首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 31 毫秒
1.
We present a hybrid magnetic/size-sorting (HMSS) chip for isolation and molecular analyses of circulating tumor cells (CTCs). The chip employs both negative and positive cell selection in order to provide high throughput, unbiased CTC enrichment. Specifically, the system utilizes a self-assembled magnet to generate high magnetic forces and a weir-style structure for cell sorting. The resulting device thus can perform multiple functions, including magnetic depletion, size-selective cell capture, and on-chip molecular staining. With such capacities, the HMSS device allowed one-step CTC isolation and single cell detection from whole blood, tested with spiked cancer cells. The system further facilitated the study of individual CTCs for heterogeneity in molecular marker expression.Circulating tumor cells (CTCs) have emerged as an important biomarker in clinical practice as well as in fundamental research.1, 2 CTCs, shed from primary tumors, have been shown to be an early harbinger of tumor expansion and metastasis3 and have been used to predict disease progression, response to treatment, relapse, and overall survival.4, 5, 6 Recent work has shown that CTCs display distinct proteomic and genetic profiles; for example, CTCs in pancreatic cancer, have increased RNA expression of Wnt, implicating this pathway in metastasis.7 Proteomic characterization of proliferative markers such as Ki-67, and hormonal markers such as androgen receptor in prostate cancer, also have been shown to be predictive of treatment outcome.8, 9Despite such clinical potential of CTCs, their routine detection and characterization still remains a significant technical challenge.10 The task requires screening of a large number of cells (e.g., > 107 cells in 10 ml blood) and enrichment of heterogeneous targets against a complex biological background. Two main methods of CTC isolation are typically used: positive and negative selection. In positive selection, CTCs are directly isolated from blood via size-based filtration11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or antibody-based capture.1, 8, 21 Negative depletion reduces abundant blood cells, often by immunomagnetic separation, for downstream CTC enrichment.22 Both approaches have been used for high throughput CTC isolation from whole blood (SI Table 1).23 Each method, however, has its own inherent limitations. Positive enrichment could be biased by its selection criteria (e.g., cell size and cell surface markers). Negative selection, albeit unbiased, often requires complex sample processing (e.g., multiple washing steps for CTC isolation) that could result in cell loss.We hypothesized that both positive and negative selection could be combined in a single platform to enable (1) highly efficient and unbiased CTC purification, and (2) in-situ molecular analyses of collected cells. As a proof-of-concept, we herein describe a hybrid magnetic/size-sorting (HMSS) system that integrates magnetic and size-based isolation into a compact microfluidic chip. The HMSS first uses a magnetic filter to deplete leukocytes through immunomagnetic capture. Samples then pass through a size-sorter region that traps individual cells at predefined locations. Since abundant leukocytes are removed by the magnetic filter, the size-sorter could have a low size cut-off (∼5 μm), which allows for the unbiased capture of even small cancer cells. Furthermore, molecular probes can be introduced to perform on-chip, multiplexed analyses at single-cell resolution. We evaluated the utility of the developed system by capturing and profiling tumor cells in whole blood. The HMSS offers the advantages of both negative and positive selection and thereby differs from the recently reported iChip system24 which can operate only in either a negative or a positive selection mode.  相似文献   

2.
Large-library fluorescent molecular arrays remain limited in sensitivity (1 × 106 molecules) and dynamic range due to background auto-fluorescence and scattering noise within a large (20–100 μm) fluorescent spot. We report an easily fabricated silica nano-cone array platform, with a detection limit of 100 molecules and a dynamic range that spans 6 decades, due to point (10 nm to 1 μm) illumination of preferentially absorbed tagged targets by singular scattering off wedged cones. Its fluorescent spot reaches diffraction-limited submicron dimensions, which are 104 times smaller in area than conventional microarrays, with comparable reduction in detection limit and amplification of dynamic range.Commercially available fluorescent micro-arrays based on target labeling, northern blot, or enzyme-linked immunosorbent assay (ELISA) are limited to a detection threshold of 1 to 10 × 106 molecules per fluorescent spot,1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23 thus requiring cell culturing or Polymerase Chain Reaction (PCR) amplification for many applications. The low sensitivity is often due to broad illumination, which creates auto-fluorescence noise. Even if point illumination and pin-hole filtering of non-focal plane noise are implemented in a confocal setup, the large and non-uniform fluorescent spots create scattering noise over each 20–100 μm element, which degrades the detection limit.4 Smaller spots can, in theory, be introduced by nano-sprays and nano-imprinting. However, directing the targets to such small areas then becomes problematic. Real-time PCR is, in principle, capable of detecting a single molecule but is limited in its target number5 and is hence slow/expensive for large-library assays. A large-library platform with much better detection limit than the current fluorescent microarrays would transform many screening assays. Ideally, this platform would not use the confocal configuration. Instead, it would direct the target molecules to a submicron spot and illuminate them with a nearby point source that does not require scanning.A promising platform is the optical fiber bundle array,6 with more than 104 fibers and targets, in principle. With its endoscopic configuration, these fiber bundles are most convenient for in situ and real-time biosensing modalities in microfluidic biochips and microfluidic 3-D cell cultures. Consequently, the optical sensing is typically carried out in the transmission mode, with the optical signals transmitted through the optical fibers to a detector. Microwell arrays at the distal end of imaging fiber, with molecular targets captured and transported to the microwells by microbeads, are the most popular among these optical fiber arrays. Although detection limit better than 1 × 106 molecules per bead has been reported, the bar-coded beads limit the target number of this platform.7, 8Our previous work9, 10 has shown that plasmonics at nanotips can enhance local electric field by three orders of magnitude. However, conduction loss and quenching of fluorescence11, 12 by the metal substrates limit the use of plasmonic enhanced fluorescence for large-library assays. Only nano-molar sensitivity has been demonstrated using plasmonics from metal coated nanocone tips.13, 14 In this paper, we will extend the conical fiber array platform not by tip plasmonics but by another optical phenomenon with induced dipoles: singular scattering off dielectric wedges and tips.15 Instead of the surface plasmon resonance on metallic nanostructures,16 field focusing at the cone tip by the dielectric media (the silica fiber) is used to produce a localized and singularly large scattering intensity at the tip. Singular scattering from a wedge or a cone has been known for decades.17, 18 It is only recently that numerical simulation19 has revealed that field focusing by this singular scattering can effect a five-order intensity enhancement that is frequency independent. This intense tip scattering produces a local light source at the tip that does not suffer from conduction loss. Unlike plasmonic metal nanostructures, the dielectric tip would also not quench the fluorescent reporters excited by the light source. In fact, it will help scatter the fluorescent signal, with Rayleigh scattering intensity scaling with respect to wavelength. We hence utilize this phenomenon for diffraction-limit fluorescent sensing/imaging for the first time here.The local light source due to tip scattering minimizes background auto-fluorescence and scattering noise, provided the target molecules preferentially diffuse towards the dielectric vertices. If the targets do not preferentially hybridize with probes at the vertices, there would be significant target loss, with a concomitant loss in sensitivity, because the vertex regions are just a small fraction of the total area. Fortunately, like electromagnetic radiation at the electrostatic limit of the Maxwell equations for sharp (sub-wavelength) vertices,20 the steady-state diffusion of molecules also obey the Laplace equation and so do the DC or AC electric potentials that drive electrophoresis and dielectrophoresis of the molecules.21 Hence, the diffusive, electrophoretic, and dielectrophoretic fluxes of target molecules are also singularly large at the vertices and there will be preferential hybridization there until the tip is saturated. Previously, we have demonstrated preferential diffusive transport of colloids to channel corners22 and dieletrophoretic trapping of bacteria23 and DNA molecules24 around sharp nanostructures like carbon nanotubes. Hence, dielectric nanotips fabricated by low-cost techniques can potentially provide the smallest fluorescent spot, which can preferentially capture target molecules and whose fluorescent image is limited in size only by the diffraction limit, without a confocal configuration.Although the scattering singularity is stronger at the conic tip, the total increase in scattering area of this singularity of measure zero is not as high as that of a sharp wedge, thus rendering the signal relatively weak. We hence employ a well-defined multi-wedged silica cone fabricated by wet-etching, with the wedges introduced by non-uniform stress formed during the fiber assembly process, to produce maximum scattering at the tip where three to four wedges converge (see inset of Fig. Fig.1A).1A). Using the reflection mode to fully exploit this singular scattering to excite fluorescent reporters at the tip and transmit the resulting signal, we report a nanocone array that can detect down to 100 molecules per cone tip with a large dynamic range from femtomolar to nanomolar concentrations. Although quantification for a single target is reported in this preliminary report, multi-target assays can readily be developed.Open in a separate windowFigure 1(A) A SEM image of the silica cone array where the single cone inset image shows three wedges converging into a 10 nm junction at the tip. (B) The optical setup of measurement. (C) The diffraction-limited fluorescent spot images.Amine-modified 35-base oligo-probes were functionalized onto both unetched silica fibers (as a control) and etched conic silica tips. The sample of 35-base ssDNA targets (corresponding to a primer for a segment of the Serotype 2 dengue genome) with a 5′ tagged Cy3 fluorophore was inserted into a microfluidic chip housing the fiber bundle (Fig. (Fig.1B)1B) and left overnight (see the supplementary material25 for exact sequence). After a standard rinsing protocol, fluorescent images were taken with an Olympus IX-71 fluorescent microscope for target concentrations ranging from 1 fM to 1 nM. A typical fluorescent image after hybridization is shown in Fig. Fig.1c,1c, where each micron-sized bright spot corresponds to a single tip in the cone array. The intensity profile shown in the supplementary material25 indicates a fluorescent spot smaller than 1 μm, indicating that the fluorescent light source is sub-wavelength and the resolution is close to diffraction limit. The size of this bright spot at the conic tip does not vary much with respect to the concentration but its intensity does, as shown in Fig. Fig.2A.2A. It was found that for flat fibers, only concentrations higher than 1 nM produced significant signals above the background. However, for etched conic fibers, 10 fM is clearly distinguishable from the background, which indicates that an improvement of sensitivity up to five orders can be realized by simply etching the flat surface into cone arrays. It also suggests very little target loss due to preferential hybridization onto the cone at sub-nM concentrations. We estimated the number of molecules per cone from the total number of molecules in target solution divided by the number of pixels on each fiber (104), which suggests less than 100 molecules per cone for a 10 fM bulk concentration, four orders better than any existing technology.Open in a separate windowFigure 2(A) Fluorescent intensity of etched conic fiber and unetched fiber for different concentrations of target molecules from 1 fM to 1 nM. (B) Fluorescent intensity increases linearly with exposure time. Non-target molecules with 1 μM concentration do not produce significant signal compared to lower concentrations of target molecules such as 1 nM and 10 nM (see the supplementary material25 for details of image analysis).Selectivity of the platform was also examined. Fig. Fig.2B2B presents the fluorescent intensity of the tips for non-target (1 μM) and target (1 nM and 10 nM) at different exposure times, which shows that fluorescent intensity increases linearly with exposure time. Beyond 5 s, saturation of images prevents further increase in the signal. For non-target, the intensity is much lower than 1 nM Target and 10 nM Target, which means non-target do not bind to the probes at the wedged tip as effectively as target molecules. Non-specific binding can be further removed by using more stringent buffers and higher flow rates.26 This platform can be extended to detect 70 000 targets, in theory, by functionalizing different probes onto each cones using localized photochemistry via masking, micro-mirror directed illumination, or direct laser writing. Extension to ELISA type protein assays is also straight forward. Integration of a transmission-mode optical fiber endoscope into a microfluidic biochip and into a 3-D cell culture for real-time monitoring of multiple molecular targets at near-single molecule resolution is currently underway.  相似文献   

3.
Plasmonic hot spots, generated by controlled 20-nm Au nanoparticle (NP) assembly, are shown to suppress fluorescent quenching effects of metal NPs, such that hair-pin FRET (Fluorescence resonance energy transfer) probes can achieve label-free ultra-sensitive quantification. The micron-sized assembly is a result of intense induced NP dipoles by focused electric fields through conic nanocapillaries. The efficient NP aggregate antenna and the voltage-tunable NP spacing for optimizing hot spot intensity endow ultra-sensitivity and large dynamic range (fM to pM). The large shear forces during assembly allow high selectivity (2-mismatch discrimination) and rapid detection (15 min) for a DNA mimic of microRNA.Irregular expressions of a panel of microRNAs (miRNA) in blood and other physiological fluids may allow early diagnosis of many diseases, including cancer and cardiovascular diseases.1 However, quantifying all relevant miRNAs (out of 1000), with similar sequences over 22 bases2 and large variations in expression level (as much as 100 fold) at small copy numbers, requires a new molecular diagnostic platform with high-sensitivity, high-selectivity, and large dynamic range. Current techniques for miRNA profiling, such as Northern blotting,3 microarray-based hybridization,4 and real-time quantitative polymerase chain reaction5 are expensive and complex. A simple and rapid miRNA array would allow broad distribution of molecular diagnostic devices for cancer and chronic diseases, eventually into homes for frequent prescreening of many diseases.At their low concentrations in untreated samples, optical sensing of miRNA is most promising. Plasmonically excited Raman scattering (SERS) and fluorescence sensors from metallic nanoparticles (NPs) or surfaces have enhanced the sensitivity of optical molecular sensors by orders of magnitude.6, 7, 8, 9 However, probe-less SERS sensing or fluorescent sensing of unlabeled targets are insufficiently specific for miRNA targets in heterogeneous samples. Plasmonic detection is also very compatible with FRET probes whose donor dye offers small light sources to excite fluorescently labelled targets upon hybridization.7, 10A particular family of FRET reporters does offer label-free sensing: hairpin oligo probes whose end-tagged fluorophores are quenched by the Au NP to which they are functionalized.11 The fluorescent signal is only detected when the hairpin is broken by the hybridizing target nucleic acid or protein (for an aptamer probe), and the more rigid paired segment separates the end fluorophore from the quenching surface to produce a fluorescent signal. It is often hoped that plasmonics on the metal surface will enhance the intensity to overcome the quenching effect, if the linearized hairpin is within the NP plasmonic penetration length. However, since fluorescent quenching decays slowly (linearly) with fluorophore-metal spacing10 whereas the plasmonic intensity decays exponentially from a flat surface, careful experimentation shows that quenching dominates and the hairpin probe actually produces a larger intensity on non-metallic surfaces,10 on which it can not function as a label-free probe. Hence, only μM limit-of-detection (LOD) has been achieved with this technique on single NPs or on flat metal surfaces,12 with expensive laser excitation and confocal detection.Plamonic hot spots formed between metal nanostructures and sharp nanocones can further amplify the plasmonic field.13, 14 The hot spot intensity decays algebraically with respect to the separation or cone tip distance and hence should dominate the linear decay of the metal quenching effect at some optimum separation.15 It is hence possible that plasmonic hot spots may allow much lower LOD with inexpensive optical instruments—ideally light-emitting diode light source and miniature camera. However, the dimension of the gaps, cones, and wedges needs to be at nanoscale, and the cost is now transferred to fabrication of such hot-spot substrates like bow-ties, double crescents, bull-eyes, etc.16 Low-cost wet-etching techniques for addressable nanocones that sustain converging plasmonic hot spots17 have been reported but the fabricated nanocones are often too non-uniform to allow precise quantification. NP monolayers have been shown to exhibit plasmonic hot spots and fluorescence enhancement.18, 19 However, the enhancement only occurs within a range of spacing between aggregated NPs, which is difficult to control and the location or even the existence of the hotspots are not known a priori.Higher sensitivity is expected if a minimum number of NPs are used in an assembly at a known location and if the NP assembly can produce crystal-like aggregates with controllable NP spacing. Induced DC and AC NP dipoles (related to dielectrophoresis) have been used to assemble NP crystals by embedded micro-electrodes to provide the requisite high field.20, 21 The resulting NP crystals are ideal for plasmonic hot spots, since the spacing of the regimented NP crystal can be controlled by the applied voltage. Conic nanocapillaries22, 23 will be used here for such field-induced NP assembly because the submicron-tip can focus the electric field into sufficient high intensity for NP assembly without embedded-electrodes. Because the field is highest at the tip due to field focusing, the micron-sized crystal would be confined to a small volume, which will be shown to be less than typical confocal volumes, at a known location. So long as the hotspots are regimented, the quantification of target molecules is determined by the total fluorescent intensity and is hence insensitive to the exact geometry of the nanocapillary.Fluorescent microscope equipped with tungsten lamp light source and normal CCD camera from Q Imaging were used for simultaneous optical and ion current measurements, as shown in Fig. Fig.1a.1a. The nanocapillaries were pulled from commercial glass capillaries using laser-assisted capillary puller. SEM image of a typical pulled glass nanocapillary in Fig. Fig.1b1b shows an inner diameter of 111 nm and cone angle of 7.3°. The capillary was inserted into a Polydimethylsiloxane chip with two reservoirs. The 20 nm Au NPs, functionalized with fluorescently labelled dsDNA, were injected into the base reservoir. With SEM imaging (Fig. S3 in the supplementary material24), the functionalized DNA is found to prevent NP aggregation even in high ionic-strength Phosphate buffered saline buffer. The NP solution is then driven into the capillary through the tip by applying a positive voltage. Fig. Fig.1c1c shows the ion current evolution over 2 h at +1 V packing voltage. The ion current increases rapidly in the first 10 min, then at a much slower rate. The rise of current indicates assembly of conductive Au NP assembly at the tip. This was confirmed by the strong fluorescence signal at the tip region during the packing process (inset of Fig. Fig.1c).1c). The one-micron region (corresponding to roughly an aggregate volume of one attoliter) near the capillary tip shows a fluorescence signal after 1 min and also appeared as a dark spot in the transmission image (supplementary material, Fig. S124). This spot darkens with longer packing time but does not grow in size, consistent with the monotonically increasing ion current with increased packing density of the NP assembly. As contrast, a strong fluorescence appeared after only 1 min of packing, but the signal became weaker after 15 min (supplementary material, Fig. S124). This reduction in fluorescence is not due to bleaching of fluorophores because we took 2 images in 15 min at 5 s exposure each and control experiments show significant bleaching only beyond an exposure time of 100 s (see supplementary material).24 Instead, the non-monotonic dependence of the fluorescence intensity with respect to time is because of the optimal hotspot spacing for highest plasmonic intensity at about 5–20 nm,25, 26, 27 which is reached at about 10 min.Open in a separate windowFigure 1Plasmonic hotspots generated at the tip of a nano-capillary. (a) Schematic of the experimental set up. (b) SEM image of glass nanocapillary shows opening at the tip with a diameter of 111 nm. (c) Current evolution during packing of fluorescently labeled gold particles at +1 V. Inset shows strong fluorescence only after 1 min of packing.The FRET probe is designed to exploit the plasmonic hotspot.24 We first electrophoretically drove the target molecules in the tip side reservoir into the nano-capillary by applying a negative voltage of −1 V. During this process, the targets are trapped within the capillary and hybridize with the hairpin probes on the Au NP in the nanocapillary. Fluorescence of the unquenched hybridized probes is too weak to be detected by our detector as shown in Fig. Fig.2b.2b. A reverse positive voltage of +1 V was then applied to the capillary to pack the Au NPs to the tip. Due to plasmonic hot spots of aggregated gold nanoparticles, the fluorescence signal is significantly enhanced at the tip and can be detected by our CCD camera, as shown in Fig. Fig.2c2c.Open in a separate windowFigure 2(a) Schematics of designed hairpin probe on gold particle. (b) Before packing gold particles, probe fluorescence signal was too weak to be detect. (c) After packing for 3 minutes, a strong fluorescence signal appears at the NP aggregate. (d) Normalized intensity (average of all pixels above a threshold (15 au) normalized with respect to the average over all pixels (with 0-250 au)) as a function of packing voltage for different samples. Black, 1 nM target ; blue, 10 pM target; purple, 10 nM 2-mismatch non-target. (e) Intensity dependence on target concentration. Measured normalized intensity before packing (black) and after packing (red), for three independent experiments with different nano-capillaries at each concentration. NT stands for non-target at 10 nM as a reference.For the same packing time, the fluorescence intensity increases initially but saturates after 10 min time of trapping (supplementary material, Fig. S2(a)24). Over 10 min of trapping with a negative voltage, we found the fluorescence intensity exhibits a maximum at a packing time of 3 min (supplementary material, Fig. S2(b)24). In later experiments, we used 10 min trapping time and 3 min packing time as standards.Fig. Fig.2d2d shows the fluorescence intensity is sensitive to the positive packing voltage at different concentration of target and non-target molecules. For target samples (1 nM and 10pM), the optimal voltage is about 1 V. We suspect that with larger voltage, the NPs are packed too tightly such that the NP spacing is smaller than the optimal distance for plasmonic hotspots. The fluorescence intensity for a nontarget with two mismatches is 7 times lower than the target even with a 10 times higher concentration (10 nM). Moreover, the optimal voltage for the non-target miRNA is reduced to 0.5 V instead 1 V for the target miRNA. Strong shear during electrophoretic packing has probably endowed this high selectivity.20Using the protocol above, the LOD and dynamic range of the target was determined (Fig. (Fig.2e).2e). The intensity at each concentration is measured with three independent experiments with different nanocapillaries to verify insensitivity with respect to the nanocapillary. The intensity increases monotonically with respect to the concentration from 1fM to 1pM. Beyond 1pM, the fluorescence signal saturates, presumably because all hairpin probes at the tip have been hybridized. At 1 fM, the fluorescent intensity is still well above the background measured from the non-target sample. Note both auto-fluorescence of gold nanoparticles and free diffusing non-target DNA molecules contribute to the background. Given the volume of tip side reservoir (∼50 μl), there are about 30 000 target molecules in the reservoir at 1 fM. However, with a short 10 min trapping time, we estimate only a small fraction of these molecules, less than 100, have been transferred from the tip reservoir into the nanocapillary.  相似文献   

4.
The 5th International Conference on Optofluidics (Optofluidics 2015) was held in Taipei, Taiwan, July 26–29, 2015. The aim of this conference was to provide a forum to promote scientific exchange and to foster closer networks and collaborative ties between leading international researchers in optics and micro/nanofluidics across various disciplines. The scope of Optofluidics 2015 was deliberately broad and interdisciplinary, encompassing the latest advances and the most innovative developments in micro/nanoscale science and technology. Topics ranged from fundamental research to its applications in chemistry, physics, biology, materials, and medicine.Approximately 300 delegates participated in Optofluidics 2015 from across the globe, including Australia, Canada, China, France, Germany, Hong Kong, India, Japan, Korea, Singapore, Taiwan, UK, and USA. In total, 242 presentations were arranged, including 10 plenary speeches, 27 keynote speeches, 65 invited talks, 33 contributed talks, and 107 poster presentations. This collection of twelve papers on this special topic spans both the fundamentals and the frontier applications of this interdisciplinary research field.Optical measurements of particle or flow and fluidic manipulation for optical applications were presented. Lin and Su1 reported a novel method to measure the depth position of rapidly moving objects inside a microfluidic channel based on the chromatic aberration effect; the depth positions of label-free particles of diameter as small as 2 μm and erythrocytes of concentration 2 × 103 cells/μl and velocity 2.78 mm/s were detected within a range ±25 μm in a simple and inexpensive manner. Sun and Huang2 demonstrated the use of a microscopic circular polariscope to measure the flow-induced birefringence in a microfluidic device that represents the kinematics of fluid motion optically; CTAB:NaSal, CPyCl:NaSal, and CPyCl:NaSal:NaCl solutions were used to investigate the strain rate and the results were compared with the μPIV diagnosis. He et al.3 studied the fundamentals, especially the thinning and opening of the oil film within each pixel of an electrowetting display; to achieve repeatable oil movement and the resulting pixel performance, a new method to fill each pixel with a controllable oil volume using an oil-droplet emulsion created with a microfluidic device was demonstrated.This special topic includes papers also on particle manipulation. Weng et al.4 evaluated the size-dependent crossing frequency of dielectrophoretically driven particles; numerical simulation using a Maxwell stress tensor and a finite element method was reported to assess the size effect. In addition to electric manipulation, magnetic driving of the particles was demonstrated. Ido et al.5 examined microswimmers of magnetic particle chains in an oscillating magnetic field experimentally and analyzed numerically with a lattice Boltzmann method, an immersed boundary method, and a discrete particle method based on simplified Stokesian dynamics. Huang et al.6 described a technique to manipulate magnetic beads and achieved a great washing efficiency with zero bead loss using an appropriate electrode design and channel height of a digital microfluidic immunoassay; a model immunoassay of human soluble tumor necrosis factor receptor I (sTNF-RI) was performed to offer an improved limit of detection (3.14 pg/ml) with a small number of magnetic beads (25 beads), decreased reagent volumes (200 nl), and decreased duration of analysis (<1 h). Chiu et al.7 reported particle separation using cross-flow filtration enhanced with hydrodynamic focusing; label-free separation of particles of diameters 2.7 and 10.6 μm at a sample throughput 10 μl/min was performed; separation of spiked human prostate cancer cell lines (PC3) cells in whole blood was also demonstrated.Chemical sensors and biosensors are covered in this special topic. Cheng et al.8 measured the chemical compounds in third-hand smoke on varied clothing fibres with an analytical balance, or nicotine and 3-ethenylpyridine (3-EP) with a surface-acoustic-wave sensor composed of coated oxidized hollow mesoporous carbon nanospheres. Pu et al.9 described a continuous glucose monitoring microsystem consisting of a three-electrode electrochemical sensor in which the working electrode (WE) was covered with a single layer of graphene and gold nanoparticles to improve the sensor performance; the results of glucose measurement were linear below concentration 162 mg/dl with a detection limit 1.44 mg/dl. Li et al.10 implemented a microfluidic device measuring the glucose concentration with integrated fibre-optic surface plasmon resonance sensor and electrode pairs for volume quantification.Implantable devices and microneedles for drug delivery and liquid transport are addressed in this special topic. Zhang et al.11 reported a flexible polyimide device seated under rabbit eyelids to deliver drug by iontophoresis; varied currents to release manganese ions (Mn2+) as tracers were investigated; the thermal effect on application of a current was studied. Lee et al.12 presented a disposable Parylene microneedle array of large aspect ratio that vibrated with a piezoelectric actuator to mimic the vibrating motion of a mosquito''s proboscis and to decrease the insertion force by 40%. Song et al.13 demonstrated microinjection into a model organism, Caenorhabditis elegans (C. elegans) on an automated device capable of loading, immobilization, injection, and sorting; with 200 worms studied, injection speed 6.6 worm/min, injection success rate 77.5%, and sorting success rate 100% were obtained.We express our gratitude for the financial support from Ministry of Science and Technology (Taiwan), Bureau of Foreign Trade (Taiwan), National Taiwan University and Research Center for Applied Sciences of Academia Sinica, and for administrative support from Instrument Technology Research Center in making Optofluidics 2015 a successful conference. Our acknowledgements include Leslie Yeo, Frederick Kontur, Christine Urso, and all staff from Biomicrofluidics for their kind assistance during the preparation, and, most importantly, all authors who have contributed their work for this special topic.  相似文献   

5.
Surface-enhanced Raman scattering (SERS) shows promise for identifying single bacteria, but the short range nature of the effect makes it most sensitive to the cell membrane, which provides limited information for species-level identification. Here, we show that a substrate based on black silicon can be used to impale bacteria on nanoscale SERS-active spikes, thereby producing spectra that convey information about the internal composition of the bacterial capsule. This approach holds great potential for the development of microfluidic devices for the removal and identification of single bacteria in important clinical diagnostics and environmental monitoring applications.Plasma etching of silicon can be used to produce inexpensive, large surface area, nano-textured surfaces known as black silicon. Recently, it has been shown that black silicon nano-needles can impale bacteria1 and that it can be used as a sensor in microfluidic devices.2 When coated by a plasmonic metal, such as gold, the nano-textured surface of black silicon is ideal for use as a surface-enhanced Raman scattering (SERS) sensing platform.3 This work aims to investigate whether gold-coated black silicon nano-needles can be used to both impale bacteria and identify them by SERS. This combination of properties would promote the development of microfluidic devices for the removal and monitoring of bacteria in a wide range of medical, environmental, and industrial applications.4Black silicon was fabricated by a reactive ion etching technique,5 resulting in pyramidal-shaped spikes of height 185 ± 30 nm, full width at half height of 54 ± 10 nm, and 10 ± 2.4 nm radius of curvature at the tip. Samples were then magnetron sputter coated with 200 nm of gold, as this coating thickness was found to provide a suitable compromise between SERS enhancement and impalement efficiency. E. coli (ATCC 25922) from −80 °C stock was isolated on a nutrient agar plate (Difco nutrient broth, Becton Dickinson) for approximately 12 h. A single E. coli colony was then inoculated from the plate into 20 ml of nutrient broth media and incubated overnight at 37 °C with orbital shaking at 200 rpm. The total biomass of overnight culture was adjusted to an optical density of 0.3 at λ = 600 nm by adding fresh sterile nutrient broth (Cary 50 spectrophotometer, Agilent). The E. coli planktonic cells were washed three times by centrifugation at 12 000 rpm (Centrifuge 5804 R, Eppendorf) for 2 min. The washed cells were then re-suspended in a low strength minimum medium (Dulbecco A, phosphate buffered saline). A volume of 100 μl of solution was pipetted onto substrates and left to incubate for 1 h on the bench. Separate sets of samples were created for scanning electron microscope (SEM) imaging, live/dead staining, and SERS. Three sets were needed as each of these measurements altered the samples and left them unsuitable for further analysis.The first set of samples was washed three times with milliQ water after incubation, allowed to dry and then immediately sputter coated with gold using the Emitech K975x (operating current 35 mA, sputter time 32 s, stage rotation 138 rpm, and vacuum of 1 × 10−2 mbar). SEM imaging was performed with a Zeiss Supra 40VP in high vacuum mode with a working distance of 5 mm and an accelerating voltage of 3 kV. Figure Figure11 shows an example of the different levels of impalement that occurred on the black silicon surface. All cells showed signs of damage, but in some cases, the damage was limited to the perimeter of the cell and the main body appeared whole. In other cases, the entire cell had collapsed onto the spikes.Open in a separate windowFIG. 1.A typical SEM image showing E. coli cells with different levels of impalement on gold-coated black silicon.The second set of samples was used for live/dead staining (Invitrogen BacLight Bacterial Viability Kit L7012) with 3.34 mM SYTO 9 (green fluorescence) and 20 mM propidium iodide (red fluorescence). Equal volumes of both dyes were mixed thoroughly in a tube and added to the sample in a ratio of 3 μl of mixed dye to 1 ml of bacterial suspension. After mixing, a volume of 100 μl of the solution was pipetted onto the substrates, which were then incubated at room temperature in the dark for 15 min, before the staining solution was removed by pipetting. The substrates were then washed three times with milliQ water and mounted on a microscope slide for fluorescence imaging. The substrates were not allowed to dry and were stored in phosphate buffered saline at 4 °C when not in use. An epifluorescence microscope (Olympus IX71) with a mercury lamp source and a 60× water immersion objective was used to collect live/dead images from the substrates. Two filter blocks were used to collect the images: U-MNIBA2 blue excitation narrow band delivered green emission (live) and U-MWIG2 green excitation wide band provided red emission (dead).The live/dead image in Figure Figure22 shows a mix of both live and dead cells on the black silicon sample. The prevalence of live cells could be due to the incomplete impalement seen under SEM for some cells. It can also be explained by the sample still being wet during live/dead staining. The cells are dried prior to imaging in the SEM and this could weaken the cell wall and allow capillary forces to draw the cells onto the spikes for impalement. This hypothesis is supported by the large number of cells on the stained sample and the presence of cell groupings and cells imaged during mid-division. If the cells were immediately impaled, then such activity would not have been visible and a greater proportion of red cells would be expected.Open in a separate windowFIG. 2.Epifluorescence image showing live (green) and dead (red) E. coli cells after incubation on gold-coated black silicon.The third set of samples was washed three times with milliQ water after incubation and allowed to dry prior to spectral analysis. SERS spectra were collected with a Renishaw inVia Raman spectrometer operating at 785 nm with a 1200 l/mm grating. Power at the sample was 150 mW focused with a 100 × /0.85 NA objective to obtain a diffraction limited laser spot. The resulting spot size (≤2 μm in diameter) is well matched to the size of the bacterial cells. Spectra were collected with three accumulations of 10 s. Data were background subtracted6 and normalised to unity for ease of plotting. A great deal of variability was observed in the resulting spectra, as shown in Figure Figure33.Open in a separate windowFIG. 3.SERS spectra of E. coli after incubation on a gold-coated black silicon substrate. The spectrum numbers represent single cells at different locations and different levels of impalement.It should be noted that E. coli SERS is known to produce a high level of variability,7–12 depending on the experimental setup.13 However, the variability seen in the SERS spectra of Fig. Fig.33 is unusual for measurements performed under consistent experimental conditions. This increased level of variability may be related to the different levels of impalement seen in Fig. Fig.1,1, which results in the probing of different internal components. SERS is a surface sensitive technique, with the signal primarily arising within 2 nm of the metal surface.14 Note that unlike apertureless nanoprobes15 or conical plasmonic nanotips,16 the SERS signal in black silicon arises primarily from “hot spots” between the spikes, where the plasmon resonance field is particularly strong.17 Therefore, depending on the depth and location of impalement, different biomolecules are expected to be excited by this novel substrate.Some peaks occur in the same position for multiple spectra (e.g., peak positions 420, 893, 1001, 1285, and 1307 cm−1), but there are also a lot of unique peaks. The vertical lines in Fig. Fig.33 indicate peaks which have appeared in the literature for SERS of E. coli.7–12 Spectrum 3 has a high proportion of peaks matching published values. This is also the case for spectrum 5, which shares a few peak positions with spectrum 3. Preliminary peak allocations have identified carbohydrates11 (420 cm−1), tyrosine11 (650 cm−1), adenine10,11 (706 and 735 cm−1), hypoxanthine7 (722 and1373 cm−1), phenylalanine9 (1001 cm−1), amide III (Ref. 10) (1285 cm−1), CH2 deformation12 (1556 cm−1), and C=C10 (1587 cm−1).Given the varying levels of impalement observed in the SEM, it appears that the spike shape and Au coating should be further optimized to ensure that the entire cell is consistently pierced and the internal biomolecules are more comprehensively probed. In this way, it may be possible to obtain a more reproducible SERS spectrum of the internal biomolecular constituents of single bacterial cells, thereby providing rapid identification for medical and environmental diagnostic applications. Given that SERS is insensitive to water,4 future work should aim to achieve impalement in an aqueous environment, so that the full capability of microfluidics can be used to separate and concentrate suspended bacteria before presenting them to the substrate for rapid analysis.4 This suggests a broad range of potential applications in the detection, monitoring, and control of bacterial contamination.  相似文献   

6.
Bipolar membranes (BMs) have interesting applications within the field of bioelectronics, as they may be used to create non-linear ionic components (e.g., ion diodes and transistors), thereby extending the functionality of, otherwise linear, electrophoretic drug delivery devices. However, BM based diodes suffer from a number of limitations, such as narrow voltage operation range and/or high hysteresis. In this work, we circumvent these problems by using a novel polyphosphonium-based BM, which is shown to exhibit improved diode characteristics. We believe that this new type of BM diode will be useful for creating complex addressable ionic circuits for delivery of charged biomolecules.Combined electronic and ionic conduction makes organic electronic materials well suited for bioelectronics applications as a technological mean of translating electronic addressing signals into delivery of chemicals and ions.1 For complex regulation of functions in cells and tissues, a chemical circuit technology is necessary in order to generate complex and dynamic signal gradients with high spatiotemporal resolution. One approach to achieve a chemical circuit technology is to use bipolar membranes (BMs), which can be used to create the ionic equivalents of diodes2, 3, 4, 5 and transistors.6, 7, 8 A BM consists of a stack of a cation- and an anion-selective membrane, and functions similar to the semiconductor PN-junction, i.e., it offers ionic current rectification9, 10 (Figure (Figure1a).1a). The high fixed charge concentration in a BM configuration make them more suited in bioelectronic applications as compared to other non-linear ionic devices, such as diodes constructed from surface charged nanopores11 or nanochannels,12 as the latter devices typically suffers from reduced performance at elevated electrolyte concentration (i.e., at physiological conditions) due to reduced Debye screening length.13 However, a unique property of most BMs, as compared to the electronic PN-junction and other ionic diodes, is the electric field enhanced (EFE) water dissociation effect.10, 14 This occurs above a threshold reverse bias voltage, typically around −1 V, as the high electric field across the ion-depleted BM interface accelerates the forward reaction rate of the dissociation of water into H+ and OH ions. As these ions migrate out from the BM, there will be an increase in the reverse bias current. The EFE water dissociation is a very interesting effect and is commonly used in industrial electrodialysis applications,15 where highly efficient water dissociating BMs are being researched.16 Also, BMs have also been utilized to generate H+ and OH ions in lab-on-a-chip applications.2, 17 However, the EFE water dissociation effect diminishes the diode property of BMs when operated outside the ±1 V window, which is unwanted in, for instance, chemical circuits and addressing matrices for delivery of complex gradients of chemical species. The effect can be suppressed by incorporating a neutral electrolyte inside the BM,10, 18 for instance, poly(ethylene glycol) (PEG).2, 6, 7 However, as previously reported,2 the PEG volume will introduce hysteresis when switching from forward to reverse bias, due to its ability to store large amounts of charges. This was circumvented by ensuring that only H+ and OH are present in the diode, which recombines into water within the PEG volume. Such diodes are well suited as integrated components in chemical circuits for pH-regulation, due to the in situ formed H+ and OH, but are less attractive if, for instance, other ions, e.g., biomolecules, are to be processed or delivered in and from the circuit. Furthermore, a PEG electrolyte introduces additional patterning layers, making device downscaling more challenging. This is undesired when designing complex, miniaturized, and large-scale ionic circuits. Thus, there is an interest in BM diodes that intrinsically do not exhibit any EFE water dissociation, are easy to miniaturize, and that turn off at relatively high speeds. It has been suggested that tertiary amines in the BM interface are important for efficient EFE water dissociation,19, 20, 21 as they function as a weak base and can therefore catalyze dissociation of water by accepting a proton. For example, anion-selective membranes that have undergone complete methylation, converting all tertiary amines to quaternary amines, shows no EFE water dissociation,19 although this effect was not permanent, as the quaternization was reversed upon application of a current. Similar results were found for anion-selective membranes containing alkali-metal complexing crown ethers as fixed charges.21 Also, EFE water dissociation was not observed or reduced in BMs with poor ion selectivity, for example, in BMs with low fixed-charge concentration5 or with predominantly secondary and tertiary amines in the anion-selective membrane,22 as the increased co-ion transport reduces the electric field at the BM interface. However, due to decreased ion selectivity, these membranes show reduced rectification. In this work, we present a non-amine based BM diode that avoids EFE water dissociation, enables easy miniaturization, and provides fast turn-off speeds and high rectification.Open in a separate windowFigure 1(a) Ionic current rectification in a BM. In forward bias, mobile ions migrate towards the interface of the BM. The changing ion selectivity causes ion accumulation, resulting in high ion concentration and high conductivity. At high ion concentration, the selectivity of the membranes fails (Donnan exclusion failure), and ions start to pass the BM. In reverse bias, the mobile ions migrate away from the BM, eventually giving a zone with low ion concentration and low conductivity. Reverse bias can cause EFE water dissociation, producing H+ and OH- ions. (b) Chemical structures of PSS, qPVBC, and PVBPPh3. (c) The device used to characterize the BMs and the BM1A, BM2A, and BM1P designs. The BM interfaces are 50 × 50 μm.An anion-selective phosphonium-based polycation (poly(vinylbenzyl chloride) (PVBC) quaternized by triphenylphospine, PVBPPh3) was synthesized and compared to the ammonium-based polycation (PVBC quaternized by dimethylbenzylamine, qPVBC) previously used in BM diodes2 and transistors,7, 8 when included in BM diode structures together with polystyrenesulfonate (PSS) as the cation-selective material (Figure (Figure1b).1b). Three types of BM diodes were fabricated using standard photolithography patterning (Figure (Figure1c),1c), either with qPVBC (BM1A and BM2A) or PVBPPh3 (BM1P) as polycation and either with (BM2A) or without PEG (BM1A and BM1P). Poly(3,4-ethylenedioxythiophene):poly(styrenesulfonate) (PEDOT:PSS) electrodes covered with aqueous electrolytes were used to convert electronic input signals into ionic currents through the BMs, according to the redox reaction PEDOT+:PSS + M+ + e ↔ PEDOT0 + M+:PSS.The rectifying behavior of the diodes was evaluated using linear sweep voltammetry (Figure (Figure2).2). The BM1A diode exhibited an increase in the reverse bias current for voltages lower than −1 V, a typical signature of EFE water dissociation,10, 14 which decreased the current rectification at ±4 V to 6.14. No such deviation in the reverse bias current was observed for BM2A and BM1P, which showed rectification ratios of 751 and 196, respectively. In fact, for BM1P, no evident EFE water dissociation was observed even at −40 V (see inset of Figure Figure2).2). Thus, the PVBPPh3 polycation allows BM diodes to operate at voltages beyond the ±1 V window with maintained high ion current rectification.Open in a separate windowFigure 2Linear sweep voltammetry from −4 to +4 V (25 mV/s) for the BM diodes. The inset shows BM1P scanning from −40 V to +4 V (250 mV/s).The dynamic performance of the diodes was tested by applying a square wave pulse from reverse bias to a forward bias voltage of 4 V with 5–90 s pulse duration (Figure (Figure3).3). To access the amount of charge injected and extracted during the forward bias and subsequent turn off, the current through the device was integrated. For BM2A, we observed that the fall time increased with the duration of the forward bias pulse. This hysteresis is due to the efficient storage of ions in the large PEG volume, with no ions crossing the BM due to Donnan exclusion failure.2 As a result, during the initial period of the return to reverse bias, a large amount of charge needs to be extracted in order to deplete the BM. After a 90 s pulse, 90.6% of the injected charge during the forward bias was extracted before turn-off. This may be contrasted with BM1P, where the fall time is hardly affected by the pulse duration, and the extracted/injected ratio is only 15.4% for a 90 s pulse. For this type of BM, the interface quickly becomes saturated with ions during forward bias, leading to Donnan exclusion failure and transport of ions across the BM.4 Thus, less charge needs to be extracted to deplete the BM, allowing for faster fall times and significantly reduced hysteresis.Open in a separate windowFigure 3Switching characteristics (5, 10, 20, 30, 60, or 90 s pulse) and ion accumulation (at 90 s pulse) of the BM2A and BM1P diodes. BM1A showed similar characteristics as BM1P when switched at ±1V (see supplementary material).24Since the neutral electrolyte is no longer required to obtain high ion current rectification over a wide potential range, the size of the BM can be miniaturized. This translates into higher component density when integrating the BM diode into ionic/chemical circuits. A miniaturized BM1P diode was constructed, where the interface of the BM was shrunk from 50 μm to 10 μm. The 10 μm device showed similar IV and switching characteristics as before (Figure (Figure4),4), but with higher ion current rectification ratio (over 800) and decreased rise/fall times (corresponding to 90%/–10% of forward bias steady state) from 10 s/12.5 s to 4 s/4 s. Since the overlap area is smaller, a probable reason for the faster switching times is the reduced amount of ions needed to saturate and deplete the BM interface. In comparison to our previous reported low hysteresis BM diode,2 this miniaturized polyphosphonium-based devices shows the same rise and fall times but increased rectification ratio.Open in a separate windowFigure 4(a) Linear sweep voltammetry and (b) switching performance of a BM1P diode with narrow junction.In summary, by using polyphosphonium instead of polyammonium as the polycation in BM ion diodes the EFE water dissociation can be entirely suppressed over a large operational voltage window, supporting the theory that a weak base, such as a tertiary amine, is needed for efficient EFE water dissociation.17, 18 As no additional neutral layer at the BM interface is needed, ion diodes that operate outside the usual EFE water dissociation window of ±1 V can be constructed using less active layers, fewer processing steps and with relaxed alignment requirement as compared to polyammonium-based devices. This enables the fabrication of ion rectification devices with an active interface as low as 10 μm. Furthermore, the exclusion of a neutral layer improves the overall dynamic performance of the BM ion diode significantly, as there is less hysteresis due to ion accumulation. Previously, the hysteresis of BM ion diodes has been mitigated by designing the diode so that only H+ and OH enters the BM, which then recombines into water.2 Such diodes also show high ion current rectification ratio and switching speed but are more complex to manufacture, and their application in organic bioelectronic systems is limited due to the H+/OH production. By instead using the polyphosphonium-based BM diode, reported here, we foresee ionic, complex, and miniaturized circuits that can include charged biomolecules as the signal carrier to regulate functions and the physiology in cell systems, such as in biomolecule and drug delivery applications, and also in lab-on-a-chip applications.  相似文献   

7.
A microfluidic device was successfully fabricated for the rapid serodiagnosis of amebiasis. A micro bead-based immunoassay was fabricated within integrated microfluidic chip to detect the antibody to Entamoeba histolytica in serum samples. In this assay, a recombinant fragment of C terminus of intermediate subunit of galactose and N-acetyl-D-galactosamine-inhibitable lectin of Entamoeba histolytica (C-Igl, aa 603-1088) has been utilized instead of the crude antigen. This device was validated with serum samples from patients with amebiasis and showed great sensitivity. The serodiagnosis can be completed within 20 min with 2 μl sample consumption. The device can be applied for the rapid and cheap diagnosis of other infectious disease, especially for the developing countries with very limited medical facilities.Entamoeba histolytica is the causative agent of amebiasis and is globally considered a leading parasitic cause of human mortality.1 It has been estimated that 50 × 106 people develop invasive disease such as amebic dysentery and amebic liver abscess, resulting in 100 000 deaths per annum.2, 3 High sensitive diagnosis method for early stage amebiasis is quite critical to prevent and cure this disease. To date, various serological tests have been used for the immune diagnosis of amebiasis, such as the indirect fluorescent antibody test (IFA) and enzyme-linked immunosorbent assay (ELISA).We have recently identified a 150-kDa surface antigen of E. histolytica as an intermediate subunit (Igl) of galactose and N-acetyl-D-galactosamine-inhibitable lectin.4, 5 In particular, it has been shown that the C-terminus of Igl (C-Igl, aa 603-1088) was an especially useful antigen for the serodiagnosis of amebiasis. ELISA using C-Igl is more specific than the traditional ELISA using crude antigen.6 However, the ELISA process usually takes several hours, which is still labor-intensive and requires experienced operators to perform. More economic and convenient filed diagnosis methods are still in need, especially for the developing countries with limited medical facilities.Among all the bioanalytical techniques, microfluidics has been attracting more and more attention because of its low reagent/power consumption, the rapid analysis speed as well as easy automation.7, 8, 9, 10, 11 Especially with the development of the fabrication technique, microfluidics chip can include valves, mixers, pumps, heating devices, and even micro sensors, so many traditional bioanalytical methods can be performed in the microfluidics. Qualitative and quantitative immune analysis on the microfluidic chip was successfully proved by plenty of research with improved sensitivity, shorten reaction time, and less sample consumption.8, 10, 11, 12, 13, 14, 15, 16, 17 Moreover, with the intervention of other physical, chemical, biology, and electronic technology, microfluidic technique has been successfully utilized in protein crystallization, protein and gene analysis, cell capture and culturing and analysis as well as in the rapid and quantitative detection of microbes.13, 14, 15, 16, 17, 18, 19, 20Herein, we report a new integrated microfluidic device, which is capable of rapid serodiagnosis of amebiasis with little sample consumption. The microfluidic device was fabricated from polydimethysiloxane (PDMS) following standard soft lithography.21, 22 The device was composed of two layers (shown in Figure Figure1)1) including upper fluidic layer (in green and blue) and bottom control layer (in red).Open in a separate windowFigure 1Structure illustration of microfluidic chip.To create the fluidic layer and the control layer, two different molds with different patterns have fabricated by photolithographic processes. The mold to create the fluidic channels was made by positive photoresist (AZ-50 XT), while the control pneumatic mold was made by negative photoresist (SU8 2025). For the chip fabrication, the fluidic layer is made from PDMS (RTV 615 A: B in ratio 5:1), and the pattern was transferred from the respective mold. The control layer is made from PDMS (RTV 615 A:B in ratio 20:1). The two layers were assembled and bonded together accurately, and there is elastic PDMS membrane about 30 μm thick between the fluidic layer channels and control layer.21, 22 The elastic membrane at the intersection can deform to block the fluid inside the fluidic channels, functioning as valves under the pressures introduced though control channels. There are two types of channels in fluidic layer, the rectangular profiled (in green, 200 μm wide, 35 μm thick) channel and round profiled channels (in blue, 200 μm wide, 25 μm center height). Because of the position of the valves on the fluidic channels, two types of valves (Figure (Figure2a)2a) were built, working as a standard valve and a sieve valve. The standard valves (on blue fluidic channels) can totally block the fluid because of the round profile of fluidic channel; the sieve valve can only half close because of the rectangular profile. The sieve valve can be used to trap the microspheres (beads) filled inside the green fluidic channels, while letting the fluid pass through. By this sieve valve, a micro column (in green) is constructed, where the entire ELISA reaction happens. The micrograph of the fabricated micro device is shown in Figure Figure2b.2b. The channels were filled with food dyes in different colors to show the relative positions of the channels. The pressures though different control channels are individually controlled by solenoid valves, connected to a computer through relay board. By programming the status (on/off) of various valves at different time periods, all the microfluidic chip operation can be digitally controlled by the computer in manual, semi-automatic, or automatic manner.Open in a separate windowFigure 2(a) Structure illustration of micro column, standard valve and sieve valve; (b) photograph of the microfluidic chip.To validate this device, 12 patient serum samples were collected. Sera from 9 patients (Nos. 1–9) with an amebic liver abscess or amebic colitis were used as symptomatic cases. The diagnosis of these patients was based on their clinical symptoms, ultrasound examination (liver abscess) and endoscopic or microscopic examination (colitis). We also identified the clinical samples using PCR amplification of rRNA genes.24 As negative control, sera obtained from 3 healthy individuals with no known history of amebiasis were mixed into pool sera. The serum was positive for E. histolytica with a titer of 1:64 (borderline positive), as determined by an indirect fluorescent-antibody (IFA) test.23, 24 In our previously study, the sensitivity and specificity of the recombinant C-Igl in the ELISA were 97% and 99%.6, 25 In the current study, the serodiagnosis of amebiasis was also examined by ELISA using C-Igl.26 The cut-off for a positive result was defined as an ELISA value > 3 SD above the mean for healthy negative controls27 (shown in Figure Figure3).3). The seropositivity to C-Igl was 100% in patients with amebiasis.Open in a separate windowFigure 3ELISA reactivity of sera from patients against C-Igl. ELISA plate was coated with 100 ng per well of C-Igl. Serum samples from patients and healthy controls were used at 1:400 dilutions. The dashed line indicates the cut-off value. Data are representative of results from three independent experiments.In the diagnosis process with microfluidic chip, the 4 micro immuno-columns filled with C-Igl-coated microspheres were the key components of the device. The C-Igl was prepared in E. coli as inclusion bodies. After expression, the recombinant protein was purified and analyzed by SDS-PAGE. The apparent molecular mass was 85 kDa.26The immune-reaction mechanism is illustrated in Figure Figure4.4. The anti-His monocolonal antibody was immobilized onto the microspheres (beads, 9 μm diameter) coated with protein A. The C-Igl was then immobilized onto the beads through the binding between the His tag and C-Igl. For the diagnosis, the microspheres immobilized with C-Igl and blocked by 5% BSA were preloaded into the columns for the rapid analysis of the patient serum samples. Generally, serum samples which were diluted 100 times were first loaded into the reaction column and incubated at room temperature for 5 min. After being washed by PBS buffer, FITC-conjugated goat anti-human polyclonal antibody was added into the column for 4 min incubation. The fluorescence image can be collected by the fluorescence microscope after the micro column was washed with PBS buffer. From loading diluted serum samples into column to collecting fluorescence images, the total time to complete the immunoassay is less than 10 min. The final fluorescence results were analyzed by Image Pro Plus 6.0.Open in a separate windowFigure 4Schematic representation of the ELISA in the chip.Different reaction conditions have been investigated to find the optimized ones. For each patient, 2 μl sample is enough for the analysis. The designed microfluidic chip with 4 micro columns is capable for 4 parallel analyses at the same time. More micro columns can be integrated into the device if more parallel tests are needed.Different incubating time for the diagnosis has also been investigated and no significant difference has been found for various time periods. It is enough to incubate the chip for only 5 min. The total diagnosis time for one sample is less than 10 min. The detection result appeared as the fluorescence intensity of the reaction column. As shown in Figure Figure5,5, the negative sample showed relatively low fluorescence intensity, because little FITC-conjugated goat anti-human polyclonal antibody could attach to the surface of microspheres; on the contrast, the positive sample showed much brighter fluorescence. The fluorescence intensity can be transferred to digital data (Table
SampleAverage scoresStandard deviation
133 790368
223 269271
339 598307
4778452
521 222197
638 878290
722 437227
836 295334
941 024396
Negative20032
Open in a separate windowOpen in a separate windowFigure 5ELISA on the chip. The signals were collected by CCD of microscope. A: negative sample; B and C: positive samples.For the heterogeneous immunoreactions, the immobilization of the immune molecules is essential for the reaction efficiency. Herein, we utilized micro columns filled with pre-modified microspheres (beads) instead of the direct surface modification for the ELISA analysis. Compared with the traditional method, diagnosis using the microfluidic device took less than 10 min with only 2 μl sample consumption and little reagent consumption. The high efficiency might be attributed to the high surface modification efficiency by using beads as well as the advantages from microfluidic device itself. The C-Igl modified microspheres can be easily prepared in 1 h and preloaded inside the micro device for convenient application. The device is made from standard soft lithography by PDMS and its throughput can be easily improved by adding more micro columns into the microfluidic device in an economic manner, which is perfect for the onsite rapid and cheap diagnosis of amebiasis. Similar methodologies can be developed for diagnosis of other infectious disease, especially for the developing countries with very limited medical facilities.  相似文献   

8.
Flow manipulation and cell immobilization for biochemical applications using thermally responsive fluids     
Anil Haraksingh Thilsted  Vahid Bazargan  Nina Piggott  Vivien Measday  Boris Stoeber 《Biomicrofluidics》2012,6(4)
A flow redirection and single cell immobilization method in a microfluidic chip is presented. Microheaters generated localized heating and induced poly(N-isopropylacrylamide) phase transition, creating a hydrogel that blocked a channel or immobilized a single cell. The heaters were activated in sets to redirect flow and exchange the fluid in which an immobilized cell was immersed. A yeast cell was immobilized in hydrogel and a 4′,6-diamidino-2-phenylindole (DAPI) fluorescent stain was introduced using flow redirection. DAPI diffused through the hydrogel and fluorescently labelled the yeast DNA, demonstrating in situ single cell biochemistry by means of immobilization and fluid exchange.The ability to control microfluidic flow is central to nearly all lab-on-a-chip processes. Recent developments in microfluidics either include microchannel based flow control in which microvalves are used to control the passage of fluid,1 or are based on discrete droplet translocation in which electric fields or thermal gradients are used to determine the droplet path.2, 3 Reconfigurable microfluidic systems have certain advantages, including the ability to adapt downstream fluid processes such as sorting to upstream conditions and events. This is especially relevant for work with individual biomolecules and high throughput cell sorting.4 Additionally, reconfigurable microfluidic systems allow for rerouting flows around defective areas for high device yield or lifetime and for increasing the device versatility as a single chip design can have a variety of applications.Microvalves often form the basis of flow control systems and use magnetic, electric, piezoelectric, and pneumatic actuation methods.5 Many of these designs require complicated fabrication steps and can have large complex structures that limit the scalability or feasability of complex microfluidic systems. Recent work has shown how phase transition of stimuli-responsive hydrogels can be used to actuate a simple valve design.6 Beebe et al. demonstrated pH actuated hydrogel valves.7 Phase transition of thermosensitive poly(N-isopropylacrylamide) (PNIPAAm) using a heater element was demonstrated by Richter et al.8 Phase transition was also achieved by using light actuation by Chen et al.9 Electric heating has shown a microflow response time of less than 33 ms.11 Previous work10 showed the use of microheaters to induce a significant shift in the viscosity of thermosensitive hydrogel to block microchannel flow and deflect a membrane, stopping flow in another microchannel. Additionally, Yu et al.12 demonstrated thermally actuated valves based on porous polymer monoliths with PNIPAAm. Krishnan and Erickson13 showed how reconfigurable optically actuated hydrogel formation can be used to dynamically create highly viscous areas and thus redirect flow with a response time of  ~ 2?s. This process can be used to embed individual biomolecules in hydrogel and suppress diffusion as also demonstrated by others.15, 16 Fiddes et al.14 demonstrated the use of hydrogels to transport immobilized biomolecules in a digital microfluidic system. While the design of Krishnan and Erickson is highly flexible, it requires the use of an optical system and absorption layer to generate a geometric pattern to redirect flow.This paper describes the use of an array of gold microheaters positioned in a single layer polydimethylsiloxane (PDMS) microfluidic network to dynamically control microchannel flow of PNIPAAm solution. Heat generation and thus PNIPAAm phase transition were localized as the microheaters were actuated using pulse width modulation (PWM) of an applied electric potential. Additionally, hydrogel was used to embed and immobilise individual cells, exchange the fluid parts of the microfluidic system in order to expose the cells to particular reagents to carry out an in situ biochemical process. The PDMS microchannel network and the microheater array are shown in Figure Figure11.Open in a separate windowFigure 1A sketch of the electrical circuit and a microscope image of the gold microheaters and the PDMS microchannels. The power to the heaters was modulated with a PWM input through a H-bridge. For clarity, the electrical circuit for only the two heaters with gelled PNIPAAm is shown (H1 and V2). There are four heaters (V1-V4) in the “vertical channels” and three heaters (H1-H3) in the “horizontal” channel.The microchannels were fabricated using a patterned mould on a silicon wafer to define PDMS microchannels, as described by DeBusschere et al.17 and based on previous work.10 A 25 × 75 mm glass microscope slide served as the remaining wall of the microchannel system as well as the substrate for the microheater array. The gold layer had a thickness of 200 nm and was deposited and patterned using E-beam evaporation and photoresist lift-off.21 The gold was patterned to function as connecting electrical conductors as well as the microheaters.It was crucial that the microheater array was aligned with an accuracy of  ~ 20μm with the PDMS microchannel network for good heat localization. The PDMS and glass lid were treated with plasma to activate the surface and alignment was carried out by mounting the microscope slide onto the condenser lens of an inverted microscope (TE-2000 Nikon Instruments). While imaging with a 4× objective, the x, y motorized stage aligned the microchannels to the heaters and the condenser lens was lowered for the glass substrate to contact the PDMS and seal the microchannels.Local phase transition of 10% w/w PNIPAAm solution in the microchannels was achieved by applying a 7 V potential through a H-bridge that received a PWM input at 500 Hz which was modulated using a USB controller (Arduino Mega 2650) and a matlab (Mathworks) GUI. The duty cycle of the PWM input was calibrated for each microheater to account for differences in heater resistances (25?Ω to 52?Ω) due to varying lengths of on-chip connections and slight fabrication inconsistencies, as well as for different flow conditions during device operation. Additionally, thermal cross-talk between heaters required decreasing the PWM input significantly when multiple heaters were activated simultaneously. This allowed confining the areas of cross-linked PNIPAAm to the microheaters, allowing the fluid in other areas to flow freely.By activating the heaters in sets, it was possible to redirect the flow and exchange the fluid in the central area. Figure Figure22 demonstrates how the flow direction in the central microchannel area was changed from a stable horizontal flow to a stable vertical flow with a 3 s response time, using only PNIPAAm phase transition. Constant pressures were applied to the inlets to the horizontal channel and to the vertical channels. Activating heaters V1-4 (Figure (Figure2,2, left) resulted in flow in the horizontal channel only. Likewise, activating heaters H1 and H2 allowed for flow in the vertical channel only. In this sequence, the fluid in the central microchannel area from one inlet was exchanged with fluid from the other inlet. Additionally, by activating heater H3, a particle could be immobilised during the exchange of fluid as shown in Figure Figure33 (top).Open in a separate windowFigure 2Switching between fluid from the horizontal and the vertical channel using hydrogel activation and flow redirection with a response time of 3 s. A pressure of 25 mbar was applied to the inlet of the horizontal channel and a pressure of 20 mbar to the vertical channel. The flow field was determined using particle image velocimetry, in which the displacement of fluorescent seed particles was determined from image pairs generated by laser pulse exposure. Processing was carried out with davis software (LaVision).Open in a separate windowFigure 3A series of microscope images near heater H3 showing: (1a)-(1c) A single yeast cell captured by local PNIPAAm phase transition and immobilized for 5 min before being released. (2a) A single yeast cell was identified for capture by embedding in hydrogel. (2b) The cell as well as the hydrogel displayed fluorescence while embedded due to the introduction of DAPI in the surrounding region. (2c) The diffusion of DAPI towards the cell as the heating power of H3 is reduced after 15 min, showing a DAPI stained yeast cell immobilized.Particle immobilisation in hydrogel and fluid exchange in the central area of the microfluidic network were used to carry out an in situ biochemical process in which a yeast cell injected through one inlet was stained in situ with a 4′,6-diamidino-2-phenylindole (DAPI) solution (Invitrogen), which attached to the DNA of the yeast cell.18 A solution of yeast cells with a concentration of 5 × 107cells/ml suspended in a 10% w/w PNIPAAm solution was injected through the horizontal channel. A solution of 2μg/l DAPI in a 10% w/w PNIPAAm solution was injected through the vertical channel. A single yeast cell was identified and captured near the central heater, and by deactivating the heaters in the vertical channel, DAPI solution was introduced in the microchannels around the hydrogel. After immobilising the cell for 15 min, the heater was deactivated, releasing the cell in the DAPI solution. This process is shown in Figure Figure33 (bottom). The sequence of the heater activation and deactivation in order to immobilize the cell and exchange the fluid is outlined in the supplementary material.21Eriksen et al.15 demonstrated the diffusion of protease K in the porous hydrogel matrix,19 and it was therefore expected that DAPI fluorescent stain (molecular weight of 350 kDa, Ref. 20) would also diffuse. DAPI diffusion is shown in Figure 3(2b) in which the yeast cell shows fluorescence while embedded in the hydrogel. The yeast cell was released by deactivating the central heater and activating all the others to suppress unwanted flow in the microchannel. As a result, the single cell was fully immersed in the DAPI solution. Immobilization of a single cell allows for selection of a cell that exhibits a certain trait and introduction of a new fluid while maintaining the cell position in the field of view of the microscope such that a biochemical response can be imaged continuously.In summary, a microfluidic chip capable of local heating was used to induce phase transition of PNIPAAm to hydrogel, blocking microchannel flow, and thereby allowing for reconfigurable flow. Additionally, the hydrogel was used to embed and immobilise a single yeast cell. DAPI fluorescent stain was introduced using flow redirection, and it stained the immobilized cell, showing diffusion into the hydrogel. The versatile design of this microfluidic chip permits flow redirection, and is suitable to carry out in situ biochemical reactions on individual cells, demonstrating the potential of this technology for forming large-scale reconfigurable microfluidic networks for biochemical applications.  相似文献   

9.
Enhancing conjugation rate of antibodies to carboxylates: Numerical modeling of conjugation kinetics in microfluidic channels and characterization of chemical over-exposure in conventional protocols by quartz crystal microbalance     
Sasan Asiaei  Brendan Smith  Patricia Nieva 《Biomicrofluidics》2015,9(6)
This research reports an improved conjugation process for immobilization of antibodies on carboxyl ended self-assembled monolayers (SAMs). The kinetics of antibody/SAM binding in microfluidic heterogeneous immunoassays has been studied through numerical simulation and experiments. Through numerical simulations, the mass transport of reacting species, namely, antibodies and crosslinking reagent, is related to the available surface concentration of carboxyl ended SAMs in a microchannel. In the bulk flow, the mass transport equation (diffusion and convection) is coupled to the surface reaction between the antibodies and SAM. The model developed is employed to study the effect of the flow rate, conjugating reagents concentration, and height of the microchannel. Dimensionless groups, such as the Damköhler number, are used to compare the reaction and fluidic phenomena present and justify the kinetic trends observed. Based on the model predictions, the conventional conjugation protocol is modified to increase the yield of conjugation reaction. A quartz crystal microbalance device is implemented to examine the resulting surface density of antibodies. As a result, an increase in surface density from 321 ng/cm2, in the conventional protocol, to 617 ng/cm2 in the modified protocol is observed, which is quite promising for (bio-) sensing applications.Microfluidics have been implemented in various bio-medical diagnostic applications, such as immunosensors and molecular diagnostic devices.1 In the last decade, a vast number of biochemical species has been detected by microfluidic-based immunosensors. Immunosensors are sensitive transducers which translate the antibody-antigen reaction to physical signals. The detection in an immunosensor is performed through immobilization of an antibody that is specific to the analyte of interest.2 The antibody is often bound to the transducing surface of the sensor covered by self-assembled monolayers (SAMs). SAMs are organic materials that form a thin, packed and robust interface on the surface of noble metals like that of gold, suitable for biosensing applications.3 Thiolic SAMs have a “head” group that shows a high affinity to being chemisorbed onto a substrate, typically gold. The SAMs'' carboxylic functional group of the “tail” end can be linked to an amine terminal of an antibody to form a SAM/antibody conjugation.3,4 The conjugation process is usually accomplished in the presence of carbodiimides, such as 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC). A yield increasing additive, N-Hydroxysuccinimide (NHS), is often used to enhance the surface loading density of the antibody.4,5A typical reaction for coupling the carboxylic acid groups of SAMs with the amine residue of antibodies in the presence of EDC/NHS is depicted in Figure Figure11.4 NHS promotes the generation of an active NHS ester (k2 reaction path). The NHS ester is capable of efficient acylation of amines, including antibodies (k3 reaction path). As a result, the amide bond formation reaction, which typically does not progress efficiently, can be enhanced using NHS as a catalyst.4Open in a separate windowFIG. 1.NHS catalyzed conjugation of antibodies to carboxylic-acid ended SAMs through EDC mediation (Adapted from G. T. Hermanson, Bioconjugate Techniques, 2nd. Edition. Copyright 2008 by Elsevier4). EDC reacts with the carboxylic acid and forms o-acylisourea, a highly reactive chemical that reacts with NHS and forms an NHS ester, which quickly reacts with an amine (i.e., antibody) to form an amide.A number of groups have studied EDC/NHS mediated conjugation reactions such as the ones depicted in Figure Figure1.1. The general stoichiometry of the reaction involves a carboxylic acid (SAM), an amine (antibody), and EDC to produce the final amide (antibody conjugated SAM) and urea. However, the recommended concentration ratio of the crosslinking reagents inside the buffer, i.e., the ratio of EDC and NHS with respect to adsorbates and each other, varies from one study to another.6 The kinetics of the reactions outlined in Figure Figure11 have also been investigated,4,6–8 but only in the absence of NHS for EDC or carboxylic acids in aqueous solutions.8 A relatively recent experimental study verified the catalytic role of the yield-increasing reagent N-hydroxybenzotriazole (HOBt), which acts similarly to NHS.7 In this study, the amide formation rate (k3 reaction path, Figure Figure1)1) was found to be dependent on the concentration of the carboxylic acid and EDC in the buffer solution, and independent of the amine and catalyst reagent concentration. The same group also showed that the amide bond formation kinetics is controlled by the reaction between the carboxylic acid and the EDC to give the O-acylisourea, which they marked as the rate-determining step (k1 reaction path, Figure Figure11).The k1 reaction path, or the conjugation reaction, is usually a lengthy process and takes between 1 and 3 h.4,9 Compared to k1, the k2 and ?k3 reactions are considerably faster. Microfluidics has the potential to enhance the kinetics of these reactions using the flow-through mode.10,11 This improvement occurs because while conventional methods rely only on diffusion as the primary reagent transport mode, microfluidics adds convection to better replenish the reagents to the reaction surfaces. However, there are many fundamental fluidic and geometrical parameters that might affect the process time and reagents consumption in a microfluidics environment, such as concentration of antibodies and reagents, flow rate, channel height, and final surface density of antibodies. A model that studies the kinetics of conjugation reaction against all these parameters would therefore be helpful for the optimization of this enhanced kinetics.There are a number of reports on numerical examination of the kinetics of binding reactions in microfluidic immunoassays.12–15 All these models developed so far couple the transport of reagents, by diffusion and convection, to the binding on the reaction surface. Myszka''s model assumes a spatially homogeneous constant concentration of reagents throughout the reaction chamber, thus fails to describe highly transport-limited conditions due to the presence of spatial heterogeneity and depletion of the bulk fluid from reagents.16,17 In transport-limited conditions, the strength of reaction is superior to the rate of transport of reagents to the reaction surface.18,19 More recently, the convection effects were included in a number of studies, describing the whole kinetic spectrum from reaction-limited conditions to transport-limited reactions.20–22 Immunoreaction kinetics has also been examined with a variety of fluid driving forces, from capillary-driven flows,20 to electrokinetic flows in micro-reaction patches,21 pressure-driven flows in a variety of geometric designs.22 Despite these comprehensive numerical investigations, the fundamental interrelations between the constitutive kinetic parameters, such as concentration, flow velocity, microchannel height, and antibody loading density, have not been studied in detail. In addition, the conjugation kinetics has not yet been exclusively examined.In this paper, a previous model for immunoreaction is modified to study the antibody/SAM conjugation reaction in a microfluidic system. Model findings are used to examine the process times recommended in the literature and possible modification scenarios are proposed. The new model connects the convective and diffusive transport of reagents in the bulk fluid to their surface reaction. The conjugation reaction is studied against fluidic and geometrical parameters such as flow rate, concentration, microchannel height and surface density of antibodies. Damköhler number is used to compare the reaction and fluidic phenomena present and justify the kinetic trends observed. Model predictions are discussed and the main finding on possible overexposure of carboxylates to crosslinking reagents, in conventional protocols, is verified by comparing the resultant antibody loading densities obtained using a quartz crystal microbalance (QCM) set up. The results demonstrate an improved receptor (antibody) loading density which is quite promising for a number of (bio-) sensing applications.23,24 Major application areas include antibody-based sensors for on-site, rapid, and sensitive analysis of pathogens such as Bacillus anthracis,23 Escherichia coli, and Listeria monocytogenes, and toxins such as fungal pathogens, viruses, mycotoxins, marine toxins, and parasites.24  相似文献   

10.
Preface to Special Topic: Microfluidics in Drug Delivery     
Brigitte Stadler 《Biomicrofluidics》2015,9(5)
In this special topic of Biomicrofluidics, the importance of microfluidics in the field of drug delivery is highlighted. Different aspects from cell-drug carrier interactions, delivery vehicle assembly to novel drug delivery devices are considered. The contributing reviews and original articles illustrate the synergistic outcomes between these two areas of research with the aim to have a positive impact on biomedical applications.Microfluidics is certainly one of the huge success stories when it comes to anticipated impact and fulfilled promises in academic research environments. Microfluidic approaches are game changers in many disciplines in natural science, including (bio)medical science. In the latter case, the fields of biosensing/diagnostics, tissue engineering, and drug discovery/delivery have benefited from concepts which allow for the fast throughput manipulation of fluids at the submillimeter length scale.A key aim in microfluidic-assisted drug discovery is the development of strategies which will facilitate the identification of potential “hits”—new drugs with the anticipated therapeutic benefit. In this context, “organ(disease)-on-chips” are considered as highly sophisticated in vitro models with lower cost and less ethical issues compared to extensive testing in animals. This technology is still very young with countless research challenges to be addressed and eventually overcome, but the few current reports are promising, and include “gut-on-chip,” “cancer-on-chip,” or “blood vessel-on-chip.” Additionally, intravenously injected drug delivery vehicles are exposed to the blood stream and the induced mechanical forces which are likely to affect their interaction with cells and tissue. Therefore, understanding the diffusion phenomena of biomolecules in microfluidic devices as reviewed by Yesil-Celiktas and coworkers in the current special content is crucial.1 What is more, the contribution by Hosta-Rigau and colleagues provides a comprehensive overview over the interaction of drug carriers and cells in microfluidic-based systems which deliver a simple, but yet more realistic model of the dynamic in vivo situation.2 Further, to illustrate the relevance of shear stress when assessing the potential of nanocarriers for drug delivery applications, we assembled novel block copolymers consisting of poly(cholesteryl acrylate) as the hydrophobic core and poly(N-isopropylacrylamide) as the hydrophilic extensions together with lipids into vesicles using the evaporation-rehydration method.3 Following on, we biologically evaluated the assemblies with applied shear stress using macrophages. In a related report by the Chakraborty group, a biocompatible acoustic microfluidic system was outlined including the effect of microbubbles with the applied acoustic field on biological cells.4From a different perspective, droplet microfluidics has become a popular method to assemble a huge diversity of particles of different size, shape, and morphology equipped with options for active or passive drug release. Microfluidics provides unique opportunities and flexibility to fabricate decent amounts of mono-disperse drug carriers using monomers, polymers, lipids, or inorganic precursor materials as building blocks. The assembly of size-tunable polymer/lipid particles by Sun et al.,5 and the fabrication of poly (lactic-co-glycolic acid) nanoparticles incorporated within poly (ethylene glycol) (PEG) microgels by the Chen group,6 provide interesting examples in this context. Further, artefacts associated with this technique have to be addressed and understood to avoid inaccurate and misleading data as reported by Litten et al.7 Microfluidic techniques can also be employed for cell encapsulation. Fan et al. demonstrated the trapping of human colon cancer cells in hydrogel particles with preserved viability and response to inflammatory stimuli.8Novel drug delivery devices which consider microfluidic concepts and set-ups are an interesting addition to traditional approaches. Implantable drug delivery systems provide an alternative to ensure constant drug level in blood without relying on the compliance of the patient while circumventing challenges involved in oral drug delivery coming from drug instability or limited absorbance among others. Yi and coworkers propose a reservoir approach in combination with a heat responsive valve towards the long term delivery of solid drugs.9 What is more, nebulizers, as alternative to inhalers for pulmonary drug delivery, suffer from miniaturization and drug degradation issues. Cortez-Jugo et al. report on a novel portable acoustomicrofluidic device, which successfully nebulized monoclonal antibodies into a fine aerosol mist including the first positive biological evaluation.10Further, combining microfluidics with sensing concepts as illustrated by Knoll and coworker11 is of importance, since the design of drug delivery vehicles strongly relies on the fundamental understanding of the interaction between biomolecules, cells, and tissue.Taken together, these articles give an overview over the use of microfluidics in the area of drug delivery, which goes beyond the assembly of drug carries, but also provides a platform for their biological evaluation or the design of entirely new drug delivery devices. I hope that this collection of articles will stimulate new ideas and future collaborations between engineers/chemists/physicist and biologists towards the common goal to provide solutions for biomedical challenges. Finally, I would like to thank Professor Leslie Yeo for the invitation to be the guest editor for this special topic, and Christine Urso and other editorial and production staffs of Biomicrofluidics for making it a reality.  相似文献   

11.
Two-dimensional and three-dimensional dynamic imaging of live biofilms in a microchannel by time-of-flight secondary ion mass spectrometry     
Xin Hua  Matthew J. Marshall  Yijia Xiong  Xiang Ma  Yufan Zhou  Abigail E. Tucker  Zihua Zhu  Songqin Liu  Xiao-Ying Yu 《Biomicrofluidics》2015,9(3)
A vacuum compatible microfluidic reactor, SALVI (System for Analysis at the Liquid Vacuum Interface), was employed for in situ chemical imaging of live biofilms using time-of-flight secondary ion mass spectrometry (ToF-SIMS). Depth profiling by sputtering materials in sequential layers resulted in live biofilm spatial chemical mapping. Two-dimensional (2D) images were reconstructed to report the first three-dimensional images of hydrated biofilm elucidating spatial and chemical heterogeneity. 2D image principal component analysis was conducted among biofilms at different locations in the microchannel. Our approach directly visualized spatial and chemical heterogeneity within the living biofilm by dynamic liquid ToF-SIMS.Mapping how metabolic pathways are interconnected and controlled at the subcellular scale within dynamic living systems continues to present a grand scientific challenge. Biofilms, consisting of aggregations of bacterial cells and extracellular polymeric substance (EPS), present an important avenue for deciphering complex microbial communities. During biofilm formation, cells assemble in a secreted polymer milieu of polysaccharides, proteins, glycolipids, and DNA.1,2 Microfluidics provides unprecedented control over flow conditions, accessibility to real-time observation, high-throughput testing, and mimics in vivo biological environments.3 An understanding of the mechanism underlying biofilm formation and the design of advanced microfluidic experiments could address challenges such as interpreting microbial community interactions, biofouling, and resistance to antimicrobial chemicals. However, only a handful of biofilm studies used microfluidic approaches that provided hydrated chemical imaging at high spatial resolution.4–7 Most studies utilized confocal microscopy,4 FTIR spectroscopy,5 or other approaches (e.g., high density interdigitated capacitors7) for biofilm monitoring. Imaging mass spectrometry has been demonstrated in biofilm studies.8,9 A coupled microfluidic-imaging mass spectrometry approach would provide the chemical molecular spatial mapping needed to better address the scientific challenge of biofilms.Recently, we developed a portable microfluidic reactor, System for Analysis at the Liquid Vacuum Interface (SALVI),10,11 which overcame the grand challenge of studying liquids with high volatility and liquid interfaces using surface sensitive vacuum instruments. SALVI enables direct imaging of liquid surfaces using electron or ion/molecular based vacuum techniques. Our microfluidic approach used a polydimethylsiloxane (PDMS) microchannel fully enclosed with a thin silicon nitride (SiN) membrane (100 nm thick). For visualization, 2 μm diameter holes were opened in the SiN membrane in vacuo. These detection windows were dynamically drilled using the time-of-flight secondary ion mass spectrometry (ToF-SIMS) primary ion beam (e.g., Bi+).12Unlike liquid sample holders for transmission electron microscopy and scanning transmission electron microscopy, SALVI is self-contained and portable.13 As a result, it can potentially be used in many finely focused analytical tool with minimal adaptation.10 The analytical performance of SALVI has been demonstrated with a variety of analytes ranging from biology to material sciences.14,15 Unlike most microfluidic applications that are only suitable under ambient conditions (e.g., separations, cell and small amount sample manipulation, and thermal flow-sensors),16–18 SALVI is compatible with both in situ ambient and in vacuo spectroscopy analysis and imaging.19 Biofilms have been successfully cultivated inside the microfluidic channel and imaged using correlative confocal laser scanning microscopy (CLSM) and ToF-SIMS.20Our approach opens a new avenue to study biological sample in their natural state. Although ToF-SIMS has been widely used for providing molecular signatures of organic and biological molecules in complex biological systems21,22 or lipid spatial mapping,23 the vacuum-based ToF-SIMS generally requires solid (either dried24 or cryo treated25) samples. Here, we report ToF-SIMS two dimensional (2D) and three dimensional (3D) chemical images of hydrated biofilms. In situ time and space-resolved identifications of fatty acid (FA) fragments characteristic of Shewanella are illustrated by 3D images reconstructed from the ToF-SIMS depth profile time series. Principal component analysis (PCA) further elucidates biofilm chemical and spatial heterogeneity and shows the key chemical component at different depth and location of the biofilm including the biofilm-surface attachment interface.For all growth experiments, two samples were cultured simultaneously. At days 5 and 6, one sample was harvested for immediate analysis, respectively, using a ToF-SIMS V spectrometer (IONTOF GmbH, Münster, Germany). Similar results were obtained from both samples, because the biofilm-attachment surface was probed. For consistency, only day 6 data are shown here, while additional data are provided in the supplementary material.28 2D and 3D image visualizations were obtained using the IONTOF instrument software. PCA was performed using MATLAB R2012a (MathWorks, Inc., Natick, MA, USA). 2D images of .bif format were converted and integrated into a matrix. Data were pretreated by normalization to total ions, square root transformation, and then mean centering.26 For m/z spectra PCA, unit mass peaks from m/z 199 to m/z 255 were used (see Figure S-228). Unit mass peaks from m/z 1–300 were also used and results are comparable (see Figure S-328). Five characteristic FA peaks (m/z 199, 213, 227, 241, and 255, corresponding to C12, C13, C14, C15, and C16 FAs) were used in image PCA.27 Images representing each PC were reconstructed from the score matrix using the red, green, and blue (RGB) color scale.Using depth profiling, we drilled through the SiN membrane and collected depth-resolved images of the live biofilm (Figure 1(a)). Our analysis of the negative ToF-SIMS spectra after SiN punch-through showed Shewanella FA fragments in the m/z 195–255 range.20 From the depth profile time series, we selected five regions (highlighted as I, II, III IV, and V) within the FA m/z range to visualize 2D spatially resolved images collected for 46 s (1000 scans) before (I), during (II), or after (III, IV, V) SiN membrane punch-through.20 When false color 2D images of FA fragments characteristic of Shewanella biofilms were selected from the dynamic depth profiling data, differences were observed (Figure 1(b)) among the five regions. Furthermore, the biofilm images after SiN membrane punch-through (III, IV, V) displayed variations across the 2 μm diameter surfaces, with C12 (m/z 199) being distributed across regions III, IV, and V and C15 (m/z 241) FAs mostly in region V (see Figure S-4 for additional FA images28). This suggested that depth-resolved chemical heterogeneities were present in the biofilm. To illustrate, we reconstructed the 2D images from depth profiling data within the biofilm region (from the beginning of III through the end of V) and show spatially resolved 3D chemical images within the entire sample (Figure 1(c) and movies S1-S328). The reconstructed 3D images revealed the heterogeneous spatial distribution overlay for C12 (red) and C15 (green) FAs during 302 s biofilm depth profiling from day 5 (Figure S-528) and day 6 (Figure 1(c)).Open in a separate windowFIG. 1.(a) ToF-SIMS depth profiling of the day 6 biofilm attached to the SiN membrane in the microfluidic channel. Five regions representing sample before SiN punch-through (I) during punch-through (II) or within the biofilm region (III, IV, and V) are illustrated. (b) 2D false color images of day 6 biofilm FAs at the five time regions highlighted in (a). (c) Reconstructed 3D day 6 biofilm images showing FA fragment distributions within the entire biofilm region (III–V, 302 s). The time axis represents depth profiling from near the SiN surface into the biofilm. (d) Spectra PCA score plot of day 6 biofilm showing the differences and similarities among selected five regions (m/z 199–255). A 95% confidence limit for each region was defined by an ellipse with the same color to the corresponding region clusters. (e) Loadings of PC1 and PC2 corresponding to (d) and the plot of PC variance contributions.Spectral PCA was used to analyze the m/z spectra. The deepest region (V) into the biofilm was the most different from the other two biofilm regions (III and IV), further confirming the heterogeneities observed in the 2D images (e.g., C12 and C15 FA fragments) contributing most to this spatial difference. In addition, C12 FA fragments played a key role in the biofilms imaged near the SiN membrane attachment surface (III and IV). When inspected individually, C12 FAs were observed throughout the entire biofilm region, suggesting that C12 FA fragments may play a role in biofilm attachment to a surface and they may be main components of EPS throughout the biofilm. In contrast, C15 FAs were more abundant deeper within the biofilm, indicating that they may be more relevant to bacteria cells themselves.Uniform sputtering rate was assumed during depth profiling. To better determine the depth and shape of the SIMS ionization crater, AFM measurements were collected using an agarose sample in the SALVI reactor as a proxy for the biofilms (Figure S-628). The AFM results showed that the 100 nm SiN was drilled through and confirmed that the biofilm interface was probed by ToF-SIMS. Ideally, real-time correlative AFM and ToF-SIMS measurements will be needed due to the self-healing property of biofilms. However, such capability is currently under development.To further analyze chemical differences within biofilms, we performed ToF-SIMS depth profiling at three locations along the microchannel; namely, the inlet, center, and outlet as illustrated in Figure S-1(b).28 At each location, we defined the five regions described in Figure 1(a), and 2D image PCA analysis was conducted on the biofilm region (from the beginning of III through the end of V) to visualize the chemical distributions on day 6. Figure 2(a) shows the loading plots for the m/z peaks that contribute to each PC image (Figure 2(b)). The first three PCs explained 93.79% of the variance within the data. For PC1, the strongest positive loading fragments were C12 and C15 FAs, which are the bright red areas in three PC1 images. The C12 FAs were the main contributor to the green regions in the PC2 image. The strongest loading for PC3 in blue was C14 FAs. Compared to PC1 and PC2, PC3 played a limited contribution to the overall spatial distribution discrimination. The merged images give a demonstration of chemical spatial distribution of key components of biofilms in the liquid microenvironment.Open in a separate windowFIG. 2.(a) Image PCA loading plots illustrating the contribution of each FA peak in the day 6 biofilm at three locations within the microfluidic channel. The variance contributions of each PC are shown at the bottom. (b) Reconstructed false-color 2D PCA images in RGB corresponding to each PC scores at these locations along the microfluidic channel. The RGB composite images of the three key PCs are depicted in the bottom. Only data within the 2 μm diameter circle were considered in analysis.Our results show that SALVI and liquid ToF-SIMS studies of live biofilms offer dynamic, depth-resolved chemical mapping and produce 2D and 3D visualizations of spatial heterogeneity within a biofilm. Chemical imaging of biofilms near the attachment interface can enhance our understanding of biofilm formation in environmental, medical, and industrial settings. Our approach provides a universal portable platform and enables in situ probing of complex living biological systems potentially across multiple time and space scales. Because of the portability and vacuum compatibility, SALVI offers a valuable linkage with proteomic mass spectrometry via microfluidics and a nondestructive package for integrative in situ analysis of live biological systems in system biology.  相似文献   

12.
Bacteria under the physical constraints of periodic micro-nanofluidic junctions reveal morphological plasticity and dynamic shifting of Min patterns     
Jie-Pan Shen  Chia-Fu Chou 《Biomicrofluidics》2014,8(4)
Morphological plasticity is an important survival strategy for bacteria adapting to stressful environments in response to new physical constraints. Here, we demonstrate Escherichia coli morphological plasticity can be induced by switching stress levels through the physical constraints of periodic micro-nanofluidic junctions. Moreover, the generation of diverse morphological aberrancies requires the intact functions of the divisome- and elongasome-directed pathways. It is also intriguing that the altered morphologies are developed in bacteria undergoing morphological reversion as stresses are removed. Cell filamentation underlies the most dominant morphological phenotypes, in which transitions between the novel pattern formations by the spatial regulators of the divisome, i.e., the Min system, are observed, suggesting their potential linkage during morphological reversion.Most bacteria have evolved sophisticated systems to manage their characteristic morphology by orchestrating the spatiotemporal synthesis of the murein sacculus (peptidoglycan exoskeleton), which is known to be the stress-bearing component of cell wall and presides over de novo generation of cell shape.1 Morphological plasticity is attributed to a bacterial survival strategy as responding to stressful environments such as innate immune effectors, antimicrobial therapy, quorum sensing, and protistan predation.2 It comes of no surprise that stress-induced diversified morphology and mechanisms, ascribed to shape control and determination, have drawn great attention in both fundamental and clinical studies.3–6 The molecular mechanism to form filamentous bacteria has been revealed that both β-lactam antibiotics3 and oxidative radicals produced by phagocytic cells5 trigger the SOS response, promoting cell elongation by inactivating cell division via the blockade of tubulin-like FtsZ, known as the divisome initiator. While apart from the scenario of length control by the divisome-directed filamentation, the elongasome assembled by proteins associated with actin-like MreB complex1,7,8 helps the insertion of peptidoglycans into lateral cell wall, suggesting the role in the determination of cell diameter during cell elongation.Recently, additional mechanisms other than the divisome/elongasome-directed pathways of shape maintenance are discovered to regenerate normal morphology de novo from wall-less lysozyme-induced (LI) spheroplasts of E. coli via a plethora types of aberrant division intermediates.9 Similar morphological reversion from different aberrant bacterial shapes has been observed as squashed wild-type bacteria generated through sub-micron constrictions are released into connected microchambers.10 Previous work using the microfluidic approach focuses on the septation accuracy and robustness of constricted bacteria,11 but the reversion process of stress-released bacteria is not well studied and analyzed. In particular, the aberrant bacterial shape is mainly branched-type with bent and curved variants in the reverting bacteria, analogous to the aberrant intermediate found in the morphological reversion of LI spheroplasts with PBP5-defective mutant.9 Since bacteria suffering from starvation12 or confronting mechanical stresses exerted by phagocytosis and protistan grazing6 can induce morphological alterations, one could manipulate the stress levels of physical constraints by adopting repeated structures of sub-micron constricted channels (nanoslits) and microchambers,10,11 to select and enrich bacteria converting to specified aberrant intermediates. The stress incurred by the nanoslit on bacteria is about the mechanical intervention over de novo synthesis of the cell wall, which is the major factor causing morphological aberrancy, while the second environmental stress comes from bacterial growth in the restricted space of microchamber as bacteria proliferate to full confluency, resulting in growth pressure of high population density, nutrient deficiency, and the size reduction of bacteria.Here, we report the selection of distinctive bacterial morphologies by size shrinkage in the outlet cross-section (W × H = 1.5 × 1.5 μm) of the terminal microchamber in the periodic structures of nanoslit-microchamber (Figs. 1(a) and 1(b)). The fluidic structures were micropatterned on fused silica wafers by photolithography, fabricated through reactive ion etching (RIE) and inductively coupled plasma (ICP) etching, and encapsulated by cover glasses coated with polydimethylsiloxane (PDMS) or polysilsesquioxane (PSQ) layer as described earlier.13,14 Two days after the outgrowth of Escherichia coli (imp4213 [MC4100 ΔlamB106 imp4213]) loaded to the microfluidic device at 25 °C, bacteria started to penetrate into the nanoslit as they proliferated to full confluency in the first microchamber (Fig. 1(c)). It takes about 10 days for bacteria traversing 500 μm long (5 repeated nanoslit-microchamber units) via proliferations and being released from the outlet of the terminal microchamber. The narrowed outlet allows only bacteria with smaller diameters to be squeezed into the spacious and nutrient-rich region, thus it acts as a spatial filter to avoid the passage of branching bacteria with cross-sectional size larger than that of the outlet. The rationale of this design is to select aberrant bacteria prone to promote de novo shape regeneration other than the branched-type, which is the dominant morphology of reverting bacteria in the prior microfluidic constriction study.10 As anticipated, the stress-released bacteria through the narrowed outlet are therefore mostly filamentous (see statistical analysis for cell morphology in the supplementary material).15 However, it is noted that the aberrant morphology of lemon-like shape with tubular poles (Figs. 1(d-1), 1(d-3), and 1(d-11)) is developed about 3 h after the stress-released bacteria escaped through the outlet. Though the generation of the lemon-like aberrancy in bacteria has been reported in PBP5/7-defective E. coli mutant subjected to a high-level inhibition of both MreB and FtsZ, while the same mutant treated with low-level MreB inhibitor, together with antagonized-FtsZ, displays filamentous shape with varying diameters,16 these morphological aberrances can be observed in our system (Figs. 1(d-2) and 1(d-12)). Besides, a high-level inhibition of MreB in E. coli with an intact divisome function is known to cause round bacteria, resembling to the cell morphology of the bacteria shown in Fig. 1(d-4). Interestingly, parallel experiments using bacteria mutants carrying impaired regulatory functions in either the divisome (Min) or the elongasome (MreB) do not develop morphological plasticity (supplementary Fig. S1).15 Taken together, the filamentous and lemon-like variants selected from our microfluidic platform, while elaborating the morphological plasticity and reverting progression, require both the functional divisome/elongasome. Alternatively, the selection by the spatial filter does not fully exclude cells with aberrant shapes such as the branched-type with initial budding (Fig. 1(d-7)), cells with asymmetric cross-section perpendicular to the longitudinal axis (Figs. 1(d-2), 1(d-8), 1(d-9), 1(d-9′), and 1(d-10)), and those resembling to the morphological phenotypes of the division intermediates reported in the LI-spheroplasts carrying genetic defects on some non-cytoskeletal proteins (Figs. 1(d-5) and 1(d-6)). In particular, intracellular vesicles and cell autolysis are observed in some reverting bacteria (Figs. 1(d-5) and 1(d-6)), which are reminiscent to the phenomena reported in the division intermediates of the LI-spheroplasts lacking stress response system (Rcs) or some accessory proteins (PBP1B and LpoB). Unlike the bacteria grow with odd shapes under the stress of nanofluidic confinement only10 (Fig. 1(c)), all the morphological aberrancy reported here are developed in the reverting bacteria, which grow in the spacious and nutrition-rich environment and are free from physical constraints. Further investigations over the expression levels of the divisome/elongasome networks and the stress-response system in bacterial cells subjected to micro-nanofluidic junctions could be insightful in understanding their role in bacterial shape control.9Open in a separate windowFIG. 1.(a) Schematics of the microfluidic device used in this study with an H-shaped geometry (left upper panel), where repeated nanoslit (L×W×H = 50×10×0.4 μm)−microchamber (L×W×H = 50×50×1.5 μm) structures are bridged between two arms of the H-shaped microchannels (left lower panel and enlarged view in right panel). (b) Top-view layout of an individual channel in (a) with close view of the outlet in the terminal microchamber (orange: nanoslits; blue: microchambers). (c) Fluorescence micrograph of E. coli imp4213 penetrating a nanoslit (scale bar: 5 μm). (d) Bright-field micrographs for various cell morphology of the selected imp4213 released from the outlet (magenta arrows: cells with vesicles; scale bar: 5 μm). (e) Sequential bright-field micrographs of morphological reversion. T1–T3 indicate the time after bacteria escaping from the outlet. T1: 3 h; T2: 6 h; T3: 24 h. Scale bar: 10 μm.During the morphological reversion, the stress-released bacteria rapidly increase their size in the first 3 h after escaping from the terminal microchamber (T1 in Fig. 1(e)). Some filamentous bacteria even grow over 50 μm long, though such a morphological phenotype implicates the cessation of functional divisome. With active growth and proliferation, the progeny of stress-released bacteria increase their population but gradually reduce their size about 6 h after being released from the constriction stress (T2 in Fig. 1(e)). Fig. Fig.22 displays the marginal histograms for different shape factors, where Fig. 2(a) is the plot of the minimal Feret diameter (cell diameter) versus Feret diameter (cell length), i.e., the shortest versus the longest distance between any two points with parallel tangents along the cell peripheral, respectively, indicating that cell diameters are larger for reverting bacteria at T1 (mean ± S.E.M. = 1.89 ± 0.08 μm) with respect to T2 (1.51 ± 0.06 μm). Moreover, the histogram of Feret diameter depicts two major populations of the cell length for reverting bacteria at T1, which mostly resume to typical cell length at T2 (the median of Feret diameter = 3.33 μm; see statistical analysis for Fig. Fig.22 in the supplementary material).15 The shape factors of circularity (4π × [area]/[perimeter]2) and aspect ratio ([major axis]/[minor axis] for the cell geometry fitted to an ellipse) confirm the existence of dual populations for bacteria at T1 as well (Fig. 2(b)). About 24 h after escaping (T3 in Fig. 1(e)), almost all the progeny of stress-released bacteria regained the rod shape.Open in a separate windowFIG. 2.Marginal histograms for shape factors measured from the reverting imp4213 at T1 and T2. (a) Minimal Feret diameter (cell diameter) versus Feret diameter (cell length). (b) Circularity versus aspect ratio. N = 366 for T1 and N = 494 for T2.The bacterial size reduction of filamentous and lemon-like shape variants, though involving negative control of the divisome positioning by the spatial regulators of MinCDE system,17 is not completely understood as to how they coordinate in aberrant geometries. Besides, the filamentation of stress-released bacteria during the period of T1 to T2 implicates the inhibition of functional divisome. With minimal perturbation of the divisome by leaky expression of GFP-MinD and MinE (imp4213/Plac-gfpmut2::minD minE), the patterning dynamics of GFP-MinD in different bacterial morphology were time-lapse imaged during morphological reversion. Intriguingly, more than the standing-wave-like pattern of MinD denoted in filamentous E. coli,18 we discovered bidirectional drifting of two standing-wave-like patterns of MinD occur in most reverting bacteria filaments (supplementary Figs. S2(a) and S2(b)).15 The bidirectional drifting in the longitudinal direction of the cells may be emanating from the cell poles (the blue upper panel of Fig. 3(a) and supplementary Fig. S2(c)15) and the cylinder region (the blue lower panel of Fig. 3(a) and supplementary Fig. S2(d)15). Furthermore, the MinD pattern transitions from the standing to traveling waves are occasionally observed (the lower panel of Figs. 3(a) and supplementary Fig. S2(e)15). Notably, the standing-wave-like MinD patterns exhibit bidirectional drifting along the cell longitudinal direction and intermittently change directions, implying the competition between coexisting MinD patterns can be supported under filamentous geometry. Despite there have been observations of multiple wave-packet of traveling waves in filamentous cells,19 the mixture of distinct wave-like MinD patterns have never been experimentally reported. While most intriguingly, multiple drifting movements of wave-like MinD patterns potentiate the mitigation of periodic minima in time-averaged Min gradient in the reverting filamentous bacteria, suggesting the disability of proper divisome positioning for recovering the typical rod shape. Apart from the wave-like movements, amoeba-like motion of Min proteins has been shown in vitro upon synthetic minimal system, but never been verified in vivo.20 Strikingly, here amoeba-like motion of MinD is the dominant mode in lemon-like bacteria and the transitions between wave-like patterns and amoeba-like motion are supported even under filamentous geometry (Figs. 3(b) and 3(c), Multimedia view).Open in a separate windowFIG. 3.Kymographs for GFP-MinD dynamics in selected imp4213 cells during morphological reversion: (a) Mixture modes of standing wave packets and traveling wave. The left panel is the stacked fluorescence micrograph displaying cell shape (scale bar = 5 μm). The kymograph is derived from the filamentous cell indicated by the green arrow (scale bar: 120 s horizontal; 5 μm vertical), where the lower panel follows the upper panel in time. The yellow windows indicate bidirectional-drifting standing wave packets, while the green indicates traveling waves (see also supplementary Fig. S2).15 (b) Sequential fluorescence micrographs of GFP-MinD in lemon-shape imp4213 show amoeba-like motion, with the first left a bright-field image (scale bar: 10 μm). (c) Mixed modes of amoeba-like motion and waves in selected filamentous imp4213 cell indicated by the green arrow in the left panel (scale bar = 5 μm). The filamentous cells depicted in (a) and (c) locate at the top region while the lemon-shape cell in (b) at the central region of the movie (time stamp in min:s). (Multimedia view) [URL: http://dx.doi.org/10.1063/1.4892860.1]In summary, we have demonstrated that the development of bacterial morphological plasticity can be stress-induced by periodic physical constraints with intact functions of the divisome and elongasome-directed pathways. Through size exclusion, the constricted outlet structure designed in our microfluidic device is useful in selecting bacteria with plethora morphological aberrancies other than the branched type. Interestingly, disparate morphological changes, rather than those being directly induced under a stressful environment, can be generated in the stress-released bacteria experiencing morphological reversion. Further, the discovery of novel transitions between the Min patterns in most reverting bacteria implicates its regulatory effect of cell filamentation. However, by exploiting the micro-nanofluidic approach, further investigations of the mechanism underlying the development of morphological plasticity in bacteria adapting to physical constraints are expected in future studies to gain more insights into the molecular basis of shape generation.  相似文献   

13.
Preface to Special Topic: Dielectrophoresis     
Ronald Pethig 《Biomicrofluidics》2010,4(2)
This Special Topic section is on dielectrophoresis, a growing area of widespread interest and relevance to the microfluidics and nanofluidics community.There was a time when the arrival of a telegram from the local post office would foreshadow a step-function change in one’s equilibrium. An internet service provider can now deliver the same effect, as illustrated by an unexpected e-mail from Leslie Yeo inquiring if I would “be interested in guest editing a special issue of Biomicrofluidics on recent advances in dielectrophoresis (DEP).” Flattery directed towards vanity can produce interesting results—which I hope this special issue of Biomicrofluidics demonstrates. The rationale for this special issue is the belief of the journal’s Editors (Dr. Chia Chang and Dr. Leslie Yeo) that dielectrophoresis is a growing area of widespread interest and relevance to the microfluidics and nanofluidics community. Papers, both fundamental and applied, were solicited from the leaders working across this broad interdisciplinary area of research. I was delighted by the positive responses of those whose invited contributions appear in this special issue—efforts certainly not motivated by vanity but through enthusiasm for the subject. Some of those invited to contribute were unable to do so because of other demands on their time. Ongoing advances being made in DEP, especially in its various applications, will surely merit another special issue in the future and hopefully include contributions from those unable to do so now.Two of the papers in this special issue address fundamental aspects of dielectrophoresis (DEP), namely the influences on DEP from electrical double-layers and from particle-particle interactions. Consideration of electrical double layers associated with charged particle surfaces is particularly important for nanoparticles because their effective polarizabilities, associated with field-induced dynamics of the counterions and co-ions in the double layer, can dominate over the intrinsic polarizability of the particle itself. This can influence, for example, to what extent the observation of changes in the DEP crossover frequency (marking the transition between positive and negative DEP) can be relied upon in new immunoassays based on the DEP behavior of functionalized nanoparticles. By considering the electrodynamics of double layers, Basuray et al.1 propose a theory to predict how the DEP crossover frequency will vary as a function of particle size and the ionic strength of the suspending electrolyte. In their paper, Sancho et al.2 derive a theoretical model to describe how particle-particle interactions (e.g., “pearl-chaining”) influence the DEP crossover frequency value. This model also describes well the changes in electrorotation and a newly observed precession effect as particles approach each other under the influence of a rotating field.DEP at the nanoscale is also addressed in contributions from the groups of Ralph Hölzel, Junya Suehiro, and Karan Kaler. Thus, Henning et al.3 describe a new method, based on the measurement of capacitance changes between planar microelectrodes, for the automatic acquisition of the DEP properties of nanoparticles without the need for labeling protocols or visual observations. Suehiro4 describes how DEP can be employed as a bottom-up approach for fabricating nanomaterial-based devices such as a carbon nanotube gas sensor and a ZnO nanowire photosensor. Kaler et al.5 describe how the DEP manipulation of miniscule amounts of polar aqueous samples, a method known as liquid-DEP, can be used for on-chip bioassays, such as nucleic acid analysis, and through parallel sample processing offer the potential for conducting automated multiplexed assays. The use of DEP to selectively trap and separate cells has been investigated over many years, and contributions from the groups of Hywel Morgan, Ana Valero, Masau Washizu, and Gerard Markx describe the latest advances and applications. Thomas et al.6 describe a new automated DEP cell trap design for the isolation, concentration, separation, and recovery of human osteoblast-like cells from a heterogeneous population. Recovery of small populations of human osteoblast-like cells with a purity of 100% is demonstrated. A cell-sorting device, based on the opposition of DEP forces that discriminates between cell types according to such properties as their membrane permittivity and cytoplasm conductivity, is described by Valeroet al.7 The versatility of the device is demonstrated by synchronizing a yeast cell culture at a particular phase of the cell cycle. Gel et al.8 describe a DEP-assisted cell trapping method for fusing pairs of cells in an array of micro-orifices. This method produces not only a high yield of viable cell fusants, but also allows for subsequent study of postfusion cell development. Zhu et al.9 describe a DEP-based microfluidic separation system in which dead and active cells can be collected from a given cell suspension, whilst at the same time eluting dormant cells. In the second paper from Gerard Markx’s group, Zhu et al.10 demonstrate that the rate-limiting resuscitation of a colony of dormant bacteria is determined by the diffusion of a resuscitation-promoting factor into the colony interior. This study involved the artificial engineering of different sizes and shapes of bacterial aggregates using DEP forces. Finally, in my own contribution,11 I have attempted to summarize the growing output of DEP publications in terms of their contributions to the theory, technology, and applications of DEP.  相似文献   

14.
Preface to Special Topic: Papers from the 2009 Conference on Advances in Microfluidics and Nanofluidics, The Hong Kong University of Science & Technology, Hong Kong, 2009     
Leslie Y. Yeo 《Biomicrofluidics》2009,3(2)
The inaugural conference on Advances in Microfluidics and Nanofluidics was held at the Hong Kong University of Science and Technology on 5–7 January 2009 and brought together leading researchers from across a wide variety of disciplines from North America, Europe, Asia, and Oceania. This Special Topic section forms the second of the two issues dedicated to original contributions covering both fundamental physicochemical aspects of microfluidics and nanofluidics as well as their applications to the miniaturization of chemical and biological systems that were presented at the conference.In the last five years, we have observed rapid growth in the microfluidics and nanofluidics community in Asia, owing largely to the substantial strategic investments by both government and industry in the region to promote the microfabrication and nanotechnology sectors.1 The organization of a regular meeting focusing on activities in the Asia-Pacific rim region was, therefore, timely, particularly to enhance dissemination of research of the highest quality within the region and to promote collaboration between researchers in the Asian community with their counterparts from Europe and the USA.Biomicrofluidics is, therefore, proud to be closely involved with the organization of the first of such conferences, Advances in Microfluidics and Nanofluidics 2009, which was kindly hosted by the Hong Kong University of Science and Technology (HKUST). As reported in the preface to the first of the two issues dedicated to invited reviews and original contributions associated with the conference,2 the meeting, which took place over three days in the breathtaking HKUST campus overlooking Clearwater Bay in Hong Kong, was a tremendous success. Together with our colleagues, the Biomicrofluidics editors are busy putting in place arrangements for a follow-up meeting in January 2011. Given the overwhelming response and positive feedback we’ve had to date, we believe that Advances in Microfluidics and Nanofluidics will form a regular event in the calendar of the Asian microfluidics and nanofluidics community in the future.It was particularly pleasing to observe the translation of fundamental and theoretical work into advanced applied chip-based platforms for a variety of practical chemical and biological applications in the talks presented at the conference. The collection of articles in this second part, in fact, provides a gist of the flavor of the multidisciplinary research spanning the entire fundamental to applied research spectrum, which is exactly the scope which the journal intends to cover.Electrokinetics continues to be a dominant theme in this issue and within the microfluidics and nanofluidics community. The article by Ng et al.3 provides experimental evidence that might put to rest a longstanding area of debate within the electrokinetics community on the role of Faradaic charging in driving electro-osmotic flow, first proposed by Ben and Chang.4 In other electrokinetics papers, the role of interfaces is explored, for example, electrowetting on the superhydrophobic nanostructured surfaces of a lotus leaf5 and droplet manipulation in an immiscible dielectric liquid continuum under an electric field.6In addition, the characterization of the surface charge density of the nanopores etched in organic foils is reported by Xue et al.,7 which provides a deeper understanding of the mechanisms by which ions are transported in nanochannels, whereas Wei and Hsiao8 present a stochastic simulation to model the condensation of linear polyelectrolyte molecules under electric fields, in which they show the marked increase in the mobility of the polyelectrolyte chain during its unfolding in free-solution electrophoresis.Continuing along the theme of numerical simulations, particulate transport in converging-diverging microchannels was studied using a Lagrangian-Eulerian finite-element model,9 and slip arising in Couette flows over superhydrophobic surfaces was studied using a hybrid multiscale simulation that interfaces molecular dynamics simulations in the near-wall region with the continuum fluid model in the bulk.10 In other numerical studies, drop coalescence11 and nanotube transport12 were studied.Complementing these fundamental studies is the use of multiphase flows in microfluidic channels to engineer scaffolds for tissue engineering in which the bubbles trapped in liquid droplets transported in microchannels were employed to produced the pores of the scaffold.13 Other practical microfluidics applications, such as chip-based enhancement of DNA hybridization through a genetic-bead-based protocol14 and an automated ELISA chip for chemical-biological analysis with an enhancement in the detection range and time,15 also constitute papers in this Special Topic section.We hope you enjoy reading the papers in this Special Topic section and that it provides you with a feel for the broad multidisciplinary spectrum across fundamental and applied microfluidic and nanofluidic research that the conference, as well as the journal, intends to span. Do watch out for the conference announcement for the next Advances in Microfluidics and Nanofluidics meeting in 2011 on the Biomicrofluidics website (http://bmf.aip.org)—hope to see you there!  相似文献   

15.
Development of vertical SU-8 microneedles for transdermal drug delivery by double drawing lithography technology     
Zhuolin Xiang  Hao Wang  Aakanksha Pant  Giorgia Pastorin  Chengkuo Lee 《Biomicrofluidics》2013,7(6)
Polymer-based microneedles have drawn much attention in transdermal drug delivery resulting from their flexibility and biocompatibility. Traditional fabrication approaches are usually time-consuming and expensive. In this study, we developed a new double drawing lithography technology to make biocompatible SU-8 microneedles for transdermal drug delivery applications. These microneedles are strong enough to stand force from both vertical direction and planar direction during penetration. They can be used to penetrate into the skin easily and deliver drugs to the tissues under it. By controlling the delivery speed lower than 2 μl/min per single microneedle, the delivery rate can be as high as 71%.Microelectromechanical systems (MEMS) technology has enabled wide range of biomedical devices applications, such as micropatterning of substrates and cells,1 microfluidics,2 molecular biology on chips,3 cells on chips,4 tissue microengineering,5 and implantable microdevices.6 Transdermal drug delivery using MEMS based devices can delivery insoluble, unstable, or unavailable therapeutic compounds to reduce the amount of those compounds used and to localize the delivery of potent compounds.7 Microneedles for transdermal drug delivery are increasingly becoming popular due to their minimally invasive procedure,8 promising chance for self-administration,9 and low injury risks.10 Moreover, since pharmaceutical and therapeutic agents can be easily transported into the body through the skin by microneedles,11, 12 the microneedles are promising to replace traditional hypodermic needles in the future. Previously, various microneedles devices for transdermal drug delivery applications have been reported. They have been successfully fabricated by different materials, including silicon,13 stainless steel,14 titanium,15 tantalum,16 and nickel.17 Although microneedles with these kinds of materials can be easily fabricated into sharp shape and offer the required mechanical strength for penetration purpose, such microneedles are prone to be damaged18 and may not be biocompatible.19 As a result, polymer based microneedles, such as SU-8,20, 21 polymethyl meth-acrylate (PMMA),22, 23 polycarbonates (PCs),24, 25 maltose,26, 27 and polylactic acid (PLA),28, 29 have caught more and more attentions in the past few years. However, in order to obtain ultra-sharp tips for penetrating the barrier layer of stratum corneum,30 conventional fabrication technologies, for instances, PDMS (Polydimethylsiloxane) molding technology,31, 32 stainless steel molding technology,33 reactive ion etching technology,34 inclined UV (Ultraviolet) exposure technology,35 and backside exposure with integrated lens technology36 are time-consuming and expensive. In this paper, we report an innovative double drawing lithography technology for scalable, reproducible, and inexpensive microneedle devices. Drawing lithography technology37 was first developed by Lee et al. They leveraged the polymers'' different viscosities under different temperatures to pattern 3D structures. However, it required that the drawing frames need to be regular cylinders, which is not proper for our devices. To solve the problem, the new double drawing lithography is developed to create sharp SU-8 tips on the top of four SU-8 pillars for penetration purpose. Drugs can flow through the sidewall gaps between the pillars and enter into the tissues under the skin surface. The experiment results indicate that the new device can have larger than 1N planar buckling force and be easily penetrated into skin for drugs delivery purpose. By delivering glucose solution inside the hydrogel, the delivering rate of the microneedles can be as high as 71% when the single microneedle delivery speed is lower than 2 μl/min.An array of 3 × 3 SU-8 supporting structures was patterned on a 140 μm thick, 6 mm × 6 mm SU-8 membrane (Fig. (Fig.1a).1a). Each SU-8 supporting structure included four SU-8 pillars and was 350 μm high. The four pillars were patterned into a tubelike shape on the membrane (Fig. (Fig.1b).1b). The inner diameter of the tube was 150 μm, while the outer diameter was 300 μm. SU-8 needles of 700 μm height were created on the top of SU-8 supporting structures to ensure the ability of transdermal penetration. Two PDMS layers were bonded with SU-8 membrane to form a sealed chamber for storing drugs from the connection tube. Once the microneedles entered into the tissue, drugs could be delivered into the body through the sidewall gaps between the pillars (Fig. (Fig.1c1c).Open in a separate windowFigure 1Schematic illustration of the SU-8 microneedles. (a) Overview of the whole device; (b) SU-8 supporting structures made of 4 SU-8 pillars; and (c) enlarged view of a single SU-8 microneedle.The fabrication process of SU-8 microneedles is shown in Fig. Fig.2.2. SU-8 microneedles fabrication started from a layer of Polyethylene Terephthalate (PET, 3M, USA) film pasted on the Si substrate by sticking the edge area with kapton tape (Fig. (Fig.2a).2a). The PET film, a kind of transparent film with poor adhesion to SU-8, was used as a sacrificial layer to dry release the final device from Si substrate. A 140 μm thick SU-8 layer was deposited on the top of this PET film. To ensure a uniform surface of this thick SU-8 layer, the SU-8 deposition was conducted in two steps coating. After exposed under 450 mJ/cm2 UV, the membrane pattern could be defined (Fig. (Fig.2b).2b). In order to ensure an even surface for following spinning process, another 350 μm SU-8 layer was directly deposited on this layer in two steps without development. With careful alignment, an exposure of 650 mJ/cm2 UV energy was performed on this 350 μm SU-8 layer to define the SU-8 supporting structures (Fig. (Fig.2c).2c). The SU-8 structure could be easily released from the PET substrate by removing the kapton tape and slightly bending the PET film. Two PDMS layers were bonded with this SU-8 structure by a method reported by Zhang et al.38 (Fig. (Fig.2d2d).Open in a separate windowFigure 2Fabrication process for SU-8 microtubes. (a) Attaching a PET film on the Si substrate; (b) exposing the first layer of SU-8 membrane without development; (c) depositing and patterning two continuous SU-8 layers as sidewall pillars; (d) releasing the SU-8 structure from the substrate and bonding it with PDMS; (e) drawing hollowed microneedles on the top of supporting structures; (f) baking and melting the hollowed microneedles to allow the SU-8 flow in the gaps between pillars; and (g) drawing second time on the top of the melted SU-8 flat surface to get microneedles.In our previous work,39 we used one time stepwise controlled drawing lithography technology for the sharp tips integration. However, since the frame used to conduct drawing process in present study is a four-pillars structure rather than a microtube, the conventional drawing process can only make a hollowed tip but not a solid tip structure (Fig. (Fig.3).3). This kind of tip was fragile and could not penetrate skin in the practical testing process. To solve the problem, we developed an innovative double drawing lithography process. After bonding released SU-8 structure with PDMS layers (Fig. (Fig.2d),2d), we used it to conduct first time stepwise controlled drawing lithography37 and got hollowed tips (Fig. (Fig.2e).2e). Briefly, the SU-8 was spun on the Si substrate and kept at 95 °C until the water inside completely vaporized. Device of SU-8 supporting structures was fixed on a precision stage. Then, the SU-8 supporting structures were immersed into the SU-8 by adjusting the precision state. The SU-8 were coated on the pillars'' surface. Then, the SU-8 supporting structures were drawn away from the interface of the liquid maltose and air. After that, the temperature and drawing speed were increased. Since the SU-8 was less viscous at higher temperature, the connection between the SU-8 supporting structures and surface of the liquid SU-8 became individual SU-8 bridge, shrank, and then broke. The end of the shrunk SU-8 bridge forms a sharp tip on the top of each SU-8 supporting structure when the connection was separated. After the hollowed tips were formed in the first step drawing process, the whole device was baked on the hotplate to melt the hollowed SU-8 tips. Melted SU-8 reflowed into the gaps between four pillars and the tips became domes (Fig. (Fig.2f).2f). Then, a second drawing process was conducted on the top of melted SU-8 to form sharp and solid tips (Fig. (Fig.2g).2g). The final fabricated device is shown in Fig. Fig.44.Open in a separate windowFigure 3A hollowed SU-8 microneedle fabricated by single drawing lithography technology (scale bar is 100 μm).Open in a separate windowFigure 4Optical images for the finished SU-8 microneedles.During the double drawing process, as long as the heated time and temperature were controlled, the SU-8 flow-in speed of SU-8 inside the gaps could be precisely determined. The relationship between baking temperature and flow-in speed was studied. As shown in Fig. Fig.5,5, the flow-in speed is positive related to the baking temperature. The explanation for this phenomena is that the SU-8''s viscosity is different under different baking temperatures.40 Generally, baked SU-8 has 3 status when temperature increases, solid, glass, and liquid. The corresponding viscosity will decrease and the SU-8 can also have higher fluidity. When the baking temperature is larger than 120 °C, the flow-in speed will increase sharply. But, if the baking temperature is higher, the SU-8 will reflow in the gaps too fast, which makes the flow-in depth hard to be controlled. There is a high chance that the whole gaps will be blocked, and no drugs can flow through these gaps any more. Considering that the total SU-8 supporting structure is only 350 μm high, we choose 125 °C as baking temperature for proper SU-8 flow-in speed and easier SU-8 flow-in depth control.Open in a separate windowFigure 5The relationship between flow-in speed and baking temperature.To ensure the adequate stiffness of the SU-8 microneedles in vertical direction, Instron Microtester 5848 (Instron, USA) was deployed to press the microneedles with the similar method reported by Khoo et al.41 As shown in Fig. Fig.6a,6a, the vertical buckling force was as much as 8.1N, which was much larger than the reported minimal required penetration force.42 However, in the previous practical testing experiments, even though the microneedles were strong enough in vertical direction, the planar shear force induced by skin deformation might also break the interface between SU-8 pillars and top tips. In our new device with four pillars supporting structure, the SU-8 could flow inside the sidewall gaps between the pillars to form anchors. These anchors could enhance microneedles'' mechanical strength and overcome the planar shear force problems. Moreover, the anchors strength could be improved by controlling the SU-8 flow-in depth. Fig. Fig.77 shows that the flow-in depth increases when the baking time increases as the baking time increases at 125 °C. Fig. Fig.6b6b shows that the corresponding planar buckling force can be improved to be larger than 1 N by increasing flow-in depth. Some sidewall gaps at bottom are kept on purpose for drugs delivery; hence, the flow-in depth is chosen as 200 μm.Open in a separate windowFigure 6(a) Measurement of the vertical buckling force. (b) The planar buckling force varies under different flow-in depth (I, II, III, and IV corresponding to the certain images in Fig. Fig.77).Open in a separate windowFigure 7Different flow-in depth inside the gaps between SU-8 pillars. (a) 0 μm; (b) 100 μm; (c) 200 μm; and (d) 350 μm (scale bar is 100 μm).The penetration capability of the 3 × 3 SU-8 microneedles array is characterized by conducting the insertion experiment on the porcine cadaver skin. 10 microneedles devices were tested and all of them were strong enough to be inserted into the tissue without any breakage. Histology images of the skin at the site of one microneedle penetration were derived to prove that the sharp conical tip was not broken during the insertion process (Fig. (Fig.8).8). It also shows penetrated evidence because the hole shape is the same as the sharp conical tip.Open in a separate windowFigure 8Histology image of individual microneedle penetration (scale bar is 100 μm).In order to verify that the drug solution can be delivered into tissue from the sidewall gaps of the microneedles, FITC (Fluorescein isothiocyanate) (Sigma Aldrich, Singapore) solution was delivered through the SU-8 microneedles after they were penetrated into the mouse cadaver skin. The representative results were then investigated via a confocal microscope (Fig. (Fig.9).9). The permeation pattern of the solution along the microchannel created by microneedles confirmed the solution delivery results. The black area was a control area without any diffused florescent solution. In contrast, the illuminated area in Fig. Fig.99 indicates the area where the solution has diffused to it. These images were taken consecutively from the skin surface down to 180 μm with 30 μm intervals. The diffusion area had a similar dimension with the inserted microneedles. It has proved that the device can be used to deliver drugs into the body.Open in a separate windowFigure 9Images of confocal microscopy to show the florescent solution is successfully delivered into the tissue underneath the skin surface. (a) 30 μm; (b) 60 μm; (c) 90 μm; (d) 120 μm; (e) 150 μm; and (f) 180 μm (scale bar is 100 μm).Due to the uneven surface of deformed skin, there is always tiny gap happened between tips of some microneedles and local surface skin. The microneedles could not be entirely inserted into the tissue. Drugs might leak to the skin surface through the sidewall gaps under certain driven pressure. Hydrogel absorption experiment was conducted to quantify the delivery rate (i.e., the ratio of solution delivered into tissues in the total delivered volume) and to optimize the delivery speed. Using hydrogel as the tissue model for quantitative analysis of microneedle releasing process was reported by Tsioris et al.43 The details are shown here. Gelatin hydrogel was prepared by boiling 70 ml DI (Deionized) water and mixing it with 7 g of KnoxTM original unflavored gelatin powder. The solution was poured into petri dish to 1 cm high. Then, the petri dish was put into a fridge for half an hour. Gelatin solution became collagen slabs. The collagen slabs were cut into 6 mm × 6 mm sections. A piece of fully stretched parafilm (Parafilm M, USA) was tightly mounted on the surface of the collagen slabs. This parafilm was used here to block the leaked solution further diffusing into the collagen slab in the delivery process. Then, the microneedles penetrated the parafilm and went into the collagen slab. Controlled by a syringe pump, 0.1 ml–0.5 mg/ml glucose solution was delivered into the collagen slab under different speeds. Methylene Blue (Sigma Aldrich, Singapore) was mixed into the solution for better inspection purpose (Fig. 10a). Then, the collagen slabs was digested in 1 mg/ml collagenase (Sigma Aldrich, Singapore) at room temperature (Fig. 10b). It took around 1 h that all the collagen slabs could be fully digested (Fig. 10d). The solution was collected to measure the glucose concentration with glucose detection kit (Abcam, Singapore). Briefly, both diluted glucose standard solution and the collected glucose solution were added into a series of wells in a well plate. Glucose assay buffer, glucose enzyme, and glucose substrate were mixed with these samples in the wells. After incubation for 30 min, their absorbance were examined by using a microplate reader at a wavelength of 450 nm. By comparing the readings with the measured concentration standard curve (Fig. 11a), the glucose concentration in the hydrogel, the glucose absorption rate in the hydrogel, and the solution delivery rate by microneedles could be measured and calculated. As shown in Fig. 11b, when the delivering speed of a single microneedle increased from 0.1 μl/min to 2 μl/min, the glucose absorption rate also increased. Most of the glucose solution from microneedles could go into the hydrogel. The delivered rate could be as high as 71%. The rest solution leaked from sidewall gaps and blocked by parafilm. However, when the delivered speed for a single microneedle was larger than 2 μl/min, the hydrogel absorption rate was saturated. More and more solution could not go into the hydrogel but leak from the sidewall gaps. Then, the delivered rate decreased. Therefore, 2 μl/min was chosen as the optimized delivery speed for the microneedle.Open in a separate windowFigure 10Glucose solution could be delivered into the hydrogel, and the collagen stabs were dissolved by collagenase.Open in a separate windowFigure 11(a) Standard curve for glucose detection; (b) glucose absorption rate and solution delivery rate in a single needle corresponding to different delivery speed.In conclusion, a drug delivery device of integrated vertical SU-8 microneedles array is fabricated based on a new double drawing lithography technology in this study. Compared with the previous biocompatible polymer-based microneedles fabrication technology, the proposed fabrication process is scalable, reproducible, and inexpensive. The fabricated microneedles are rather strong along both vertical and planar directions. It is proved that the microneedles were penetrated into the pig skin easily. The feasibility of drug delivery using SU-8 microneedles is confirmed by FITC fluorescent delivery experiment. In the hydrogel absorption experiment, by controlling the delivery speed under 2 μl/min per microneedle, the delivery rate provided the microneedle is as high as 71%. In the next step, the microneedles will be further integrated with microfluidics on a flexible substrate, forming a skin-patch like drug delivery device, which may potentially demonstrate a self-administration function. When patients need an injection treatment at home, they can easily use such a device just like using an adhesive bandage strip.  相似文献   

16.
Magnetographic array for the capture and enumeration of single cells and cell pairs     
C. Wyatt Shields  IV  Carissa E. Livingston  Benjamin B. Yellen  Gabriel P. López  David M. Murdoch 《Biomicrofluidics》2014,8(4)
We present a simple microchip device consisting of an overlaid pattern of micromagnets and microwells capable of capturing magnetically labeled cells into well-defined compartments (with accuracies >95%). Its flexible design permits the programmable deposition of single cells for their direct enumeration and pairs of cells for the detailed analysis of cell-cell interactions. This cell arraying device requires no external power and can be operated solely with permanent magnets. Large scale image analysis of cells captured in this array can yield valuable information (e.g., regarding various immune parameters such as the CD4:CD8 ratio) in a miniaturized and portable platform.The emergent need for point-of-care devices has spurred development of simplified platforms to organize cells across well-defined templates.1 These devices employ passive microwells, immunospecific adhesive islands, and electric, optical, and acoustic traps to manipulate cells.2–6 In contrast, magnetic templating can control the spatial organization of cells through its ability to readily program ferromagnetic memory states.7 While it has been applied to control the deposition of magnetic beads,8–13 it has not been used to direct the deposition of heterogeneous cell pairs, which may help provide critical insight into the function of single cells.14,15 As such, we developed a simple magnetographic device capable of arraying single cells and pairs of cells with high fidelity. We show this magnetic templating tool can use immunospecific magnetic labels for both the isolation of cells from blood and their organization into spatially defined wells.We used standard photolithographic techniques to fabricate the microchips (see supplementary material16). Briefly, an array of 10 × 30 μm cobalt micromagnets were patterned by a photolithographic liftoff process and overlaid with a pattern of dumbbell-shaped microwells formed in SU-8 photoresist (Fig. 1(a)). The micromagnets were designed to produce a predominantly vertical field in the microwells by aligning the ends of the micromagnet at the center of each well of the dumbbell. These features were deposited across an area of ≈400 mm2 (>50 000 well pairs per microchip) (Fig. 1(b)). Depending on the programmed magnetization state with respect to the external field, magnetic beads or cells were attracted to one pole and repelled by the other pole of each micromagnet, leading to a biased deposition (Fig. 1(c)).12Open in a separate windowFIG. 1.Magnetographic array for single cell analysis. (a) SEM image of the dumbbell-shaped well pairs for capturing magnetically labelled cells. (b) Photograph of the finished device. (c) An array of well pairs displaying a pitch of 60 × 120 μm before (top) and 10 min after the deposition of magnetic beads (bottom).To demonstrate the capability of the array to capture cells into a format amenable for rapid image processing, we organized CD3+ lymphocytes using only hand-held permanent magnets. We isolated CD3+ lymphocytes from blood via positive selection using anti-CD3 magnetic nanoparticles (EasySep™, STEMCELL Technologies) with purities confirmed by flow cytometry (97.8%; see supplementary material16). We then stained 1 × 106 CD3+ cells with anti-CD8 Alexa-488 and anti-CD4 Alexa-647 (5 μl of each antibody in 100 μl for 20 min; BD Bioscience) to determine the CD4:CD8 ratio, a prognostic ratio for assessing the immune system.17,18Variably spaced neodymium magnets (0.5 in. × 0.5 in. × 1 in.; K&J Magnetics, Inc.) were fixed on either side of the microchip to generate a tunable magnetic field (0–400 G; Fig. 2(a)). Using this setup, fluorescently labeled cells were deposited, and the populations of CD4+ and CD8+ cells were indiscriminately arrayed, imaged, and enumerated using ImageJ. The resulting CD4:CD8 ratio of 1.84 ± 0.18 (Fig. 2(b)) was confirmed by flow cytometry with a high correlation (5.4% difference; Fig. 2(c)), indicating the magnetographic microarray can pattern cells for the rapid and accurate assessment of critical phenotypical parameters without complex equipment (e.g., function generators or flow cytometers).Open in a separate windowFIG. 2.CD8 analysis of CD3+ lymphocytes. (a) Photograph of the magnetographic device activated by permanent magnets (covered with green tape). The CD4:CD8 ratio determined by the (b) magnetographic microarray and (c) and (d) flow cytometry was 1.84 and 1.74, respectively.More complex operations, such as the programmed deposition of cell pairs, can be achieved by leveraging the switchable, bistable magnetization of the micromagnets for the detailed studies of cell-cell interactions (Figs. 3(a)–3(d)).12 For these studies, a 200 G horizontal field generated from an electromagnetic coil was used to magnetize the micromagnets.19 We then captured different concentrations of magnetic beads as surrogates for cells (8.4 μm polystyrene, Spherotech, Inc.) and found that higher bead concentrations did not affect the capture accuracy (>95%; see supplementary material16).Open in a separate windowFIG. 3.Programmed pairing of magnetic beads and CD3+ lymphocytes. (a) Schematic of the magnetographic cell pair isolations. (b) Polarized micromagnets isolate cells of one type to one side in a vertical magnetic field and then cells of a second type to the other side when the field is reversed. (c) Fluorescent image of magnetically trapped green stained (top) and red stained (bottom) cell pairs. (d) SEM image of magnetically labeled cells in the microwells. (e) Capture accuracy of magnetic bead pairs. (Each color (and shape) represents the field strength of the reversed field.) (f) Change in the capture accuracy (loss) of initially captured beads after reversing the magnetic field. The capture accuracy of (g) magnetically labeled cell pairs and (h) the second magnetically labeled cell (for (e)–(h): n = 5; time starts from the deposition of the second set of cells or beads).The opposite side of each micromagnet was then populated with the second (yellow fluorescent) bead by reversing the direction of the applied magnetic field. We tested several field strengths (i.e., 10, 25, 40, or 55 G) to optimize the conditions for isolating the desired bead in the opposite well without ejecting the first bead. If the field strength was too large, the previously deposited beads could be ejected from their wells due to the repulsive magnetic force overcoming gravity.12 As shown in Figure 3(e), increasing the field strength from 10 to 25 G significantly increased the capture accuracy at 60 min from the deposition of the second bead (p < 0.01), but increases from 25 to 55 G did not affect the capture accuracy (p > 0.10). As shown in Figure 3(f), higher field strengths (i.e., 40 and 55 G) resulted in lower capture accuracies compared to lower field strengths (i.e., 10 and 25 G) (p < 0.01), which was primarily due to ejection of the initially captured beads when the micromagnets reversed their polarity.We then arranged pairs of membrane dyed (calcein AM, Invitrogen; PKH26, Sigma) magnetically labeled CD3+ lymphocytes. First, red stained cells (150 μl of 2 × 104 cells/ml) were deposited on the microchip in the presence of 250 G vertical magnetic field. After 20 min, the field was reversed (i.e., to 40, 55, and 70 G) and green stained cells (150 μl of 2 × 104 cells/ml) were deposited on the microchip with images taken in 10 min intervals. Fluorescence images were overlaid (Fig. 3(c)) and the capture accuracy of cell pairs was determined (ImageJ).As seen in Figure 3(g), the capture accuracy of pairs of CD3+ lymphocytes was lower than that of magnetic beads (Fig. 3(e)). However, as shown in Figure 3(h), the second set of cells (green fluorescent) exhibited an average capture accuracy of 91.8% ± 1.9%. This indicates that the lower capture accuracy of cell pairs was either due to the ejection of initially captured (red fluorescent) cells or the migration of initially captured cells through the connecting channel, resulting from their relatively high deformability compared to magnetic beads.In summary, we developed a simple device capable of organizing magnetic particles, cells, and pairs of cells into well-defined compartments. A major advantage of this system is the use of specific magnetic labels to both isolate cells and program their deposition. While the design of this device does not enable dynamic control of the spacing between captured cell pairs as does some dielectrophoresis-based devices,20 it can easily capture cells with high fidelity using only permanent magnets and has clinical relevance in the assessment of immune parameters. These demonstrations potentiate a relatively simple and robust device where highly organized spatial arrangement of cells facilitates rapid and accurate analyses towards a functional and low-cost point-of-care device.  相似文献   

17.
A high-throughput cellulase screening system based on droplet microfluidics     
Raluca Ostafe  Radivoje Prodanovic  W. Lloyd Ung  David A. Weitz  Rainer Fischer 《Biomicrofluidics》2014,8(4)
A new ultra-high-throughput screening assay for the detection of cellulase activity was developed based on microfluidic sorting. Cellulase activity is detected using a series of coupled enzymes leading to the formation of a fluorescent product that can be detected on a chip. Using this method, we have achieved up to 300-fold enrichments of the active population of cells and greater than 90% purity after just one sorting round. In addition, we proved that we can sort the cellulase-expressing cells from mixtures containing less than 1% active cells.Cellulases are important enzymes with numerous applications across multiple industries, including biofuel, pulp, paper, textile and laundry, food, feed, brewing, and agriculture.1 Most cellulases have low activity and stability, so improving these properties would have substantial impact on numerous industrial processes.Enzymatic properties can be improved by protein engineering2 but the limiting step is the screening process. Classical screening uses microtiter plates (MTPs), where each well contains cells expressing a single type of mutant enzyme. However, this type of screening is the bottleneck in directed evolution, because a maximum number of 105 clones can be screened over the course of weeks or even months3 and large quantities of reagents and consumables are needed. High-throughput screening methods based on either fluorescence activated cell sorting (FACS)4–7 or microfluidic devices8 increase the number of clones that can be screened and reduce the amount of consumables required. Here, we demonstrate the use of a high-throughput screening system for cellulases by combining lab-on-chip sorting devices with an emulsion-based fluorescent assay previously developed for use in flow cytometry.5Water–in-oil emulsions are needed to maintain the connection between genotype and phenotype by compartmentalizing individual cells expressing a mutant enzyme together with the components of the fluorescence assay corresponding to the enzyme activity.7 For FACS, double emulsions (water-in-oil-in-water) are required because the instrument''s mobile phase is an aqueous solution. Such double emulsions can be produced by stirring or agitation,9,10 but the resulting emulsions are polydisperse and multiple water droplets may be scattered within a single oil droplet. In addition, large droplets tend to produce more fluorescence because there are more substrate molecules available for conversion into the fluorescent product. The emulsions are produced in bulk, so each droplet will be detected at a different time point from the start of the reaction. This means that increased fluorescence may result because an enzyme has worked on the substrate for a longer amount of time, and the fluorescence of the droplet may plateau before sorting as the enzyme consumes all the available substrate. Cell loading is difficult to control because the average number of cells per droplet scales with droplet volume. Also, if several inner droplets, containing cells with different activities, are encapsulated within the same outer droplet, false positives may occur upon sorting. Consequently, it is impossible to differentiate fluorescence changes due to enzyme activity from those due to other effects using polydisperse double emulsions in FACS, but it is possible to achieve plus/minus screening,4 separating cells with activity from those without.Droplet microfluidics overcomes many of the drawbacks of high-throughput enzyme sorting with FACS. Both the size and composition of the droplets can be tuned precisely. Furthermore, once the enzyme is mixed with the substrate, the incubation time can be controlled and all compartments will have the same conditions in terms of concentration and total number of substrate molecules. Although cell loading is still subject to Poisson statistics, the probability for cells to be loaded into a given droplet is the same and can be adjusted by tuning the input cell density. These characteristics make the microfluidic method more sensitive, flexible, and quantitative at detecting changes in enzyme activity than the FACS-based sorting of double emulsions.Here, we report a method in which droplet microfluidics is used to sort libraries containing different percentages of cells expressing cellulase activity and demonstrate enrichment of the cells expressing active cellulases. The entire process is summarized in Figure Figure11.Open in a separate windowFIG. 1.General overview of cellulase screening using droplet microfluidics. In the emulsification device, suspensions of yeast surface displayed libraries are co-flowed with the substrate solution at equal flow rates to a drop-forming junction where they mix. A stream of perfluorinated oil then breaks the aqueous mixture into monodisperse water-in-oil emulsions. Within each droplet, the cellulase reaction starts after compartmentalization and the fluorescent product is formed by a coupled enzymatic cascade in droplets containing cells that express the active enzyme. After a fixed incubation time, the emulsion droplets are re-injected into a microfluidic sorting device, where they are analyzed and sorted based on their fluorescence.To detect cellulase activity, we designed an assay that uses a chain of coupled enzymatic reactions to yield fluorescence corresponding to cellulase activity without needing artificial substrates (which may lead to confounding effects, such as improved binding of the enzyme specifically to the artificial compound but not the natural substrate). In this method, cellulase hydrolyzes cellulose, its natural substrate, into monosaccharides and oligosaccharides that are further detected by the enzymatic cascade5 (Figure (Figure11).Based on previous FACS experiments, no difference in activity can be detected between the positive and the negative droplets before 2 h incubation time.5 Based on these observations, we expected the cells to require more than 2 h of incubation in droplets for the reaction to develop.Emulsions were formed using a co-flow flow-focusing Polydimethylsiloxane device prepared by soft lithography as previously described8 and using fluorocarbon oil containing 1% (v/v) Krytox-PEG-Krytox detergent synthesized as reported in an earlier study.11,14 The solutions, one containing library cells (S. cerevisiae YPH500 cells, Agilent Technologies, Santa Clara, USA) and the other with the substrate,14 were mixed at the same flow rate, giving a one-to-one mixing ratio. The library cells were a defined mixture of cells transformed with cel5A pESC-Trp (positive cells) or empty pESC-Trp (negative cells). The two solutions therefore mixed just prior to encapsulation, minimizing the chance that fluorescent products would enter neighboring droplets. The substrate solution contained carboxymethyl cellulose (CMC), which has a high viscosity. To prevent fluctuations in the flow of substrate during the emulsification process, we optimized the flow rate and the concentration of CMC and found that a CMC concentration of 0.33% (w/v) produced monodisperse emulsions.We discovered that the HOx required for the enzymatic cascade causes droplet coalescence. HOx alone was sufficient to cause the observed change in droplet stability because droplets containing only hexose oxidase in buffer exhibited the same amount of coalescence as those containing the full set of assay components. We hypothesized that the enzyme might be surface active, disturbing the emulsion interface, but emulsions of an inactivated form of the enzyme were stable (Figure 2(a)). One possible explanation is that active HOx may interact with the detergent through the active site. Adding bovine serum albumin (BSA), which is known to have a stabilizing effect,12 to the mixture improved droplet stability (Figure 2(a)). Emulsions of the assay mixture with BSA were stable for more than 1 day at room temperature.Open in a separate windowFIG. 2.(a) Transmission light micrographs of water-in-perfluorinated-oil emulsions produced using the microfluidic emulsification devices after 2 h incubation at room temperature. The emulsions contain 3 U/ml HOx either in its native form (left image), inactivated by heating at 99 °C for 20 min (middle image), or supplemented with 1 mg/ml BSA (right image). (b) Images of the results of the agar plate Congo Red cellulase assay before and after sorting, with the percentage of positive colonies indicated. The cells expressing cellulase activity show clear hallos.The time required for the cellulase reaction to produce detectable quantities of fluorescent product was monitored using the droplet screening instrument. These devices proved to have a higher sensitivity than the FACS system because the optics are designed for the droplet size selected for the assay. We were able to detect cellulase activity just 20 min after the compartmentalization of cells. This shorter incubation time allowed us to couple the emulsification device directly to the droplet sorting device using a short piece of tubing. The rate of emulsion flow and the dimensions of the tube set the droplet incubation time.Using the optimized conditions, we used droplet microfluidics to sort cellulase-expressing cells from a set of reference libraries. The reference libraries were created by mixing different concentrations of positive S. cerevisiae YPH500 cells expressing Cel5A cellulase and negative S. cerevisiae YPH500 cells transformed with the pESC-Trp empty vector. The mixed populations were emulsified together with the assay components in water-in-perfluorinated-oil emulsions and incubated at room temperature for 20 min. The gated population was sorted and the cells were spread on yeast nitrogen base casaminoacids (YNB CAA) Glu agar plates. An aliquot of the reference library was also plated on agar plates prior to sorting. Approximately, 100 cells before and after sorting were transferred to YNB CAA CMC Gal/Raf induction plates, and the Congo red assay13 was used to detect cells expressing cellulase. In this assay, colonies of positive cells developed transparent halos around them.14 The results before and after sorting are presented in Figure 2(b).We enriched cellulase-expressing cells from a pool of negative cells, regardless of the starting concentration of positive cells. We were able to isolate the cellulase-expressing cells even when starting from a low percentage of active cells (0.1%). We obtained high enrichment factors of up to 300 when starting from low concentrations of positive cells, and we were able to sort to a purity of greater than 90%. These results exceed those obtained by comparable experiments using FACS.5In conclusion, we developed a high-throughput screening system for cellulase activity based on droplet microfluidics. We optimized the emulsification conditions to produce highly stable and monodisperse droplets. The low dispersity of the emulsion enables the sensitive, tunable, and quantitative detection of cellulase activity. In addition, we substantially reduced the reaction time needed for the development of a fluorescent product from 2 h to 20 min. As a result, we sorted reference libraries of cellulases with various ratios of positive to negative cells, and regardless of the starting population of positive cells we were always able to enrich the active population to a higher purity than that obtained by FACS.  相似文献   

18.
Polyphosphonium-based ion bipolar junction transistors     
Erik O. Gabrielsson  Klas Tybrandt  Magnus Berggren 《Biomicrofluidics》2014,8(6)
Advancements in the field of electronics during the past few decades have inspired the use of transistors in a diversity of research fields, including biology and medicine. However, signals in living organisms are not only carried by electrons but also through fluxes of ions and biomolecules. Thus, in order to implement the transistor functionality to control biological signals, devices that can modulate currents of ions and biomolecules, i.e., ionic transistors and diodes, are needed. One successful approach for modulation of ionic currents is to use oppositely charged ion-selective membranes to form so called ion bipolar junction transistors (IBJTs). Unfortunately, overall IBJT device performance has been hindered due to the typical low mobility of ions, large geometries of the ion bipolar junction materials, and the possibility of electric field enhanced (EFE) water dissociation in the junction. Here, we introduce a novel polyphosphonium-based anion-selective material into npn-type IBJTs. The new material does not show EFE water dissociation and therefore allows for a reduction of junction length down to 2 μm, which significantly improves the switching performance of the ion transistor to 2 s. The presented improvement in speed as well the simplified design will be useful for future development of advanced iontronic circuits employing IBJTs, for example, addressable drug-delivery devices.There has been a recent interest in developing diodes1–4 and transistors4–8 that conduct and modulate ion currents. Such non-linear iontronic components are, for example, interesting as they allow further control of ions in, for instance, electrophoretic drug delivery devices. A range of microfabricated diodes,9–11 transistors,12,13 and circuits9,14 has been constructed using ion-selective membranes. These membranes contain fixed charges of either polarity, compensated by mobile ions of opposite charge (counter-ions). When immersed in an electrolyte, counter-ions can move through the membrane, while ions with the same charge as the fixed charges (co-ions) are repelled. This renders the membrane selective for the counter-ion and can therefore be considered as p- or n-type ion conductors. By combining two membranes of opposite polarity, a bipolar membrane (BM) configuration is obtained15 (Figure 1(a)). The BM junction can be biased by an ion current in the reverse and forward directions, respectively.16,17 This modulates the ion concentration inside the BM, and thus the ionic conductivity, which then results in an current rectification.2,18 In the three-terminal ion bipolar junction transistor12 (IBJT), an ion-selective base (B) is connected to oppositely selective emitter (E) and collector (C), forming two BM configurations (EB and BC) (Figure 1(b)). pnp- and npn-IBJTs have been constructed14 from photolithography patterned poly(styrene sulfonate) (PSS, p-selective) and quaternized poly(vinylbenzyl chloride) (n-selective) as emitter, collector, and base. In these devices, a neutral poly(ethylene glycol) (PEG) electrolyte is typically inserted into the junction to separate the base from the emitter and collector,12 in order to avoid19 electric field enhanced (EFE) water dissociation16 (Figure 1(a)). EFE water dissociation is typically observed in BMs20 and produces water ions inside the BM under reverse bias, which prevents proper IBJT operation. In PEG-IBJTs, the current between the emitter and collector (IC) is thus modulated by controlling the ion concentration inside the PEG-junction.21 Ions are injected or extracted into the junction depending on the bias of the base (VEB). In a npn-IBJT, a positive bias is typically applied between emitter and collector (VEC), thus allowing anions to migrate from the emitter to the collector. In the cut-off mode (Figure 1(c)), a negative bias VEB is applied, resulting in reverse bias of both EB and BC. Cations in the junction will migrate into the base, while anions will primarily migrate into the collector, due to the higher collector bias. This base current (IB) will extract ions from the junction, which decreases the ionic conductivity in the junction resulting in a low IC. Eventually, the resistive characteristics for ion charge transport, between the emitter and collector, will be entirely dominated by the junction. This gives that most of the applied VEC is consumed across the junction with only a minimal voltage potential drop across the emitter and base terminals.Open in a separate windowFIG. 1.(a) The modes of operation for a BM; forward bias (high conduction and ion accumulation), reverse bias (low conduction and ion depletion), and EFE water dissociation (high conduction, formation of ions). (b) Illustrations of an npn-IBJT, with anion-selective emitter (E) and collector (C) forming a junction with a cation-selective base (B). (c) In cut-off mode, the base and collector extract ions from the junction, prohibiting co-ion migration through the base. (d) In active mode, the forward biased EB injects ions into the base, thus allowing anions from the emitter to migrate as co-ions through the base into the collector.In the active-mode of the npn-IBJT (Figure 1(d)), the VEB bias at the base is reversed (i.e., now positive). This causes injection of cations, from the base, and anions, from the emitter, into the junction. As the ion concentration increases, anions from the emitter can start to drift across the junction to the collector, thus a high IC is obtained. The high concentration of ions inside the junction is reflected in a low resistive value for ion transport. This now causes the voltage to drop over the emitter and collector terminals, thus lowering the EB forward bias and the injection of ions from the base. At the collector-junction interface, the extraction of anions produces an ion depletion zone and a corresponding voltage drop. Thus, in the active-mode, the applied VEC is primarily consumed across the emitter and collector terminals and also at the collector-junction interface.The switching speed of an IBJT should be strongly correlated to the distance separating the emitter and collector,14 as this length determines the volume that needs to be filled or emptied with ions causing modulation of ions in the junction. To achieve a fast-switching IBJT, the junction volume, i.e., the collector-emitter separation, should be as small as possible. However, EFE water dissociation must be avoided since this process ruin the IBJT operation. EFE water dissociation is, in part, driven by the appearance of a large potential drop across a small distance, as occurring at the interface of a BM under reverse bias, producing a high electric field that accelerates the forward reaction rate of water auto-dissociation.16 Miniaturization of the collector-emitter distance is therefore problematic, as the separation inside the EB and BC BMs evidently also mush shrink, resulting in higher reverse bias electric fields across the BMs and thus promoting EFE water dissociation. The problem of EFE water dissociation in an IBJT primarily manifests itself in the cut-off mode, as water ions are generated in the reversed biased EB and BC BMs. These ions produce an elevated cut-off IC, and hence deteriorate the IBJTs on–off performance. Here, we report an IBJT, in which the EFE water dissociation is avoided by the use of a novel polyphosphonium-based anion-selective material, which previously has been shown to prevent EFE water dissociation in BM diodes.11 This allows the collector and emitter to directly contact the base without an intermediate PEG-layer. Without the need for a PEG-separator inside the BMs, the collector-emitter distance is reduced to only 2 μm.Polyphosphonium-based npn-IBJTs were produced following the same manufacturing protocol as was reported for polyphosphonium-based ion diodes.11 Conjugated polymer electrodes and cation-selective base was patterned from ∼200 nm thick poly(3,4-ethylenedioxythiophene):polystyrene sulfonate film on polyethylene terephthalate-sheets using photolithography and dry-etching. The base was rendered electronically insulating by chemical overoxidation via exposure to sodium hypochlorite through a mask. A 2 μm thick SU8-layer was patterned on-top of this configuration, with an opening defining the actual junction. 1 μm thick polyphosphonium-based anion-selective emitter and collector were deposited and patterned using photolithography and dry-etching, to overlap with the base at the opening of the SU8. Finally, a second 10 μm thick layer of SU8 was used to seal the junction. The membranes were hydrated by incubation in dH2O for 24 h before any measurements were carried out. Aqueous 0.1M NaCl electrolytes were used during the measurement. All electrical measurements were performed using a Keithley 2602 source meter.The switching characteristics of the npn-IBJT were obtained by applying VEC of 10 V and alternating VEB at ±3 V for various duration of time, see Figure Figure2.2. A periodic 5 s switching with 8 Hz measurement rate was used to record the dynamics of the turn-on/off characteristics of the device. When VEB switches from −3 to +3 V, there is a quick increase in the IB, as ions from the base and emitter migrate into the emitter/base junction. After a delay of ∼0.25 s, IC starts to increase due to the increased ion concentration in the emitter/base junction and the subsequent diffusion of anions into the base. As the IC increases, the IB decreases as the voltage drop between the emitter and base decreases, and after ∼2 s IC reaches 90% of the steady state on-current level. For longer on-switching times, the IB and IC stay stable over 30 s, after which a small increase is observed. This current-drift in both IB and IC is likely due to the contribution of co-ion migration. As cations from the base migrate into the emitter as co-ions, the conductivity in the emitter increases, leading to an increased IC value. This increases the ion concentration at the base, which gives less selective ion injection and thus more cation injection from the base, i.e., a higher IB.Open in a separate windowFIG. 2.Emitter-collector current response as the IBJT is switched between cut-off (VEB=−3 V) and active mode (VEB = 3 V) for VEC = 10 V, at 5 s and 120 s periods.As VEB is switched back to −3 V, there is a sharp negative peak in IE as ions are extracted from the junction, which occur mainly through the base (cations) and collector (anions) terminals. As the ion concentration in the base drops, IC decreases. The transistor turns off to 10% of the value of the steady state on-current within ∼2 s, regardless of the duration of the on-state. The constant turn-off time indicates that ions are not accumulating to a significant extent inside the junction during the on-steady state but are instead constantly transported out of the junction. When all co-ions have been extracted from the junction, the Donnan exclusion prevents subsequent injection of anions into the base, and IC is therefore low. The on/off ratio of IC reaches above 100.A transfer curve was obtained by scanning VEB between −3 and +3 V while keeping VEC at 10 V (Figure 3(a)). As expected, both IC and IB remain low for negative VEB. In this range, both EB and BC are biased in reverse direction. As VEB turns positive, the EB configuration is switched into forward bias and ions are injected into the junction. This leads to a linear increase in IC vs. VEB. For the reverse scan, a minor hysteresis is observed for both the IC and IB scans, again probably due to the contribution of co-ion migration due to long time operation of the device.Open in a separate windowFIG. 3.Transfer and output curves. (a) The transfer curve is low for negative VEB and increases linearly for positive VEB with approximately zero threshold. (b) The output curves show IC saturating with respect of VEC for positive VEB.The transistor output characteristics were obtained by scanning VEC at different VEB values (Figure 3(b)). The saturation regime, i.e., the bias mode was both EB and BC are in forward bias, was avoided as this has negative impact on the stability of the device. As reported for previous IBJT devices, the output characteristics show a clear saturation behaviour of IC across the entire range of VEC. Further, the IC increases linearly with VEB. The increase of both IC and IB when operating for extended periods of time in the active mode is again attributed to the addition and inclusion of co-ions in the junction. The current gain (IC/IB) at VEC = 10 V decreases with VEB and reaches 43.9, 17.9, and 10.7 for VEB = 1 V, 2 V, and 3 V, respectively. For higher base bias voltages, the ion concentration increases in the junction and thus the injection selectivity decreases.In comparison with previously reported IBJTs,12,14,21 the lack of a neutral electrolyte layer in the junction has an overall positive effect on the device characteristics. Main performance improvements are found in a decrease in the turn-on time from 9 s (for npn-IBJT21) to 2 s, for devices with comparable junction widths and heights. The main contribution to the improved switching speed is likely the decreased length between the emitter and collector. Interestingly, simulations have shown that an extended space charge region (ESCR), for a PEG-IBJT in cut-off mode, can extend several micrometers away from the collector.22 Thus, a PEG-IBJT with an emitter-collector separation of single micrometers should show an increased cut-off current due to the ESCR overlapping in the junction. However, by omitting the PEG in the junction, the ESCR is reduced due to screening from the fixed charges in the BM layers. This enables the IBJT, reported here, to operate with retained low cut-off currents. On-off ratios and ion current gains are approximately equal to previous IBJTs,12,14,21 at above 100 and 10, respectively. The on–off ratio and ion current gain are more dependent on the selectivity of the membranes and the charge of the junction.Further, the need to separate the layers in a PEG-IBJT puts high demands on the patterning resolution and alignment accuracy to reduce the separation between emitter/collector and base. As polyphosphonium allows the IBJT to be built without separation of layers, miniaturization of the junction is relatively easier to obtain. The switching speed can potentially be further improved by retaining the base material between the emitter and collector (see Figure 1(b)), thus allowing for a more direct pathway for IC. This design would, however, require a much more accurate layer alignment or that the base patterned on top of the emitter and collector layers. In general, such modifications of device geometry are simpler to accomplish with the non-EFE water dissociating polyphosphonium as fewer active layers are used, suggesting a further use of polyphosphonium to improve switching speed and miniaturization of IBJTs. Such further advancement in IBJT performance would be welcomed, for example, in the continued work towards complex ionic circuits14 to regulate signalling in bioelectronics and in drug delivery applications, in which generation of dynamic and complex gradients, at high spatial resolution, is of generic interest.  相似文献   

19.
Preface to Special Topic: Papers from the 13th International Conference on Surface and Colloid Science (ICSCS) and the 83rd ACS Colloid and Surface Science Symposium, Columbia University, New York, 2009     
Leslie Y. Yeo 《Biomicrofluidics》2010,4(1)
This Special Topic section is a compilation of several original contributions covering both fundamental and practical aspects of electrokinetic microfluidic phenomena that were presented during the Electrokinetics and Microfluidics sessions held at the conference.Electrokinetics is currently the mechanism of choice for the manipulation of fluids as well as colloidal and biological particles at microscale and nanoscale dimensions.1 The popularity of electrokinetics is perhaps not so surprising as electrodes are easy to fabricate and embed into microfluidic chips, thus allowing the entire fluid and particle actuation mechanism to be completely integrated into the device. In addition, driving microfluidics with electric fields is relatively straightforward and allows for precise actuation. Nevertheless, considerable challenges remain in understanding the complex mechanisms associated with the hydrodynamics of conducting and dielectric fluids and particles under the influence of electric fields. Concomitantly, there has been an exponential increase in research and development in this field along both fundamental and applied themes in the past five years.This sustained growth in the microfluidics community of electrokinetics research has led to a sequel to the first Electrokinetic Phenomena and Microfluidics session at the 82nd ACS Colloid and Surface Science Symposium in Raleigh, NC, in 2008, and which we hope will now be a regular feature at successive ACS Colloid and Surface Science meetings. This year at the combined 2009 13th International Conference on Surface and Colloid Science (ICSCS) and the 83rd ACS Colloid and Surface Science Symposium in New York, the Electrokinetics and Microfluidics symposium proved to be extremely popular, with three keynote lectures presented by Professor Howard Stone, Professor Hsueh-Chia Chang, and Professor Thomas Healy, and 44 oral presentations. In both 2008 and 2009, Biomicrofluidics has organized a special issue to cover some of the contributions reported at these meetings.2The growing interest in using electric fields to manipulate biological entities such as cells, DNA, and even single molecules is reflected in this year’s collection of papers with dielectrophoretic (DEP) phenomena comprising the bulk of the contributions. In Ref. 3, a new theory to describe Stern layer conductance along the surface of nanocolloids is proposed, forming the basis for the derivation of a more accurate prediction of the DEP crossover frequency. This theory is then employed to determine the conformation and, hence, optimum coverage of oligonucleotides on the surface of nanocolloid functionalized molecular probes during DNA hybridization under the influence of DEP, which can be exploited for biomolecular sensing. Other fundamental DEP papers include the investigation of particle motion under DEP induced optically via a photoconductor, in which Zhu et al.4 characterized the frequency dependence of the motion through the synchronous velocity spectra of the particles, and a numerical study of particle trapping at the throat of converging-diverging microchannels under the influence of negative DEP using a transient arbitrary Lagrangian–Eulerian finite element method.5 A more practical implementation is, on the other hand, reported by Yang et al.6 in which the negative DEP is exploited to separate colorectal cancer cells from other cells in a microfluidic device as a demonstration of a portable cancer detection tool.Continuing along the separation theme, but with regard to DNA separation using pulsed-field gel electrophoresis aided by sparse but regularly ordered microfabricated arrays of nanoposts, is a Brownian dynamics simulation model reported by Ou et al.7 in which DNA channeling, which predicts that the motion of DNA is undisturbed by the presence of arrays for large spacing to DNA equilibrium size ratios and when the field lines are straight, is predicted, consistent with experimental observations. In another fundamental paper, a direct numerical simulation model is presented to predict the current-voltage relationship across conducting pores along cell membranes, which is of fundamental importance in the electroporation process.8We hope that you will enjoy reading the contributions in this special topic and that it encourages you to participate in future Electrokinetics and Microfluidics meetings at the ACS Colloid and Surface Science Symposia, which we definitely hope will continue on a regular basis.  相似文献   

20.
An on-chip study on the influence of geometrical confinement and chemical gradient on cell polarity     
Wenfu Zheng  Yunyan Xie  Kang Sun  Dong Wang  Yi Zhang  Chen Wang  Yong Chen  Xingyu Jiang 《Biomicrofluidics》2014,8(5)
  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号