We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Application of microfluidic technology to pancreatic islet research: first decade of endeavor

    Yong Wang

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    ,
    Joe F Lo

    Department of Bioengineering, University of Illinois at Chicago

    ,
    Joshua E Mendoza-Elias

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    Department of Bioengineering, University of Illinois at Chicago

    ,
    Adeola F Adewola

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    ,
    Tricia A Harvat

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    ,
    Katie P Kinzer

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    ,
    Dongyoung Lee

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    Department of Bioengineering, University of Illinois at Chicago

    ,
    Meirigeng Qi

    Department of Transplant/Surgery, University of Illinois at Chicago, 840 South Wood Street, Clinical Science Building, Suite 402, Chicago, Illinois 60612, USA

    ,
    David T Eddington

    Department of Bioengineering, University of Illinois at Chicago

    &
    Published Online:https://doi.org/10.4155/bio.10.131

    β-cells respond to blood glucose by secreting insulin to maintain glucose homeostasis. Perifusion enables manipulation of biological and chemical cues in elucidating the mechanisms of β-cell physiology. Recently, microfluidic devices made of polydimethylsiloxane and Borofloat glass have been developed as miniaturized perifusion setups and demonstrated distinct advantages over conventional techniques in resolving rapid secretory and metabolic waveforms intrinsic to β-cells. In order to enhance sensing and monitoring capabilities, these devices have been integrated with analytical tools to increase assay throughput. The spatio-temporal resolutions of these analyses have been improved through enhanced flow control, valves and compartmentalization. For the first time, this review provides an overview of current devices used in islet studies and analyzes their strengths and experimental suitability. To realize the potential of microfluidic islet applications, it is essential to bridge the gap in design and application between engineers and biologists through the creation of standardized bioassays and user-friendly interfaces.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    Bibliography

    • Lacy PE, Walker MM, Fink CJ. Perifusion of isolated rat islets in vitro. Participation of the microtubular system in the biphasic release of insulin. Diabetes21(10),987–998 (1972).
    • Hoshi M, Shreeve WW. Release and production of insulin by isolated, perifused rat pancreatic islets. Control by glucose. Diabetes22(1),16–24 (1973).
    • Lacy LR, Knudson MM, Williams JJ, Richards JS, Midgley AR Jr. Progesterone metabolism by the ovary of the pregnant rat: discrepancies in the catabolic regulation model. Endocrinology99(4),929–934 (1976).
    • Weaver DC, McDaniel ML, Naber SP, Barry CD, Lacy PE. Alloxan stimulation and inhibition of insulin release from isolated rat islets of Langerhans. Diabetes27(12),1205–1214 (1978).
    • Liu WW, Goodhouse J, Jeon NL, Enquist LW. A microfluidic chamber for analysis of neuron-to-cell spread and axonal transport of an α-herpesvirus. PLoS One3(6),e2382 (2008).
    • Mosadegh B, Huang C, Park JW et al. Generation of stable complex gradients across two-dimensional surfaces and three-dimensional gels. Langmuir23(22),10910–10912 (2007).
    • Taylor AM, Rhee SW, Jeon NL. Microfluidic chambers for cell migration and neuroscience research. Methods Mol. Biol.321,167–177 (2006).
    • Oppegard SC, Nam KH, Carr JR, Skaalure SC, Eddington DT. Modulating temporal and spatial oxygenation over adherent cellular cultures. PLoS ONE4(9),e6891 (2009).
    • Craighead H. Future lab-on-a-chip technologies for interrogating individual molecules. Nature442(7101),387–393 (2006).
    • 10  El-Ali J, Sorger PK, Jensen KF. Cells on chips. Nature442(7101),403–411 (2006).
    • 11  Mark D, Haeberle S, Roth G, von Stetten F, Zengerle R. Microfluidic lab-on-a-chip platforms: requirements, characteristics and applications. Chem. Soc. Rev.39(3),1153–1182 (2010).
    • 12  Beebe DJ, Mensing GA, Walker GM. Physics and applications of microfluidics in biology. Annu. Rev. Biomed. Eng.4,261–286 (2002).
    • 13  Figeys D, Pinto D. Lab-on-a-chip: a revolution in biological and medical sciences. Anal. Chem.72(9),330A–335A (2000).
    • 14  Weibel DB, Diluzio WR, Whitesides GM. Microfabrication meets microbiology. Nat. Rev. Microbiol.5(3),209–218 (2007).
    • 15  McDonald JC, Whitesides GM. Poly(dimethylsiloxane) as a material for fabricating microfluidic devices. Acc. Chem. Res.35(7),491–499 (2002).
    • 16  McDonald JC, Duffy DC, Anderson JR et al. Fabrication of microfluidic systems in poly(dimethylsiloxane). Electrophoresis21(1),27–40 (2000).
    • 17  Dishinger JF, Kennedy RT. Serial immunoassays in parallel on a microfluidic chip for monitoring hormone secretion from living cells. Anal. Chem.79(3),947–954 (2007).
    • 18  Toriello NM, Douglas ES, Thaitrong N et al. Integrated microfluidic bioprocessor for single-cell gene expression analysis. Proc. Natl Acad. Sci. USA105(51),20173–20178 (2008).
    • 19  Friis P, Hoppe K, Leistiko O, Mogensen KB, Hubner J, Kutter JP. Monolithic integration of microfluidic channels and optical waveguides in silica on silicon. Appl. Opt.40(34),6246–6251 (2001).
    • 20  McCreedy T. Fabrication techniques and materials commonly used for the production of microreactors and micro total analytical systems. Trends Analyt. Chem.19(6),396–401 (2000).
    • 21  Dishinger JF, Reid KR, Kennedy RT. Quantitative monitoring of insulin secretion from single islets of Langerhans in parallel on a microfluidic chip. Anal. Chem.81(8),3119–3127 (2009).▪▪ Reported the second generation microfluidic chip based on [17]. The device improved assay throughputs by simultaneous 15-channel analysis of online insulin secretion in parallel and showed a decent temporal insulin resolution from a single islet. This device offered a novel research platform to study the dynamics of islet insulin secretion.
    • 22  Simpson PT WA, Mathies RA. Microfabrication technology for the production of capillary array electrophoresis chips. Biomed Microdevices1(1),7–26 (1998).
    • 23  Fu J, Mao P, Han J. Continuous-flow bioseparation using microfabricated anisotropic nanofluidic sieving structures. Nat. Protoc.4(11),1681–1698 (2009).
    • 24  Simpson PC, Roach D, Woolley AT et al. High-throughput genetic analysis using microfabricated 96-sample capillary array electrophoresis microplates. Proc. Natl Acad. Sci. USA95(5),2256–2261 (1998).
    • 25  Pamme N. Continuous flow separations in microfluidic devices. Lab Chip7(12),1644–1659 (2007).
    • 26  Sato K, Hibara A, Tokeshi M, Hisamoto H, Kitamori T. Microchip-based chemical and biochemical analysis systems. Adv. Drug Deliv. Rev.55(3),379–391 (2003).
    • 27  Haeberle S, Zengerle R. Microfluidic platforms for lab-on-a-chip applications. Lab Chip7(9),1094–1110 (2007).
    • 28  Ottino JM, Wiggins S. Applied physics. Designing optimal micromixers. Science305(5683),485–486 (2004).
    • 29  Jeong GS, Chung S, Kim CB, Lee SH. Applications of micromixing technology. Analyst135(3),460–473 (2010).
    • 30  Thompson DM, King KR, Wieder KJ, Toner M, Yarmush ML, Jayaraman A. Dynamic gene expression profiling using a microfabricated living cell array. Anal. Chem.76(14),4098–4103 (2004).
    • 31  Situma C, Hashimoto M, Soper SA. Merging microfluidics with microarray-based bioassays. Biomol. Eng.23(5),213–231 (2006).
    • 32  Wang J. Electrochemical detection for microscale analytical systems: a review. Talanta56(2),223–231 (2002).
    • 33  Sikanen T, Franssila S, Kauppila TJ, Kostiainen R, Kotiaho T, Ketola RA. Microchip technology in mass spectrometry. Mass Spectrom. Rev.29(3),351–391 (2010).
    • 34  Lefebvre PJ, Paolisso G, Scheen AJ, Henquin JC. Pulsatility of insulin and glucagon release: physiological significance and pharmacological implications. Diabetologia30(7),443–452 (1987).
    • 35  Polonsky KS, Given BD, Van Cauter E. Twenty-four-hour profiles and pulsatile patterns of insulin secretion in normal and obese subjects. J. Clin. Invest.81(2),442–448 (1988).
    • 36  Ashcroft FM, Harrison DE, Ashcroft SJ. Glucose induces closure of single potassium channels in isolated rat pancreatic β-cells. Nature312(5993),446–448 (1984).
    • 37  Keizer J, Magnus G. ATP-sensitive potassium channel and bursting in the pancreatic β cell. A theoretical study. Biophys. J.56(2),229–242 (1989).
    • 38  Henquin JC. Triggering and amplifying pathways of regulation of insulin secretion by glucose. Diabetes49(11),1751–1760 (2000).
    • 39  Gilon P, Shepherd RM, Henquin JC. Oscillations of secretion driven by oscillations of cytoplasmic Ca2+ as evidences in single pancreatic islets. J. Biol. Chem.268(30),22265–22268 (1993).
    • 40  Beauvois MC, Merezak C, Jonas JC, Ravier MA, Henquin JC, Gilon P. Glucose-induced mixed [Ca2+]c oscillations in mouse β-cells are controlled by the membrane potential and the SERCA3 Ca2+-ATPase of the endoplasmic reticulum. Am. J. Physiol., Cell Physiol.290(6),C1503–1511 (2006).
    • 41  Shackman JG, Dahlgren GM, Peters JL, Kennedy RT. Perfusion and chemical monitoring of living cells on a microfluidic chip. Lab Chip5(1),56–63 (2005).▪▪ Describes an optimized microfluidics based on [42] that enabled perifusion of islets, improved temporal resolution of monitoring insulin release and easy control of the cellular environment.
    • 42  Roper MG, Shackman JG, Dahlgren GM, Kennedy RT. Microfluidic chip for continuous monitoring of hormone secretion from live cells using an electrophoresis-based immunoassay. Anal. Chem.75(18),4711–4717 (2003).▪ Reported the development and evaluation of one of earliest microfluidic devices used for islet study with the capability of online electrophoresis insulin assay. The device demonstrated the feasibility of using micro-scale system for monitoring β-cells, but with some limitations.
    • 43  Rocheleau JV, Walker GM, Head WS, McGuinness OP, Piston DW. Microfluidic glucose stimulation reveals limited coordination of intracellular Ca2+ activity oscillations in pancreatic islets. Proc. Natl Acad. Sci. USA101(35),12899–12903 (2004).▪▪ Developed a six-port microfluidic device for the purpose of studying the communication of calcium signaling across the islet and demonstrated that calcium communication depends on a delicate interaction between the degree of coupling and KATPactivation. This device provides a unique experimental setup and offers advantages over conventional approaches for the study of islet physiology.
    • 44  Adewola AF, Lee D, Harvat T et al. Microfluidic perifusion and imaging device for multiparametric islet function assessment. Biomed. Microdevices,12(3),409–417 (2010).▪ Reported a second generation microfluidics based on the device developed in [45]. With simple design geometry, it improved flow distribution within the device and enabled the creation of user’s defined chemical gradient with decent spatiol-temporal analytical resolutions.
    • 45  Mohammed JS, Wang Y, Harvat TA, Oberholzer J, Eddington DT. Microfluidic device for multimodal characterization of pancreatic islets. Lab Chip9(1),97–106 (2009).▪▪ Described a three-layer microfluidic with a unique islet immobilization approach. The device is particularly useful for real-time, intact human islet evaluation prior to islet transplantation and drug screen.
    • 46  Chen D, Du W, Liu Y et al. The chemistrode: a droplet-based microfluidic device for stimulation and recording with high temporal, spatial and chemical resolution. Proc. Natl Acad. Sci. USA105(44),16843–16848 (2008).▪▪ Developed a ‘chemistrode’, a plug-based microfluidic device that enables the stimulation, recording and analysis of molecular signals with high spatial and temporal resolution. This device provides a creative research platform for the understanding of β-cell physiology.
    • 47  Zhang X, Roper MG. Microfluidic perfusion system for automated delivery of temporal gradients to islets of Langerhans. Anal. Chem.81(3),1162–1168 (2009).▪▪ Described a three-layer microfluidics that is capable of delivering stimulant waveforms to islet cells using on-chip pumps and mixing channels. This perifusion system is suitable for studying islet physiology.
    • 48  Zhang X, Grimley A, Bertram R, Roper MG. Microfluidic system for generation of sinusoidal glucose waveforms for entrainment of islets of Langerhans. Anal. Chem.82(15),6704–6711 (2010).
    • 49  Kovachev N, Canals A, Escarpa A. Fast and selective microfluidic chips for electrochemical antioxidant sensing in complex samples. Anal. Chem.82(7),2925–2931 (2010).
    • 50  Slater GW, Tessier F, Kopecka K. The electroosmotic flow (EOF). Methods Mol. Biol.583,121–134 (2010).
    • 51  Lorenz RM, Edgar JS, Jeffries GD, Chiu DT. Microfluidic and optical systems for the on-demand generation and manipulation of single femtoliter-volume aqueous droplets. Anal. Chem.78(18),6433–6439 (2006).
    • 52  Fidalgo LM, Abell C, Huck WT. Surface-induced droplet fusion in microfluidic devices. Lab Chip7(8),984–986 (2007).
    • 53  Tice JD, Lyon AD, Ismagilov RF. Effects of viscosity on droplet formation and mixing in microfluidic channels. Anal. Chim. Acta507(1),73–77 (2004).
    • 54  Easley CJ, Rocheleau JV, Head WS, Piston DW. Quantitative measurement of zinc secretion from pancreatic islets with high temporal resolution using droplet-based microfluidics. Anal. Chem.81(21),9086–9095 (2009).▪▪ Reported a novel passive microfluidic droplet sampling method for quantifying islet zinc secretion with improved temporal resolution. In addition, the system is useful for studying other β-cell dynamic profiles.
    • 55  Pollack MG, Shenderov AD, Fair RB. Electrowetting-based actuation of droplets for integrated microfluidics. Lab Chip2(2),96–101 (2002).
    • 56  Paik P, Pamula VK, Fair RB. Rapid droplet mixers for digital microfluidic systems. Lab Chip3(4),253–259 (2003).
    • 57  Roman GT, Kennedy RT. Fully integrated microfluidic separations systems for biochemical analysis. J. Chromatogr. A1168(1–2),170–188 (2007).
    • 58  Kuswandi B, Nuriman, Huskens J, Verboom W. Optical sensing systems for microfluidic devices: a review. Anal. Chim. Acta601(2),141–155 (2007).
    • 59  Gilon P, Arredouani A, Gailly P, Gromada J, Henquin JC. Uptake and release of Ca2+ by the endoplasmic reticulum contribute to the oscillations of the cytosolic Ca2+ concentration triggered by Ca2+ influx in the electrically excitable pancreatic β-cell. J. Biol. Chem.274(29),20197–20205 (1999).
    • 60  Nunemaker CS, Dishinger JF, Dula SB et al. Glucose metabolism, islet architecture, and genetic homogeneity in imprinting of [Ca2+](i) and insulin rhythms in mouse islets. PLoS One4(12),e8428 (2009).
    • 61  Benninger RK, Zhang M, Head WS, Satin LS, Piston DW. Gap junction coupling and calcium waves in the pancreatic islet. Biophys. J.95(11),5048–5061 (2008).
    • 62  Rocheleau JV, Remedi MS, Granada B et al. Critical role of gap junction coupled KATP channel activity for regulated insulin secretion. PLoS Biol.4(2),e26 (2006).
    • 63  Richards-Williams C, Contreras JL, Berecek KH, Schwiebert EM. Extracellular ATP and zinc are co-secreted with insulin and activate multiple P2X purinergic receptor channels expressed by islet β-cells to potentiate insulin secretion. Purinergic Signal4(4),393–405 (2008).
    • 64  Pound LD, Sarkar SA, Benninger RK et al. Deletion of the mouse Slc30a8 gene encoding zinc transporter-8 results in impaired insulin secretion. Biochem. J.421(3),371–376 (2009)
    • 65  Qian WJ, Peters JL, Dahlgren GM, Gee KR, Kennedy RT. Simultaneous monitoring of Zn2+ secretion and intracellular Ca2+ from islets and islet cells by fluorescence microscopy. Biotechniques37(6),922–924, 926, 928–930 (2004).
    • 66  Qian WJ, Gee KR, Kennedy RT. Imaging of Zn2+ release from pancreatic β-cells at the level of single exocytotic events. Anal. Chem.75(14),3468–3475 (2003).
    • 67  Qian WJ, Aspinwall CA, Battiste MA, Kennedy RT. Detection of secretion from single pancreatic β-cells using extracellular fluorogenic reactions and confocal fluorescence microscopy. Anal. Chem.72(4),711–717 (2000).
    • 68  Sturis J, Pugh WL, Tang J, Ostrega DM, Polonsky JS, Polonsky KS. Alterations in pulsatile insulin secretion in the Zucker diabetic fatty rat. Am. J. Physiol.267(2 Pt 1),E250–E259 (1994).
    • 69  Ritzel RA, Veldhuis JD, Butler PC. Glucose stimulates pulsatile insulin secretion from human pancreatic islets by increasing secretory burst mass: dose-response relationships. J. Clin. Endocrinol. Metab.88(2),742–747 (2003).
    • 70  Morioka T, Asilmaz E, Hu J et al. Disruption of leptin receptor expression in the pancreas directly affects β cell growth and function in mice. J. Clin. Invest.117(10),2860–2868 (2007).
    • 71  Ueki K, Okada T, Hu J et al. Total insulin and IGF-I resistance in pancreatic β cells causes overt diabetes. Nat. Genet.38(5),583–588 (2006).
    • 72  Reid KR, Kennedy RT. Continuous operation of microfabricated electrophoresis devices for 24 hours and application to chemical monitoring of living cells. Anal. Chem.81(16),6837–6842 (2009).
    • 73  Gerin I, Dolinsky VW, Shackman JG et al. LXRb is required for adipocyte growth, glucose homeostasis, and β cell function. J. Biol. Chem.280(24),23024–23031 (2005).
    • 74  Ohta M, Nelson D, Nelson J, Meglasson MD, Erecinska M. Oxygen and temperature dependence of stimulated insulin secretion in isolated rat islets of Langerhans. J. Biol. Chem.265(29),17525–17532 (1990).
    • 75  Curry DL, Curry KP. Hypothermia and insulin secretion. Endocrinology87(4),750–755 (1970).
    • 76  Dionne KE, Colton CK, Yarmush ML. A microperifusion system with environmental control for studying insulin secretion by pancreatic tissue. Biotechnol. Prog.7(4),359–368 (1991).
    • 77  Charati SG SS. Diffusion of gases in silicone polymers: molecular dynamics simulations. Macromol.31,5529–5535 (1998).
    • 78  Merkel TC BV, Nagai K, Freeman BD, Pinnau I. Gas sorption, diffusion and permeation in poly(dimethylsiloxane). J. Polym. Sci. B Polym. Phy.38(415–434) (2000).
    • 79  Zhang Y, Park S, Yang S, Wang TH. An all-in-one microfluidic device for parallel DNA extraction and gene analysis. Biomed. Microdevices DOI: 10.1007/s10544-010-9458-6 (2010) (Epub ahead of print).
    • 80  Shen F, Du W, Kreutz JE, Fok A, Ismagilov RF. Digital PCR on a SlipChip. Lab Chip DOI: 10.1039/c004521g (Epub ahead of print) (2010).
    • 81  Ito T, Inoue A, Sato K, Hosokawa K, Maeda M. Autonomous polymer loading and sample injection for microchip electrophoresis. Anal. Chem.77(15),4759–4764 (2005).
    • 82  Vigolo D, Rusconi R, Piazza R, Stone HA. A portable device for temperature control along microchannels. Lab Chip10(6),795–798 (2010).
    • 83  Yamamoto T, Fujii T, Nojima T. PDMS-glass hybrid microreactor array with embedded temperature control device. Application to cell-free protein synthesis. Lab Chip2(4),197–202 (2002).
    • 84  Tallarek U, Rapp E, Scheenen T, Bayer E, Van As H. Electroosmotic and pressure-driven flow in open and packed capillaries: velocity distributions and fluid dispersion. Anal. Chem.72(10),2292–2301 (2000).
    • 85  Lacey ME, Webb AG, Sweedler JV. Monitoring temperature changes in capillary electrophoresis with nanoliter-volume NMR thermometry. Anal. Chem.72(20),4991–4998 (2000).
    • 86  Davis KL, Liu KL, Lanan M, Morris MD. Spatially resolved temperature measurements in electrophoresis capillaries by Raman thermometry. Anal. Chem.65(3),293–298 (1993).
    • 87  Ross D, Gaitan M, Locascio LE. Temperature measurement in microfluidic systems using a temperature-dependent fluorescent dye. Anal. Chem.73(17),4117–4123 (2001).
    • 88  Rocheleau JV, Piston DW. Chapter 4: combining microfluidics and quantitative fluorescence microscopy to examine pancreatic islet molecular physiology. Methods Cell Biol.89,71–92 (2008).