Skip to main content
Log in

Recent trends in microelectrode array technology for in vitro neural interface platform

  • Review Article
  • Published:
Biomedical Engineering Letters Aims and scope Submit manuscript

Abstract

Microelectrode array (MEA) technology is a widely used platform for the study of in vitro neural networks as it can either record or stimulate neurons by accessing multiple sites of neural circuits simultaneously. Unlike intracellular recording techniques, MEAs form noninvasive interface with cells so that they provides relatively long time window for studying neural circuits. As the technology matured, there have been various engineering solutions to meet the requirements in diverse application areas of MEAs: High-density MEAs, high-throughput platforms, flexible electrodes, monitoring subthreshold activity, co-culture platforms, and surface micropatterning. The MEA technology has been applied to neural network analysis, drug screening and neural prostheses studies. In this paper, the MEA technology is reviewed and the future prospect is discussed.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

Refrerences

  1. Thomas CA Jr, Springer PA, Loeb GE, Berwald-Netter Y, Okun LM. A miniature microelectrode array to monitor the bioelectric activity of cultured cells. Exp Cell Res. 1972; 74(1):61–6.

    Google Scholar 

  2. Pine J. Recording action potentials from cultured neurons with extracellular microcircuit electrodes. J Neurosci Methods. 1980; 2(1):19–31.

    Google Scholar 

  3. Gross GW. Simultaneous single unit recording in vitro with a photoetched laser deinsulated gold multimicroelectrode surface. IEEE Trans Biomed Eng. 1979; 26(5):273–9.

    Google Scholar 

  4. Novak JL, Wheeler BC. Recording from the Aplysia abdominal ganglion with a planar microelectrode array. IEEE Trans Biomed Eng. 1986; 33(2):196–202.

    Google Scholar 

  5. Boppart SA, Wheeler BC, Wallace CS. A flexible perforated microelectrode array for extended neural recordings. IEEE Trans Biomed Eng. 1992; 39(1):37–42.

    Google Scholar 

  6. Fromherz P. Electrical interfacing of nerve cells and semiconductor chips. ChemPhysChem. 2002; 3(3):276–84.

    Google Scholar 

  7. Stett A, Egert U, Guenther E, Hofmann F, Meyer T, Nisch W, Haemmerle H. Biological application of microelectrode arrays in drug discovery and basic research. Anal Bioanal Chem. 2003; 377(3):486–95.

    Google Scholar 

  8. Fromherz P, Offenhausser A, Vetter T, Weis J. A neuron-silicon junction: a Retzius cell of the leech on an insulated-gate fieldeffect transistor. Science. 1991; 252(5010):1290–3.

    Google Scholar 

  9. Guo J, Yuan J, Chan M. Modeling of the cell-electrode interface noise for microelectrode arrays. IEEE Trans Biomed Circuits Syst. 2012; 6(6):605–13.

    Google Scholar 

  10. Spira ME, Hai A. Multi-electrode array technologies for neuroscience and cardiology. Nat Nanotechnol. 2013; 8(2):83–94.

    Google Scholar 

  11. Buitenweg JR, Rutten WL, Marani E. Modeled channel distributions explain extracellular recordings from cultured neurons sealed to microelectrodes. IEEE Trans Biomed Eng. 2002; 49(12 Pt 2):1580–90.

    Google Scholar 

  12. Cohen A, Shappir J, Yitzchaik S, Spira ME. Reversible transition of extracellular field potential recordings to intracellular recordings of action potentials generated by neurons grown on transistors. Biosens Bioelectron. 2008; 23(6):811–9.

    Google Scholar 

  13. Hai A, Shappir J, Spira ME. In-cell recordings by extracellular microelectrodes. Nat Methods. 2010; 7(3):200–2.

    Google Scholar 

  14. Buitenweg JR, Rutten WL, Marani E. Extracellular stimulation window explained by a geometry-based model of the neuronelectrode contact. IEEE Trans Biomed Eng. 2002; 49(12 Pt 2):1591–9.

    Google Scholar 

  15. Chang JC, Brewer GJ, Wheeler BC. Microelectrode array recordings of patterned hippocampal neurons for four weeks. Biomed Microdevices. 2000; 2(4):245–53.

    Google Scholar 

  16. Nam Y, Chang JC, Wheeler BC, Brewer GJ. Gold-coated microelectrode array with thiol linked self-assembled monolayers for engineering neuronal cultures. IEEE Trans Biomed Eng. 2004; 51(1):158–65.

    Google Scholar 

  17. Xie C, Lin Z, Hanson L, Cui Y, Cui B. Intracellular recording of action potentials by nanopillar electroporation. Nat Nanotechnol. 2012; 7(3):185–90.

    Google Scholar 

  18. Kim R, Hong N, Nam Y. Gold nanograin microelectrodes for neuroelectronic interfaces. Biotechnol J. 2013; 8(2):206–14.

    Google Scholar 

  19. Kim JH, Kang G, Nam Y, Choi YK. Surface-modified microelectrode array with flake nanostructure for neural recording and stimulation. Nanotechnology. 2010; 21(8):853–3.

    Google Scholar 

  20. Seker E, Berdichevsky Y, Begley MR, Reed ML, Staley KJ, Yarmush ML. The fabrication of low-impedance nanoporous gold multiple-electrode arrays for neural electrophysiology studies. Nanotechnology. 2010; 21(12):1255–4.

    Google Scholar 

  21. Wang K, Fishman HA, Dai H, Harris JS. Neural stimulation with a carbon nanotube microelectrode array. Nano Lett. 2006; 6(9):2043–8.

    Google Scholar 

  22. Suzuki I, Fukuda M, Shirakawa K, Jiko H, Gotoh M. Carbon nanotube multi-electrode array chips for noninvasive real-time measurement of dopamine, action potentials, and postsynaptic potentials. Biosens Bioelectron. 2013; 49:270–5.

    Google Scholar 

  23. Mathieson K, Kachiguine S, Adams C, Cunningham W, Gunning D, O’Shea V, Smith KM, Chichilnisky EJ, Litke AM, Sher A, Rahman M. Large-area microelectrode arrays for recording of neural signals. IEEE Trans Nucl Sci. 2004; 51(5):2027–31.

    Google Scholar 

  24. Egert U, Schlosshauer B, Fennrich S, Nisch W, Fejtl M, Knott T, Muller T, Hammerle H. A novel organotypic long-term culture of the rat hippocampus on substrate-integrated multielectrode arrays. Brain Res Brain Res Protoc. 1998; 2(4):229–42.

    Google Scholar 

  25. Prohaska OJ, Olcaytug F, Pfundner P, Dragaun H. Thin-film multiple electrode probes: Possibilities and limitations. IEEE Trans Biomed Eng. 1986; 33(2):223–9.

    Google Scholar 

  26. Gabay T, Ben-David M, Kalifa I, Sorkin R, Abrams ZR, Ben-Jacob E, Hanein Y. Electro-chemical and biological properties of carbon nanotube based multi-electrode arrays. Nanotechnology. 2007; 18(3):0352–1.

    Google Scholar 

  27. Bruggemann D, Wolfrum B, Maybeck V, Mourzina Y, Jansen M, Offenhausser A. Nanostructured gold microelectrodes for extracellular recording from electrogenic cells. Nanotechnology. 2011; 22(26):2651–4.

    Google Scholar 

  28. Drake KL, Wise KD, Farraye J, Anderson DJ, BeMent SL. Performance of planar multisite microprobes in recording extracellular single-unit intracortical activity. IEEE Trans Biomed Eng. 1988; 35(9):719–32.

    Google Scholar 

  29. Jun SB, Hynd MR, Dowell-Mesfin N, Smith KL, Turner JN, Shain W, Kim SJ. Low-density neuronal networks cultured using patterned poly-l-lysine on microelectrode arrays. J Neurosci Methods. 2007; 160(2):317–26.

    Google Scholar 

  30. Gross GW, Wen WY, Lin JW. Transparent indium-tin oxide electrode patterns for extracellular, multisite recording in neuronal cultures. J Neurosci Methods. 1985; 15(3):243–52.

    Google Scholar 

  31. Heuschkel MO, Fejtl M, Raggenbass M, Bertrand D, Renaud P. A three-dimensional multi-electrode array for multi-site stimulation and recording in acute brain slices. J Neurosci Methods. 2002; 114(2):135–48.

    Google Scholar 

  32. Oka H, Shimono K, Ogawa R, Sugihara H, Taketani M. A new planar multielectrode array for extracellular recording: application to hippocampal acute slice. J Neurosci Methods. 1999; 93(1):61–7.

    Google Scholar 

  33. Liang G, Guvanasen GS, Xi L, Tuthill C, Nichols TR, DeWeerth SP. A PDMS-based integrated stretchable microelectrode array (isMEA) for neural and muscular surface interfacing. IEEE Trans Biomed Circuits Syst. 2013; 7(1):1–10.

    Google Scholar 

  34. Nam Y, Musick K, Wheeler BC. Application of a PDMS microstencil as a replaceable insulator toward a single-use planar microelectrode array. Biomed Microdevices. 2006; 8(4):375–81.

    Google Scholar 

  35. Blau A, Murr A, Wolff S, Sernagor E, Medini P, Iurilli G, Ziegler C, Benfenati F. Flexible, all-polymer microelectrode arrays for the capture of cardiac and neuronal signals. Biomaterials. 2011; 32(7):1778–86.

    Google Scholar 

  36. Yu F, Zhao Y, Gu J, Quigley KL, Chi NC, Tai YC, Hsiai TK. Flexible microelectrode arrays to interface epicardial electrical signals with intracardial calcium transients in zebrafish hearts. Biomed Microdevices. 2012; 14(2):357–66.

    Google Scholar 

  37. Potter SM, DeMarse TB. A new approach to neural cell culture for long-term studies. J Neurosci Methods. 2001; 110(1–2):17–24.

    Google Scholar 

  38. Goyal G, Nam Y. Neuronal micro-culture engineering by microchannel devices of cellular scale dimensions. Biomed Eng Lett. 2011; 1(2):89–98.

    Google Scholar 

  39. Gesteland RC, Howland B, Lettvin JY, Pitts WH. Comments on Microelectrodes. Proc IRE. 1959; 47(11):1856–62.

    Google Scholar 

  40. Maher MP, Pine J, Wright J, Tai YC. The neurochip: a new multielectrode device for stimulating and recording from cultured neurons. J Neurosci Methods. 1999; 87(1):45–56.

    Google Scholar 

  41. Park S, Song YJ, Boo H, Chung TD. Nanoporous Pt microelectrode for neural stimulation and recording: In vitro characterization. J Phys Chem C. 2010; 114(19):8721–6.

    Google Scholar 

  42. Cui XY, Martin DC. Fuzzy gold electrodes for lowering impedance and improving adhesion with electrodeposited conducting polymer films. Sensor Actuat A Phys. 2003; 103(3):384–94.

    Google Scholar 

  43. Czeschik A, Offenhäusser A, Wolfrum B. Fabrication of MEA-based nanocavity sensor arrays for extracellular recording of action potentials. Phy Status Solidi A. 2014; 211(6):1462–6.

    Google Scholar 

  44. Keefer EW, Botterman BR, Romero MI, Rossi AF, Gross GW. Carbon nanotube coating improves neuronal recordings. Nat Nanotechnol. 2008; 3(7):434–9.

    Google Scholar 

  45. Fuchsberger K, Le Goff A, Gambazzi L, Toma FM, Goldoni A, Giugliano M, Stelzle M, Prato M. Multiwalled carbon-nanotube-functionalized microelectrode arrays fabricated by microcontact printing: platform for studying chemical and electrical neuronal signaling. Small. 2011; 7(4):524–30.

    Google Scholar 

  46. Maybeck V, Edgington R, Bongrain A, Welch JO, Scorsone E, Bergonzo P, Jackman RB, Offenhausser A. Boron-doped nanocrystalline diamond microelectrode arrays monitor cardiac action potentials. Adv Healthc Mater. 2014; 3(2):283–9.

    Google Scholar 

  47. Cui X, Lee VA, Raphael Y, Wiler JA, Hetke JF, Anderson DJ, Martin DC. Surface modification of neural recording electrodes with conducting polymer/biomolecule blends. J Biomed Mater Res. 2001; 56(2):261–72.

    Google Scholar 

  48. Ludwig KA, Langhals NB, Joseph MD, Richardson-Burns SM, Hendricks JL, Kipke DR. Poly(3,4-ethylenedioxythiophene) (PEDOT) polymer coatings facilitate smaller neural recording electrodes. J Neural Eng. 2011; 8(1):0140–1.

    Google Scholar 

  49. Venkatraman S, Hendricks J, King ZA, Sereno AJ, Richardson-Burns S, Martin D, Carmena JM. In vitro and in vivo evaluation of PEDOT microelectrodes for neural stimulation and recording. IEEE Trans Neural Syst Rehabil Eng. 2011; 19(3):307–16.

    Google Scholar 

  50. Gerwig R, Fuchsberger K, Schroeppel B, Link GS, Heusel G, Kraushaar U, Schuhmann W, Stett A, Stelzle M. PEDOT-CNT composite microelectrodes for recording and electrostimulation applications: Fabrication, morphology, and electrical properties. Front Neuroeng. 2012; 5–8.

    Google Scholar 

  51. Zhou H, Cheng X, Rao L, Li T, Duan YY. Poly(3,4-ethylenedioxythiophene)/multiwall carbon nanotube composite coatings for improving the stability of microelectrodes in neural prostheses applications. Acta Biomater. 2013; 9(5):6439–49.

    Google Scholar 

  52. Deng M, Yang X, Silke M, Qiu W, Xu M, Borghs G, Chen H. Electrochemical deposition of polypyrrole/graphene oxide composite on microelectrodes towards tuning the electrochemical properties of neural probes. Sensor Actuat B Chem. 2011; 158(1):176–84.

    Google Scholar 

  53. Kim W, Ng JK, Kunitake ME, Conklin BR, Yang P. Interfacing silicon nanowires with mammalian cells. J Am Chem Soc. 2007; 129(23):7228–9.

    Google Scholar 

  54. Robinson JT, Jorgolli M, Shalek AK, Yoon MH, Gertner RS, Park H. Vertical nanowire electrode arrays as a scalable platform for intracellular interfacing to neuronal circuits. Nat Nanotechnol. 2012; 7(3):180–4.

    Google Scholar 

  55. Lin ZC, Xie C, Osakada Y, Cui Y, Cui B. Iridium oxide nanotube electrodes for sensitive and prolonged intracellular measurement of action potentials. Nat Commun. 2014; 5–3206.

    Google Scholar 

  56. Nam Y, Branch DW, Wheeler BC. Epoxy-silane linking of biomolecules is simple and effective for patterning neuronal cultures. Biosens Bioelectron. 2006; 22(5):589–97.

    Google Scholar 

  57. Kang K, Lee S, Kim R, Choi IS, Nam Y. Electrochemically driven, electrode-addressable formation of functionalized polydopamine films for neural interfaces. Angew Chem Int Ed Engl. 2012; 51(52):13101–4.

    Google Scholar 

  58. Eversmann B, Jenkner M, Hofmann F, Paulus C, Brederlow R, Holzapfl B, Fromherz P, Merz M, Brenner M, Schreiter M, Gabl R, Plehnert K, Steinhauser M, Eckstein G, Schmitt-Landsiedel D, Thewes R. A 128 × 128 CMOS biosensor array for extracellular recording of neural activity. IEEE J Solid-State Circuits. 2003; 38(12):2306–17.

    Google Scholar 

  59. Lambacher A, Vitzthum V, Zeitler R, Eickenscheidt M, Eversmann B, Thewes R, Fromherz P. Identifying firing mammalian neurons in networks with high-resolution multitransistor array (MTA). Appl Phys A-Mater. 2011; 102(1):1–11.

    Google Scholar 

  60. Zeck G, Lambacher A, Fromherz P. Axonal transmission in the retina introduces a small dispersion of relative timing in the ganglion cell population response. PLoS One. 2011; 6(6):e20810.

    Google Scholar 

  61. Eickenscheidt M, Jenkner M, Thewes R, Fromherz P, Zeck G. Electrical stimulation of retinal neurons in epiretinal and subretinal configuration using a multicapacitor array. J Neurophysiol. 2012; 107(10):2742–55.

    Google Scholar 

  62. Berdondini L, Imfeld K, Maccione A, Tedesco M, Neukom S, Koudelka-Hep M, Martinoia S. Active pixel sensor array for high spatio-temporal resolution electrophysiological recordings from single cell to large scale neuronal networks. Lab Chip. 2009; 9(18):2644–51.

    Google Scholar 

  63. Imfeld K, Neukom S, Maccione A, Bornat Y, Martinoia S, Farine PA, Koudelka-Hep M, Berdondini L. Large-scale, highresolution data acquisition system for extracellular recording of electrophysiological activity. IEEE Trans Biomed Eng. 2008; 55(8):2064–73.

    Google Scholar 

  64. Berdondini L, van der Wal PD, de Rooij NF, Koudelka-Hep M. Development of an electroless post-processing technique for depositing gold as electrode material on CMOS devices. Sensor Actuat B Chem. 2004; 99(2–3):505–10.

    Google Scholar 

  65. Ferrea E, Maccione A, Medrihan L, Nieus T, Ghezzi D, Baldelli P, Benfenati F, Berdondini L. Large-scale, highresolution electrophysiological imaging of field potentials in brain slices with microelectronic multielectrode arrays. Front Neural Circuits. 2012; 6–80.

    Google Scholar 

  66. Maccione A, Hennig MH, Gandolfo M, Muthmann O, van Coppenhagen J, Eglen SJ, Berdondini L, Sernagor E. Following the ontogeny of retinal waves: pan-retinal recordings of population dynamics in the neonatal mouse. J Physiol. 2014; 592(Pt 7):1545–63.

    Google Scholar 

  67. Frey U, Sedivy J, Heer F, Pedron R, Ballini M, Mueller J, Bakkum D, Hafizovic S, Faraci FD, Greve F, Kirstein K-U, Hierlemann A. Switch-matrix-based high-density microelectrode array in CMOS technology. IEEE J Solid-St Circ. 2010; 45(2):467–82.

    Google Scholar 

  68. Frey U, Egert U, Heer F, Hafizovic S, Hierlemann A. Microelectronic system for high-resolution mapping of extracellular electric fields applied to brain slices. Biosens Bioelectron. 2009; 24(7):2191–8.

    Google Scholar 

  69. Muller J, Bakkum DJ, Hierlemann A. Sub-millisecond closedloop feedback stimulation between arbitrary sets of individual neurons. Front Neural Circuits. 2013; 6–121.

    Google Scholar 

  70. Bakkum DJ, Frey U, Radivojevic M, Russell TL, Muller J, Fiscella M, Takahashi H, Hierlemann A. Tracking axonal action potential propagation on a high-density microelectrode array across hundreds of sites. Nat Commun. 2013; 4–2181.

    Google Scholar 

  71. Ballini M, Muller J, Livi P, Chen Y, Frey U, Shadmani A, Jones IL, Gong W, Fiscella M, Radivojevic M, Bakkum D, Stett A, Heer F, Hierlemann A. A 1024-channel CMOS microelectrode-array system with 26′400 electrodes for recording and stimulation of electro-active cells in-vitro. VLSI Circuits (VLSIC), 2013 Symposium on; 2013: IEEE;C54–C5.

    Google Scholar 

  72. Park JW, Kim HJ, Kang MW, Jeon NL. Advances in microfluidics-based experimental methods for neuroscience research. Lab Chip. 2013; 13(4):509–21.

    Google Scholar 

  73. Taylor AM, Blurton-Jones M, Rhee SW, Cribbs DH, Cotman CW, Jeon NL. A microfluidic culture platform for CNS axonal injury, regeneration and transport. Nat Methods. 2005; 2(8):599–605.

    Google Scholar 

  74. Campenot RB. Local control of neurite development by nerve growth factor. Proc Natl Acad Sci USA. 1977; 74(10):4516–9.

    Google Scholar 

  75. Park J, Koito H, Li J, Han A. Multi-compartment neuron-glia co-culture platform for localized CNS axon-glia interaction study. Lab Chip. 2012; 12(18):3296–304.

    Google Scholar 

  76. Peyrin JM, Deleglise B, Saias L, Vignes M, Gougis P, Magnifico S, Betuing S, Pietri M, Caboche J, Vanhoutte P, Viovy JL, Brugg B. Axon diodes for the reconstruction of oriented neuronal networks in microfluidic chambers. Lab Chip. 2011; 11(21):3663–73.

    Google Scholar 

  77. Dworak BJ, Wheeler BC. Novel MEA platform with PDMS microtunnels enables the detection of action potential propagation from isolated axons in culture. Lab Chip. 2009; 9(3):404–10.

    Google Scholar 

  78. Morales R, Riss M, Wang L, Gavin R, Del Rio JA, Alcubilla R, Claverol-Tinture E. Integrating multi-unit electrophysiology and plastic culture dishes for network neuroscience. Lab Chip. 2008; 8(11):1896–905.

    Google Scholar 

  79. Pan L, Alagapan S, Franca E, Brewer GJ, Wheeler BC. Propagation of action potential activity in a predefined microtunnel neural network. J Neural Eng. 2011; 8(4):0460–1.

    Google Scholar 

  80. Brewer GJ, Boehler MD, Leondopulos S, Pan L, Alagapan S, DeMarse TB, Wheeler BC. Toward a self-wired active reconstruction of the hippocampal trisynaptic loop: DG-CA3. Front Neural Circuits. 2013; 7–165.

    Google Scholar 

  81. Honegger T, Scott MA, Yanik MF, Voldman J. Electrokinetic confinement of axonal growth for dynamically configurable neural networks. Lab Chip. 2013; 13(4):589–98.

    Google Scholar 

  82. Kanagasabapathi TT, Franco M, Barone RA, Martinoia S, Wadman WJ, Decre MM. Selective pharmacological manipulation of cortical-thalamic co-cultures in a dual-compartment device. J Neurosci Methods. 2013; 214(1):1–8.

    Google Scholar 

  83. Kanagasabapathi TT, Ciliberti D, Martinoia S, Wadman WJ, Decre MM. Dual-compartment neurofluidic system for electrophysiological measurements in physically segregated and functionally connected neuronal cell culture. Front Neuroeng. 2011; 4–13.

    Google Scholar 

  84. Takayama Y, Moriguchi H, Kotani K, Suzuki T, Mabuchi K, Jimbo Y. Network-wide integration of stem cell-derived neurons and mouse cortical neurons using microfabricated coculture devices. Biosystems. 2012; 107(1):1–8.

    Google Scholar 

  85. Wheeler BC, Brewer GJ. Designing neural networks in culture: Experiments are described for controlled growth, of nerve cells taken from rats, in predesigned geometrical patterns on laboratory culture dishes. Proc IEEE Inst Electr Electron Eng. 2010; 98(3):398–406.

    Google Scholar 

  86. Chang JC, Brewer GJ, Wheeler BC. Modulation of neural network activity by patterning. Biosens Bioelectron. 2001; 16(7–8):527–33.

    Google Scholar 

  87. Segev R, Benveniste M, Hulata E, Cohen N, Palevski A, Kapon E, Shapira Y, Ben-Jacob E. Long term behavior of lithographically prepared in vitro neuronal networks. Phys Rev Lett. 2002; 88(11):1181–2.

    Google Scholar 

  88. Cheng J, Zhu G, Wu L, Du X, Zhang H, Wolfrum B, Jin Q, Zhao J, Offenhausser A, Xu Y. Photopatterning of selfassembled poly (ethylene) glycol monolayer for neuronal network fabrication. J Neurosci Methods. 2013; 213(2):196–203.

    Google Scholar 

  89. Boehler MD, Leondopulos SS, Wheeler BC, Brewer GJ. Hippocampal networks on reliable patterned substrates. J Neurosci Methods. 2012; 203(2):344–53.

    Google Scholar 

  90. James CD, Spence AJ, Dowell-Mesfin NM, Hussain RJ, Smith KL, Craighead HG, Isaacson MS, Shain W, Turner JN. Extracellular recordings from patterned neuronal networks using planar microelectrode arrays. IEEE Trans Biomed Eng. 2004; 51(9):1640–8.

    Google Scholar 

  91. Jungblut M, Knoll W, Thielemann C, Pottek M. Triangular neuronal networks on microelectrode arrays: an approach to improve the properties of low-density networks for extracellular recording. Biomed Microdevices. 2009; 11(6):1269–78.

    Google Scholar 

  92. Marconi E, Nieus T, Maccione A, Valente P, Simi A, Messa M, Dante S, Baldelli P, Berdondini L, Benfenati F. Emergent functional properties of neuronal networks with controlled topology. PLoS One. 2012; 7(4):e34648.

    Google Scholar 

  93. Kang K, Choi IS, Nam Y. A biofunctionalization scheme for neural interfaces using polydopamine polymer. Biomaterials. 2011; 32(27):6374–80.

    Google Scholar 

  94. Suzuki M, Ikeda K, Yamaguchi M, Kudoh SN, Yokoyama K, Satoh R, Ito D, Nagayama M, Uchida T, Gohara K. Neuronal cell patterning on a multi-electrode array for a network analysis platform. Biomaterials. 2013; 34(21):5210–7.

    Google Scholar 

  95. Jimbo Y, Robinson HP, Kawana A. Simultaneous measurement of intracellular calcium and electrical activity from patterned neural networks in culture. IEEE Trans Biomed Eng. 1993; 40(8):804–10.

    Google Scholar 

  96. Suzuki I, Sugio Y, Jimbo Y, Yasuda K. Stepwise pattern modification of neuronal network in photo-thermally-etched agarose architecture on multi-electrode array chip for individual-cell-based electrophysiological measurement. Lab Chip. 2005; 5(3):241–7.

    Google Scholar 

  97. Kang G, Lee JH, Lee CS, Nam Y. Agarose microwell based neuronal micro-circuit arrays on microelectrode arrays for high throughput drug testing. Lab Chip. 2009; 9(22):3236–42.

    Google Scholar 

  98. Herzog N, Shein-Idelson M, Hanein Y. Optical validation of in vitro extra-cellular neuronal recordings. J Neural Eng. 2011; 8(5):0560–8.

    Google Scholar 

  99. Tchumatchenko T, Newman JP, Fong MF, Potter SM. Delivery of continuously-varying stimuli using channelrhodopsin-2. Front Neural Circuits. 2013; 7–184.

    Google Scholar 

  100. Yakushenko A, Gong Z, Maybeck V, Hofmann B, Gu E, Dawson M, Offenhausser A, Wolfrum B. On-chip optical stimulation and electrical recording from cells. J Biomed Opt. 2013; 18(11):1114–2.

    Google Scholar 

  101. Morefield SI, Keefer EW, Chapman KD, Gross GW. Drug evaluations using neuronal networks cultured on microelectrode arrays. Biosens Bioelectron. 2000; 15(7–8):383–96.

    Google Scholar 

  102. Selinger JV, Pancrazio JJ, Gross GW. Measuring synchronization in neuronal networks for biosensor applications. Biosens Bioelectron. 2004; 19(7):675–83.

    Google Scholar 

  103. Novellino A, Scelfo B, Palosaari T, Price A, Sobanski T, Shafer TJ, Johnstone AF, Gross GW, Gramowski A, Schroeder O, Jugelt K, Chiappalone M, Benfenati F, Martinoia S, Tedesco MT, Defranchi E, D’Angelo P, Whelan M. Development of micro-electrode array based tests for neurotoxicity: assessment of interlaboratory reproducibility with neuroactive chemicals. Front Neuroeng. 2011; 4–4.

    Google Scholar 

  104. Goo YS, Ye JH, Lee S, Nam Y, Ryu SB, Kim KH. Retinal ganglion cell responses to voltage and current stimulation in wild-type and rd1 mouse retinas. J Neural Eng. 2011; 8(3):0350–3.

    Google Scholar 

  105. Sekirnjak C, Hottowy P, Sher A, Dabrowski W, Litke AM, Chichilnisky EJ. Electrical stimulation of mammalian retinal ganglion cells with multielectrode arrays. J Neurophysiol. 2006; 95(6):3311–27.

    Google Scholar 

  106. Sekirnjak C, Hottowy P, Sher A, Dabrowski W, Litke AM, Chichilnisky EJ. High-resolution electrical stimulation of primate retina for epiretinal implant design. J Neurosci. 2008; 28(17):4446–56.

    Google Scholar 

  107. Gautam V, Rand D, Hanein Y, Narayan KS. A polymer optoelectronic interface provides visual cues to a blind retina. Adv Mater. 2014; 26(11):1751–6.

    Google Scholar 

  108. Lambacher A, Jenkner M, Merz M, Eversmann B, Kaul RA, Hofmann F, Thewes R, Fromherz P. Electrical imaging of neuronal activity by multi-transistor-array (MTA) recording at 7.8 μm resolution. Appl Phys A. 2004; 79(7):1607–11.

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yoonkey Nam.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, R., Joo, S., Jung, H. et al. Recent trends in microelectrode array technology for in vitro neural interface platform. Biomed. Eng. Lett. 4, 129–141 (2014). https://doi.org/10.1007/s13534-014-0130-6

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13534-014-0130-6

Keywords

Navigation