You are on page 1of 19

Biomolecular Engineering 23 (2006) 213–231

www.elsevier.com/locate/geneanabioeng

Review
Merging microfluidics with microarray-based bioassays
Catherine Situma, Masahiko Hashimoto, Steven A. Soper *
Center for Bio-Modular Multi-Scale Systems, Department of Chemistry,
Louisiana State University, Baton Rouge, LA 70803, United States
Received 8 February 2006; received in revised form 10 March 2006; accepted 10 March 2006

Abstract
Microarray technologies provide powerful tools for biomedical researchers and medicine, since arrays can be configured to monitor the
presence of molecular signatures in a highly parallel fashion and can be configured to search either for nucleic acids (DNA microarrays) or proteins
(antibody-based microarrays) as well as different types of cells. Microfluidics on the other hand, provides the ability to analyze small volumes
(micro-, nano- or even pico-liters) of sample and minimize costly reagent consumption as well as automate sample preparation and reduce sample
processing time. The marriage of microarray technologies with the emerging field of microfluidics provides a number of advantages such as,
reduction in reagent cost, reductions in hybridization assay times, high-throughput sample processing, and integration and automation capabilities
of the front-end sample processing steps. However, this potential marriage is also fraught with some challenges as well, such as developing low-
cost manufacturing methods of the fluidic chips, providing good interfaces to the macro-world, minimizing non-specific analyte/wall interactions
due to the high surface-to-volume ratio associated with microfluidics, the development of materials that accommodate the optical readout phases of
the assay and complete integration of peripheral components (optical and electrical) to the microfluidic to produce autonomous systems
appropriate for point-of-care testing. In this review, we provide an overview and recent advances on the coupling of DNA, protein and cell
microarrays to microfluidics and discuss potential improvements required for the implementation of these technologies into biomedical and
clinical applications.
# 2006 Published by Elsevier B.V.

Keywords: Microfluidics; Microarrays; DNA diagnostics; Protein arrays; Cell arrays

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
1.1. What are microarrays? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
1.2. Microfluidics: basic fabrication and operation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 214
1.3. Why merge microarray technology with microfluidics? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 215
2. Microfluidics and microarrays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217
2.1. Support materials for microarray production . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 217
2.2. Probe array preparation techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 221
2.3. Hybridization speed enhancement in microfluidic devices . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222
2.4. Applications of microfluidic-based microarrays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 225
2.4.1. DNA microarray systems. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.4.2. Protein microarray systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
2.4.3. Cell-based microarray systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
3. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
Acknowledgement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 228

* Corresponding author. Tel.: +1 225 578 1527; fax: +1 225 578 3458.
E-mail address: chsope@lsu.edu (S.A. Soper).

1389-0344/$ – see front matter # 2006 Published by Elsevier B.V.


doi:10.1016/j.bioeng.2006.03.002
214 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

1. Introduction applied in proteomics for the analysis and quantification of a


large number of proteins comprising a proteome. Microarray
The timely and correct expression of a large number of genes applications in protein analyses includes screening binding
and the products they produce, such as mRNAs and/or proteins, specificities of a protein expression library (Postier Bradley
is a critical component of normal development and main- et al., 2003), high-throughput antibody screening (Bussow
tenance of all living organisms. Any disruptions or changes in et al., 2000), or the detection of protein–protein interactions,
genes and the products they express are responsible for many enzymatic targets and protein-small molecule interactions
diseases that possess a genetic basis. Biomedical and clinical (MacBeath and Schreiber, 2000).
diagnosis/prognosis evolves and advances not only through the Not only can DNAs and proteins be successfully printed
compilation of knowledge accumulated by the discovery of new onto substrates in a microarray formats, but cells have also been
biomarkers, but also through the development of new patterned onto substrates in a microarray format, which has
technologies that can read the presence or absence of those advanced the study of cellular functions and metabolism,
biomarkers as well as the amount of those markers found in a identification of the phenotype of a cell in a population of
clinical sample. Unfortunately, the traditional tools used to heterogeneously transfected cells (Wu et al., 2002), the
assay gene expression, such as SAGE (serial analysis of gene detection of toxic agents and high-throughput screening of
expression) requires steps that are not conducive to analyzing combinatorial libraries/gene products (Kapur, 2005).
large numbers of genes simultaneously, which is typically Many complex diseases and traits are caused by rare alleles,
required in many multi-gene related diseases, such as cancer. which can only be detected by sequencing complete genomic
Thus, the emergence of highly parallel processing techniques, regions in multiple individuals first for the identification of the
such as microarrays, has enabled researchers to study thousands variants and then for disease diagnostics. Progress in
of genes simultaneously for both discovery and diagnostic microarray systems, which enable highly multiplexed genotyp-
applications. ing, is therefore very useful for genome-wide single nucleotide
polymorphism (SNP) genotyping. There exists a database of
1.1. What are microarrays? hundreds of SNPs/single base differences in the human
genome, which can be used as genetic markers. As such,
A microarray consists of a support onto which hundreds to microarrays can be used in genotyping assays and also in
thousands of different molecular reporter probes are attached or comparative genomic hybridization (CGH), a technique that
immobilized at fixed locations in either a two-dimensional or reveals how many copies of given genes are actually in a sample
three-dimensional format. The multiplexing capabilities of being interrogated. Microarrays have been demonstrated
microarray-based assays are produced by spatially encoding the widely in SNP genotyping applications (Baner et al., 2003;
array, in which each location on the array is used as a reporter of Erali et al., 2003; Grimm et al., 2004; Ishikawa et al., 2005; Lin
a specific analyte. Due to the similar nature in which each assay et al., 2004; Lovmar et al., 2003; Meaburn et al., 2006).
at a location is carried out, highly reproducible and quantitative
information can be obtained from each address of the array. The 1.2. Microfluidics: basic fabrication and operation
probes that can be used can consist of antibodies to recognize
specific proteins or oligonucleotides/PCR products that Microfluidics is an emerging and relatively new technology
recognize unique DNA sequences through Watson–Crick base platform that typically requires a multi-disciplinary approach
pairing. for building effective systems through the interface of physics,
DNA microarrays consist of oligonucleotides that are chemistry, engineering and biochemistry. These devices usually
attached onto supports and are used to interrogate the presence consist of structures that are fabricated in the micrometer-scale
or the amount of analyte (i.e., targets or DNAs) in sample and can be designed to produce diagnostically useful systems
solutions. DNA microarray assays are based on the principle of for potential point-of-care measurements. Systems based upon
specific molecular recognition events that occur for nucleic acids microfluidic operation possess a unique set of potential
through interactions between complementary sequences. Selec- advantages, such as reduction in reagent cost, enhancement
tivity is imparted by targets and probes that are non- of assay speed, potential for mass production of devices at low
complementary and thus do not form thermodynamically strong cost and the ability to integrate several processing steps into a
hybrids, which can be selectively tuned out through stringent single system (i.e., high levels of system automation).
washing with buffers or temperature regulation. Because Decreased reagent consumption eventually results in the
microarrays allow one to investigate large numbers of analytes reduction of assay cost, which could translate into large-scale
or targets rapidly and efficiently, researchers have applied this molecular screening of certain diseases for early detection. The
technology not only in gene expression analysis (Zhang et al., accelerated analysis time is directly gained from the reduced
1991), but also in drug screening/therapeutic studies (Lebrun, footprint of these devices. Other benefits of microfluidic-based
2005), proteomics (MacBeath, 2002; MacBeath and Schreiber, systems include high-throughput biological screening by
2000), cell analysis (McClain et al., 2003), and DNA sequencing/ parallel processing of multiple samples achieved with multiple
fragment analysis (Pease et al., 1994). fluidic paths and the minimization of cross contamination, since
Although microarray technology was initially fostered in different samples flow through separate fluidic processors in
genomic applications, its basic operational concepts have been closed architectures.
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 215

Microfluidic devices can be fabricated from a variety of Using UV laser ablation or micromilling, a polymer
materials using different techniques. Based primarily on the substrate can be exposed to laser radiation or a milling bit
fabrication techniques developed in the microelectronics sector, resulting in the direct writing of the microstructures into the
silicon and glass have traditionally been used to create polymer part. In the case of laser ablation, the laser photons,
microfluidic devices (Li et al., 1999; McClain et al., 2003). typically from a pulsed excimer laser, is focused directly on the
Microfabrication in glass/silicon substrates has been accom- polymer. Fluidic channels are produced either by moving the
plished through the use of lithography, plasma etching or reactive polymer substrate or by moving a focused laser beam across the
ion etching techniques (Boerner et al., 1996; He et al., 1999). The surface. This method offers the ability to form complex features
particularly attractive advantages of glass and quartz include with various geometries, even in the third dimension, because
their well defined surfaces and excellent optical properties, the patterning beam can be moved both horizontally and
which are highly desired for signal readout of microarrays by vertically on the substrates. In either case, fluidic prototypes
fluorescence. However, the fabrication processes involved are can be rapidly produced using direct write techniques to
fairly time consuming, labor intensive and relatively expensive. optimize the device performance prior to mass production using
Polymers are rapidly evolving as alternative substrate micro-replication techniques.
materials for many microfluidic applications due to their diverse Poly(dimethylsiloxane) (PDMS) fabrication technology,
properties that can be selected to suite the particular application sometimes called soft lithography, was developed by White-
need and the ability to microfabricate structures in a high sides and co-workers and has been widely adopted for the
production mode and at low cost. The fabrication of the fabrication of different microfluidic networks (Anderson et al.,
prerequisite fluidic networks on polymer substrates can be 2000a; Duffy et al., 1998, 1999; McDonald and Whitesides,
achieved by lithography, UV laser ablation, hot embossing, 2002; Sia and Whitesides, 2003). Fabrication of microfluidic
injection molding, or direct micromilling techniques (Ford et al., structures using PDMS technology involves pouring a solution
1999; Friedrich et al., 1997; Grass et al., 2001; Ihlemann and containing the PDMS prepolymer and a curing agent over a
Rubahn, 2000; Martynova et al., 1997; Qi et al., 2002; Roberts relief (mold) containing the prerequisite microstructures
et al., 1997; Rossier et al., 1999; Soper et al., 2000). Hot followed by curing to cross-link the polymer. This technology
embossing and/or injection molding involve the use of mold first requires creation of positive reliefs using a variety of
masters containing the desired microstructures to stamp (reverse- techniques (Duffy et al., 1998; Becker and Gartner, 2000; Love
duplicate) patterns into the required substrate. The patterns et al., 2001), such as wet etching of silicon or glass following
poised on the mold masters can be made either by micro- photolithography, micromachining metals or reactive ion
machining or by a lithography-based approach, such as LiGA; etching of silicon. A solution containing the pre-polymer
German acronym for lithography (Lithograhie), electroplating and curing agent is then cast against the relief and the cross-
(Electroformung) and molding (Abformung). A schematic of the linked polymer conforms to the shape of the relief. After
LiGA processing steps using X-ray lithography to build the casting, the polymer is simply removed from the relief resulting
microstructures on the mold master is shown in Fig. 1. The in a replica that contains the network of microfluidic channels.
important aspect of this technology is that the difficult fabrication Finally, the PDMS replicate can be plasma oxidized and sealed
steps, lithography and electroplating, are performed but once and to other surfaces by conformal contact to enclose the fluidic
then, parts are micro-replicated from this mold master using network. Plasma oxidation also renders the PDMS channels
either injection molding or hot embossing. For hot embossing, hydrophilic so that they can easily be filled with aqueous
three steps are required: (1) First, the polymer substrates are solutions. Besides plasma oxidation, Quake and coworkers has
placed between two metal plates, one containing the micro- also described a method for sealing fluidic networks created in
structure topology, and are heated above their glass transition PDMS, which involves the use of two slabs (Unger et al., 2000);
temperature. (2) A force is applied on the softened polymer by one slab consists of an excess of base while the other contains
pressing with the heated mold master under vacuum. (3) The the curing agent. These two are then brought into conformal
metal plates and polymer substrate are cooled below the contact followed by curing.
polymer’s glass transition temperature and the metal plates are
withdrawn from the polymer part. Due to the simplicity of the 1.3. Why merge microarray technology with microfluidics?
process and the fact that the mold master only requires one
lithography step to fabricate it, the procedure is very cost For conventional microarrays, several shortcomings exist
effective in that it can mass produce thousands of parts and the including: (1) The hybridization (analyte binding) process on
whole production process can be completed in <5 min. supports requires long incubation times to produce the optimal
Examples of polymer parts embossed from a mold master are signal because of slow diffusional kinetics as the target
depicted in Fig. 1. In addition, the process is conducive to molecules must travel to the arrayed probes situated on a
replicating parts in a variety of materials from the same mold surface. This process is typically diffusion-controlled and the
master as long as the glass transition temperature of the substrate fact that reactions on the surface are slow due to local target
is below the melting point of the master. Once the fluidic network depletion near the probe environment. (2) The consumption of
is formed, a cover plate of the same material can be sealed to the large amounts of precious sample material for interrogating the
substrate to enclose the fluidic channels using a low-temperature array due to the large area occupied by high density arrays
sealing process. (1 cm2). In order to completely cover the array, the volume of
216 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

Fig. 1. Processing steps required to prepare a mold master using X-ray LiGA and molding finished parts. The process starts with a lithography step using a mask that
transfers the desired pattern into a photoresist. Following development of the resist, the underlying metal layer is exposed, which serves as a plating base for another
metal that fills the voids left by the resist that was removed due to exposure to the patterning radiation. The deposited metal forms the desired microstructures. The
unexposed resist is then removed, forming the finished mold master. This mold master is then available for micro-replicating parts in polymers or other materials using
hot-embossing or injection molding. Shown on the right in this figure is a picture of the mold master and some parts that have been prepared from the mold master.
Adopted with permission from Ford et al. (1999).

sample analyzed must be such that the fluid covers every The linkage chemistry of the probe to the support can undergo
element of the array. In addition, the large lateral distances that solution-dependent cleavage over the extended incubation
must be traveled by low concentration species for high density times, lowering the reproducibility and sensitivity of the assay.
arrays in order to find their appropriate binding partners can add For example, siloxane-based chemistry is typically used to
to the incubation time, slowing down assay processing rate. (3) tether molecules to glass, and this is susceptible to hydrolytic
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 217

cleavage, especially at high or low pH values. Therefore, any molecules interrogated is low for microfluidic-based devices,
process that can enhance the rate of producing the maximum which typically have interrogation volumes that are <1 nL. In
signal for surface-confined association events can provide a addition, the substrate selected for the microfluidic must
number of important benefits, such as reducing the develop- accommodate the sensitivity requirements for readout. For
ment time and its associated consequences. example, while polymers are attractive due to their low cost and
The use of microfluidic platforms can directly address many high production capabilities using micro-replication methods,
of the issues cited above for microarrays and thus, the merging many polymers display poor optical properties that are not
of microfluidics with microarrays can provide some unique conducive to the readout modality required for the array.
opportunities that are not obtainable when arrays are configured Judicious selection of the polymer must be undertaken, since
into conventional planar 2D formats. For example, microfluidic several materials do display excellent properties for optical
addressing of the array can accelerate hybridization speeds due readout, especially when using visible or near-IR fluorescence
to the enhanced mass transport of target molecules and reduced (Shadpour et al., 2006). In order to realize cost-savings in terms
diffusional distances required of targets when delivered in a of reagent consumption using microchips, precious reagents
micro-scale flow-through hybridization chamber. In addition, can be distributed to multi-channel chips by using low-volume
microfluidics can offer the potential for parallel processing of fluidic distribution networks (Fu et al., 2002; Liu et al., 2003a).
multiple samples. Other advantages offered by the incorpora- Another issue that should be considered is that while the
tion of microfluidics into the microarray includes reduced footprint of microfluidic chips have been substantially reduced,
reagent and sample volumes and the integration capabilities of these devices must be interfaced to the macro-world. In most
sample pre-processing prior to incubation with the array, which cases, microfluidic chip loading, both with reagents and
can provide high levels of system automation and also, provide samples, is accomplished using conventional pipettes with fluid
closed architectures for sample processing minimizing con- movement actuated either electrokinetically or hydrodynami-
tamination effects that can give rise to false positives, which can cally. In addition, readout of molecular association events must
be a significant issue for diagnostic applications. be accomplished using techniques such as laser-induced
Since microfluidics is a relatively new field of research, there fluorescence. In most cases, these operational processes are
are likely to be challenges when attempting to merge it to a carried out using off-chip apparati, such as mechanical pumps,
relatively mature technology platform, such as microarrays. For high voltage power supplies and optical readers, similar to
instance, there is a need for the development or discovery of those used for reading conventional 2D microarrays. While
new materials that are easily fabricated on the size-scale efforts have been initiated to package these components into a
demanded by the microfluidic application and at the same time, complete and autonomous system with a footprint similar to
offer modification chemistries that allow for the stable that of the fluidic elements, these efforts are still in their
attachment of probes. In addition, the fabrication technologies infancy. In this review, we will focus primarily on miniaturiza-
as well as the material used for the device must be low cost if tion efforts focused on the fluidic elements. For information on
intended for diagnostic screening using microarray-based packaging and integration of ancillary components to the fluidic
assays. elements, the reader is referred to the following references
Even though miniaturized devices offer the advantages (Auroux et al., 2002; Burns et al., 1998, 1996; Liu et al., 2004a;
associated with the small scale reactors and fluidic vias used in Pal et al., 2005; Reyes et al., 2002).
their implementation, the high surface area to volume ratio This review will detail the current research that involves
associated with these small scales produce surface effects that coupling microarrays to microfluidics with emphasis on DNA
dominate volume effects. As such, there is the potential for arrays, protein arrays and cell-based arrays with concluding
increased surface adsorption of solution molecules that can lead remarks on potential improvements required for future
to high-levels of non-specific adsorption, providing poor developments that will allow the benefits of microfluidics/
microarray spot definition or even fast positive results. Methods microarray devices to be fully utilized for applications in
to ameliorate these non-specific adsorption artifacts must be diagnostics and the understanding of biological function for
employed, which can consist of selecting the appropriate discovery-based projects. Information will be presented on the
substrate for the microfluidic that demonstrates minimal non- different types of substrates that can be used for microarrays
specific adsorption for the intended application or to use that are also conducive to microfluidic fabrication and
dynamic or covalent coating procedures of the fluidic substrate operation as well as the attachment chemistries that can be
(Gong et al., 2005; Henry et al., 2000; Hu et al., 2004; Lee et al., employed for probe tethering to the surface. Novel topogra-
2003; Lu et al., 2005; McCarley et al., 2005; Nuzzo, 2003; phical architectures for microarrays configured into micro-
Sebra et al., 2005). fluidics will also be presented.
While microfluidics can potentially provide the use of
reduced sample and reagent volumes due to the small size of 2. Microfluidics and microarrays
these elements, the detection of molecules in dilute solutions
will require highly sensitive detection devices, maybe even at 2.1. Support materials for microarray production
the single molecule level. This is so because the number of
molecules sampled scales as the cube of the volume analyzed Microarrays require the use of substrates with well-
and hence, with low concentration samples the number of established surface modification/probe attachment chemistries
218 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

Fig. 2. Various modification strategies that can be used to attach probes to different solid supports. (A) Glutardialdehyde linkage of amine-containing
oligonucleotides to glass surfaces. (B) Covalent immobilization of oligonucleotide probes to PC. (C) UV activation of PMMA with the subsequent attachment
of primary amine containing oligonucleotides. Adopted with permission from Joos et al. (1997), Lenigk et al. (2002a) and Situma et al. (2005).
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 219

Fig. 2. (Continued ).

that provide high probe densities on the surface of the substrate, surface forming an array of spots with high affinity for target
maximize probe accessibility by its binding partner, provide analytes. The micro-porous silicon was fabricated by means of
robust attachment chemistries between the probe and substrate anodic dissolution in hydrofluoric acid producing rigid sponge-
that are stable and easy to implement, and minimize non- like structures with high uniformity and mechanical stability as
specific interactions. Glass and quartz have been widely used as well as high surface area due to the three-dimensional network
typical support materials since their physical/chemical proper- formed following anodic dissolution. The microfluidic proper-
ties are well investigated including, surface modification ties of these substrates were then utilized for arraying of
chemistries (Halliwell and Cass, 2001; Joos et al., 1997). antibodies that were able to capture antigen binding partners
Glass is particularly well-established in the literature as a with detection levels of 70 pM.
substrate material for microarray applications due to its Recently, polymers have generated attention from research-
superior optical properties and the presence of silanol groups ers as potential supports for the production of microarrays
that can be easily functionalized using cross linking agents, because of their low cost and also, they offer a diverse range of
such as glutardialdehyde or an amino-silane reagent, to allow chemical structures to allow the implementation of different
stable attachment of DNA or protein probes directly to the glass immobilization strategies of probes to the support. Polymers
using siloxane-based chemistries (see Fig. 2A). In addition, the that have been used for microarray applications include;
functional group density can be relatively high, with levels poly(dimethylsiloxane) (PDMS), polycarbonate (PC), poly(-
approaching 10 9 mol cm 2 (Horvath and Dolnik, 2006). methyl methacrylate) (PMMA), polystyrene, poly(ethylene
An example of microarrays fabricated on silicon is the work terephthalate) (PET) and cellulose acetate. PDMS has been
reported by Ressine, who used a pore chip protein array (PCPA) widely used because it is inexpensive, and has high optical
based on the use of micro-porous silicon as the matrix for transparency. Other substrates that have been reported in the
immobilizing antibodies (Ressine et al., 2003). The capture literature as substrates for microarrays include gold (Csaki
antibodies were anchored onto the surface by physical et al., 2001; Steel et al., 2000), filter membranes (Liu et al.,
adsorption. The probes were micro-dispensed onto the chip 2004b), and optical fibers (Ahn and Walt, 2005).
220 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

The use of microbeads as supports for the immobilization of immobilized onto beads through streptavidin–biotin conjuga-
probes has also been reported (Marquette and Blum, 2004; Sato tion for interrogation with DNA probes that were transported
et al., 2001). Bead arrays have been used extensively because via pneumatic pumping. This dynamic DNA hybridization
they are inexpensive, efficient, require mild operating condi- offered flexibility such that the type/amount of beads, DNA
tions and allow multiplexed assays when each bead is used to targets or probes could be changed as needed. Beads conjugated
capture a specific analyte. However, the challenges that occur in with DNA probes have also been placed in a capillary tube
these multiplexed formats hinge on the ability to decode the producing linear probe arrays (Kohara et al., 2002). Most
specific identity of the probe on any particular microbead. recently, Lilliehorn et al. have demonstrated a method of
Thus, bead-based assays have focused predominately on their generating arrays in microfluidic devices based on ultrasonic
use in low density array applications. Microbeads do, however, trapping of microbeads using acoustic forces in standing waves
have the advantage of enhanced surface area compared to a (Lilliehorn et al., 2005). They produced protein arrays for
planar support, which increases the binding capacity of the simultaneous analysis of different sets of bead-based biotin–
probe and thus, increasing the dynamic range of the assay. In avidin samples. The antibody/antigen interactions were
work reported by Marquette et al., a method of arraying beads performed on chemically activated microbead clusters in a
bearing active enzymes, antibodies and oligonucleotides was laminar streaming microfluidic system. This involved the use of
presented (Roberts et al., 1997). A picture of the bead-based different antigen-specific bead clusters trapped in an array
microarray system is shown in Fig. 3. The beads were spotted pattern followed by addressing each bead cluster with
and dried on a poly(vinylchloride) (PVC) master and individual sample streams using acoustic forces. A model
subsequently transferred to a PDMS interface by direct bioassay using fluorescein-labeled avidin was bound to biotin-
molding of the polymer on the mask (Marquette and Blum, coated beads using this technique, which showed its potential
2004). This technique enabled the development of low density for parallel protein analysis in microfluidic devices.
arrays that were read out using chemiluminescence detection. Microbeads have also been used in conjunction with
Fan et al. have also described a dynamic DNA hybridization quantum dots for readout. For example, the ‘‘Q bead
protocol that incorporated paramagnetic beads as a transpor- technology’’ uses multi-colored microbeads labeled with
table support (Fan et al., 1999). Target molecules were quantum dots for specific biological assays. This technology

Fig. 3. Low density bead arrays fabricated in PDMS. The microbeads were made from Sepharose and were spotted onto a polyvinylchloride (PVC) master. (A) The
Sepharose beads could be derivatized with a variety of different probes, such as nucleic acids, antibodies or glucose oxidase (Gox). The readout of an association event
was performed using a chemiluminescent reaction with horseradish peroxidase (HRP) that was strapped to the target via a biotin/streptavidin linkage. (B) The beads
containing the appropriate probes were spotted onto the PVC master, which was used to form the fluidic network with the entrapped beads. A PDMS solution is poured
over the PVC master and following drying, it is peeled from the PVC master. The bottom image is generated by a chemiluminescence response following
hybridization of a poly d(T)22 oligonucleotide containing HRP. Adopted with permission from Marquette and Blum (2004).
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 221

involves the insertion of a combination of various quantum dots Pre-synthesized probes can be covalently or physically
into an array of beads with each bead serving as a recognition arrayed onto a variety of chip surfaces using several different
substrate for specific molecules. Since each bead can be chemistries (Gingeras et al., 1987; Okamoto et al., 2000).
individually prepared with different probes, there is an obvious Examples of several of these chemistries can be seen in Fig. 2.
advantage of flexibility. An example of the use of quantum dot Lenigk et al. (2002a) demonstrated the use of PC substrates
encoded micro-sphere hybridization assays have been described coated with bi-functional linker molecules for the immobiliza-
by Xu et al., who applied this technology for multiplexed SNP tion of pre-synthesized amine-terminated oligonucleotide
genotyping (Xu et al., 2003). Several companies have also probes (see Fig. 2B). Probe immobilization procedures were
developed special encoding techniques for bead-based hybridi- also studied on four different polymers (polystyrene, PC,
zation assays, such as Illumina (http://www.illumina.com) and PMMA and polypropylene) as possible support candidates for
Becton Dickinson (http://www.bdbiosciences.com). microarrays with three different immobilization procedures
(Surmodics, cetyltrimethylammonium bromide (CTAB) and
2.2. Probe array preparation techniques Reacti-bind procedures). These authors showed that different
polymers exhibited different immobilization efficiencies
DNA microarrays, which utilize short (<25 bp) oligonu- depending on the immobilization procedure used (Liu and
cleotide probes, can either be synthesized directly on the Rauch, 2003). Soper and coworkers described a functionaliza-
substrate or pre-synthesized separately and then micro-printed tion protocol that yielded amine-terminated functionalities on
or spotted onto the support in an orderly or fixed manner, PMMA (Henry et al., 2000). This protocol was later used for the
typically in a two-dimensional format. Identification of the covalent attachment of probe oligonucleotides for microarray
presence of a particular DNA sequence is transduced by a hybridization assays (Hashimoto et al., 2006; Waddell et al.,
recognition signal produced through an association event 2000; Wang et al., 2003). Recently, this same group has
between the surface-tethered probe and its solution comple- described a simple UV photo-modification protocol for PMMA
ment and locating the location of the signal in the two- and PC substrates that generate surface-confined carboxylate
dimensional array, which allows for highly parallel processing groups, which can be subsequently used for the immobilization
of multiple targets. High quality probe arrays are usually of amine-terminated nucleic acid probes through an amide-
desired to improve the sensitivity of the microarray assay. The bond as the linking chemistry (Situma et al., 2005). The photo-
probe array quality depends intimately on the probe attachment modification process for array fabrication involved only three
chemistry and deposition (spotting) technique used. Any non- steps (see Fig. 2C): (1) broadband UV exposure of the polymer
uniform surface morphology that might arise during surface surface; (2) carbodiimide coupling of amine-terminated
activation or spotting can lead to variances in the array element oligonucleotide probes to the surface (via an amide bond);
shape and probe surface concentration uniformity across the (3) washing of the surface. Other probe attachment techniques
particular element, which eventually affects the data quality. on PMMA surfaces have been presented by Fixe et al.
The availability of these surface attached probes to interact with (2004a,b). They developed a chemical procedure to functio-
their complementary targets during hybridization is also highly nalize PMMA by reaction of the substrate with hexamethylene
desirable. The surface probe density as well as its spacing from diamine to yield an aminated surface for immobilizing DNA
the surface will determine the extent to which the immobilized probes, which was accomplished using a glutardialdehyde
probes are able to capture their targets from solution because, cross-linking agent for the attachment of amino-modified
other than thermodynamic conditions, the immobilized probes oligonucleotide probes via a Schiff base.
can also be kinetically or sterically inaccessible (Peterson et al., Probe arrays can also be generated by use of PDMS-assisted
2001). techniques, which is based on the properties of PDMS to seal
Direct on-chip probe synthesis is achieved by combining both reversibly and irreversibly with other substrates. Lee et al.
solid-phase chemistry, photolabile protecting groups and constructed 1D and 2D DNA microarrays on gold films through
photolithography (Pease et al., 1994; Fodor et al., 1991; the use of parallel microfluidic channels fabricated in PDMS
Southern et al., 1994). An example of this is the Affymetrix (Lee et al., 2001). A PDMS polymer sheet containing parallel
microarray technology, whose Genechips are produced by microchannels was physically attached to a gold film modified
photolithography (Kim, 2003). These on-chip probe synthesis with self-assembled monolayers. Single stranded DNA
protocols allow for the manufacture of high density micro- (ssDNA) probes were directed through these channels with
arrays. Moorcroft et al. have recently described an in situ each channel containing a unique probe and allowed to
synthesis technique of oligonucleotide probes onto PDMS immobilize onto the gold surface after which the PDMS was
surfaces, which is a common substrate frequently used in removed from the gold surface. These ssDNA probes were used
microfluidic applications, using phosphoramidite chemistry for the interrogation of complementary targets, which were
(Moorcroft et al., 2005). The PDMS surface was activated by directed onto the probe array surface by using a second set of
UV/ozone exposure followed by vapor phase deposition of PDMS fluidic channels placed perpendicular to the probe
glycidoxypropyl trimethloxysilane, which was then reacted arrays. This stenciling technique has been adopted for
with polyethylene glycol spacers that aided in oligonucleotide patterning protein antigens on gold surfaces (Kanda et al.,
coupling and the minimization of non-specific adsorption of 2004) and DNA probe arrays onto PMMA surfaces (Situma
targets to the PDMS surface. et al., 2005).
222 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

Su et al. demonstrated the use of microfluidic networks molecule of 30 bases is approximately 2.4  10 7 cm2 s 1 at
fabricated in PDMS to prepare peptide microarrays on gold by room temperature and in free solution. Therefore, for a
using self-assembled monolayers (SAMs) of alkanethiols (Su molecule to travel laterally 1 cm across the array would require
et al., 2005). Microchannels fabricated in PDMS were used to approximately 289 h!
immobilize a linear array of cysteine-terminated peptides onto Several techniques have been employed to enhance mass
the SAMs containing maleimide groups. The PDMS stencil was transport of analytes and reduce diffusional distances using
then reapplied to the SAM in a perpendicular direction to shallow microchannels (<100 mm) to reduce the hybridization
introduce enzyme solutions so that each solution could interact incubation time. One such technique is the use of continuous
with an identical linear array of immobilized peptides. The set flow of targets within microchannels by hydrodynamic
of peptides used enabled a multi-analyte assay selective for pumping (Hashimoto et al., 2006; Wang et al., 2003; Cheek
kinase and phosphatase enzymes. Another example is that et al., 2001; Noerholm et al., 2004). Forced flow of the target
demonstrated by Tani et al., who used PDMS microchannels solution over the probe array surface provides enhanced mass
connected via perforated microwells to a silicon substrate for transport of targets to the surface-immobilized probes since the
the immobilization of Escherichia coli sensor strains (Tani transport does not depend solely on diffusion, but also on
et al., 2004). The sensor strains were mixed with agarose and convection leading to reduced hybridization times. In addition,
then injected into the channels and immobilized by gelation. the use of shallow fluidic channels reduces the diffusional
Probe arrays have also been constructed in 3D ordered distances that must be traveled to reach the probe. Hydro-
microchannels (Benoit et al., 2001; Cheek et al., 2001). These dynamic flow has been shown to reduce the hybridization time
uniform porous substrates enhance probe immobilization to less than 1 min (Wang et al., 2003).
because the effective surface area available for probe Introduction of high sample velocities can induce exten-
immobilization is increased compared to that of planar 2D sional strain to solution targets reducing hybridization time and
surfaces. They also enable uniform spot shapes to be achieved efficiency as well (Chung et al., 2004, 2003; Vanderhoeven
since the evaporation of probe solutions are greatly reduced et al., 2004). Chung et al. demonstrated that long DNA targets
during the immobilization process. Probes used in these flow- tend to change into a super coil form in solution that reduces
through geometries eventually increase the sensitivity and hybridization efficiencies due to the inaccessibility of the
reduce hybridization assay analysis times as well. Hydrogel recognition sequence in the target. Therefore, extensional strain
microstructures can also provide an ideal 3D aqueous can help to uncoil the long DNA molecules improving
microenvironment for the capture and encapsulation of hybridization efficiency (Chung et al., 2004). The authors
biomolecules. Koh et al. fabricated arrays of hydrogel were able to show a nearly nine-fold increase in hybridization
microstructures of different cell phenotypes using microfluidics signal due to strain-induced extension of a 1.4 kbp single-
made from PDMS (Koh et al., 2003). They used three different stranded DNA target by introducing a taper within the fluidic
phenotypes of cells; fibroblasts, hepatocytes and macrophages, channel. Vanderhoeven et al. also developed a shear-driven flow
for the fabrication of multi-phenotype cell microarrays. This system that induced enhanced lateral transport rates in
group was able to uniformly distribute and control the number hybridization chambers increasing hybridization efficiencies
of cells in one hydrogel microstructure by changing the cell as well (Vanderhoeven et al., 2004).
density in the precursor conditions. This reduced the sample Another approach for reducing hybridization time is by
volume of a cell containing precursor solutions when compared using active mixing of samples. While the use of devices with
to conventional cell patterning techniques, which involve the small dimensions reduces the target diffusional lengths, the
use of spin coating procedures. fluidic flow within these devices are uniaxial and laminar due to
their low Reynolds numbers. Mixing in such flows is due
2.3. Hybridization speed enhancement in microfluidic primarily to molecular diffusion (Yaralioglu et al., 2004). The
devices hybridization rate can be improved by active mixing because it
can replenish the target concentration in the vicinity of each
In all microarray applications, whether they are DNA or probe due to depletion created through binding. Active mixing
protein-based arrays, the target is typically found in solution also relieves the dependence of only lateral diffusion to
and must diffuse to the surface to interact with its probe, since replenish target near its probe.
the probe is chemically tethered to the surface and thus, Mixing-enhanced hybridization can be described as a three-
immobile. In addition, it must move laterally to find the location step process: (1) transport (diffusion and convective flow) of
of its appropriate binding partner in 2D gridded arrays. As such, targets in the solution to the diffusion (stagnant) boundary
in many cases, several hours of incubation time are required to layer; (2) transport (primarily diffusion) of target within the
allow probe saturation to be obtained due to the slow mass diffusion boundary layer to the probes on the chip surface; (3)
transfer of target to its appropriate probe. For example, with a reaction of target with probes on the surface (Liu et al., 2003b).
1 cm2 array area and a low target concentration in the solution, Conventional microstreaming has the ability to provide rapid
depletion of target near the probe recognition element of the lateral mass transport and enhanced vertical mass transport of
two-dimensional array will require new target to diffuse the target DNA in solution. This combination results in rapid
laterally to ‘‘find’’ its appropriate probe. As a reference, the transport of targets in solution to the diffusion boundary layer,
translational diffusion coefficient for a single-stranded DNA which allows a continuous replenishment of DNA targets
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 223

around probes depleted of complementary targets. This flows caused by the shearing interaction of the fluid in the plug
phenomenon was later used to perform DNA hybridizations with the walls of the microchannel.
with electrochemical detection on a fully integrated biochip, Lenigk et al. demonstrated the use of an integrated cartridge,
which consisted of fluidic components that were used to which was comprised of fluidic channel networks and a
enhance target cell capture and accelerate hybridization micropump for the oscillation of the hybridization mixture
reactions (Liu et al., 2004a). (Lenigk et al., 2002b). Accelerated hybridization times were
Yaralioglu et al. demonstrated the use of ultrasonic realized in these channels due to the ability to pass a large
transducers for mixing by an acoustic streaming mechanism number of target molecules per unit time over a probe spot
(Yaralioglu et al., 2004). They showed that when reagents were during filling of the channel. This model was then used for the
localized within discrete droplets in a microchannel with a low simultaneous detection of four pathogenic bacteria surrogate
Reynolds number, they could be mixed quickly by re-circulating strains in a parallel format.

Fig. 4. (A) Schematic of an array of electrodes fabricated on Si. The 25 electrodes, made from Pt, are 80 mm in diameter and cover an area of 1 cm2. The insulating
layer consists of silicon nitride layer and over the Pt electrodes was a gel permeation layer consisting of agarose gel loaded with streptavidin. The bottom panel shows
a cross-section of one element of the array with the immobilized DNA probes. The probes are attached to the permeation layer via a biotin–streptavidin association.
(B) Electric field enhanced transport of a poly d(T)12 target to the Pt electrode surface with different bias voltages applied to the Pt electrode: C1: positive potential;
C2: negative potential; C3: no potential. R3 and R5 correspond to biotinylated and non-biotinylated probes, respectively. Adopted with permission from Sosnowski
et al. (1997).
224
Table 1
Abbreviated list of literature reports highlighting the coupling of DNA microarrays to microfluidics
Reference Sample Support Analysis speed Detection Functions Applications/achievements
volume material (min) limits integrated
Lilliehorn et al. (2005) 1 ml Glass n/a n/a Ultrasonic microtransducers A model bioassay of a multiplexed bead-trapping
microarray consisting of piezoelectric microtranducers
integrated in microfluidic channels
Lenigk et al. (2002a) 25 ml PC 30 n/a Integrated air pump for (i) Accelerated hybridization process by sample oscillation;
sample oscillation (ii) application in single nucleotide polymorphism detection,
detection of pathogen bacteria surrogate
Wei et al. (2005) 1 ml PMMA 8 19 amol n/a Enhanced microarray processing speed in
microfluidics by ‘‘shuttle hybridization’’

C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231


Anderson et al. (2000b) 15 ml PC 20 n/a Multiple enzymatic reactions Parallel multi-step molecular operations and hybridization
and reagent handling assays integrated on a miniaturized device
Dimalanta et al. (2004) ml PDMS n/a 25 pg/ml DNA elongation, deposition Restriction enzyme mapping and identification
and digestion of human genomic DNA molecules
13
Ali et al. (2003) 1.5 ml Silicon <10 10 M Fluid transport (i) Chip-based sensor array incorporated DNA
capture probes into microporous microbeads;
(ii) multiplexed oligonucleotide microarray assays
Wang et al. (2003) 0.2 ml PMMA 5 1 mutant in 10,000 n/a Detection of point mutations in certain gene fragments
wild type copies using universal zipcode arrays
Hashimoto et al. (2006) 0.1 ml PMMA/PC <20 1 mutant in 100 Ligase detection reaction/ Flow throughput biochip for mutational analysis
wild type copies hybridization assay
Liu et al. (2003b) 1 ml PC 60 n/a Fully integrated for cell capture/ Pathogenic bacterial DNA detection/single nucleotide
cell preconcentration/purification/ polymorphism genetic analysis
cell lysis and PCR amplification/
DNA hybridization
Erickson et al. (2004) nl Glass/PDMS 5 50 pM Synchronized online DNA (i) Reduced sample volumes; (ii) rapid removal
hybridization, washing of nonspecifically adsorbed materials;
and detection (iii) enhanced hybridization
Benoit et al. (2001) 0.5 ml Glass 180 40 pM n/a Multiplexed DNA detection
Lee et al. (2001) 1 ml Gold/PDMS 60 20 fmol n/a Parallel detection of specific adsorptions of DNA /RNA.
Cheek et al. (2001) 50 ml Glass 180 250 amol n/a Flow-through chemiluminescence detection of multiple
enzymes in a microarray format
Dimalanta et al. (2004) 10 ml PMMA/glass/ 120 n/a n/a (i) Hybrid multiplexed detection of campylobacter DNA;
PDMS (ii) effective mechanical protection and reduced
sample evaporation
Noerholm et al. (2004) 10 ml PC 300 10 nM Temperature control Online monitoring of DNA hybridization assays
and liquid handling
Yuen et al. (2003) 50 ml PDMS/glass 120 1.25 pg/ml n/a Enhanced hybridizations by two- to five-fold through
sample fluid circulation and mixing
Chung et al. (2004) n/a PMMA/glass 30 n/a n/a Enhanced hybridization efficiencies via strain rate
n/a: not available.
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 225

Immunoassays based on solid supported lipid bilayers for multivalent


Bynum and Gordon used a microfluidic platform consisting
of a two-axis centrifuge, whose fluidic chambers rotated in a

(i) Fabrication of arrays of microwells for patterning proteins;


Polyethylene glycol-based microstructures used to immobilize

binding between surface-bound ligands and aqueous receptors


planetary relationship to produce a radial gravitational field to
produce effective mixing based on the premise that this process

cells with potential applications in cell-based biosensors


would overcome surface and viscous forces in microchambers

On chip bioassays for carrying out genotoxicity tests


(Bynum and Gordon, 2004). Yuen and co-workers have also

Low density protein arrays patterned on gold by


On-chip transformation of Escherichia coli cells

(i) Multiplexed fabrication of cell phenotypes;


developed a microchip device for closed loop fluidic circulation

(ii) selective adhering of cells onto the wells


and mixing (Liu et al., 2004b). The device consisted of two

using fluidic networks containing SAMs


interconnected reaction chambers molded in PDMS with a
standard microscope glass slide. Mixing and circulation of fluid
samples was accomplished by rotation of a magnetic stirring

Profiling expression dynamics

(ii) reduced sample volumes


bar driven by a standard magnetic stirrer (Yuen et al., 2003).

Applications/achievements

in agarose hydrogel wells


Other examples of convective mixing involve cavitational
microstreaming, which has been reported to accelerate
hybridization kinetics by nearly five-fold (Liu et al., 2003b).
In this process, tiny air-filled chambers were added to a backing
slide; the air bubbles resting on a surface were set into vibration
by a sound field generating steady circulatory flows that
resulted in convective flows producing rapid mixing and
therefore, enhancing hybridization kinetics.

cell monitoring of expression events


Accelerated hybridization achieved by mixing has also been

stimulation and non-destructive


described by Wei et al., who used fluidic channels that were

High-throughput molecular
scrambled into discrete plugs to induce droplet mixing (Wei
et al., 2005). The plugs were thoroughly mixed by the natural
re-circulating flows shuttled back and forth along the channels
sweeping over the array probes, which reduced the hybridiza-
tion times to 500 s with reduced sample volumes of 1 ml.
integrated
Functions

DNA hybridizations can also be accelerated by electro-


kinetic delivery of samples (Edman et al., 1997; Erickson et al.,
n/a

n/a

n/a

n/a
n/a

n/a

n/a
2004; Sosnowski et al., 1997). Electrokinetic transport of
targets offers the advantages of controlled fluidic flow and the
2  107 cells/ml
Abbreviated list publications outlining the coupling of protein/cell microarrays to microfluidics

capability of miniaturization without having to use pumps or

1–106 cells/ml
10–30 ng/ml
active valves. The electrokinetic control allows rapid and
Detection

230 pmol
105 cells

efficient mass transfer of samples and therefore, hybridization


1 ng/ml
limits

equilibrium is rapidly attained leading to reduced reaction


n/a

n/a
times. Fig. 4 shows an example of electrokinetic addressing of a
Analysis speed

DNA microarray.

2.4. Applications of microfluidic-based microarrays


(min)
360

n/a

n/a

n/a

n/a

n/a
60

10

A comprehensive description of applications involving the


Glass/silicon/PDMS

use of microfluidics coupled to DNA microarrays (Liu et al.,


2004a; Lilliehorn et al., 2005; Lenigk et al., 2002a; Hashimoto
Silicon/PDMS
Silicon/PDMS

et al., 2006; Wang et al., 2003; Lee et al., 2001; Benoit et al.,
Gold/PDMS

2001; Cheek et al., 2001; Koh et al., 2003; Noerholm et al.,


material
Support

PDMS

PDMS

PDMS

PDMS

2004; Chung et al., 2003; Yuen et al., 2003; Wei et al., 2005;
Erickson et al., 2004; Ali et al., 2003; Anderson et al., 2000b;
Dimalanta et al., 2004; Keramas et al., 2004) is summarized in
Sample
volume

0.2 ml

10 ml

Table 1 while those of protein and cell microarrays (Kanda


5 ml

2 ml

3 ml
n/a

n/a

n/a

et al., 2004; Tani et al., 2004; Koh et al., 2003; Khademhosseini


et al., 2004; Nagamine et al., 2005; Ostuni et al., 2001;
Thompson et al. (2004)
Nagamine et al. (2005)

Thompson et al., 2004; Yang et al., 2001) are summarized in


Ostuni et al. (2001)

Kanda et al. (2004)


Yang et al. (2001)

n/a: not available.


Tani et al. (2004)

Koh et al. (2003)

Table 2. The information provided illustrates a full range of


Khademhosseini
et al. (2004)

microarrays performed in microfluidic devices detailing the


Reference

important achievements obtained in terms of volume reduction,


Table 2

analysis speed, applications, integration, automation and limit


of detection.
226 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

A standard microarray experiment typically involves stenciling techniques are effective in building low to medium
several steps: (1) extraction/purification of the targets from density arrays. In the case of direct in situ synthesis, high
the cells following lysis; (2) in the case of mRNAs, reverse density arrays can be produced (300,000 features on a
transcription into cDNAs; (3) labeling of the solution targets 1.28 cm  1.28 cm chip of 25-mer oligonucleotide probes)
with reporter molecules, such as fluorescent dyes, which can be (Lipshutz et al., 1999).
added during PCR amplification in the case of DNAs; (4) The advantages exhibited by microfluidics, including their
incubation of the targets with the arrayed probes. The probes ability to integrate several processing steps into a fully
are patterned onto derivatized/treated substrates either by functional system, has attracted several examples where the
printing or direct synthesis as mentioned previously. Printing biological sample is fully processed in the microfluidic prior to
or spotting is accomplished through the use of robotic arrayers hybridization interrogation. Systems have been reported that
that are designed to dispense small volumes of solutions utilize microfluidics to process the biological sample and
(Cheung et al., 1999; Duggan et al., 1999) or by ink-jet printing prepare it for structure analysis using either DNA, protein or
(Okamoto et al., 2000). Other than spotting/printing, micro- cell-based microarrays. In the following sections, examples
fluidics can also be used for deposition of pre-synthesized will be given from the literature in which sample-preprocessing
probes onto supports as well via the stenciling techniques using microfluidics was invoked prior to interrogation of the
described above. In most cases, printing, spotting and targets using a microarray.

Fig. 5. (A) Conceptual schematic of the PCR/LDR/universal array assay format. The zip code probes and complements contain no sequence homology to the DNA
target being interrogated for point mutations. In the absence of a successful ligation event, LDR primers bearing zip code complements do hybridize to their zip code
probes, but produce no fluorescence signature since the LDR primer containing the fluorescent label is not present. (B) Schematic representation of the CFPCR/
CFLDR and zip code array biochips. The CFPCR/CFLDR microchip possessed channels that were 100 mm in width and 50 mm in depth with a 400 mm inter-channel
spacing. The total length of the thermal cycling channel was 2.28 m and consisted of a 30-cycle PCR (1.57 m long) and a 13-cycle LDR (0.71 m long). The top
inset represents the PMMA zip code array microfluidic chip. The bottom inset is an enlarged schematic of the Y-shaped passive micromixer for mixing the PCR
product with the LDR cocktail. Adopted with permission from Hashimoto et al. (2006).
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 227

Fig. 5. (Continued ).

2.4.1. DNA microarray systems PMMA chips were integrated into a single unit by using PDMS-
An attractive feature of microfluidic systems is their ability to assisted interconnect technology. The PC chip was used for
process the sample in a close architecture and with high levels of performing the LDR on chip while the PMMA chip was used for
automation. This provides the ability to not only reduce detection of these LDR products in a microarray format.
processing time, but also minimize sample contamination and Hydrodynamic pumping of fluids on this 3D microfluidic
simplify assay implementation and cost. Recently, a flow through network enabled an LDR/hybridization assay to be carried out
microchip assembly containing two biochips was fabricated to with applications in the detection of low-abundant point
carry out a sequential ligase detection reaction (LDR) followed mutations in K-ras genes in less that 20 min (total processing
by an hybridization assay (Hashimoto et al., 2006). The system time). This processing time was significantly reduced compared
(see Fig. 5) was used to detect rare point mutations in genomic to previous work performed, which involved carrying out a PCR/
DNA that possess high diagnostic value for colorectal cancers. LDR with array readout using conventional bench-top equip-
The LDR generates products that bear a fluorescence label and ment, which required approximately 270 min.
also, a short sequence appended to the DNA primers involved in
the ligation reaction that directs a successfully formed ligation 2.4.2. Protein microarray systems
product to a location on the array. The tethered probes are Protein arrays are based on biospecific binding, e.g. antibody/
complements to the short sequence found on one of the LDR antigen interactions. Yang et al. designed a multivalent immuno-
primers. This ‘‘universal array format’’ consisting of zip code assay for monitoring the binding of bivalent anti-dinitrophenyl
probes and zip code complements allows for the hybridization (anti-DNP) antibodies to phospholipid bilayer surfaces contain-
detection of a variety of biologically derived products. PC and ing dinitrophenyl haptens (Yang et al., 2001). These assays were
228 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

conducted inside linear arrays of bilayer-coated microchannels intended assay, which are very useful for transitioning high end
formed by placing patterned PDMS molds into conformal molecular analysis to a point-of-care format. Parallelization
contact with borosilicate cover slips. Twelve microchannels were capability of microfluidics on a single chip is also important,
arrayed onto a single chip to allow heterogeneous assays to be since different samples can be simultaneously processed with
performed rapidly with reduced protein sample volumes. highly controlled reaction conditions allowing high-throughput
analysis as well as reducing inter-sample variability. Also, the
2.4.3. Cell-based microarray systems closed architecture of the microfluidic provides reduction in
The ability of microfluidics to enhance controlled cellular possible sample contamination, which will have important
interactions and increase response times has also resulted in clinical ramifications in the area of DNA forensics. The use of
their incorporation into cell studies. Cell studies provide inexpensive polymers as the substrate for microfluidic devices
information on the dynamics of gene expression or their allows one to employ these systems as single-use devices,
morphological characteristics. Arrays of different phenotypes which will be particularly attractive to eliminate cross-
of cells can be useful in clinical diagnostics as well. Thompson contaminations during operation, eliminating false positive
et al. reported a living cell array platform for the simultaneous results due to sample carryover. Coupling microfluidics to DNA
stimulation and monitoring of gene expression in living cells arrays, protein arrays or cell arrays therefore will have a major
(Thompson et al., 2004). They used a PDMS/glass chamber impact on understanding many fundamental biological
coated with fibronectin to promote cell attachment. This activities, since these technology platforms make high end
platform was used to control dynamic inputs and measure the technologies for studying these processes available to a large
outputs from adherent mammalian cells. One advantage offered research community due to the simplicity of their operation and
by this device over conventional cell microarrays was its ability the low cost of their production.
to simultaneously screen many conditions and efficiently Another challenge associated with coupling microfluidics and
explore the large parameter space relating to stimulation of microarrays is the ability to manipulate microfluidics in a
certain cell responses. complicated fully integrated fluidic network. With this, many
Protein and cell microarrays have allowed the reduction in multi-step microarray hybridization processes that require
sample and reagent volumes and analysis speed to be achieved, manual handling can be eliminated when all the processes are
however, they are still in their infancy in terms of development integrated into a single system. These automated platforms may
and as such, are faced with several operational challenges. For eventually lead to improved reproducibility of microarray results
example, it is difficult to observe low abundant targets by direct with acceptable array-to-array coefficients of variation. Micro-
molecular association assays because the trace amount of target fluidics has also improved microarray sensitivities enabling
has to compete with the mixed population of higher abundant detection of even low expressed genes, however, array analyses
interferents for binding onto the recognition element situated still lacks an efficient data analysis tool (bioinformatics) for the
on the support (MacBeath, 2002). Also, high content analysis of generated microarray information. Improved bioinformatics
cell microarrays in living cells require the imaging time of an tools and fully integrated microfluidic devices will therefore
entire slide at sufficient time-resolution so as not to obscure enable microarrays to be fully exploited in gene expression
transitory phenotypes of interest (Wheeler et al., 2005). analysis and clinical diagnostics with reduced analysis time.
While microfluidics can offer some attractive advantages for
3. Conclusions microarray-based assays, these systems still depend on
conventional peripheral equipment for their operation, such
Microfluidics has advanced the rapid growth in biochip as sample/reagent loading, fluid actuation and readout. It is
applications due in part to their ability to pre-process the sample clear that complete packaging of these peripherals into a system
in an automated fashion prior to incubation with the array and that is on the same size as the fluidic element will allow the
the fact that the operational characteristics of the array can be creation of instruments that can be envisioned for potential
improved when scaled to the micrometer domain. One point-of-care testing. This realization will provide increased
particularly compelling advantage associated with the marriage accessibility of microarray technologies to a potentially larger
of microfluidics with microarrays is the reduction in reaction user base and expand on their capabilities.
times exhibited in all applications ranging from DNA arrays,
protein arrays and cell bioassays. The reduced reaction times Acknowledgements
are attributed to improved kinetics compared to conventional
non-microfluidic techniques and often times is a consequence The authors would like to thank the financial support of this
of enhanced mass transport to the surface tethered recognition work through the US National Institutes of Health (R01-
elements by the target and small diffusional distances. EB00215), the US National Science Foundation (EPS-
Successful integration of sample pre-treatment processes, such 0346411) and the State of Louisiana Board of Regents.
as cell culture/lysis, sample purification, pre-concentration and
amplification, with the hybridization reaction on a single References
microfluidic platform has shown significant reduction in
analysis time, improved sensitivities, fully automated sample Ahn, S., Walt, D.R., 2005. Detection of Salmonella spp. Using microsphere-
processing, and the reduction in hardware demands for the based, fiber-optic DNA microarrays. Anal. Chem. 77 (15), 5041–5047.
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 229

Ali, M.F., Kirby, R., Goodey, A.P., Rodriguez, M.D., Ellington, A.D., Neikirk, Erali, M., Schmidt, B., Lyon, E., Wittwer, C., 2003. Evaluation of electronic
D.P., McDevitt, J.T., 2003. DNA hybridization and discrimination of single- microarrays for genotyping factor V, factor II, and MTHFR. Clin. Chem. 49
nucleotide mismatches using chip-based microbead arrays. Anal. Chem. 75 (5), 732–739.
(18), 4732–4739. Erickson, D., Liu, X., Krull, U., Li, D., 2004. Electrokinetically controlled DNA
Anderson, J.R., Chiu, D.T., Jackman, R.J., Cherniavskaya, O., McDonald, J.C., hybridization microfluidic chip enabling rapid target analysis. Anal. Chem.
Wu, H., Whitesides, S.H., Whitesides, G.M., 2000a. Fabrication of topo- 76 (24), 7269–7277.
logically complex three-dimensional microfluidic systems in PDMS by Fan, Z.H., Mangru, S., Granzow, R., Heaney, P., Ho, W., Dong, Q., Kumar, R.,
rapid prototyping. Anal. Chem. 72 (14), 3158–3164. 1999. Dynamic DNA hybridization on a chip using paramagnetic beads.
Anderson, R.C., Su, X., Bogdan, G.J., Fenton, J., 2000b. A miniature integrated Anal. Chem. 71 (21), 4851–4859.
device for automated multistep genetic assays. Nucleic Acids Res. 28 (12) Fixe, F., Dufva, M., Telleman, P., Christensen, C.B.V., 2004a. Functionalization
e60, ii–vi. of poly(methyl methacrylate) (PMMA) as a substrate for DNA microarrays.
Auroux, P.-A., Iossifidis, D., Reyes, D.R., Manz, A., 2002. Micro-total analysis Nucleic Acids Res. 32 (1), e9/1–e9/8.
systems. 2. Analytical standard operations and applications. Anal. Chem. 74 Fixe, F., Dufva, M., Telleman, P., Christensen, C.B.V., 2004b. One-step
(12), 2637–2652. immobilization of aminated and thiolated DNA onto poly(methylmetha-
Baner, J., Isaksson, A., Waldenstroem, E., Jarvius, J., Landegren, U., Nilsson, crylate) (PMMA) substrates. Lab. Chip 4 (3), 191–195.
M., 2003. Parallel gene analysis with allele-specific padlock probes and tag Fodor, S.P.A., Read, J.L., Pirrung, M.C., Stryer, L., Lu, A.T., Solas, D., 1991.
microarrays. Nucleic Acids Res. 31 (17) e103/1-e03/7. Light-directed, spatially addressable parallel chemical synthesis. Science
Becker, H., Gartner, C., 2000. Polymer microfabrication methods for micro- (Washington, DC, United States) 251 (24), 767–773.
fluidic analytical applications. Electrophoresis 21 (1), 12–26. Ford, S.M., Davies, J., Kar, B., Qi, S.D., McWhorter, S., Soper, S.A., Malek,
Benoit, V., Steel, A., Torres, M., Yu, Y.-Y., Yang, H., Cooper, J., 2001. C.K., 1999. Micromachining in plastics using X-ray lithography for the
Evaluation of three-dimensional microchannel glass biochips for multi- fabrication of microelectrophoresis devices. J. Biomech. Eng. 121 (1), 13–
plexed nucleic acid fluorescence hybridization assays. Anal. Chem. 73 (11), 21.
2412–2420. Friedrich, C.R., Coane, P.J., Vasile, M.J., 1997. Micromilling development and
Boerner, M.W., Kohl, M., Pantenburg, F.J., Bacher, W., Hein, H., Schomburg, applications for microfabrication. Microelectron. Eng. 35 (1-4, Micro- and
W.K., 1996. Sub-micron LIGA process for movable microstructures. Nano-Engineering 96), 367–372.
Microelectron. Eng. 30 (1–4), 505–508. Fu, A.Y., Chou, H.-P., Spence, C., Arnold, F.H., Quake, S.R., 2002.
Burns, M.A., Johnson, B.N., Brahmasandra, S.N., Handique, K., Webster, J.R., An integrated microfabricated cell sorter. Anal. Chem. 74 (11), 2451–
Krishnan, M., Sammarco, T.S., Man, P.M., Jones, D., Heldsinger, D., 2457.
Mastrangelo, C.H., Burke, D.T., 1998. An integrated nanoliter DNA Gingeras, T.R., Kwoh, D.Y., Davis, G.R., 1987. Hybridization properties of
analysis device. Science 282 (5388), 484–487. immobilized nucleic acids. Nucleic Acids Res. 15 (13), 5373–5390.
Burns, M.A., Mastrangelo, C.H., Sammarco, T.S., Man, F.P., Webster, J.R., Gong, Y.-K., Luo, L., Petit, A., Zukor, D.J., Huk, O.L., Antoniou, J., Winnik,
Johnsons, B.N., Foerster, B., Jones, D., Fields, Y., Kaiser, A.R., Burke, D.T., F.M., Mwale, F., 2005. Adhesion of human U937 macrophages to phos-
1996. Microfabricated structures for integrated DNA analysis. Proc. Natl. phorylcholine-coated surfaces. J. Biomed. Mater. Res. 72A (1), 1–9.
Acad. Sci. U.S.A. 93 (11), 5556–5561. Grass, B., Neyer, A., Johnck, M., Siepe, D., Eisenbeiss, F., Weber, G.,
Bussow, K., Nordhoff, E., Lubbert, C., Lehrach, H., Walter, G., 2000. A human Hergenroder, R., 2001. A new PMMA-microchip device for isotachophor-
cDNA library for high-throughput protein expression screening. Genomics esis with integrated conductivity detector. Sens. Actuators B 72 (3), 249–
65 (1), 1–8. 258.
Bynum, M.A., Gordon, G.B., 2004. Hybridization enhancement using micro- Grimm, V., Ezaki, S., Susa, M., Knabbe, C., Schmid, R.D., Bachmann, T.T.,
fluidic planetary centrifugal mixing. Anal. Chem. 76 (23), 7039–7044. 2004. Use of DNA microarrays for rapid genotyping of TEM beta-lacta-
Cheek, B.J., Steel, A.B., Torres, M.P., Yu, Y.-Y., Yang, H., 2001. Chemilumi- mases that confer resistance. J. Clin. Microbiol. 42 (10), 4918 [Erratum to
nescence detection for hybridization assays on the flow-thru chip, a three- document cited in CA142:087118].
dimensional microchannel biochip. Anal. Chem. 73 (24), 5777–5783. Halliwell, C.M., Cass, A.E.G., 2001. A factorial analysis of silanization
Cheung, V.G., Morley, M., Aguilar, F., Massimi, A., Kucherlapati, R., Childs, conditions for the immobilization of oligonucleotides on glass surfaces.
G., 1999. Making and reading microarrays. Nat. Genet. 21 (Suppl. 1), 15– Anal. Chem. 73 (11), 2476–2483.
19. Hashimoto, M., Barany, F., Soper, S.A., 2006. Polymerase chain reaction/ligase
Chung, Y.-C., Lin, Y.-C., Hsu, Y.-L., Chang, W.-N.T., Shiu, M.-Z., 2004. The detection reaction/hybridization assays using flow-through microfluidic
effect of velocity and extensional strain rate on enhancing DNA hybridiza- devices for the detection of low-abundant DNA point mutations. Biosens.
tion. J. Micromech. Microeng. 14 (10), 1376–1383. Bioelectron. 21, 1915–1923.
Chung, Y.-C., Lin, Y.-C., Shiu, M.-Z., Chang, W.-N.T., 2003. Microfluidic chip He, B., Tan, L., Regnier, F., 1999. Microfabricated filters for microfluidic
for fast nucleic acid hybridization. Lab. Chip 3 (4), 228–233. analytical systems. Anal. Chem. 71 (7), 1464–1468.
Csaki, A., Moller, R., Straube, W., Kohler, J.M., Fritzsche, W., 2001. DNA Henry, A.C., Tutt, T.J., Galloway, M., Davidson, Y.Y., McWhorter, C.S., Soper,
monolayer on gold substrates characterized by nanoparticle labeling and S.A., McCarley, R.L., 2000. Surface modification of poly(methyl metha-
scanning force microscopy. Nucleic Acids Res. 29 (16), e81/1–e81/5. crylate) used in the fabrication of microanalytical devices. Anal. Chem. 72
Dimalanta, E.T., Lim, A., Runnheim, R., Lamers, C., Churas, C., Forrest, D.K., (21), 5331–5337.
de Pablo, J.J., Graham, M.D., Coppersmith, S.N., Goldstein, S., Schwartz, Horvath, J., Dolnik, V., 2006. Polymer wall coatings for capillary electrophor-
D.C., 2004. A microfluidic system for large DNA molecule arrays. Anal. esis. Electrophoresis 22, 644–655.
Chem. 76 (18), 5293–5301. Hu, S., Ren, X., Bachman, M., Sims, C.E., Li, G.P., Allbritton, N.L., 2004.
Duffy, D.C., McDonald, J.C., Schueller, O.J.A., Whitesides, G.M., 1998. Rapid Surface-directed, graft polymerization within microfluidic channels. Anal.
prototyping of microfluidic systems in poly(dimethylsiloxane). Anal. Chem. 76 (7), 1865–1870.
Chem. 70 (23), 4974–4984. Ihlemann, J., Rubahn, K., 2000. Excimer laser micro-machining: fabrica-
Duffy, D.C., Schueller, O.J.A., Brittain, S.T., Whitesides, G.M., 1999. Rapid tion and applications of dielectric masks. Appl. Surf. Sci. 154–155,
prototyping of microfluidic switches in poly(dimethylsiloxane) and their 587–592.
actuation by electro-osmotic flow. J. Micromech. Microeng. 9 (3), 211–217. Ishikawa, S., Komura, D., Tsuji, S., Nishimura, K., Yamamoto, S., Panda, B.,
Duggan, D.J., Bittner, M., Chen, Y., Meltzer, P., Trent, J.M., 1999. Expression Huang, J., Fukayama, M., Jones, K.W., Aburatani, H., 2005. Allelic dosage
profiling using cDNA microarrays. Nat. Genet. 21 (Suppl 1), 10–14. analysis with genotyping microarrays. Biochem. Biophys. Res. Commun.
Edman, C.F., Raymond, D.E., Wu, D.J., Tu, E., Sosnowski, R.G., Butler, W.F., 333 (4), 1309–1314.
Nerenberg, M., Heller, M., 1997. Electric field directed nucleic acid Joos, B., Kuster, H., Cone, R., 1997. Covalent attachment of hybridizable
hybridization on microchips. Nucleic Acids Res. 25 (24), 4907–4914. oligonucleotides to glass supports. Anal. Biochem. 247 (1), 96–101.
230 C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231

Kanda, V., Kariuki, J.K., Harrison, D.J., McDermott, M.T., 2004. Label-free capture and type-specific primer extension. J. Clin. Microbiol. 41 (11),
reading of microarray-based immunoassays with surface plasmon reso- 5153–5158.
nance imaging. Anal. Chem. 76 (24), 7257–7262. Lu, H.H., Cooper, J.A., Manuel, S., Freeman, J.W., Attawia, M.A., Ko, F.K.,
Kapur, R., 2005. Whole cell microarrays. Microarray Technol. Appl. 335–343. Laurencin, C.T., 2005. Anterior cruciate ligament regeneration using
Keramas, G., Perozziello, G., Geschke, O., Christensen, C.B.V., 2004. Devel- braided biodegradable scaffolds: in vitro optimization studies. Biomaterials
opment of a multiplex microarray microsystem. Lab. Chip 4 (2), 152–158. 26 (23), 4805–4816.
Khademhosseini, A., Yeh, J., Jon, S., Eng, G., Suh, K.Y., Burdick, J.A., Langer, MacBeath, G., 2002. Protein microarrays and proteomics. Nat. Genet. Suppl.
R., 2004. Molded polyethylene glycol microstructures for capturing cells 32, 526–532.
within microfluidic channels. Lab. Chip 4 (5), 425–430. MacBeath, G., Schreiber, S.L., 2000. Printing proteins as microarrays for high-
Kim, H.-L., 2003. Comparison of oligonucleotide-microarray and serial ana- throughput function determination. Science (Washington, DC) 289 (5485),
lysis of gene expression (SAGE) in transcript profiling analysis of mega- 1760–1763.
karyocytes derived from CD34+ cells. Exp. Mol. Med. 35 (5), 460–466. Marquette, C.A., Blum, L.J., 2004. Direct immobilization in poly(dimethylsi-
Koh, W.-G., Itle, L.J., Pishko, M.V., 2003. Molding of hydrogel microstructures loxane) for DNA, protein and enzyme fluidic biochips. Anal. Chim. Acta
to create multiphenotype cell microarrays. Anal. Chem. 75 (21), 5783– 506 (2), 127–132.
5789. Martynova, L., Locascio, L.E., Gaitan, M., Kramer, G.W., Christensen, R.G.,
Kohara, Y., Noda, H., Okano, K., Kambara, H., 2002. DNA probes on beads MacCrehan, W.A., 1997. Fabrication of plastic microfluid channels by
arrayed in a capillary, bead-array, exhibited high hybridization perfor- imprinting methods. Anal. Chem. 69 (23), 4783–4789.
mance. Nucleic Acids Res. 30 (16), e87/1–e87/7. McCarley, R.L., Vaidya, B., Wei, S., Smith, A.F., Patel, A.B., Feng, J., Murphy,
Lebrun, S.J., 2005. The use of microarrays for highly sensitive, multiplex M.C., Soper, S.A., 2005. Resist-free patterning of surface architectures in
immunoassays in biomarker discovery and drug efficacy studies. Pharma- polymer-based microanalytical devices. J. Am. Chem. Soc. 127 (3), 842–
ceut. Discov. Suppl. 53–56. 843.
Lee, H.J., Goodrich, T.T., Corn, R.M., 2001. SPR imaging measurements of 1-D McClain, M.A., Culbertson, C.T., Jacobson, S.C., Allbritton, N.L., Sims, C.E.,
and 2-D DNA microarrays created from microfluidic channels on gold thin Ramsey, J.M., 2003. Microfluidic devices for the high-throughput chemical
films. Anal. Chem. 73 (22), 5525–5531. analysis of cells. Anal. Chem. 75 (21), 5646–5655.
Lee, K.-B., Yoon, K.R., Woo, S.I., Choi, I.S., 2003. Surface modification of McDonald, J.C., Whitesides, G.M., 2002. Poly(dimethylsiloxane) as a material
poly(glycolic acid) (PGA) for biomedical applications. J. Pharm. Sci. 92 (5), for fabricating microfluidic devices. Acc. Chem. Res. 35 (7), 491–499.
933–937. Meaburn, E., Butcher, L.M., Schalkwyk, L.C., Plomin, R., 2006. Genotyping
Lenigk, R., Liu, R.H., Athavale, M., Chen, Z., Ganser, D., Yang, J., Rauch, C., pooled DNA using 100 K SNP microarrays: a step towards genome wide
Liu, Y., Chan, B., Yu, H., Ray, M., Marrero, R., Grodzinski, P., 2002a. association scans. Nucleic Acids Res. 34 (4), e27.
Plastic biochannel hybridization devices: a new concept for microfluidic Moorcroft, M.J., Meuleman, W.R.A., Latham, S.G., Nicholls, T.J., Egeland,
DNA arrays. Anal. Biochem. 311 (1), 40–49. R.D., Southern, E.M., 2005. In situ oligonucleotide synthesis on poly(-
Lenigk, R., Liu, R.H., Athavale, M., Chen, Z.J., Ganser, D., Yang, J.N., Rauch, dimethylsiloxane): a flexible substrate for microarray fabrication. Nucleic
C., Liu, Y.J., Chan, B., Yu, H.N., Ray, M., Marrero, R., Grodzinski, P., Acids Res. 33 (8), e75/1–e75/10.
2002b. Plastic biochannel hybridization devices: a new concept for micro- Nagamine, K., Onodera, S., Torisawa, Y., Yasukawa, T., Shiku, H., Matsue, T.,
fluidic DNA arrays. Anal. Biochem. 311 (1), 40–49. 2005. On-chip transformation of bacteria. Anal. Chem. 77 (13), 4278–4281.
Li, J., Thibault, P., Bings, N.H., Skinner, C.D., Wang, C., Colyer, C., Harrison, Noerholm, M., Bruus, H., Jakobsen, M.H., Telleman, P., Ramsing, N.B., 2004.
J., 1999. Integration of microfabricated devices to capillary electrophoresis– Polymer microfluidic chip for online monitoring of microarray hybridiza-
electrospray mass spectrometry using a low dead volume connection: tions. Lab. Chip 4 (1), 28–37.
application to rapid analyses of proteolytic digests. Anal. Chem. 71 (15), Nuzzo, R.G., 2003. Biomaterials: stable antifouling surfaces. Nat. Mater. 2 (4),
3036–3045. 207–208.
Lilliehorn, T., Nilsson, M., Simu, U., Johansson, S., Almqvist, M., Nilsson, J., Okamoto, T., Suzuki, T., Yamamoto, N., 2000. Microarray fabrication with
Laurell, T., 2005. Dynamic arraying of microbeads for bioassays in micro- covalent attachment of DNA using bubble jet technology. Nat. Biotechnol.
fluidic channels. Sens. Actuators B 106 (2), 851–858. 18 (4), 438–441.
Lin, M., Wei, L.-J., Sellers, W.R., Lieberfarb, M., Wong, W.H., Li, C., 2004. Ostuni, E., Chen, C.S., Ingber, D.E., Whitesides, G.M., 2001. Selective
dChipSNP: significance curve and clustering of SNP-array-based loss-of- deposition of proteins and cells in arrays of microwells. Langmuir 17
heterozygosity data. Bioinformatics 20 (8), 1233–1240. (9), 2828–2834.
Lipshutz, R.J., Fodor, S.P.A., Gingeras, T.R., Lockhart, D.J., 1999. High density Pal, R., Yang, M., Lin, R., Johnson, B.N., Srivastava, N., Razzacki, S.Z.,
synthetic oligonucleotide arrays. Nat. Genet. 21 (Suppl. 1), 20–24. Chomistek, K.J., Heldsinger, D.C., Haque, R.M., Ugaz, V.M., Thwar,
Liu, J., Hansen, C., Quake, S.R., 2003a. Solving the ‘‘world-to-chip’’ interface P.K., Chen, Z., Alfano, K., Yim, M.B., Krishnan, M., Fuller, A.O.,
problem with a microfluidic matrix. Anal. Chem. 75 (18), 4718–4723. Larson, R.G., Burke, D.T., Burns, M.A., 2005. An integrated micro-
Liu, R.H., Lenigk, R., Druyor-Sanchez, R.L., Yang, J., Grodzinski, P., 2003b. fluidic device for influenza and other genetic analyses. Lab. Chip 5 (10),
Hybridization enhancement using cavitation microstreaming. Anal. Chem. 1024–1032.
75 (8), 1911–1917. Pease, A.C., Solas, D., Sullivan, E.J., Cronin, M.T., Holmes, C.P., Fodor, S.P.,
Liu, Y., Rauch, C.B., 2003. DNA probe attachment on plastic surfaces and 1994. Light-generated oligonucleotide arrays for rapid DNA sequence
microfluidic hybridization array channel devices with sample oscillation. analysis. Proc. Natl. Acad. Sci. U.S.A. 91 (11), 5022–5026.
Anal. Biochem. 317 (1), 76–84. Peterson, A.W., Heaton, R.J., Georgiadis, R.M., 2001. The effect of surface
Liu, R.H., Yang, J., Lenigk, R., Bonanno, J., Grodzinski, P., 2004a. Self- probe density on DNA hybridization. Nucleic Acids Res. 29 (24), 5163–
contained, fully integrated biochip for sample preparation, polymerase 5168.
chain reaction amplification, and DNA microarray detection. Anal. Chem. Postier Bradley, L., Wang, H.-L., Singh, A., Impson, L., Andrews Heather, L.,
76 (7), 1824–1831. Klahn, J., Li, H., Risinger, G., Pesta, D., Deyholos, M., Galbraith David, W.,
Liu, J., Walker, N., Waalkes, M.P., 2004b. Hybridization buffer systems impact Sherman Louis, A., Burnap Robert, L., 2003. The construction and use of
the quality of filter array data. J. Pharmacol. Toxicol. Methods 50 (1), 67– bacterial DNA microarrays based on an optimized two-stage PCR strategy.
71. BMC Genom. Electr. Resour. 4 (1), 23.
Love, J.C., Wolfe, D.B., Jacobs, H.O., Whitesides, G.M., 2001. Microscope Qi, S., Liu, X., Ford, S., Barrows, J., Thomas, G., Kelly, K., McCandless, A.,
projection photolithography for rapid prototyping of masters with micron- Lian, K., Goettert, J., Soper, S.A., 2002. Microfluidic devices fabricated
scale features for use in soft lithography. Langmuir 17 (19), 6005–6012. in poly(methyl methacrylate) using hot-embossing with integrated sam-
Lovmar, L., Fock, C., Espinoza, F., Bucardo, F., Syvanen, A.-C., Bondeson, K., pling capillary and fiber optics for fluorescence detection. Lab. Chip 2
2003. Microarrays for genotyping human group A rotavirus by multiplex (2), 88–95.
C. Situma et al. / Biomolecular Engineering 23 (2006) 213–231 231

Ressine, A., Ekstrom, S., Marko-Varga, G., Laurell, T., 2003. Macro-/nanopor- Tani, H., Maehana, K., Kamidate, T., 2004. Chip-based bioassay using bacterial
ous silicon as a support for high-performance protein microarrays. Anal. sensor strains immobilized in three-dimensional microfluidic network.
Chem. 75 (24), 6968–6974. Anal. Chem. 76 (22), 6693–6697.
Reyes, D.R., Iossifidis, D., Auroux, P.-A., Manz, A., 2002. Micro-total analysis Thompson, D.M., King, K.R., Wieder, K.J., Toner, M., Yarmush, M.L., Jayara-
systems. 1. Introduction, theory, and technology. Anal. Chem. 74 (12), man, A., 2004. Dynamic gene expression profiling using a microfabricated
2623–2636. living cell array. Anal. Chem. 76 (14), 4098–5103.
Roberts, M.A., Rossier, J.S., Bercier, P., Girault, H., 1997. UV laser machined Unger, M.A., Chou, H.-P., Thorsen, T., Scherer, A., Quake, S.R., 2000.
polymer substrates for the development of microdiagnostic systems. Anal. Monolithic microfabricated valves and pumps by multilayer soft lithogra-
Chem. 69 (11), 2035–2042. phy. Science (Washington, DC) 288 (5463), 113–116.
Rossier, J.S., Schwarz, A., Reymond, F., Ferrigno, R., Bianchi, F., Girault, H.H., Vanderhoeven, J., Pappaert, K., Dutta, B., Vanhummelen, P., Baron, G.V.,
1999. Microchannel networks for electrophoretic separations. Electrophor- Desmet, G., 2004. Exploiting the benefits of miniaturization for the
esis 20 (4–5), 727–731. enhancement of DNA microarrays. Electrophoresis 25 (21–22), 3677–3686.
Sato, K., Tokeshi, M., Kimura, H., Kitamori, T., 2001. Determination of Waddell, E., Wang, Y., Stryjewski, W., McWhorter, S., Henry, A.C., Evans, D.,
carcinoembryonic antigen in human sera by integrated bead-bed immu- McCarley, R.L., Soper, S.A., 2000. High-resolution near-infrared imaging
noassay in a microchip for cancer diagnosis. Anal. Chem. 73 (6), 1213– of DNA microarrays with time-resolved acquisition of fluorescence life-
1218. times. Anal. Chem. 72 (24), 5907–5917.
Sebra, R.P., Masters, K.S., Bowman, C.N., Anseth, K.S., 2005. Surface grafted Wang, Y., Vaidya, B., Farquar, H.D., Stryjewski, W., Hammer, R.P., McCarley,
antibodies: controlled architecture permits enhanced antigen detection. R.L., Soper, S.A., Cheng, Y.-W., Barany, F., 2003. Microarrays assembled in
Langmuir 21 (24), 10907–10911. microfluidic chips fabricated from poly(methyl methacrylate) for the detec-
Shadpour, H., Musyimi, H., Chen, J., Soper, S.A., 2006. Physiochemical tion of low-abundant DNA mutations. Anal. Chem. 75 (5), 1130–1140.
properties of various polymer substrates and their effects on microchip Wei, C.-W., Cheng, J.-Y., Huang, C.-T., Yen, M.-H., Young, T.-H., 2005. Using
electrophoresis performance. J. Chromatogr. A 1111 (2), 238–251. a microfluidic device for 1 ml DNA microarray hybridization in 500 s.
Sia, S.K., Whitesides, G.M., 2003. Microfluidic devices fabricated in poly(- Nucleic Acids Res. 33 (8), e78.
dimethylsiloxane) for biological studies. Electrophoresis 24 (21), 3563– Wheeler, D.B., Carpenter, A.E., Sabatini, D.M., 2005. Cell microarrays and
3576. RNA interference chip away at gene function. Nat. Genet. 37 (Suppl.), S25–
Situma, C., Wang, Y., Hupert, M., Barany, F., McCarley, R.L., Soper, S.A., S30.
2005. Fabrication of DNA microarrays onto poly(methyl methacrylate) with Wu, R.Z., Bailey, S.N., Sabatini, D.M., 2002. Cell-biological applications of
ultraviolet patterning and microfluidics for the detection of low-abundant transfected-cell microarrays. Trends Cell Biol. 12 (10), 485–488.
point mutations. Anal. Biochem. 340 (1), 123–135. Xu, H., Sha, M.Y., Wong, E.Y., Uphoff, J., Xu, Y., Treadway, J.A., Truong, A.,
Soper, S.A., Ford, S.M., Qi, S., McCarley, R.L., Kelly, K., Murphy, M.C., 2000. O’Brien, E., Asquith, S., Stubbins, M., Spurr, N.K., Lai, E.H., Mahoney, W.,
Polymeric microelectromechanical systems. Anal. Chem. 72 (19), 643A– 2003. Multiplexed SNP genotyping using the Qbead system: a quantum dot-
651A. encoded microsphere-based assay. Nucleic Acids Res. 31 (8), e43/1–e43/10.
Sosnowski, R.G., Tu, E., Butlter, W.F., O’Connell, J.P., Heller, M.J., 1997. Yang, T., Jung, S.-Y., Mao, H., Cremer, P.S., 2001. Fabrication of phospholipid
Rapid determination of single base mismatch mutations in DNA hybrids by bilayer-coated microchannels for on-chip immunoassays. Anal. Chem. 73
direct electric field control. Proc. Natl. Acad. Sci. U.S.A. 94 (4), 1119–1123. (2), 165–169.
Southern, E.M., Case-Green, S.C., Elder, J.K., Johnson, M., Mir, K.U., Wang, Yaralioglu, Goksen, G., Wygant, Ira, O., Marentis, Theodore, C., Khuri-Yakub,
L., Williams, J.C., 1994. Arrays of complementary oligonucleotides for Butrus, T., 2004. Ultrasonic mixing in microfluidic channels using inte-
analyzing the hybridization behavior of nucleic acids. Nucleic Acids Res. grated transducers. Anal. Chem. 76 (13), 3694–3698.
22 (8), 1368–1373. Yuen, P.K., Li, G., Bao, Y., Mueller, U.R., 2003. Microfluidic devices for fluidic
Steel, A.B., Levicky, R.L., Herne, T.M., Tarlov, M.J., 2000. Immobilization of circulation and mixing improve hybridization signal intensity on DNA
nucleic acids at solid surfaces: effect of oligonucleotide length on layer arrays. Lab. Chip 3 (1), 46–50.
assembly. Biophys. J. 79 (2), 975–981. Zhang, Y., Coyne, M.Y., Will, S.G., Levenson, C.H., Kawasaki, E.S., 1991.
Su, J., Bringer, M.R., Ismagilov, R.F., Mrksich, M., 2005. Combining micro- Single-base mutational analysis of cancer and genetic diseases using
fluidic networks and peptide arrays for multi-enzyme assays. J. Am. Chem. membrane bound modified oligonucleotides. Nucleic Acids Res. 19 (14),
Soc. 127 (20), 7280–7281. 3929–3933.

You might also like