L., Leester-Schaedel, M., Eds.; Volume 8066.
51. Martinez, A.W.; Phillips, S.T.; Butte, M.J.; Whitesides, G.M. Patterned paper as a platform for inexpensive,
low-volume, portable bioassays. Angew. Chem. Int. Ed. 2007, 46, 1318–1320. [CrossRef] [PubMed]
52. Martinez, A.W.; Phillips, S.T.; Whitesides, G.M.; Carrilho, E. Diagnostics for the developing world:
Microfluidic paper-based analytical devices. Anal. Chem. 2010, 82, 3–10. [CrossRef] [PubMed]
31160
Sensors 2015, 15, 31142–31170
53. Mukhopadhyay, R. When microfluidic devices go bad. Anal. Chem. 2005, 77, 429A–432A. [CrossRef]
[PubMed]
54. Zhou, J.; Khodakov, D.A.; Ellis, A.V.; Voelcker, N.H. Surface modification for PDMS-based microfluidic
devices. Electrophoresis 2012, 33, 89–104. [CrossRef] [PubMed]
55. Barbulovic-Nad, I.; Wheeler, A.R. Cell Assays in Microfluidics. In Encyclopedia of Microfluidics and
Nanofluidics; Springer: Berlin, Germany, 2008; pp. 209–216.
56. Xiong, B.; Ren, K.; Shu, Y.; Chen, Y.; Shen, B.; Wu, H. Recent developments in microfluidics for cell studies.
Adv. Mater. 2014, 26, 5525–5532. [CrossRef] [PubMed]
57. Borenstein, J.T.; Vunjak-Novakovic, G. Engineering tissue with BioMEMS. IEEE Pulse 2011, 2, 28–34.
[CrossRef] [PubMed]
58. Tourovskaia, A.; Fauver, M.; Kramer, G.; Simonson, S.; Neumann, T. Tissue-engineered microenvironment
systems for modeling human vasculature. Exp. Biol. Med. 2014, 239, 1264–1271. [CrossRef] [PubMed]
59. Theberge, A.B.; Yu, J.; Young, E.W.K.; Ricke, W.A.; Bushman, W.; Beebe, D.J. Microfluidic multiculture assay
to analyze biomolecular signaling in angiogenesis. Anal. Chem. 2015, 87, 3239–3246. [CrossRef] [PubMed]
60. Guo, Q.; Duffy, S.P.; Matthews, K.; Santoso, A.T.; Scott, M.D.; Ma, H. Microfluidic analysis of red blood cell
deformability. J. Biomech. 2014, 47, 1767–1776. [CrossRef] [PubMed]
61. Grosberg, A.; Alford, P.W.; McCain, M.L.; Parker, K.K. Ensembles of engineered cardiac tissues for
physiological and pharmacological study: Heart on a chip. Lab Chip 2011, 11, 4165–4173. [CrossRef]
[PubMed]
62. Yeom, E.; Kang, Y.J.; Lee, S. Changes in velocity profile according to blood viscosity in a microchannel.
Biomicrofluidics 2014, 8, 034110:1–034110:11. [CrossRef] [PubMed]
63. Yeom, E.; Jun Kang, Y.; Joon Lee, S. Hybrid system for ex vivo hemorheological and hemodynamic analysis:
A feasibility study. Sci. Rep. 2015, 5, 1–15. [CrossRef] [PubMed]
64. Tomaiuolo, G.; Lanotte, L.; D’Apolito, R.; Cassinese, A.; Guido, S. Microconfined flow behavior of red blood
cells. Med. Eng. Phys. 2015. [CrossRef] [PubMed]
65. Li, L.; Lv, X.; Ostrovidov, S.; Shi, X.; Zhang, N.; Liu, J. Biomimetic microfluidic device for in vitro
antihypertensive drug evaluation. Mol. Pharm. 2014, 11, 2009–2015. [CrossRef] [PubMed]
66. McCain, M.L.; Sheehy, S.P.; Grosberg, A.; Goss, J.A.; Parker, K.K. Recapitulating maladaptive, multiscale
remodeling of failing myocardium on a chip. Proc. Natl. Acad. Sci. USA 2013, 110, 9770–9775. [CrossRef]
[PubMed]
67. Giridharan, G.A.; Nguyen, M.-D.; Estrada, R.; Parichehreh, V.; Hamid, T.; Ismahil, M.A.; Prabhu, S.D.;
Sethu, P. Microfluidic cardiac cell culture model (µCCCM). Anal. Chem. 2010, 82, 7581–7587. [CrossRef]
[PubMed]
68. Yasotharan, S.; Pinto, S.; Sled, J.G.; Bolz, S.-S.; Günther, A. Artery-on-a-chip platform for automated,
multimodal assessment of cerebral blood vessel structure and function. Lab Chip 2015, 15, 2660–2669.
[CrossRef] [PubMed]
69. Ryu, H.; Oh, S.; Lee, H.J.; Lee, J.Y.; Lee, H.K.; Jeon, N.L. Engineering a blood vessel network module for
body-on-a-chip applications. J. Lab. Autom. 2015, 20, 296–301. [CrossRef] [PubMed]
70. Hattori, K.; Munehira, Y.; Kobayashi, H.; Satoh, T.; Sugiura, S.; Kanamori, T. Microfluidic perfusion culture
chip providing different strengths of shear stress for analysis of vascular endothelial function. J. Biosci.
Bioeng. 2014, 118, 327–332. [CrossRef] [PubMed]
71. Hald, E.S.; Steucke, K.E.; Reeves, J.A.; Win, Z.; Alford, P.W. Long-term vascular contractility assay using
genipin-modified muscular thin films. Biofabrication 2014, 6, 045005:1–045005:11. [CrossRef] [PubMed]
72. Dominical, V.M.; Vital, D.M.; O’Dowd, F.; Saad, S.T.O.; Costa, F.F.; Conran, N. In vitro microfluidic model
for the study of vaso-occlusive processes. Exp. Hematol. 2015, 43, 223–228. [CrossRef] [PubMed]
73. Harris, D.G.; Benipal, P.K.; Cheng, X.; Burdorf, L.; Azimzadeh, A.M.; Pierson, R.N. Four-dimensional
characterization of thrombosis in a live-cell, shear-flow assay: Development and application to
xenotransplantation. PLoS ONE 2015, 10, e0123015. [CrossRef] [PubMed]
74. Hu, R.; Li, F.; Lv, J.; He, Y.; Lu, D.; Yamada, T.; Ono, N. Microfluidic analysis of pressure drop and flow
behavior in hypertensive micro vessels. Biomed. Microdevices 2015, 17, 1387–2176. [CrossRef] [PubMed]
75. Huh, D.; Matthews, B.D.; Mammoto, A.; Montoya-Zavala, M.; Hsin, H.Y.; Ingber, D.E. Reconstituting
organ-level lung functions on a chip. Science 2010, 328, 1662–1668. [CrossRef] [PubMed]
31161
Sensors 2015, 15, 31142–31170
76. Bol, L.; Galas, J.-C.; Hillaireau, H.; Potier, I.L.; Nicolas, V.; Haghiri-Gosnet, A.-M.; Fattal, E.; Taverna, M.
A microdevice for parallelized pulmonary permeability studies. Biomed. Microdevices 2014, 16, 277–285.
[CrossRef] [PubMed]
77. Kao, Y.-C.; Hsieh, M.-H.; Liu, C.-C.; Pan, H.-J.; Liao, W.-Y.; Cheng, J.-Y.; Kuo, P.-L.; Lee, C.-H. Modulating
chemotaxis of lung cancer cells by using electric fields in a microfluidic device. Biomicrofluidics 2014, 8,
024107:1–024107:12. [CrossRef] [PubMed]
78. Sellgren, K.L.; Butala, E.J.; Gilmour, B.P.; Randell, S.H.; Grego, S. A biomimetic multicellular model of the
airways using primary human cells. Lab Chip 2014, 14, 3349–3358. [CrossRef] [PubMed]
79. Ling, T.-Y.; Liu, Y.-L.; Huang, Y.-K.; Gu, S.-Y.; Chen, H.-K.; Ho, C.-C.; Tsao, P.-N.; Tung, Y.-C.; Chen, H.-W.;
Cheng, C.-H.; et al. Differentiation of lung stem/progenitor cells into alveolar pneumocytes and induction
of angiogenesis within a 3D gelatin—Microbubble scaffold. Biomaterials 2014, 35, 5660–5669. [CrossRef]
[PubMed]
80. Punde, T.H.; Wu, W.-H.; Lien, P.-C.; Chang, Y.-L.; Kuo, P.-H.; Chang, M.D.-T.; Lee, K.-Y.; Huang, C.-D.;
Kuo, H.-P.; Chan, Y.-F.; et al. A biologically inspired lung-on-a-chip device for the study of protein-induced
lung inflammation. Integr. Biol. 2015, 7, 162–169. [CrossRef] [PubMed]
81. Wu, J.; Hillier, C.; Komenda, P.; Lobato de Faria, R.; Levin, D.; Zhang, M.; Lin, F. A microfluidic platform for
evaluating neutrophil chemotaxis induced by sputum from COPD patients. PLoS ONE 2015, 10, e0126523.
[CrossRef] [PubMed]
82. Cortez-Jugo, C.; Qi, A.; Rajapaksa, A.; Friend, J.R.; Yeo, L.Y. Pulmonary monoclonal antibody delivery via
a portable microfluidic nebulization platform. Biomicrofluidics 2015, 9, 1–10. [CrossRef] [PubMed]
83. Rochow, N.; Manan, A.; Wu, W.-I.; Fusch, G.; Monkman, S.; Leung, J.; Chan, E.; Nagpal, D.; Predescu, D.;
Brash, J.; et al. An integrated array of microfluidic oxygenators as a neonatal lung assist device: In vitro
characterization and in vivo demonstration. Artif. Organs 2014, 38, 856–866. [CrossRef] [PubMed]
84. Li, E.; Xu, Z.; Liu, F.; Wang, H.; Wen, J.; Shao, S.; Zhang, L.; Wang, L.; Liu, C.; Lu, J.; et al. Continual
exposure to cigarette smoke extracts induces tumor-Like transformation of human nontumor bronchial
epithelial cells in a microfluidic chip. J. Thorac. Oncol. 2014, 9, 1091–1100. [CrossRef] [PubMed]
85. Felder, M.; Stucki, A.O.; Stucki, J.D.; Geiser, T.; Guenat, O.T. The potential of microfluidic lung epithelial
wounding: Towards in vivo-like alveolar microinjuries. Integr. Biol. 2014, 6, 1132–1140. [CrossRef]
[PubMed]
86. Eleftheriadou, I.; Trabalza, A.; Ellison, S.M.; Gharun, K.; Mazarakis, N.D. Specific retrograde transduction
of spinal motor neurons using lentiviral vectors targeted to presynaptic NMJ receptors. Mol. Ther. 2014, 22,
1285–1298. [CrossRef] [PubMed]
87. Neumann, S.; Campbell, G.E.; Szpankowski, L.; Goldstein, L.S.B.; Encalada, S.E. Characterizing the
composition of molecular motors on moving axonal cargo using “cargo mapping” analysis. J. Vis. Exp.
2014, 1–2. [CrossRef] [PubMed]
88. Wang, T.; Martin, S.; Papadopulos, A.; Harper, C.B.; Mavlyutov, T.A.; Niranjan, D.; Glass, N.R.;
Cooper-White, J.J.; Sibarita, J.-B.; Choquet, D.; et al. Control of autophagosome axonal retrograde flux
by presynaptic activity unveiled using botulinum neurotoxin type A. J. Neurosci. 2015, 35, 6179–6194.
[CrossRef] [PubMed]
89. Zhao, X.; Zhou, Y.; Weissmiller, A.M.; Pearn, M.L.; Mobley, W.C.; Wu, C. Real-time imaging of axonal
transport of quantum dot-labeled BDNF in primary neurons. J. Vis. Exp. 2014, 91, e51899. [CrossRef]
[PubMed]
90. Robertson, G.; Bushell, T.J.; Zagnoni, M. Chemically induced synaptic activity between mixed primary
hippocampal co-cultures in a microfluidic system. Integr. Biol. 2014, 6, 636–644. [CrossRef] [PubMed]
91. Xu, H.; Ferreira, M.M.; Heilshorn, S.C. Small-molecule axon-polarization studies enabled by a shear-free
microfluidic gradient generator. Lab Chip 2014, 14, 2047–2056. [CrossRef] [PubMed]
92. An, Q.; Fillmore, H.L.; Vouri, M.; Pilkington, G.J. Brain tumor cell line authentication, an efficient alternative
to capillary electrophoresis by using a microfluidics-based system. Neuro. Oncol. 2014, 16, 265–273.
[CrossRef] [PubMed]
93. Pollen, A.A.; Nowakowski, T.J.; Shuga, J.; Wang, X.; Leyrat, A.A.; Lui, J.H.; Li, N.; Szpankowski, L.;
Fowler, B.; Chen, P.; et al. Low-coverage single-cell mRNA sequencing reveals cellular heterogeneity and
activated signaling pathways in developing cerebral cortex. Nat. Biotechnol. 2014, 32, 1053–1058. [CrossRef]
[PubMed]
31162
Sensors 2015, 15, 31142–31170
94. Kerman, B.E.; Kim, H.J.; Padmanabhan, K.; Mei, A.; Georges, S.; Joens, M.S.; Fitzpatrick, J.A.J.; Jappelli, R.;
Chandross, K.J.; August, P.; et al. In vitro myelin formation using embryonic stem cells. Development 2015,
142, 2213–2225. [CrossRef] [PubMed]
95. Nery, F.C.; da Hora, C.C.; Yaqub, U.; Zhang, X.; McCarthy, D.M.; Bhide, P.G.; Irimia, D.; Breakefield, X.O.
New methods for investigation of neuronal migration in embryonic brain explants. J. Neurosci. Methods
2015, 239, 80–84. [CrossRef] [PubMed]
96. Wu, K.-Y.; He, M.; Hou, Q.-Q.; Sheng, A.-L.; Yuan, L.; Liu, F.; Liu, W.-W.; Li, G.; Jiang, X.-Y.; Luo, Z.-G.
Semaphorin 3A activates the guanosine triphosphatase Rab5 to promote growth cone collapse and organize
callosal axon projections. Sci. Signal. 2014, 7, 1–13. [CrossRef] [PubMed]
97. Rajbhandari, L.; Tegenge, M.A.; Shrestha, S.; Ganesh Kumar, N.; Malik, A.; Mithal, A.; Hosmane, S.;
Venkatesan, A. Toll-like receptor 4 deficiency impairs microglial phagocytosis of degenerating axons. Glia
2014, 62, 1982–1991. [CrossRef] [PubMed]
98. Fournier, A.J.; Rajbhandari, L.; Shrestha, S.; Venkatesan, A.; Ramesh, K.T. In vitro and in situ visualization
of cytoskeletal deformation under load: Traumatic axonal injury. FASEB J. 2014, 28, 5277–5287. [CrossRef]
[PubMed]
99. Brown, J.A.; Sherrod, S.D.; Goodwin, C.R.; Brewer, B.; Yang, L.; Garbett, K.A.; Li, D.; McLean, J.A.;
Wikswo, J.P.; Mirnics, K. Metabolic consequences of interleukin-6 challenge in developing neurons and
astroglia. J. Neuroinflamm. 2014, 11, 1–12. [CrossRef] [PubMed]
100. Sun, M.; Kaplan, S.V.; Gehringer, R.C.; Limbocker, R.A.; Johnson, M.A. Localized drug application and
sub-second voltammetric dopamine release measurements in a brain slice perfusion device. Anal. Chem.
2014, 86, 4151–4156. [CrossRef] [PubMed]
101. Lin, X.; Wang, S.; Yu, X.; Liu, Z.; Wang, F.; Li, W.T.; Cheng, S.H.; Dai, Q.; Shi, P. High-throughput mapping
of brain-wide activity in awake and drug-responsive vertebrates. Lab Chip 2015, 15, 680–689. [CrossRef]
[PubMed]
102. Ruiz, A.; Joshi, P.; Mastrangelo, R.; Francolini, M.; Verderio, C.; Matteoli, M. Testing Aβ toxicity on primary
CNS cultures using drug-screening microfluidic chips. Lab Chip 2014, 14, 2860–2866. [CrossRef] [PubMed]
103. Booth, R.; Kim, H. Permeability analysis of neuroactive drugs through a dynamic microfluidic in vitro
blood–brain barrier model. Ann. Biomed. Eng. 2014, 42, 2379–2391. [CrossRef] [PubMed]
104. Zhao, Y.; Abdelfattah, A.S.; Zhao, Y.; Ruangkittisakul, A.; Ballanyi, K.; Campbell, R.E.; Harrison, D.J.
Microfluidic cell sorter-aided directed evolution of a protein-based calcium ion indicator with an inverted
fluorescent response. Integr. Biol. 2014, 6, 714–725. [CrossRef] [PubMed]
105. Coquinco, A.; Kojic, L.; Wen, W.; Wang, Y.T.; Jeon, N.L.; Milnerwood, A.J.; Cynader, M. A microfluidic
based in vitro model of synaptic competition. Mol. Cell. Neurosci. 2014, 60, 43–52. [CrossRef] [PubMed]
106. Deleglise, B.; Magnifico, S.; Duplus, E.; Vaur, P.; Soubeyre, V.; Belle, M.; Vignes, M.; Viovy, J.-L.; Jacotot, E.;
Peyrin, J.-M.; et al. B-amyloid induces a dying-back process and remote trans-synaptic alterations in a
microfluidic-based reconstructed neuronal network. Acta Neuropathol. Commun. 2014, 2, 1–9. [CrossRef]
[PubMed]
107. Zhu, L.; Xu, M.; Yang, M.; Yang, Y.; Li, Y.; Deng, J.; Ruan, L.; Liu, J.; Du, S.; Liu, X.; et al. An ALS-mutant
TDP-43 neurotoxic peptide adopts an anti-parallel -structure and induces TDP-43 redistribution. Hum. Mol.
Genet. 2014, 23, 6863–6877. [CrossRef] [PubMed]
108. Chang, T.C.; Mikheev, A.M.; Huynh, W.; Monnat, R.J.; Rostomily, R.C.; Folch, A. Parallel microfluidic
chemosensitivity testing on individual slice cultures. Lab Chip 2014, 14, 4540–4551. [CrossRef] [PubMed]
109. Mu, X.; Zheng, W.; Xiao, L.; Zhang, W.; Jiang, X. Engineering a 3D vascular network in hydrogel for
mimicking a nephron. Lab Chip 2013, 13, 1612–1618. [CrossRef] [PubMed]
110. Baudoin, R.; Alberto, G.; Legendre, A.; Paullier, P.; Naudot, M.; Fleury, M.J.; Jacques, S.; Griscom, L.;
Leclerc, E. Investigation of expression and activity levels of primary rat hepatocyte detoxication genes
under various flow rates and cell densities in microfluidic biochips. Biotechnol. Prog. 2014, 30, 401–410.
[CrossRef] [PubMed]
111. Lee, P.J.; Hung, P.J.; Lee, L.P. An artificial liver sinusoid with a microfluidic endothelial-like barrier for
primary hepatocyte culture. Biotechnol. Bioeng. 2007, 97, 1340–1346. [CrossRef] [PubMed]
112. Silva, P.N.; Green, B.J.; Altamentova, S.M.; Rocheleau, J.V. A microfluidic device designed to induce media
flow throughout pancreatic islets while limiting shear-induced damage. Lab Chip 2013, 13, 4374–4384.
[CrossRef] [PubMed]
31163
Sensors 2015, 15, 31142–31170
113. Jun, Y.; Kim, M.J.; Hwang, Y.H.; Jeon, E.A.; Kang, A.R.; Lee, S.-H.; Lee, D.Y. Microfluidics-generated
pancreatic islet microfibers for enhanced immunoprotection. Biomaterials 2013, 34, 8122–8130. [CrossRef]
[PubMed]
114. Lee, D.; Wang, Y.; Mendoza-Elias, J.E.; Adewola, A.F.; Harvat, T.A.; Kinzer, K.; Gutierrez, D.; Qi, M.;
Eddington, D.T.; Oberholzer, J. Dual microfluidic perifusion networks for concurrent islet perifusion and
optical imaging. Biomed. Microdevices 2012, 14, 7–16. [CrossRef] [PubMed]
115. Zhang, M.Y.; Lee, P.J.; Hung, P.J.; Johnson, T.; Lee, L.P.; Mofrad, M.R.K. Microfluidic environment for high
density hepatocyte culture. Biomed. Microdevices 2008, 10, 117–121. [CrossRef] [PubMed]
116. Baudoin, R.; Griscom, L.; Prot, J.M.; Legallais, C.; Leclerc, E. Behavior of HepG2/C3A cell cultures in a
microfluidic bioreactor. Biochem. Eng. J. 2011, 53, 172–181. [CrossRef]
117. Illa, X.; Vila, S.; Yeste, J.; Peralta, C.; Gracia-Sancho, J.; Villa, R. A novel modular bioreactor to in vitro study
the hepatic sinusoid. PLoS ONE 2014, 9, e111864. [CrossRef] [PubMed]
118. Li, C.Y.; Stevens, K.R.; Schwartz, R.E.; Alejandro, B.S.; Huang, J.H.; Bhatia, S.N. Micropatterned cell-cell
interactions enable functional encapsulation of primary hepatocytes in hydrogel microtissues. Tissue Eng.
Part A 2014, 20, 2200–2212. [CrossRef] [PubMed]
119. Khetani, S.R.; Bhatia, S.N. Microscale culture of human liver cells for drug development. Nat. Biotechnol.
2008, 26, 120–126. [CrossRef] [PubMed]
120. Domansky, K.; Inman, W.; Serdy, J.; Dash, A.; Lim, M.H.M.; Griffith, L.G. Perfused multiwell plate for 3D
liver tissue engineering. Lab Chip 2010, 10, 51–58. [CrossRef] [PubMed]
121. Jang, K.-J.; Suh, K.-Y. A multi-layer microfluidic device for efficient culture and analysis of renal tubular
cells. Lab Chip 2010, 10, 36–42. [CrossRef] [PubMed]
122. Ju, X.; Li, D.; Gao, N.; Shi, Q.; Hou, H. Hepatogenic differentiation of mesenchymal stem cells using
microfluidic chips. Biotechnol. J. 2008, 3, 383–391. [CrossRef] [PubMed]
123. Huang, H.-C.; Chang, Y.-J.; Chen, W.-C.; Harn, H.I.-C.; Tang, M.-J.; Wu, C.-C. Enhancement of renal
epithelial cell functions through microfluidic-based coculture with adipose-derived stem cells. Tissue Eng.
Part A 2013, 19, 2024–2034. [CrossRef] [PubMed]
124. Ghazalli, N.; Mahdavi, A.; Feng, T.; Jin, L.; Kozlowski, M.T.; Hsu, J.; Riggs, A.D.; Tirrell, D.A.; Ku, H.T.
Postnatal pancreas of mice contains tripotent progenitors capable of giving rise to duct, acinar, and
endocrine cells in vitro. Stem Cells Dev. 2015. [CrossRef] [PubMed]
125. Sheng, W.; Ogunwobi, O.O.; Chen, T.; Zhang, J.; George, T.J.; Liu, C.; Fan, Z.H. Capture, release and culture
of circulating tumor cells from pancreatic cancer patients using an enhanced mixing chip. Lab Chip 2014,
14, 89–98. [CrossRef] [PubMed]
126. Thege, F.I.; Lannin, T.B.; Saha, T.N.; Tsai, S.; Kochman, M.L.; Hollingsworth, M.A.; Rhim, A.D.; Kirby, B.J.
Microfluidic immunocapture of circulating pancreatic cells using parallel EpCAM and MUC1 capture:
Characterization, optimization and downstream analysis. Lab Chip 2014, 14, 1775–1784. [CrossRef]
[PubMed]
127. Rhim, A.D.; Thege, F.I.; Santana, S.M.; Lannin, T.B.; Saha, T.N.; Tsai, S.; Maggs, L.R.; Kochman, M.L.;
Ginsberg, G.G.; Lieb, J.G.; et al. Detection of circulating pancreas epithelial cells in patients with pancreatic
cystic lesions. Gastroenterology 2014, 146, 647–651. [CrossRef] [PubMed]
128. Kim, H.J.; Ingber, D.E. Gut-on-a-chip microenvironment induces human intestinal cells to undergo villus
differentiation. Integr. Biol. 2013, 5, 1130–1140. [CrossRef] [PubMed]
129. Esch, M.B.; Sung, J.H.; Yang, J.; Yu, C.; Yu, J.; March, J.C.; Shuler, M.L. On chip porous polymer
membranes for integration of gastrointestinal tract epithelium with microfluidic “body-on-a-chip” devices.
Biomed. Microdevices 2012, 14, 895–906. [CrossRef] [PubMed]
130. Kim, S.H.; Lee, J.W.; Choi, I.; Kim, Y.C.; Lee, J.B.; Sung, J.H. A microfluidic device with 3D hydrogel villi
scaffold to simulate intestinal absorption. J. Nanosci. Nanotechnol. 2013, 13, 7220–7228. [CrossRef] [PubMed]
131. Legendre, A.; Baudoin, R.; Alberto, G.; Paullier, P.; Naudot, M.; Bricks, T.; Brocheton, J.; Jacques, S.;
Cotton, J.; Leclerc, E. Metabolic characterization of primary rat hepatocytes cultivated in parallel
microfluidic biochips. J. Pharm. Sci. 2013, 102, 3264–3276. [CrossRef] [PubMed]
132. Kimura, H.; Yamamoto, T.; Sakai, H.; Sakai, Y.; Fujii, T. An integrated microfluidic system for long-term
perfusion culture and on-line monitoring of intestinal tissue models. Lab Chip 2008, 8, 741–746. [CrossRef]
[PubMed]
31164
Sensors 2015, 15, 31142–31170
133. Pasirayi, G.; Scott, S.M.; Islam, M.; O’Hare, L.; Bateson, S.; Ali, Z. Low cost microfluidic cell culture array
using normally closed valves for cytotoxicity assay. Talanta 2014, 129, 491–498. [CrossRef] [PubMed]
134. Chang, R.; Emami, K.; Wu, H.; Sun, W. Biofabrication of a three-dimensional liver micro-organ as an in vitro
drug metabolism model. Biofabrication 2010, 2, 1–11. [CrossRef] [PubMed]
135. Chang, R.C.; Emami, K.; Jeevarajan, A.; Wu, H.; Sun, W. Microprinting of liver micro-organ for drug
metabolism study. Methods Mol. Biol. 2011, 671, 219–238. [PubMed]
136. Ramadan, Q.; Jafarpoorchekab, H.; Huang, C.; Silacci, P.; Carrara, S.; Koklü, G.; Ghaye, J.; Ramsden, J.;
Ruffert, C.; Vergeres, G.; et al. NutriChip: Nutrition analysis meets microfluidics. Lab Chip 2013, 13, 196–203.
[CrossRef] [PubMed]
137. Snouber, L.C.; Bunescu, A.; Naudot, M.; Legallais, C.; Brochot, C.; Dumas, M.E.; Elena-Herrmann, B.;
Leclerc, E. Metabolomics-on-a-chip of hepatotoxicity induced by anticancer drug flutamide and its active
metabolite hydroxyflutamide using HepG2/C3a microfluidic biochips. Toxicol. Sci. 2013, 132, 8–20.
[CrossRef] [PubMed]
138. Dhumpa, R.; Truong, T.M.; Wang, X.; Roper, M.G. Measurement of the entrainment window of islets of
Langerhans by microfluidic delivery of a chirped glucose waveform. Integr. Biol. 2015, 7, 1061–1067.
[CrossRef] [PubMed]
139. Lomasney, A.R.; Yi, L.; Roper, M.G. Simultaneous monitoring of insulin and islet amyloid polypeptide
secretion from islets of langerhans on a microfluidic device. Anal. Chem. 2013, 85, 7919–7925. [CrossRef]
[PubMed]
140. Lo, J.F.; Wang, Y.; Blake, A.; Yu, G.; Harvat, T.A.; Jeon, H.; Oberholzer, J.; Eddington, D.T. Islet
preconditioning via multimodal microfluidic modulation of intermittent hypoxia. Anal. Chem. 2012, 84,
1987–1993. [CrossRef] [PubMed]
141. Ferrell, N.; Ricci, K.B.; Groszek, J.; Marmerstein, J.T.; Fissell, W.H. Albumin handling by renal tubular
epithelial cells in a microfluidic bioreactor. Biotechnol. Bioeng. 2012, 109, 797–803. [CrossRef] [PubMed]
142. McAuliffe, G.J.; Chang, J.Y.; Glahn, R.P.; Shuler, M.L. Development of a gastrointestinal tract microscale cell
culture analog to predict drug transport. Mol. Celullar Biomech. 2008, 5, 119–132.
143. Nourmohammadzadeh, M.; Lo, J.F.; Bochenek, M.; Mendoza-Elias, J.E.; Wang, Q.; Li, Z.; Zeng, L.; Qi, M.;
Eddington, D.T.; Oberholzer, J.; et al. Microfluidic array with integrated oxygenation control for real-time
live-cell imaging: Effect of hypoxia on physiology of microencapsulated pancreatic islets. Anal. Chem. 2013,
85, 11240–11249. [CrossRef] [PubMed]
144. Bricks, T.; Paullier, P.; Legendre, A.; Fleury, M.J.; Zeller, P.; Merlier, F.; Anton, P.M.; Leclerc, E. Development
of a new microfluidic platform integrating co-cultures of intestinal and liver cell lines. Toxicol. Vitr. 2014,
28, 885–895. [CrossRef] [PubMed]
145. Kimura, H.; Ikeda, T.; Nakayama, H.; Sakai, Y.; Fujii, T. An on-chip small intestine-liver model for
pharmacokinetic studies. J. Lab. Autom. 2015, 20, 265–273. [CrossRef] [PubMed]
146. Matharu, Z.; Patel, D.; Gao, Y.; Haque, A.; Zhou, Q.; Revzin, A. Detecting transforming growth factor-β
release from liver cells using an aptasensor integrated with microfluidics. Anal. Chem. 2014, 86, 8865–8872.
[CrossRef] [PubMed]
147. Zilberman, Y.; Sonkusale, S.R. Microfluidic optoelectronic sensor for salivary diagnostics of stomach cancer.
Biosens. Bioelectron. 2015, 67, 465–471. [CrossRef] [PubMed]
148. Das, R.; Murphy, R.G.; Seibel, E.J. Beyond isolated cells: Microfluidic transport of large tissue for pancreatic
cancer diagnosis. Proc. SPIE 2015, 9320, 1–29.
149. Leonard, E.F. Technical approaches toward ambulatory ESRD therapy. Seminars Dial. 2009, 22, 658–660.
[CrossRef] [PubMed]
150. Leonard, E.F.; Cortell, S.; Jones, J. The path to wearable ultrafiltration and dialysis devices. Blood Purification
2011, 31, 92–95. [CrossRef] [PubMed]
151. Kim, H.J.; Huh, D.; Hamilton, G.; Ingber, D.E. Human gut-on-a-chip inhabited by microbial flora that
experiences intestinal peristalsis-like motions and flow. Lab Chip 2012, 12, 2165–2174. [CrossRef] [PubMed]
152. Huh, D.; Kim, H.J.; Fraser, J.P.; Shea, D.E.; Khan, M.; Bahinski, A.; Hamilton, G.A.; Ingber, D.E.
Microfabrication of human organs-on-chips. Nat. Protoc. 2013, 8, 2135–2157. [CrossRef] [PubMed]
31165
Sensors 2015, 15, 31142–31170
153. Legendre, A.; Jacques, S.; Dumont, F.; Cotton, J.; Paullier, P.; Fleury, M.J.; Leclerc, E. Investigation of
the hepatotoxicity of flutamide: Pro-survival/apoptotic and necrotic switch in primary rat hepatocytes
characterized by metabolic and transcriptomic profiles in microfluidic liver biochips. Toxicol. Vitr. 2014, 28,
1075–1087. [CrossRef] [PubMed]
154. Leclerc, E.; Hamon, J.; Claude, I.; Jellali, R.; Naudot, M.; Bois, F. Investigation of acetaminophen toxicity in
HepG2/C3a microscale cultures using a system biology model of glutathione depletion. Cell Biol. Toxicol.
2015, 31, 173–185. [CrossRef] [PubMed]
155. Snouber, L.C.; Jacques, S.; Monge, M.; Legallais, C.; Leclerc, E. Transcriptomic analysis of the effect of
ifosfamide on MDCK cells cultivated in microfluidic biochips. Genomics 2012, 100, 27–34. [CrossRef]
[PubMed]
156. Snouber, L.C.; Letourneur, F.; Chafey, P.; Broussard, C.; Monge, M.; Legallais, C.; Leclerc, E. Analysis of
transcriptomic and proteomic profiles demonstrates improved Madin-Darby canine kidney cell function in
a renal microfluidic biochip. Biotechnol. Prog. 2011, 28, 474–484. [CrossRef] [PubMed]
157. Snouber, L.C.; Aninat, C.; Grsicom, L.; Madalinski, G.; Brochot, C.; Poleni, P.E.; Razan, F.; Guillouzo, C.G.;
Legallais, C.; Corlu, A.; et al. Investigation of ifosfamide nephrotoxicity induced in a liver-kidney co-culture
biochip. Biotechnol. Bioeng. 2013, 110, 597–608. [CrossRef] [PubMed]
158. Mahler, G.J.; Esch, M.B.; Glahn, R.P.; Shuler, M.L. Characterization of a gastrointestinal tract microscale cell
culture analog used to predict drug toxicity. Biotechnol. Bioeng. 2009, 104, 193–205. [CrossRef] [PubMed]
159. Leclerc, E.; Hamon, J.; Legendre, A.; Bois, F.Y. Integration of pharmacokinetic and NRF2 system biology
models to describe reactive oxygen species production and subsequent glutathione depletion in liver
microfluidic biochips after flutamide exposure. Toxicol. Vitr. 2014, 28, 1230–1241. [CrossRef] [PubMed]
160. Jang, K.-J.; Mehr, A.P.; Hamilton, G.A.; McPartlin, L.A.; Chung, S.; Suh, K.-Y.; Ingber, D.E. Human
kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr. Biol. 2013,
5, 1119–1129. [CrossRef] [PubMed]
161. Lang, J.D.; Berry, S.M.; Powers, G.L.; Beebe, D.J.; Alarid, E.T. Hormonally responsive breast cancer cells in
a microfluidic co-culture model as a sensor of microenvironmental activity. Integr. Biol. 2013, 5, 807–816.
[CrossRef] [PubMed]
162. Kim, B.J.; Hannanta-anan, P.; Chau, M.; Kim, Y.S.; Swartz, M.A.; Wu, M. Cooperative roles of SDF-1α and
EGF gradients on tumor cell migration revealed by a robust 3D microfluidic model. PLoS ONE 2013, 8,
e68422. [CrossRef] [PubMed]
163. Huang, H.-Y.; Wu, T.-L.; Huang, H.-R.; Li, C.-J.; Fu, H.-T.; Soong, Y.-K.; Lee, M.-Y.; Yao, D.-J. Isolation of
motile spermatozoa with a microfluidic chip having a surface-modified microchannel. J. Lab. Autom. 2014,
19, 91–99. [CrossRef] [PubMed]
164. Tung, C.; Hu, L.; Fiore, A.G.; Ardon, F.; Hickman, D.G.; Gilbert, R.O.; Suarez, S.S.; Wu, M. Microgrooves
and fluid flows provide preferential passageways for sperm over pathogen Tritrichomonas foetus. Proc.
Natl. Acad. Sci. USA 2015, 112, 5431–5436. [CrossRef] [PubMed]
165. Ferrie, A.M.; Wang, C.; Deng, H.; Fang, Y. A label-free optical biosensor with microfluidics identifies an
intracellular signalling wave mediated through the β2-adrenergic receptor. Integr. Biol. 2013, 5, 1253–1261.
[CrossRef] [PubMed]
166. Ges, I.A.; Brindley, R.L.; Currie, K.P.M.; Baudenbacher, F.J. A microfluidic platform for chemical stimulation
and real time analysis of catecholamine secretion from neuroendocrine cells. Lab Chip 2013, 13, 4663–4673.
[CrossRef] [PubMed]
167. Pires, N.M.; Dong, T. Detection of stress hormones by a microfluidic-integrated polycarbazole/fullerene
photodetector. In Proceedings of the 35th Annual International Conference of the IEEE Engineering in
Medicine and Biology Society (EMBC), Osaka, Japan, 3–7 July 2013.
168. Broccardo, C.J.; Schauer, K.L.; Kohrt, W.M.; Schwartz, R.S.; Murphy, J.P.; Prenni, J.E. Multiplexed analysis
of steroid hormones in human serum using novel microflow tile technology and LC-MS/MS. J. Chromatogr.
B Anal. Technol. Biomed. Life Sci. 2013, 934, 16–21. [CrossRef] [PubMed]
169. Kim, J.; Abdulwahab, S.; Choi, K.; Lafrenière, N.M.; Mudrik, J.M.; Gomaa, H.; Ahmado, H.; Behan, L.-A.;
Casper, R.F.; Wheeler, A.R. A microfluidic technique for quantification of steroids in core needle biopsies.
Anal. Chem. 2015, 87, 4688–4695. [CrossRef] [PubMed]
31166
Sensors 2015, 15, 31142–31170
170. Kaushik, A.; Yndart, A.; Jayant, R.D.; Sagar, V.; Atluri, V.; Bhansali, S.; Nair, M. Electrochemical sensing
method for point-of-care cortisol detection in human immunodeficiency virus-infected patients. Int. J.
Nanomedicine 2015, 10, 677–685. [PubMed]
171. Selimovic, A.; Erkal, J.L.; Spence, D.M.; Martin, R.S. Microfluidic device with tunable post arrays and
integrated electrodes for studying cellular release. Analyst 2014, 139, 5686–5694. [CrossRef] [PubMed]
172. Shamsi, M.H.; Choi, K.; Ng, A.H.C.; Wheeler, A.R. A digital microfluidic electrochemical immunoassay.
Lab Chip 2014, 14, 547–554. [CrossRef] [PubMed]
173. Arends, F.; Sellner, S.; Seifert, P.; Gerland, U.; Rehberg, M.; Lieleg, O. A microfluidics approach to study the
accumulation of molecules at basal lamina interfaces. Lab Chip 2015, 15, 3326–3334. [CrossRef] [PubMed]
174. Zhu, B.; Smith, J.; Yarmush, M.L.; Nahmias, Y.; Kirby, B.J.; Murthy, S.K. Microfluidic enrichment of mouse
epidermal stem cells and validation of stem cell proliferation in vitro. Tissue Eng. Part C Methods 2013, 19,
765–773. [CrossRef] [PubMed]
175. Jean, L.; Yang, L.; Majumdar, D.; Gao, Y.; Shi, M.; Brewer, B.M.; Li, D.; Webb, D.J. The Rho family GEF Asef2
regulates cell migration in three dimensional (3D) collagen matrices through myosin II. Cell Adh. Migr.
2014, 8, 460–467. [CrossRef] [PubMed]
176. Liu, G.; Smith, K.; Kaya, T. Implementation of a microfluidic conductivity sensor—A potential sweat
electrolyte sensing system for dehydration detection. Conf. Proc. IEEE Eng. Med. Biol. Soc. 2014. [CrossRef]
177. Rose, D.P.; Ratterman, M.E.; Griffin, D.K.; Hou, L.; Kelley-Loughnane, N.; Naik, R.R.; Hagen, J.A.;
Papautsky, I.; Heikenfeld, J. Adhesive RFID sensor patch for monitoring of sweat electrolytes. IEEE Trans.
Biomed. Eng. 2015, 62, 1457–1465. [CrossRef] [PubMed]
178. Li, Y.; Wang, S.; Huang, R.; Huang, Z.; Hu, B.; Zheng, W.; Yang, G.; Jiang, X. Evaluation of the effect of the
structure of bacterial cellulose on full thickness skin wound repair on a microfluidic chip. Biomacromolecules
2015, 16, 780–789. [CrossRef] [PubMed]
179. Lo, J.F.; Brennan, M.; Merchant, Z.; Chen, L.; Guo, S.; Eddington, D.T.; DiPietro, L.A. Microfluidic wound
bandage: Localized oxygen modulation of collagen maturation. Wound Repair Regen. 2013, 21, 226–234.
[CrossRef] [PubMed]
180. Berthier, E.; Young, E.W.K.; Beebe, D. Engineers are from PDMS-land, biologists are from Polystyrenia.
Lab Chip 2012, 12, 1224–1237. [CrossRef] [PubMed]
181. Zheng, W.; Jiang, B.; Wang, D.; Zhang, W.; Wang, Z.; Jiang, X. A microfluidic flow-stretch chip for
investigating blood vessel biomechanics. Lab Chip 2012, 12, 3441–3450. [CrossRef] [PubMed]
182. Zheng, W.; Jiang, B.; Hao, Y.; Zhao, Y.; Zhang, W.; Jiang, X. Screening reactive oxygen species scavenging
properties of platinum nanoparticles on a microfluidic chip. Biofabrication 2014, 6, 045004:1–045004:11.
[CrossRef] [PubMed]
183. Nalayanda, D.D.; Wang, Q.; Fulton, W.B.; Wang, T.-H.; Abdullah, F. Engineering an artificial
alveolar-capillary membrane: A novel continuously perfused model within microchannels. J. Pediatr. Surg.
2010, 45, 45–51. [CrossRef] [PubMed]
184. Nalayanda, D.D.; Puleo, C.M.; Fulton, W.B.; Wang, T.-H.; Abdullah, F. Characterization of pulmonary cell
growth parameters in a continuous perfusion microfluidic environment. Exp. Lung Res. 2007, 33, 321–335.
[CrossRef] [PubMed]
185. Nichols, J.E.; Niles, J.A.; Vega, S.P.; Argueta, L.B.; Eastaway, A.; Cortiella, J. Modeling the lung: Design and
development of tissue engineered macro- and micro-physiologic lung models for research use. Exp. Biol.
Med. 2014, 239, 1135–1169. [CrossRef] [PubMed]
186. Zeng, L.; Qiu, L.; Yang, X.; Zhou, Y.; Du, J.; Wang, H.; Sun, J.; Yang, C.; Jiang, J. Isolation of lung multipotent
stem cells using a novel microfluidic magnetic activated cell sorting system. Cell Biol. Int. 2015. [CrossRef]
[PubMed]
187. Hoganson, D.M.; Pryor, H.I., II; Bassett, E.K.; Spool, I.D.; Vacanti, J.P. Lung assist device technology with
physiologic blood flow developed on a tissue engineered scaffold platform. Lab Chip 2011, 11, 700–707.
[CrossRef] [PubMed]
188. Kniazeva, T.; Hsiao, J.C.; Charest, J.L.; Borenstein, J.T. A microfluidic respiratory assist device with high gas
permeance for artificial lung applications. Biomed. Microdevices 2011, 13, 315–323. [CrossRef] [PubMed]
189. Kniazeva, T.; Epshteyn, A.A.; Hsiao, J.C.; Kim, E.S.; Kolachalama, V.B.; Charest, J.L.; Borenstein, J.T.
Performance and scaling effects in a multilayer microfluidic extracorporeal lung oxygenation device.
Lab Chip 2012, 12, 1686–1695. [CrossRef] [PubMed]
31167
Sensors 2015, 15, 31142–31170
190. Kovach, K.M.; LaBarbera, M.A.; Moyer, M.C.; Cmolik, B.L.; van Lunteren, E.; Sen Gupta, A.; Capadona, J.R.;
Potkay, J.A. In vitro evaluation and in vivo demonstration of a biomimetic, hemocompatible, microfluidic
artificial lung. Lab Chip 2015, 15, 1366–1375. [CrossRef] [PubMed]
191. Taylor, A.M.; Blurton-Jones, M.; Rhee, S.W.; Cribbs, D.H.; Cotman, C.W.; Jeon, N.L. A microfluidic culture
platform for CNS axonal injury, regeneration and transport. Nat. Methods 2005, 2, 599–605. [CrossRef]
[PubMed]
192. Booth, R.; Kim, H. Characterization of a microfluidic in vitro model of the blood-brain barrier (µBBB).
Lab Chip 2012, 12, 1784–1792. [CrossRef] [PubMed]
193. Shintu, L.; Baudoin, R.; Navratil, V.; Prot, J.-M.; Pontoizeau, C.; Defernez, M.; Blaise, B.J.; Domange, C.;
Péry, A.R.; Toulhoat, P.; et al. Metabolomics-on-a-chip and predictive systems toxicology in microfluidic
bioartificial organs. Anal. Chem. 2012, 84, 1840–1848. [CrossRef] [PubMed]
194. Baudoin, R.; Legendre, A.; Jacques, S.; Cotton, J.; Bois, F.; Leclerc, E. Evaluation of a liver microfluidic
biochip to predict in vivo clearances of seven drugs in rats. J. Pharm. Sci. 2014, 103, 706–718. [CrossRef]
[PubMed]
195. Van Midwoud, P.M.; Verpoorte, E.; Groothuis, G.M.M. Microfluidic devices for in vitro studies on liver drug
metabolism and toxicity. Integr. Biol. 2011, 3, 509–521. [CrossRef] [PubMed]
196. Russell, S.J.; El-Khatib, F.H.; Sinha, M.; Magyar, K.L.; McKeon, K.; Goergen, L.G.; Balliro, C.; Hillard, M.A.;
Nathan, D.M.; Damiano, E.R. Outpatient glycemic control with a bionic pancreas in type 1 diabetes. N. Engl.
J. Med. 2014, 371, 313–325. [CrossRef] [PubMed]
197. Xu, S.; Zhang, Y.; Jia, L.; Mathewson, K.E.; Jang, K.-I.; Kim, J.; Fu, H.; Huang, X.; Chava, P.; Wang, R.; et al.
Soft microfluidic assemblies of sensors, circuits, and radios for the skin. Science 2014, 344, 70–74. [CrossRef]
[PubMed]
198. Sonner, Z.; Wilder, E.; Heikenfeld, J.; Kasting, G.; Beyette, F.; Swaile, D.; Sherman, F.; Joyce, J.; Hagen, J.;
Kelley-Loughnane, N.; et al. The microfluidics of the eccrine sweat gland, including biomarker partitioning,
transport, and biosensing implications. Biomicrofluidics 2015. [CrossRef] [PubMed]
199. Scannell, J.W.; Blanckley, A.; Boldon, H.; Warrington, B. Diagnosing the decline in pharmaceutical R&D
efficiency. Nat. Rev. Drug Discov. 2012, 11, 191–200. [PubMed]
200. Esch, M.B.; Mahler, G.J.; Stokol, T.; Shuler, M.L. Body-on-a-chip simulation with gastrointestinal tract and
liver tissues suggests that ingested nanoparticles have the potential to cause liver injury. Lab Chip 2014, 14,
3081–3092. [CrossRef] [PubMed]
201. Shuler, M.L. Modeling life. Ann. Biomed. Eng. 2012, 40, 1399–1407. [CrossRef] [PubMed]
202. Akagi, T.; Kato, K.; Kobayashi, M.; Kosaka, N.; Ochiya, T.; Ichiki, T. On-Chip Immunoelectrophoresis of
Extracellular Vesicles Released from Human Breast Cancer Cells. PLoS ONE 2015, 10, e0123603. [CrossRef]
[PubMed]
203. Huang, N.-T.; Chen, W.; Oh, B.-R.; Cornell, T.T.; Shanley, T.P.; Fu, J.; Kurabayashi, K. An integrated
microfluidic platform for in situ cellular cytokine secretion immunophenotyping. Lab Chip 2012, 12,
4093–4101. [CrossRef] [PubMed]
204. Esch, M.B.; King, T.L.; Shuler, M.L. The role of body-on-a-chip devices in drug and toxicity studies.
Annu. Rev. Biomed. Eng. 2011, 13, 55–72. [CrossRef] [PubMed]
205. Vunjak-Novakovic, G.; Bhatia, S.; Chen, C.; Hirschi, K. HeLiVa platform: Integrated heart-liver-vascular
systems for drug testing in human health and disease. Stem Cell Res. Ther. 2013. [CrossRef] [PubMed]
206. Prot, J.M.; Maciel, L.; Bricks, T.; Merlier, F.; Cotton, J.; Paullier, P.; Bois, F.Y.; Leclerc, E. First pass intestinal
and liver metabolism of paracetamol in a microfluidic platform coupled with a mathematical modeling as
a means of evaluating ADME processes in humans. Biotechnol. Bioeng. 2014, 111, 2027–2040. [CrossRef]
[PubMed]
207. Maschmeyer, I.; Lorenz, A.K.; Schimek, K.; Hasenberg, T.; Ramme, A.P.; Hübner, J.; Lindner, M.; Drewell, C.;
Bauer, S.; Thomas, A.; et al. A four-organ-chip for interconnected long-term co-culture of human intestine,
liver, skin and kidney equivalents. Lab Chip 2015, 15, 2688–2699. [CrossRef] [PubMed]
208. Maschmeyer, I.; Hasenberg, T.; Jaenicke, A.; Lindner, M.; Lorenz, A.K.; Zech, J.; Garbe, L.-A.; Sonntag, F.;
Hayden, P.; Ayehunie, S.; et al. Chip-based human liver–intestine and liver–skin co-cultures—A first step
toward systemic repeated dose substance testing in vitro. Eur. J. Pharm. Biopharm. 2015. [CrossRef]
[PubMed]
31168
Sensors 2015, 15, 31142–31170
209. Frey, O.; Misun, P.M.; Fluri, D.A.; Hengstler, J.G.; Hierlemann, A. Reconfigurable microfluidic hanging drop
network for multi-tissue interaction and analysis. Nat. Commun. 2014, 5, 1–11.
210. Kim, J.-Y.; Fluri, D.A.; Kelm, J.M.; Hierlemann, A.; Frey, O. 96-Well format-based microfluidic platform for
parallel interconnection of multiple multicellular spheroids. J. Lab. Autom. 2015, 20, 274–282. [CrossRef]
[PubMed]
211. Kim, J.-Y.; Fluri, D.A.; Marchan, R.; Boonen, K.; Mohanty, S.; Singh, P.; Hammad, S.; Landuyt, B.;
Hengstler, J.G.; Kelm, J.M.; et al. 3D spherical microtissues and microfluidic technology for multi-tissue
experiments and analysis. J. Biotechnol. 2015, 205, 24–35. [CrossRef] [PubMed]
212. Sung, J.H.; Kam, C.; Shuler, M.L. A microfluidic device for a pharmacokinetic–pharmacodynamic (PK–PD)
model on a chip. Lab Chip 2010, 10, 446–455. [CrossRef] [PubMed]
213. Sung, J.H.; Srinivasan, B.; Esch, M.B.; McLamb, W.T.; Bernabini, C.; Shuler, M.L.; Hickman, J.J. Using
physiologically-based pharmacokinetic-guided “body-on-a-chip” systems to predict mammalian response
to drug and chemical exposure. Exp. Biol. Med. 2014, 239, 1225–1239. [CrossRef] [PubMed]
214. Wikswo, J.P.; Curtis, E.L.; Eagleton, Z.E.; Evans, B.C.; Kole, A.; Hofmeister, L.H.; Matloff, W.J. Scaling
and systems biology for integrating multiple organs-on-a-chip. Lab Chip 2013, 13, 3496–3511. [CrossRef]
[PubMed]
215. Huh, D.; Hamilton, G.A.; Ingber, D.E. From 3D cell culture to organs-on-chips. Trends Cell Biol. 2011, 21,
745–754. [CrossRef] [PubMed]
216. Dance, A. Correction for Dance, News Feature: Building benchtop human models. Proc. Natl. Acad.
Sci. USA 2015, 112, 6773–6775. [CrossRef] [PubMed]
217. Chen, Y.-C.; Allen, S.G.; Ingram, P.N.; Buckanovich, R.; Merajver, S.D.; Yoon, E. Single-cell migration chip
for chemotaxis-based microfluidic selection of heterogeneous cell populations. Sci. Rep. 2015, 5, 1–13.
[CrossRef] [PubMed]
218. Lee, H.; Park, W.; Ryu, H.; Jeon, N.L. A microfluidic platform for quantitative analysis of cancer
angiogenesis and intravasationa. Biomicrofluidics 2014. [CrossRef] [PubMed]
219. Mattei, F.; Schiavoni, G.; De Ninno, A.; Lucarini, V.; Sestili, P.; Sistigu, A.; Fragale, A.; Sanchez, M.;
Spada, M.; Gerardino, A.; et al. A multidisciplinary study using in vivo tumor models and microfluidic
cell-on-chip approach to explore the cross-talk between cancer and immune cells. J. Immunotoxicol. 2014,
11, 337–346. [CrossRef] [PubMed]
220. Zhang, Y.; Zhou, L.; Qin, L. High-throughput 3D cell invasion chip enables accurate cancer metastatic
assays. J. Am. Chem. Soc. 2014, 136, 15257–15262. [CrossRef] [PubMed]
221. Zou, H.; Yue, W.; Yu, W.-K.; Liu, D.; Fong, C.-C.; Zhao, J.; Yang, M. Microfluidic platform for studying
chemotaxis of adhesive cells revealed a gradient-dependent migration and acceleration of cancer stem cells.
Anal. Chem. 2015, 87, 7098–7108. [CrossRef] [PubMed]
222. Riahi, R.; Yang, Y.L.; Kim, H.; Jiang, L.; Wong, P.K.; Zohar, Y. A microfluidic model for organ-specific
extravasation of circulating tumor cells. Biomicrofluidics 2014, 8, 024103. [CrossRef] [PubMed]
223. Wang, X.-Y.; Pei, Y.; Xie, M.; Jin, Z.-H.; Xiao, Y.-S.; Wang, Y.; Zhang, L.-N.; Li, Y.; Huang, W.-H. An artificial
blood vessel implanted three-dimensional microsystem for modeling transvascular migration of tumor
cells. Lab Chip 2015, 15, 1178–1187. [CrossRef] [PubMed]
224. Casavant, B.P.; Strotman, L.N.; Tokar, J.J.; Thiede, S.M.; Traynor, A.M.; Ferguson, J.S.; Lang, J.M.; Beebe, D.
Paired diagnostic and pharmacodynamic analysis of rare non-small cell lung cancer cells enabled by the
VerIFAST platform. Lab Chip 2014, 14, 99–105. [CrossRef] [PubMed]
225. Park, J.-M.; Kim, M.S.; Moon, H.-S.; Yoo, C.E.; Park, D.; Kim, Y.J.; Han, K.-Y.; Lee, J.-Y.; Oh, J.H.; Kim, S.S.;
et al. Fully automated circulating tumor cell isolation platform with large-volume capacity based on
lab-on-a-disc. Anal. Chem. 2014, 86, 3735–3742. [CrossRef] [PubMed]
226. Watanabe, M.; Serizawa, M.; Sawada, T.; Takeda, K.; Takahashi, T.; Yamamoto, N.; Koizumi, F.; Koh, Y. A
novel flow cytometry-based cell capture platform for the detection, capture and molecular characterization
of rare tumor cells in blood. J. Transl. Med. 2014, 12, 1–12. [CrossRef] [PubMed]
227. Yu, I.F.; Yu, Y.H.; Chen, L.Y.; Fan, S.K.; Chou, H.Y.E.; Yang, J.T. A portable microfluidic device for the rapid
diagnosis of cancer metastatic potential which is programmable for temperature and CO2 . Lab Chip 2014,
14, 3621–3628. [CrossRef] [PubMed]
31169
Sensors 2015, 15, 31142–31170
228. Galletti, G.; Sung, M.S.; Vahdat, L.T.; Shah, M.A.; Santana, S.M.; Altavilla, G.; Kirby, B.J.; Giannakakou, P.
Isolation of breast cancer and gastric cancer circulating tumor cells by use of an anti HER2-based
microfluidic device. Lab Chip 2014, 14, 147–156. [CrossRef] [PubMed]
229. Sarioglu, A.F.; Aceto, N.; Kojic, N.; Donaldson, M.C.; Zeinali, M.; Hamza, B.; Engstrom, A.; Zhu, H.;
Sundaresan, T.K.; Miyamoto, D.T.; et al. A microfluidic device for label-free, physical capture of circulating
tumor cell clusters. Nat. Methods 2015, 12, 685–691. [CrossRef] [PubMed]
230. Lu, Y.-T.; Zhao, L.; Shen, Q.; Garcia, M.A.; Wu, D.; Hou, S.; Song, M.; Xu, X.; OuYang, W.-H.;
OuYang, W.W.-L.; et al. NanoVelcro chip for CTC enumeration in prostate cancer patients. Methods 2013,
64, 144–152. [CrossRef] [PubMed]
231. Huang, M.-Y.; Liu, H.-C.; Yen, L.-C.; Chang, J.-Y.; Huang, J.-J.; Wang, J.-Y.; Lin, S.-R. Decreasing relapse in
colorectal cancer patients treated with cetuximab by using the activating KRAS detection chip. Tumor Biol.
2014, 35, 9639–9647. [CrossRef] [PubMed]
232. Xue, P.; Wu, Y.; Guo, J.; Kang, Y. Highly efficient capture and harvest of circulating tumor cells on a
microfluidic chip integrated with herringbone and micropost arrays. Biomed. Microdevices 2015. [CrossRef]
[PubMed]
233. Zhang, Z.; Shiratsuchi, H.; Lin, J.; Chen, G.; Reddy, R.M.; Azizi, E.; Fouladdel, S.; Chang, A.C.; Lin, L.;
Jiang, H.; et al. Expansion of CTCs from early stage lung cancer patients using a microfluidic co-culture
model. Oncotarget 2014, 5, 12383–12397. [CrossRef] [PubMed]
234. Huang, T.; Jia, C.-P.; Jun-Yang; Sun, W.-J.; Wang, W.-T.; Zhang, H.-L.; Cong, H.; Jing, F.-X.; Mao, H.-J.;
Jin, Q.-H.; et al. Highly sensitive enumeration of circulating tumor cells in lung cancer patients using a
size-based filtration microfluidic chip. Biosens. Bioelectron. 2014, 51, 213–218. [CrossRef] [PubMed]
235. Ying, L.; Zhu, Z.; Xu, Z.; He, T.; Li, E.; Guo, Z.; Liu, F.; Jiang, C.; Wang, Q. Cancer associated
fibroblast-derived hepatocyte growth factor inhibits the paclitaxel-induced apoptosis of lung cancer A549
cells by up-regulating the PI3K/Akt and GRP78 signaling on a microfluidic platform. PLoS ONE 2015, 10,
e0129593. [CrossRef] [PubMed]
236. Ruppen, J.; Wildhaber, F.D.; Strub, C.; Hall, S.R.R.; Schmid, R.A.; Geiser, T.; Guenat, O.T. Towards
personalized medicine: Chemosensitivity assays of patient lung cancer cell spheroids in a perfused
microfluidic platform. Lab Chip 2015, 15, 3076–3085. [CrossRef] [PubMed]
237. Jeon, J.S.; Zervantonakis, I.K.; Chung, S.; Kamm, R.D.; Charest, J.L. In vitro model of tumor cell
extravasation. PLoS ONE 2013, 8, e56910. [CrossRef] [PubMed]
238. Bersini, S.; Jeon, J.S.; Dubini, G.; Arrigoni, C.; Chung, S.; Charest, J.L.; Moretti, M.; Kamm, R.D. A
microfluidic 3D in vitro model for specificity of breast cancer metastasis to bone. Biomaterials 2014, 35,
2454–2461. [CrossRef] [PubMed]
239. Jeon, J.S.; Bersini, S.; Gilardi, M.; Dubini, G.; Charest, J.L.; Moretti, M.; Kamm, R.D. Human 3D vascularized
organotypic microfluidic assays to study breast cancer cell extravasation. Proc. Natl. Acad. Sci. USA 2015,
112, 214–219. [CrossRef] [PubMed]
240. Wikswo, J.P.; Block, F.E.; Cliffel, D.E.; Goodwin, C.R.; Marasco, C.C.; Markov, D.A.; McLean, D.L.;
McLean, J.A.; McKenzie, J.R.; Reiserer, R.S.; et al. Engineering challenges for instrumenting and controlling
integrated organ-on-chip systems. IEEE Trans. Biomed. Eng. 2013, 60, 682–690. [CrossRef] [PubMed]
241. Hayes, D.F.; Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Miller, M.C.; Matera, J.; Allard, W.J.;
Doyle, G.V.; Terstappen, L.W.W.M. Circulating tumor cells at each follow-up time point during therapy of
metastatic breast cancer patients predict progression-free and overall survival. Clin. Cancer Res. 2006, 12,
4218–4224. [CrossRef] [PubMed]
© 2015 by the authors; licensee MDPI, Basel, Switzerland. This article is an open
access article distributed under the terms and conditions of the Creative Commons by
Attribution (CC-BY) license (http://creativecommons.org/licenses/by/4.0/).
31170