Microfluidics

Microfluidics refers to the behaviour, precise control, and manipulation of fluids that are geometrically constrained to a small scale (typically sub-millimeter) at which capillary penetration governs mass transport. It is a multidisciplinary field that involves engineering, physics, chemistry, biochemistry, nanotechnology, and biotechnology. It has practical applications in the design of systems that process low volumes of fluids to achieve multiplexing, automation, and high-throughput screening. Microfluidics emerged in the beginning of the 1980s and is used in the development of inkjet printheads, DNA chips, lab-on-a-chip technology, micro-propulsion, and micro-thermal technologies.

Typically, micro means one of the following features:

  • Small volumes (μL, nL, pL, fL)
  • Small size
  • Low energy consumption
  • Microdomain effects

Typically microfluidic systems transport, mix, separate, or otherwise process fluids. Various applications rely on passive fluid control using capillary forces, in the form of capillary flow modifying elements, akin to flow resistors and flow accelerators. In some applications, external actuation means are additionally used for a directed transport of the media. Examples are rotary drives applying centrifugal forces for the fluid transport on the passive chips. Active microfluidics refers to the defined manipulation of the working fluid by active (micro) components such as micropumps or microvalves. Micropumps supply fluids in a continuous manner or are used for dosing. Microvalves determine the flow direction or the mode of movement of pumped liquids. Often, processes normally carried out in a lab are miniaturised on a single chip, which enhances efficiency and mobility, and reduces sample and reagent volumes.

Microscale behaviour of fluids

Silicone rubber and glass microfluidic devices. Top: a photograph of the devices. Bottom: Phase contrast micrographs of a serpentine channel ~15 μm wide.

The behaviour of fluids at the microscale can differ from "macrofluidic" behaviour in that factors such as surface tension, energy dissipation, and fluidic resistance start to dominate the system. Microfluidics studies how these behaviours change, and how they can be worked around, or exploited for new uses.[1][2][3][4][5]

At small scales (channel size of around 100 nanometers to 500 micrometers) some interesting and sometimes unintuitive properties appear. In particular, the Reynolds number (which compares the effect of the momentum of a fluid to the effect of viscosity) can become very low. A key consequence is co-flowing fluids do not necessarily mix in the traditional sense, as flow becomes laminar rather than turbulent; molecular transport between them must often be through diffusion.[6]

High specificity of chemical and physical properties (concentration, pH, temperature, shear force, etc.) can also be ensured resulting in more uniform reaction conditions and higher grade products in single and multi-step reactions.[7][8]

Key application areas

Microfluidic structures include micropneumatic systems, i.e. microsystems for the handling of off-chip fluids (liquid pumps, gas valves, etc.), and microfluidic structures for the on-chip handling of nanoliter (nl) and picoliter (pl) volumes.[9] To date, the most successful commercial application of microfluidics is the inkjet printhead.[10] Additionally, microfluidic manufacturing advances mean that makers can produce the devices in low-cost plastics[11] and automatically verify part quality.[12]

Advances in microfluidics technology are revolutionizing molecular biology procedures for enzymatic analysis (e.g., glucose and lactate assays), DNA analysis (e.g., polymerase chain reaction and high-throughput sequencing), proteomics, and in chemical synthesis.[13][14] The basic idea of microfluidic biochips is to integrate assay operations such as detection, as well as sample pre-treatment and sample preparation on one chip.[15][16]

An emerging application area for biochips is clinical pathology, especially the immediate point-of-care diagnosis of diseases.[17] In addition, microfluidics-based devices, capable of continuous sampling and real-time testing of air/water samples for biochemical toxins and other dangerous pathogens, can serve as an always-on "bio-smoke alarm" for early warning.

Microfluidic technology has led to the creation of powerful tools for biologists to control the complete cellular environment, leading to new questions and discoveries. Many diverse advantages of this technology for microbiology are listed below:

  • General single cell studies including growth [18][19]
  • Cellular aging: microfluidic devices such as the "mother machine" allow tracking of thousands of individual cells for many generations until they die.[18]
  • Microenvironmental control: ranging from mechanical environment [20] to chemical environment [21][22]
  • Precise spatiotemporal concentration gradients by incorporating multiple chemical inputs to a single device [23]
  • Force measurements of adherent cells or confined chromosomes: objects trapped in a microfluidic device can be directly manipulated using optical tweezers or other force-generating methods [24]
  • Confining cells and exerting controlled forces by coupling with external force-generation methods such as Stokes flow, optical tweezer, or controlled deformation of the PDMS (Polydimethylsiloxane) device [24][25][26]
  • Electric field integration [26]
  • Plant on a chip and plant tissue culture [27]
  • Antibiotic resistance: microfluidic devices can be used as heterogeneous environments for microorganisms. In a heterogeneous environment, it is easier for a microorganism to evolve. This can be useful for testing the acceleration of evolution of a microorganism / for testing the development of antibiotic resistance.

Some of these areas are further elaborated in the sections below.

Open microfluidics

In open microfluidics, at least one boundary of the system is removed, exposing the fluid to air or another interface (i.e. liquid).[28][29][30] Advantages of open microfluidics include accessibility to the flowing liquid for intervention, larger liquid-gas surface area, and minimized bubble formation.[28][30][31] Another advantage of open microfluidics is the ability to integrate open systems with surface-tension driven fluid flow, which eliminates the need for external pumping methods such as peristaltic or syringe pumps.[32] Open microfluidic devices are also easy and inexpensive to fabricate by milling, thermoforming, and hot embossing.[33][34][35][36] In addition, open microfluidics eliminates the need to glue or bond a cover for devices, which could be detrimental to capillary flows. Examples of open microfluidics include open-channel microfluidics, rail-based microfluidics, paper-based, and thread-based microfluidics.[28][32][37] Disadvantages to open systems include susceptibility to evaporation,[38] contamination,[39] and limited flow rate.[30]

Continuous-flow microfluidics

Continuous flow microfluidics rely on the control of a steady state liquid flow through narrow channels or porous media predominantly by accelerating or hindering fluid flow in capillary elements.[13] In paper based microfluidics, capillary elements can be achieved through the simple variation of section geometry. In general, the actuation of liquid flow is implemented either by external pressure sources, external mechanical pumps, integrated mechanical micropumps, or by combinations of capillary forces and electrokinetic mechanisms.[40][41] Continuous-flow microfluidic operation is the mainstream approach because it is easy to implement and less sensitive to protein fouling problems. Continuous-flow devices are adequate for many well-defined and simple biochemical applications, and for certain tasks such as chemical separation, but they are less suitable for tasks requiring a high degree of flexibility or fluid manipulations. These closed-channel systems are inherently difficult to integrate and scale because the parameters that govern flow field vary along the flow path making the fluid flow at any one location dependent on the properties of the entire system. Permanently etched microstructures also lead to limited reconfigurability and poor fault tolerance capability. Computer-aided design automation approaches for continuous-flow microfluidics have been proposed in recent years to alleviate the design effort and to solve the scalability problems.[42]

Process monitoring capabilities in continuous-flow systems can be achieved with highly sensitive microfluidic flow sensors based on MEMS technology, which offers resolutions down to the nanoliter range.

Droplet-based microfluidics

Droplet-based microfluidics is a subcategory of microfluidics in contrast with continuous microfluidics; droplet-based microfluidics manipulates discrete volumes of fluids in immiscible phases with low Reynolds number and laminar flow regimes. Interest in droplet-based microfluidics systems has been growing substantially in past decades. Microdroplets allow for handling miniature volumes (μl to fl) of fluids conveniently, provide better mixing, encapsulation, sorting, and sensing, and suit high throughput experiments.[19] Exploiting the benefits of droplet-based microfluidics efficiently requires a deep understanding of droplet generation [44] to perform various logical operations[45][46] such as droplet motion, droplet sorting, droplet merging, and droplet breakup.[47]

Digital microfluidics

Alternatives to the above closed-channel continuous-flow systems include novel open structures, where discrete, independently controllable droplets are manipulated on a substrate using electrowetting. Following the analogy of digital microelectronics, this approach is referred to as digital microfluidics. Le Pesant et al. pioneered the use of electrocapillary forces to move droplets on a digital track.[48] The "fluid transistor" pioneered by Cytonix[49] also played a role. The technology was subsequently commercialised by Duke University. By using discrete unit-volume droplets,[44] a microfluidic function can be reduced to a set of repeated basic operations, i.e., moving one unit of fluid over one unit of distance. This "digitisation" method facilitates the use of a hierarchical and cell-based approach for microfluidic biochip design. Therefore, digital microfluidics offers a flexible and scalable system architecture as well as high fault-tolerance capability. Moreover, because each droplet can be controlled independently, these systems also have dynamic reconfigurability, whereby groups of unit cells in a microfluidic array can be reconfigured to change their functionality during the concurrent execution of a set of bioassays. Although droplets are manipulated in confined microfluidic channels, since the control on droplets is not independent, it should not be confused as "digital microfluidics". One common actuation method for digital microfluidics is electrowetting-on-dielectric (EWOD).[50] Many lab-on-a-chip applications have been demonstrated within the digital microfluidics paradigm using electrowetting. However, recently other techniques for droplet manipulation have also been demonstrated using magnetic force,[51] surface acoustic waves, optoelectrowetting, mechanical actuation,[52] etc.

Paper-based microfluidics

Paper-based microfluidic devices fill a growing niche for portable, cheap, and user-friendly medical diagnostic systems.[53] Paper based microfluidics rely on the phenomenon of capillary penetration in porous media.[54] To tune fluid penetration in porous substrates such as paper in two and three dimensions, the pore structure, wettability and geometry of the microfluidic devices can be controlled while the viscosity and evaporation rate of the liquid play a further significant role. Many such devices feature hydrophobic barriers on hydrophilic paper that passively transport aqueous solutions to outlets where biological reactions take place.[55] Current applications include portable glucose detection[56] and environmental testing,[57] with hopes of reaching areas that lack advanced medical diagnostic tools.

Microfluidic-assisted magnetophoresis

One major area of application for microfluidic devices is the separation and sorting of different fluids or cell types. Recent developments in the microfluidics field have seen the integration of microfluidic devices with magnetophoresis: the migration of particles by a magnetic field.[58] This can be accomplished by sending a fluid containing at least one magnetic component through a microfluidic channel that has a magnet positioned along the length of the channel. This creates a magnetic field inside the microfluidic channel which draws magnetically active substances towards it, effectively separating the magnetic and non-magnetic components of the fluid. This technique can be readily utilized in industrial settings where the fluid at hand already contains magnetically active material. For example, a handful of metallic impurities can find their way into certain consumable liquids, namely milk and other dairy products.[59] Conveniently, in the case of milk, many of these metal contaminants exhibit paramagnetism. Therefore, before packaging, milk can be flowed through channels with magnetic gradients as a means of purifying out the metal contaminants.

Other, more research-oriented applications of microfluidic-assisted magnetophoresis are numerous and are generally targeted towards cell separation. The general way this is accomplished involves several steps. First, a paramagnetic substance (usually micro/nanoparticles or a paramagnetic fluid)[60] needs to be functionalized to target the cell type of interest. This can be accomplished by identifying a transmembranal protein unique to the cell type of interest and subsequently functionalizing magnetic particles with the complementary antigen or antibody.[61][62][63][64][65] Once the magnetic particles are functionalized, they are dispersed in a cell mixture where they bind to only the cells of interest. The resulting cell/particle mixture can then be flowed through a microfluidic device with a magnetic field to separate the targeted cells from the rest.

Conversely, microfluidic-assisted magnetophoresis may be used to facilitate efficient mixing within microdroplets or plugs. To accomplish this, microdroplets are injected with paramagnetic nanoparticles and are flowed through a straight channel which passes through rapidly alternating magnetic fields. This causes the magnetic particles to be quickly pushed from side to side within the droplet and results in the mixing of the microdroplet contents.[64] This eliminates the need for tedious engineering considerations that are necessary for traditional, channel-based droplet mixing. Other research has also shown that the label-free separation of cells may be possible by suspending cells in a paramagnetic fluid and taking advantage of the magneto-Archimedes effect.[66][67] While this does eliminate the complexity of particle functionalization, more research is needed to fully understand the magneto-Archimedes phenomenon and how it can be used to this end. This is not an exhaustive list of the various applications of microfluidic-assisted magnetophoresis; the above examples merely highlight the versatility of this separation technique in both current and future applications.

DNA chips (microarrays)

Early biochips were based on the idea of a DNA microarray, e.g., the GeneChip DNAarray from Affymetrix, which is a piece of glass, plastic or silicon substrate, on which pieces of DNA (probes) are affixed in a microscopic array. Similar to a DNA microarray, a protein array is a miniature array where a multitude of different capture agents, most frequently monoclonal antibodies, are deposited on a chip surface; they are used to determine the presence and/or amount of proteins in biological samples, e.g., blood. A drawback of DNA and protein arrays is that they are neither reconfigurable nor scalable after manufacture. Digital microfluidics has been described as a means for carrying out Digital PCR.

Molecular biology

In addition to microarrays, biochips have been designed for two-dimensional electrophoresis,[68] transcriptome analysis,[69] and PCR amplification.[70] Other applications include various electrophoresis and liquid chromatography applications for proteins and DNA, cell separation, in particular, blood cell separation, protein analysis, cell manipulation and analysis including cell viability analysis [19] and microorganism capturing.[16]

Evolutionary biology

By combining microfluidics with landscape ecology and nanofluidics, a nano/micro fabricated fluidic landscape can be constructed by building local patches of bacterial habitat and connecting them by dispersal corridors. The resulting landscapes can be used as physical implementations of an adaptive landscape,[71] by generating a spatial mosaic of patches of opportunity distributed in space and time. The patchy nature of these fluidic landscapes allows for the study of adapting bacterial cells in a metapopulation system. The evolutionary ecology of these bacterial systems in these synthetic ecosystems allows for using biophysics to address questions in evolutionary biology.

Cell behavior

The ability to create precise and carefully controlled chemoattractant gradients makes microfluidics the ideal tool to study motility,[72] chemotaxis and the ability to evolve / develop resistance to antibiotics in small populations of microorganisms and in a short period of time. These microorganisms including bacteria [73] and the broad range of organisms that form the marine microbial loop,[74] responsible for regulating much of the oceans' biogeochemistry.

Microfluidics has also greatly aided the study of durotaxis by facilitating the creation of durotactic (stiffness) gradients.

Cellular biophysics

By rectifying the motion of individual swimming bacteria,[75] microfluidic structures can be used to extract mechanical motion from a population of motile bacterial cells.[76] This way, bacteria-powered rotors can be built.[77][78]

Optics

The merger of microfluidics and optics is typical known as optofluidics. Examples of optofluidic devices are tunable microlens arrays[79][80] and optofluidic microscopes.

Microfluidic flow enables fast sample throughput, automated imaging of large sample populations, as well as 3D capabilities.[81][82] or superresolution.[83]

High Performance Liquid Chromatography (HPLC)

HPLC in the field of microfluidics comes in two different forms. Early designs included running liquid through the HPLC column then transferring the eluted liquid to microfluidic chips and attaching HPLC columns to the microfluidic chip directly.[84] The early methods had the advantage of easier detection from certain machines like those that measure fluorescence.[85] More recent designs have fully integrated HPLC columns into microfluidic chips. The main advantage of integrating HPLC columns into microfluidic devices is the smaller form factor that can be achieved, which allows for additional features to be combined within one microfluidic chip. Integrated chips can also be fabricated from multiple different materials, including glass and polyimide which are quite different from the standard material of PDMS used in many different droplet-based microfluidic devices.[86][87] This is an important feature because different applications of HPLC microfluidic chips may call for different pressures. PDMS fails in comparison for high-pressure uses compared to glass and polyimide. High versatility of HPLC integration ensures robustness by avoiding connections and fittings between the column and chip.[88] The ability to build off said designs in the future allows the field of microfluidics to continue expanding its potential applications.

The potential applications surrounding integrated HPLC columns within microfluidic devices have proven expansive over the last 10–15 years. The integration of such columns allows for experiments to be run where materials were in low availability or very expensive, like in biological analysis of proteins. This reduction in reagent volumes allows for new experiments like single-cell protein analysis, which due to size limitations of prior devices, previously came with great difficulty.[89] The coupling of HPLC-chip devices with other spectrometry methods like mass-spectrometry allow for enhanced confidence in identification of desired species, like proteins.[90] Microfluidic chips have also been created with internal delay-lines that allow for gradient generation to further improve HPLC, which can reduce the need for further separations.[91] Some other practical applications of integrated HPLC chips include the determination of drug presence in a person through their hair[92] and the labeling of peptides through reverse phase liquid chromatography.[93]

Acoustic droplet ejection (ADE)

Acoustic droplet ejection uses a pulse of ultrasound to move low volumes of fluids (typically nanoliters or picoliters) without any physical contact. This technology focuses acoustic energy into a fluid sample to eject droplets as small as a millionth of a millionth of a litre (picoliter = 10−12 litre). ADE technology is a very gentle process, and it can be used to transfer proteins, high molecular weight DNA and live cells without damage or loss of viability. This feature makes the technology suitable for a wide variety of applications including proteomics and cell-based assays.

Fuel cells

Microfluidic fuel cells can use laminar flow to separate the fuel and its oxidant to control the interaction of the two fluids without the physical barrier that conventional fuel cells require.[94][95][96]

Astrobiology

To understand the prospects for life to exist elsewhere in the universe, astrobiologists are interested in measuring the chemical composition of extraplanetary bodies.[97] Because of their small size and wide-ranging functionality, microfluidic devices are uniquely suited for these remote sample analyses.[98][99][100] From an extraterrestrial sample, the organic content can be assessed using microchip capillary electrophoresis and selective fluorescent dyes.[101] These devices are capable of detecting amino acids,[102] peptides,[103] fatty acids,[104] and simple aldehydes, ketones,[105] and thiols.[106] These analyses coupled together could allow powerful detection of the key components of life, and hopefully inform our search for functioning extraterrestrial life.[107]

Future directions

  • Microfluidic drug assays:[108]
  • On-chip characterization:[109]
  • Microfluidics in the classroom: On-chip acid-base titrations[110]
  • Sepsis detection in minutes not days.
  • Unlocking multi-angle imaging for microfluidic devices [111]

See also

References

  1. Terry SC, Jerman JH, Angell JB (December 1979). "A gas chromatographic air analyzer fabricated on a silicon wafer". IEEE Transactions on Electron Devices. 26 (12): 1880–6. doi:10.1109/T-ED.1979.19791.
  2. Kirby BJ (2010). Micro- and Nanoscale Fluid Mechanics: Transport in Microfluidic Devices. Cambridge University Press.
  3. Karniadakis GM, Beskok A, Aluru N (2005). Microflows and Nanoflows. Springer Verlag.
  4. Bruus H (2007). Theoretical Microfluidics. Oxford University Press.
  5. Shkolnikov V (2019). Principles of Microfluidics. ISBN 978-1790217281.
  6. Tabeling P (2005). Introduction to Microfluidics. Oxford University Press.
  7. Chokkalingam V, Weidenhof B, Krämer M, Maier WF, Herminghaus S, Seemann R (July 2010). "Optimized droplet-based microfluidics scheme for sol-gel reactions". Lab on a Chip. 10 (13): 1700–5. doi:10.1039/b926976b. PMID 20405061.
  8. Shestopalov I, Tice JD, Ismagilov RF (August 2004). "Multi-step synthesis of nanoparticles performed on millisecond time scale in a microfluidic droplet-based system" (PDF). Lab on a Chip. 4 (4): 316–21. doi:10.1039/b403378g. PMID 15269797.
  9. Nguyen NT, Wereley S (2006). Fundamentals and Applications of Microfluidics. Artech House.
  10. DeMello AJ (July 2006). "Control and detection of chemical reactions in microfluidic systems". Nature. 442 (7101): 394–402. Bibcode:2006Natur.442..394D. doi:10.1038/nature05062. PMID 16871207.
  11. Pawell RS, Inglis DW, Barber TJ, Taylor RA (2013). "Manufacturing and wetting low-cost microfluidic cell separation devices". Biomicrofluidics. 7 (5): 56501. doi:10.1063/1.4821315. PMC 3785532. PMID 24404077.
  12. Pawell RS, Taylor RA, Morris KV, Barber TJ (2015). "Automating microfluidic part verification". Microfluidics and Nanofluidics. 18 (4): 657–665. doi:10.1007/s10404-014-1464-1.
  13. Konda A, Morin SA (June 2017). "Flow-directed synthesis of spatially variant arrays of branched zinc oxide mesostructures". Nanoscale. 9 (24): 8393–8400. doi:10.1039/C7NR02655B. PMID 28604901.
  14. Cheng JJ, Nicaise SM, Berggren KK, Gradečak S (January 2016). "Dimensional Tailoring of Hydrothermally Grown Zinc Oxide Nanowire Arrays". Nano Letters. 16 (1): 753–9. Bibcode:2016NanoL..16..753C. doi:10.1021/acs.nanolett.5b04625. PMID 26708095.
  15. Herold KE (2009). Rasooly A (ed.). Lab-on-a-Chip Technology: Fabrication and Microfluidics. Caister Academic Press. ISBN 978-1-904455-46-2.
  16. Herold KE (2009). Rasooly A (ed.). Lab-on-a-Chip Technology: Biomolecular Separation and Analysis. Caister Academic Press. ISBN 978-1-904455-47-9.
  17. Barrett MP, Cooper JM, Regnault C, Holm SH, Beech JP, Tegenfeldt JO, Hochstetter A (October 2017). "Microfluidics-Based Approaches to the Isolation of African Trypanosomes". Pathogens. 6 (4): 47. doi:10.3390/pathogens6040047. PMC 5750571. PMID 28981471.
  18. Wang P, Robert L, Pelletier J, Dang WL, Taddei F, Wright A, Jun S (June 2010). "Robust growth of Escherichia coli". Current Biology. 20 (12): 1099–103. doi:10.1016/j.cub.2010.04.045. PMC 2902570. PMID 20537537.
  19. Chokkalingam V, Tel J, Wimmers F, Liu X, Semenov S, Thiele J, et al. (December 2013). "Probing cellular heterogeneity in cytokine-secreting immune cells using droplet-based microfluidics". Lab on a Chip. 13 (24): 4740–4. doi:10.1039/C3LC50945A. PMID 24185478.
  20. Manbachi A, Shrivastava S, Cioffi M, Chung BG, Moretti M, Demirci U, et al. (May 2008). "Microcirculation within grooved substrates regulates cell positioning and cell docking inside microfluidic channels". Lab on a Chip. 8 (5): 747–54. doi:10.1039/B718212K. PMC 2668874. PMID 18432345.
  21. Yliperttula M, Chung BG, Navaladi A, Manbachi A, Urtti A (October 2008). "High-throughput screening of cell responses to biomaterials". European Journal of Pharmaceutical Sciences. 35 (3): 151–60. doi:10.1016/j.ejps.2008.04.012. PMID 18586092.
  22. Gilbert DF, Mofrad SA, Friedrich O, Wiest J (February 2019). "Proliferation characteristics of cells cultured under periodic versus static conditions". Cytotechnology. 71 (1): 443–452. doi:10.1007/s10616-018-0263-z. PMC 6368509. PMID 30515656.
  23. Chung BG, Manbachi A, Saadi W, Lin F, Jeon NL, Khademhosseini A (2007). "A gradient-generating microfluidic device for cell biology". Journal of Visualized Experiments. 7 (7): 271. doi:10.3791/271. PMC 2565846. PMID 18989442.
  24. Pelletier J, Halvorsen K, Ha BY, Paparcone R, Sandler SJ, Woldringh CL, et al. (October 2012). "Physical manipulation of the Escherichia coli chromosome reveals its soft nature". Proceedings of the National Academy of Sciences of the United States of America. 109 (40): E2649-56. Bibcode:2012PNAS..109E2649P. doi:10.1073/pnas.1208689109. PMC 3479577. PMID 22984156.
  25. Amir A, Babaeipour F, McIntosh DB, Nelson DR, Jun S (April 2014). "Bending forces plastically deform growing bacterial cell walls". Proceedings of the National Academy of Sciences of the United States of America. 111 (16): 5778–83. arXiv:1305.5843. Bibcode:2014PNAS..111.5778A. doi:10.1073/pnas.1317497111. PMC 4000856. PMID 24711421.
  26. Choi JW, Rosset S, Niklaus M, Adleman JR, Shea H, Psaltis D (March 2010). "3-dimensional electrode patterning within a microfluidic channel using metal ion implantation" (PDF). Lab on a Chip. 10 (6): 783–8. doi:10.1039/B917719A. PMID 20221568.
  27. Yetisen AK, Jiang L, Cooper JR, Qin Y, Palanivelu R, Zohar Y (May 2011). "A microsystem-based assay for studying pollen tube guidance in plant reproduction". J. Micromech. Microeng. 25 (5): 054018. Bibcode:2011JMiMi..21e4018Y. doi:10.1088/0960-1317/21/5/054018.
  28. Berthier J, Brakke KA, Berthier E (2016-08-01). Open Microfluidics. doi:10.1002/9781118720936. ISBN 9781118720936.
  29. Pfohl T, Mugele F, Seemann R, Herminghaus S (December 2003). "Trends in microfluidics with complex fluids". Chemphyschem. 4 (12): 1291–8. doi:10.1002/cphc.200300847. PMID 14714376.
  30. Kaigala GV, Lovchik RD, Delamarche E (November 2012). "Microfluidics in the "open space" for performing localized chemistry on biological interfaces". Angewandte Chemie. 51 (45): 11224–40. doi:10.1002/anie.201201798. PMID 23111955.
  31. Li C, Boban M, Tuteja A (April 2017). "Open-channel, water-in-oil emulsification in paper-based microfluidic devices". Lab on a Chip. 17 (8): 1436–1441. doi:10.1039/c7lc00114b. PMID 28322402.
  32. Casavant BP, Berthier E, Theberge AB, Berthier J, Montanez-Sauri SI, Bischel LL, et al. (June 2013). "Suspended microfluidics". Proceedings of the National Academy of Sciences of the United States of America. 110 (25): 10111–6. Bibcode:2013PNAS..11010111C. doi:10.1073/pnas.1302566110. PMC 3690848. PMID 23729815.
  33. Guckenberger DJ, de Groot TE, Wan AM, Beebe DJ, Young EW (June 2015). "Micromilling: a method for ultra-rapid prototyping of plastic microfluidic devices". Lab on a Chip. 15 (11): 2364–78. doi:10.1039/c5lc00234f. PMC 4439323. PMID 25906246.
  34. Truckenmüller R, Rummler Z, Schaller T, Schomburg WK (2002-06-13). "Low-cost thermoforming of micro fluidic analysis chips". Journal of Micromechanics and Microengineering. 12 (4): 375–379. Bibcode:2002JMiMi..12..375T. doi:10.1088/0960-1317/12/4/304. ISSN 0960-1317.
  35. Jeon JS, Chung S, Kamm RD, Charest JL (April 2011). "Hot embossing for fabrication of a microfluidic 3D cell culture platform". Biomedical Microdevices. 13 (2): 325–33. doi:10.1007/s10544-010-9496-0. PMC 3117225. PMID 21113663.
  36. Young EW, Berthier E, Guckenberger DJ, Sackmann E, Lamers C, Meyvantsson I, et al. (February 2011). "Rapid prototyping of arrayed microfluidic systems in polystyrene for cell-based assays". Analytical Chemistry. 83 (4): 1408–17. doi:10.1021/ac102897h. PMC 3052265. PMID 21261280.
  37. Bouaidat S, Hansen O, Bruus H, Berendsen C, Bau-Madsen NK, Thomsen P, et al. (August 2005). "Surface-directed capillary system; theory, experiments and applications". Lab on a Chip. 5 (8): 827–36. doi:10.1039/b502207j. PMID 16027933.
  38. Kachel S, Zhou Y, Scharfer P, Vrančić C, Petrich W, Schabel W (February 2014). "Evaporation from open microchannel grooves". Lab on a Chip. 14 (4): 771–8. doi:10.1039/c3lc50892g. PMID 24345870.
  39. Ogawa M, Higashi K, Miki N (August 2015). "Development of hydrogel microtubes for microbe culture in open environment". Conference Proceedings. 2015 (6): 5896–9. doi:10.3390/mi8060176. PMC 6190135.
  40. Chang HC, Yeo L (2009). Electrokinetically Driven Microfluidics and Nanofluidics. Cambridge University Press.
  41. fluid transistor Archived July 8, 2011, at the Wayback Machine
  42. Tseng T, Li M, Freitas DN, McAuley T, Li B, Ho T, Araci IE, Schlichtmann U (2018). "Columba 2.0: A Co-Layout Synthesis Tool for Continuous-Flow Microfluidic Biochips". IEEE Transactions on Computer-Aided Design of Integrated Circuits and Systems. 37 (8): 1588–1601. doi:10.1109/TCAD.2017.2760628.
  43. Churchman AH (2018). "Data associated with 'Combined flow-focus and self-assembly routes for the formation of lipid stabilized oil-shelled microbubbles'". University of Leeds. doi:10.5518/153. Cite journal requires |journal= (help)
  44. Chokkalingam V, Herminghaus S, Seemann R (2008). "Self-synchronizing Pairwise Production of Monodisperse Droplets by Microfluidic Step Emulsification". Applied Physics Letters. 93 (25): 254101. Bibcode:2008ApPhL..93y4101C. doi:10.1063/1.3050461. Archived from the original on 2013-01-13.
  45. Teh SY, Lin R, Hung LH, Lee AP (February 2008). "Droplet microfluidics". Lab on a Chip. 8 (2): 198–220. doi:10.1039/B715524G. PMID 18231657.
  46. Prakash M, Gershenfeld N (February 2007). "Microfluidic bubble logic". Science. 315 (5813): 832–5. Bibcode:2007Sci...315..832P. CiteSeerX 10.1.1.673.2864. doi:10.1126/science.1136907. PMID 17289994.
  47. Samie M, Salari A, Shafii MB (May 2013). "Breakup of microdroplets in asymmetric T junctions". Physical Review. E, Statistical, Nonlinear, and Soft Matter Physics. 87 (5): 053003. Bibcode:2013PhRvE..87e3003S. doi:10.1103/PhysRevE.87.053003. PMID 23767616.
  48. Le Pesant et al., Electrodes for a device operating by electrically controlled fluid displacement, U.S. Pat. No. 4,569,575, Feb. 11, 1986.
  49. NSF Award Search: Advanced Search Results
  50. Junghoon Lee, Chang-Jin Kim (June 2000). "Surface-tension-driven microactuation based on continuous electrowetting". Journal of Microelectromechanical Systems. 9 (2): 171–180. doi:10.1109/84.846697. ISSN 1057-7157.
  51. Zhang Y, Nguyen NT (March 2017). "Magnetic digital microfluidics - a review". Lab on a Chip. 17 (6): 994–1008. doi:10.1039/c7lc00025a. hdl:10072/344389. PMID 28220916.
  52. Shemesh J, Bransky A, Khoury M, Levenberg S (October 2010). "Advanced microfluidic droplet manipulation based on piezoelectric actuation". Biomedical Microdevices. 12 (5): 907–14. doi:10.1007/s10544-010-9445-y. PMID 20559875.
  53. Berthier J, Brakke KA, Berthier E (2016). Open Microfluidics. John Wiley & Sons, Inc. pp. 229–256. doi:10.1002/9781118720936.ch7. ISBN 9781118720936.
  54. Liu M, Suo S, Wu J, Gan Y, Ah Hanaor D, Chen CQ (March 2019). "Tailoring porous media for controllable capillary flow". Journal of Colloid and Interface Science. 539: 379–387. Bibcode:2019JCIS..539..379L. doi:10.1016/j.jcis.2018.12.068. PMID 30594833.
  55. Galindo-Rosales, Francisco José (2017-05-26). Complex Fluid-Flows in Microfluidics. Springer. ISBN 9783319595931.
  56. Martinez AW, Phillips ST, Butte MJ, Whitesides GM (2007). "Patterned paper as a platform for inexpensive, low-volume, portable bioassays". Angewandte Chemie. 46 (8): 1318–20. doi:10.1002/anie.200603817. PMC 3804133. PMID 17211899.
  57. Park TS, Yoon J (2015-03-01). "Smartphone Detection of Escherichia coli From Field Water Samples on Paper Microfluidics". IEEE Sensors Journal. 15 (3): 1902. Bibcode:2015ISenJ..15.1902P. doi:10.1109/JSEN.2014.2367039.
  58. Munaz A, Shiddiky MJ, Nguyen NT (May 2018). "Recent advances and current challenges in magnetophoresis based micro magnetofluidics". Biomicrofluidics. 12 (3): 031501. doi:10.1063/1.5035388. PMC 6013300. PMID 29983837.
  59. Dibaji S, Rezai P (2020-06-01). "Triplex Inertia-Magneto-Elastic (TIME) sorting of microparticles in non-Newtonian fluids". Journal of Magnetism and Magnetic Materials. 503: 166620. doi:10.1016/j.jmmm.2020.166620. ISSN 0304-8853.
  60. Alnaimat F, Dagher S, Mathew B, Hilal-Alnqbi A, Khashan S (November 2018). "Microfluidics Based Magnetophoresis: A Review". Chemical Record. 18 (11): 1596–1612. doi:10.1002/tcr.201800018. PMID 29888856.
  61. Dibaji S, Rezai P (2020-06-01). "Triplex Inertia-Magneto-Elastic (TIME) sorting of microparticles in non-Newtonian fluids". Journal of Magnetism and Magnetic Materials. 503: 166620. doi:10.1016/j.jmmm.2020.166620. ISSN 0304-8853.
  62. Unni M, Zhang J, George TJ, Segal MS, Fan ZH, Rinaldi C (March 2020). "Engineering magnetic nanoparticles and their integration with microfluidics for cell isolation". Journal of Colloid and Interface Science. 564: 204–215. doi:10.1016/j.jcis.2019.12.092. PMC 7023483. PMID 31911225.
  63. Xia N, Hunt TP, Mayers BT, Alsberg E, Whitesides GM, Westervelt RM, Ingber DE (December 2006). "Combined microfluidic-micromagnetic separation of living cells in continuous flow". Biomedical Microdevices. 8 (4): 299–308. doi:10.1007/s10544-006-0033-0. PMID 17003962.
  64. Pamme N (January 2006). "Magnetism and microfluidics". Lab on a Chip. 6 (1): 24–38. doi:10.1039/B513005K. PMID 16372066.
  65. Song K, Li G, Zu X, Du Z, Liu L, Hu Z (March 2020). "The Fabrication and Application Mechanism of Microfluidic Systems for High Throughput Biomedical Screening: A Review". Micromachines. 11 (3): 297. doi:10.3390/mi11030297. PMC 7143183. PMID 32168977.
  66. Gao Q, Zhang W, Zou H, Li W, Yan H, Peng Z, Meng G (2019). "Label-free manipulation via the magneto-Archimedes effect: fundamentals, methodology and applications". Materials Horizons. 6 (7): 1359–1379. doi:10.1039/C8MH01616J. ISSN 2051-6347.
  67. Akiyama Y, Morishima K (2011-04-18). "Label-free cell aggregate formation based on the magneto-Archimedes effect". Applied Physics Letters. 98 (16): 163702. doi:10.1063/1.3581883. ISSN 0003-6951.
  68. Fan H, Das C, Chen H (2009). "Two-Dimensional Electrophoresis in a Chip". In Herold KE, Rasooly A (eds.). Lab-on-a-Chip Technology: Biomolecular Separation and Analysis. Caister Academic Press. ISBN 978-1-904455-47-9.
  69. Bontoux N, Dauphinot L, Potier MC (2009). "Elaborating Lab-on-a-Chips for Single-cell Transcriptome Analysis". In Herold KE, Rasooly A (eds.). Lab-on-a-Chip Technology: Biomolecular Separation and Analysis. Caister Academic Press. ISBN 978-1-904455-47-9.
  70. Cady NC (2009). "Microchip-based PCR Amplification Systems". Lab-on-a-Chip Technology: Biomolecular Separation and Analysis. Caister Academic Press. ISBN 978-1-904455-47-9.
  71. Keymer JE, Galajda P, Muldoon C, Park S, Austin RH (November 2006). "Bacterial metapopulations in nanofabricated landscapes". Proceedings of the National Academy of Sciences of the United States of America. 103 (46): 17290–5. Bibcode:2006PNAS..10317290K. doi:10.1073/pnas.0607971103. PMC 1635019. PMID 17090676.
  72. Hochstetter A, Stellamanns E, Deshpande S, Uppaluri S, Engstler M, Pfohl T (April 2015). "Microfluidics-based single cell analysis reveals drug-dependent motility changes in trypanosomes" (PDF). Lab on a Chip. 15 (8): 1961–8. doi:10.1039/C5LC00124B. PMID 25756872.
  73. Ahmed T, Shimizu TS, Stocker R (November 2010). "Microfluidics for bacterial chemotaxis". Integrative Biology. 2 (11–12): 604–29. doi:10.1039/C0IB00049C. hdl:1721.1/66851. PMID 20967322.
  74. Seymour JR, Simó R, Ahmed T, Stocker R (July 2010). "Chemoattraction to dimethylsulfoniopropionate throughout the marine microbial food web". Science. 329 (5989): 342–5. Bibcode:2010Sci...329..342S. doi:10.1126/science.1188418. PMID 20647471.
  75. Galajda P, Keymer J, Chaikin P, Austin R (December 2007). "A wall of funnels concentrates swimming bacteria". Journal of Bacteriology. 189 (23): 8704–7. doi:10.1128/JB.01033-07. PMC 2168927. PMID 17890308.
  76. Angelani L, Di Leonardo R, Ruocco G (January 2009). "Self-starting micromotors in a bacterial bath". Physical Review Letters. 102 (4): 048104. arXiv:0812.2375. Bibcode:2009PhRvL.102d8104A. doi:10.1103/PhysRevLett.102.048104. PMID 19257480.
  77. Di Leonardo R, Angelani L, Dell'arciprete D, Ruocco G, Iebba V, Schippa S, et al. (May 2010). "Bacterial ratchet motors". Proceedings of the National Academy of Sciences of the United States of America. 107 (21): 9541–5. arXiv:0910.2899. Bibcode:2010PNAS..107.9541D. doi:10.1073/pnas.0910426107. PMC 2906854. PMID 20457936.
  78. Sokolov A, Apodaca MM, Grzybowski BA, Aranson IS (January 2010). "Swimming bacteria power microscopic gears". Proceedings of the National Academy of Sciences of the United States of America. 107 (3): 969–74. Bibcode:2010PNAS..107..969S. doi:10.1073/pnas.0913015107. PMC 2824308. PMID 20080560.
  79. Grilli S, Miccio L, Vespini V, Finizio A, De Nicola S, Ferraro P (May 2008). "Liquid micro-lens array activated by selective electrowetting on lithium niobate substrates". Optics Express. 16 (11): 8084–93. Bibcode:2008OExpr..16.8084G. doi:10.1364/OE.16.008084. PMID 18545521.
  80. Ferraro P, Miccio L, Grilli S, Finizio A, De Nicola S, Vespini V (2008). "Manipulating Thin Liquid Films for Tunable Microlens Arrays". Optics and Photonics News. 19 (12): 34. doi:10.1364/OPN.19.12.000034.
  81. Pégard NC, Toth ML, Driscoll M, Fleischer JW (December 2014). "Flow-scanning optical tomography". Lab on a Chip. 14 (23): 4447–50. doi:10.1039/C4LC00701H. PMC 5859944. PMID 25256716.
  82. Pégard NC, Fleischer JW (2012). "3D microfluidic microscopy using a tilted channel". Biomedical Optics and 3-D Imaging. pp. BM4B.4. doi:10.1364/BIOMED.2012.BM4B.4. ISBN 978-1-55752-942-8.
  83. Lu C, Pégard NC, Fleischer JW (22 April 2013). "Flow-based structured illumination". Applied Physics Letters. 102 (16): 161115. doi:10.1063/1.4802091.
  84. Kim JY, Cho SW, Kang DK, Edel JB, Chang SI, deMello AJ, O'Hare D (September 2012). "Lab-chip HPLC with integrated droplet-based microfluidics for separation and high frequency compartmentalisation". Chemical Communications. Cambridge, England. 48 (73): 9144–6. doi:10.1039/c2cc33774f. PMID 22871959.
  85. Ochoa A, Álvarez-Bohórquez E, Castillero E, Olguin LF (May 2017). "Detection of Enzyme Inhibitors in Crude Natural Extracts Using Droplet-Based Microfluidics Coupled to HPLC". Analytical Chemistry. 89 (9): 4889–4896. doi:10.1021/acs.analchem.6b04988. PMID 28374582.
  86. Gerhardt RF, Peretzki AJ, Piendl SK, Belder D (December 2017). "Seamless Combination of High-Pressure Chip-HPLC and Droplet Microfluidics on an Integrated Microfluidic Glass Chip". Analytical Chemistry. 89 (23): 13030–13037. doi:10.1021/acs.analchem.7b04331. PMID 29096060.
  87. Killeen K, Yin H, Sobek D, Brennen R, Van de Goor T (October 2003). Chip-LC/MS: HPLC-MS using polymer microfluidics (PDF). 7th lnternatonal Conference on Miniaturized Chemical and Blochemlcal Analysts Systems. Proc MicroTAS. Squaw Valley, Callfornla USA. pp. 481–484.
  88. Vollmer M, Hörth P, Rozing G, Couté Y, Grimm R, Hochstrasser D, Sanchez JC (March 2006). "Multi-dimensional HPLC/MS of the nucleolar proteome using HPLC-chip/MS". Journal of Separation Science. 29 (4): 499–509. doi:10.1002/jssc.200500334. PMID 16583688.
  89. Reichmuth DS, Shepodd TJ, Kirby BJ (May 2005). "Microchip HPLC of peptides and proteins". Analytical Chemistry. 77 (9): 2997–3000. doi:10.1021/ac048358r. PMID 15859622.
  90. Hardouin J, Duchateau M, Joubert-Caron R, Caron M (2006). "Usefulness of an integrated microfluidic device (HPLC-Chip-MS) to enhance confidence in protein identification by proteomics". Rapid Communications in Mass Spectrometry : RCM. 20 (21): 3236–44. doi:10.1002/rcm.2725. PMID 17016832.
  91. Brennen RA, Yin H, Killeen KP (December 2007). "Microfluidic gradient formation for nanoflow chip LC". Analytical Chemistry. 79 (24): 9302–9. doi:10.1021/ac0712805. PMID 17997523.
  92. Zhu KY, Leung KW, Ting AK, Wong ZC, Ng WY, Choi RC, Dong TT, Wang T, Lau DT, Tsim KW (March 2012). "Microfluidic chip based nano liquid chromatography coupled to tandem mass spectrometry for the determination of abused drugs and metabolites in human hair". Analytical and Bioanalytical Chemistry. 402 (9): 2805–15. doi:10.1007/s00216-012-5711-6. PMID 22281681.
  93. Polat AN, Kraiczek K, Heck AJ, Raijmakers R, Mohammed S (November 2012). "Fully automated isotopic dimethyl labeling and phosphopeptide enrichment using a microfluidic HPLC phosphochip". Analytical and Bioanalytical Chemistry. 404 (8): 2507–12. doi:10.1007/s00216-012-6395-7. PMID 22975804.
  94. Santiago JG. "Water Management in PEM Fuel Cells". Stanford Microfluidics Laboratory. Archived from the original on 28 June 2008.
  95. Tretkoff E (May 2005). "Building a Better Fuel Cell Using Microfluidics". APS News. 14 (5): 3.
  96. Allen J. "Fuel Cell Initiative at MnIT Microfluidics Laboratory". Michigan Technological University.
  97. "NASA Astrobiology Strategy, 2015" (PDF). Archived from the original (PDF) on 2016-12-22.
  98. Beebe DJ, Mensing GA, Walker GM (2002). "Physics and applications of microfluidics in biology". Annual Review of Biomedical Engineering. 4: 261–86. doi:10.1146/annurev.bioeng.4.112601.125916. PMID 12117759.
  99. Theberge AB, Courtois F, Schaerli Y, Fischlechner M, Abell C, Hollfelder F, Huck WT (August 2010). "Microdroplets in microfluidics: an evolving platform for discoveries in chemistry and biology" (PDF). Angewandte Chemie. 49 (34): 5846–68. doi:10.1002/anie.200906653. PMID 20572214.
  100. van Dinther AM, Schroën CG, Vergeldt FJ, van der Sman RG, Boom RM (May 2012). "Suspension flow in microfluidic devices--a review of experimental techniques focussing on concentration and velocity gradients". Advances in Colloid and Interface Science. 173: 23–34. doi:10.1016/j.cis.2012.02.003. PMID 22405541.
  101. Mora MF, Greer F, Stockton AM, Bryant S, Willis PA (November 2011). "Toward total automation of microfluidics for extraterrestial in situ analysis". Analytical Chemistry. 83 (22): 8636–41. doi:10.1021/ac202095k. PMID 21972965.
  102. Chiesl TN, Chu WK, Stockton AM, Amashukeli X, Grunthaner F, Mathies RA (April 2009). "Enhanced amine and amino acid analysis using Pacific Blue and the Mars Organic Analyzer microchip capillary electrophoresis system". Analytical Chemistry. 81 (7): 2537–44. doi:10.1021/ac8023334. PMID 19245228.
  103. Kaiser RI, Stockton AM, Kim YS, Jensen EC, Mathies RA (2013). "On the Formation of Dipeptides in Interstellar Model Ices". The Astrophysical Journal. 765 (2): 111. Bibcode:2013ApJ...765..111K. doi:10.1088/0004-637X/765/2/111. ISSN 0004-637X.
  104. Stockton AM, Tjin CC, Chiesl TN, Mathies RA (July 2011). "Analysis of carbonaceous biomarkers with the Mars Organic Analyzer microchip capillary electrophoresis system: carboxylic acids". Astrobiology. 11 (6): 519–28. Bibcode:2011AsBio..11..519S. doi:10.1089/ast.2011.0634. PMID 21790324.
  105. Stockton AM, Tjin CC, Huang GL, Benhabib M, Chiesl TN, Mathies RA (November 2010). "Analysis of carbonaceous biomarkers with the Mars Organic Analyzer microchip capillary electrophoresis system: aldehydes and ketones". Electrophoresis. 31 (22): 3642–9. doi:10.1002/elps.201000424. PMID 20967779.
  106. Mora MF, Stockton AM, Willis PA (2015). "Analysis of thiols by microchip capillary electrophoresis for in situ planetary investigations". Methods in Molecular Biology. New York, NY: Humana Press. 1274: 43–52. doi:10.1007/978-1-4939-2353-3_4. ISBN 9781493923526. PMID 25673481.
  107. Bowden SA, Wilson R, Taylor C, Cooper JM, Parnell J (January 2007). "The extraction of intracrystalline biomarkers and other organic compounds from sulphate minerals using a microfluidic format – a feasibility study for remote fossil-life detection using a microfluidic H-cell". International Journal of Astrobiology. 6 (1): 27–36. Bibcode:2007IJAsB...6...27B. doi:10.1017/S147355040600351X. ISSN 1475-3006.
  108. Regnault C, Dheeman DS, Hochstetter A (June 2018). "Microfluidic Devices for Drug Assays". High-Throughput. 7 (2): 18. doi:10.3390/ht7020018. PMC 6023517. PMID 29925804.
  109. Greener J, Tumarkin E, Debono M, Kwan CH, Abolhasani M, Guenther A, Kumacheva E (January 2012). "Development and applications of a microfluidic reactor with multiple analytical probes". The Analyst. 137 (2): 444–50. Bibcode:2012Ana...137..444G. doi:10.1039/C1AN15940B. PMID 22108956.
  110. Greener J, Tumarkin E, Debono M, Dicks AP, Kumacheva E (February 2012). "Education: a microfluidic platform for university-level analytical chemistry laboratories". Lab on a Chip. 12 (4): 696–701. doi:10.1039/C2LC20951A. PMID 22237720.
  111. Hochstetter A (December 2019). "Presegmentation Procedure Generates Smooth-Sided Microfluidic Devices: Unlocking Multiangle Imaging for Everyone?". ACS Omega. 4 (25): 20972–20977. doi:10.1021/acsomega.9b02139. PMC 6921255. PMID 31867488.

Further reading

Review papers

Books

  • Bruus H (2008). Theoretical Microfluidics. Oxford University Press. ISBN 978-0199235094.
  • Herold KE, Rasooly A (2009). Lab-on-a-Chip Technology: Fabrication and Microfluidics. Caister Academic Press. ISBN 978-1-904455-46-2.
  • Kelly R, ed. (2012). Advances in Microfluidics. Richland, Washington, USA: Pacific Northwest National Laboratory. ISBN 978-953-510-106-2.
  • Tabeling P (2006). Introduction to Microfluidics. Oxford University Press. ISBN 978-0-19-856864-3.
  • Jenkins G, Mansfield CD (2012). Microfluidic Diagnostics. Humana Press. ISBN 978-1-62703-133-2.
  • Li X, Zhou Y, eds. (2013). Microfluidic devices for biomedical applications. Woodhead Publishing. ISBN 978-0-85709-697-5.

Education

This article is issued from Wikipedia. The text is licensed under Creative Commons - Attribution - Sharealike. Additional terms may apply for the media files.