Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Glial responses to implanted electrodes in the brain

A Publisher Correction to this article was published on 07 December 2017

This article has been updated

Abstract

The use of implants that can electrically stimulate or record electrophysiological or neurochemical activity in nervous tissue is rapidly expanding. Despite remarkable results in clinical studies and increasing market approvals, the mechanisms underlying the therapeutic effects of neuroprosthetic and neuromodulation devices, as well as their side effects and reasons for their failure, remain poorly understood. A major assumption has been that the signal-generating neurons are the only important target cells of neural-interface technologies. However, recent evidence indicates that the supporting glial cells remodel the structure and function of neuronal networks and are an effector of stimulation-based therapy. Here, we reframe the traditional view of glia as a passive barrier, and discuss their role as an active determinant of the outcomes of device implantation. We also discuss the implications that this has on the development of bioelectronic medical devices.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Traditional electrode arrays incite gliosis.
Fig. 2: In vivo multiphoton imaging of the glial response to the implantation of a multielectrode array.
Fig. 3: Evidence for a negative impact of increased gliosis on recording quality.
Fig. 4: Potential mechanisms of the active modulation of neurotransmission by glia.
Fig. 5: Next-generation arrays mitigate gliosis.
Fig. 6: Opportunities for further enquiry in device design.
Fig. 7: Opportunities for further biological enquiry.

Similar content being viewed by others

Change history

  • 07 December 2017

    In the version of this Review Article originally published, in Fig. 4b, the label ‘Glutamate’ was mistakenly duplicated and an arrow between a purinergic P2 receptor and a glutamate transporter was missing. The figure has now been updated in all versions of the Review Article.

References

  1. Kasthuri, N. & Lichtman, J. W. Neurocartography. Neuropsychopharmacology 35, 342–343 (2010).

  2. Oberlaender, M. et al. Three-dimensional axon morphologies of individual layer 5 neurons indicate cell type-specific intracortical pathways for whisker motion and touch. Proc. Natl Acad. Sci USA 108, 4188–4193 (2011).

    Article  PubMed Central  PubMed  Google Scholar 

  3. Kubota, Y. Untangling GABAergic wiring in the cortical microcircuit. Curr. Opin. Neurobiol. 26, 7–14 (2014).

    Article  CAS  PubMed  Google Scholar 

  4. Zhang, Y. & Barres, B. A. Astrocyte heterogeneity: an underappreciated topic in neurobiology. Curr. Opin. Neurobiol. 20, 588–594 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Miocinovic, S., Somayajula, S., Chitnis, S. & Vitek, J. L. History, applications, and mechanisms of deep brain stimulation. JAMA Neurol. 70, 163–171 (2013).

    Article  PubMed  Google Scholar 

  6. Herrington, T. M. et al. Mechanisms of deep brain stimulation. J. Neurophysiol. 115, 19–38 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Borton, D., Micera, S., Millán, J. del R. & Courtine, G. Personalized neuroprosthetics. Sci. Transl. Med. 5, 210rv2 (2013).

  8. Jennings, J. H. & Stuber, G. D. Tools for resolving functional activity and connectivity within intact neural circuits. Curr. Biol. 24, R41–R50 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Henze, D. A. et al. Intracellular features predicted by extracellular recordings in the hippocampus in vivo. J. Neurophysiol. 84, 390–400 (2000).

    CAS  PubMed  Google Scholar 

  10. Prasad, A. et al. Comprehensive characterization and failure modes of tungsten microwire arrays in chronic neural implants. J. Neural Eng. 9, 56015 (2012).

    Article  Google Scholar 

  11. Barrese, J. C. et al. Failure mode analysis of silicon-based intracortical microelectrode arrays in non-human primates. J. Neural Eng. 10, 66014 (2013).

    Article  Google Scholar 

  12. Ludwig, K. A. et al. Poly(3,4-ethylenedioxythiophene) (PEDOT) polymer coatings facilitate smaller neural recording electrodes. J. Neural Eng. 8, 14001 (2011).

    Article  Google Scholar 

  13. Perge, J. A. et al. Intra-day signal instabilities affect decoding performance in an intracortical neural interface system. J. Neural Eng. 10, 36004 (2013).

    Article  Google Scholar 

  14. Jackson, A. & Fetz, E. E. Compact movable microwire array for long-term chronic unit recording in cerebral cortex of primates. J. Neurophysiol. 98, 3109–3118 (2007).

    Article  PubMed  Google Scholar 

  15. Liu, X. et al. Stability of the interface between neural tissue and chronically implanted intracortical microelectrodes. IEEE Trans. Rehabil. Eng. 7, 315–326 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. McCreery, D. B., Yuen, T. G. H., Agnew, W. F. & Bullara, L. A. A characterization of the effects on neuronal excitability due to prolonged microstimulation with chronically implanted microelectrodes. IEEE Trans. Biomed. Eng. 44, 931–939 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. McCreery, D. B., Agnew, W. F. & Bullara, L. A. The effects of prolonged intracortical microstimulation on the excitability of pyramidal tract neurons in the cat. Ann. Biomed. Eng. 30, 107–119 (2002).

    Article  PubMed  Google Scholar 

  18. Prasad, A. et al. Abiotic-biotic characterization of Pt/Ir microelectrode arrays in chronic implants. Front. Neuroeng. 7, 2 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Cahoy, J. D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Pannasch, U. & Rouach, N. Emerging role for astroglial networks in information processing: from synapse to behavior. Trends Neurosci. 36, 405–417 (2013).

    Article  CAS  PubMed  Google Scholar 

  21. Virchow, R. Gesammelte Abhandlungen zur Wissenschaftlichen Medicin (Meidinger, Frankfurt, 1856).

    Google Scholar 

  22. Khakh, B. S. & Sofroniew, M. V. Diversity of astrocyte functions and phenotypes in neural circuits. Nat. Neurosci. 18, 942–952 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  23. Burda, J. E., Bernstein, A. M. & Sofroniew, M. V. Astrocyte roles in traumatic brain injury. Exp. Neurol. 275, 305–315 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Liddelow, S. A. et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 541, 481–487 (2017).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  25. Sofroniew, M. V. Astrogliosis. Cold Spring Harb. Perspect. Biol. 7, a020420 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Kozai, T. D. Y., Jaquins-Gerstl, A. S., Vazquez, A. L., Michael, A. C. & Cui, X. T. Brain tissue responses to neural implants impact signal sensitivity and intervention strategies. ACS Chem. Neurosci. 6, 48–67 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Buffo, A., Rolando, C. & Ceruti, S. Astrocytes in the damaged brain: molecular and cellular insights into their reactive response and healing potential. Biochem. Pharmacol. 79, 77–89 (2010).

    Article  CAS  PubMed  Google Scholar 

  28. Kozai, T. D. Y. et al. Reduction of neurovascular damage resulting from microelectrode insertion into the cerebral cortex using in vivo two-photon mapping. J. Neural Eng. 7, 46011 (2010).

    Article  CAS  Google Scholar 

  29. Perry, V. H. & Teeling, J. Microglia and macrophages of the central nervous system: the contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin. Immunopathol. 35, 601–612 (2013).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  30. Sofroniew, M. V. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 32, 638–647 (2009).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  31. Chew, S. S. L., Johnson, C. S., Green, C. R. & Danesh-Meyer, H. V. Role of connexin43 in central nervous system injury. Exp. Neurol. 225, 250–261 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Pannasch, U. et al. Astroglial networks scale synaptic activity and plasticity. Proc. Natl Acad. Sci. USA 108, 8467–8472 (2011).

    Article  PubMed Central  PubMed  Google Scholar 

  33. Liu, J. Y. W. et al. Neuropathology of the blood–brain barrier and pharmaco-resistance in human epilepsy. Brain 135, 3115–3133 (2012).

    Article  PubMed  Google Scholar 

  34. Kozai, T. D. Y., Eles, J. R., Vazquez, A. L. & Cui, X. T. Two-photon imaging of chronically implanted neural electrodes: sealing methods and new insights. J. Neurosci. Methods 258, 46–55 (2016).

    Article  PubMed  Google Scholar 

  35. Jorfi, M., Skousen, J. L., Weder, C. & Capadona, J. R. Progress towards biocompatible intracortical microelectrodes for neural interfacing applications. J. Neural Eng. 12, 11001 (2015).

    Article  Google Scholar 

  36. Saxena, T. et al. The impact of chronic blood–brain barrier breach on intracortical electrode function. Biomaterials 34, 4703–4713 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Nolta, N. F., Christensen, M. B., Crane, P. D., Skousen, J. L. & Tresco, P. A. BBB leakage, astrogliosis, and tissue loss correlate with silicon microelectrode array recording performance. Biomaterials 53, 753–762 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Bekar, L. et al. Adenosine is crucial for deep brain stimulation–mediated attenuation of tremor. Nat. Med. 14, 75–80 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Lempka, S. F., Miocinovic, S., Johnson, M. D., Vitek, J. L. & McIntyre, C. C. In vivo impedance spectroscopy of deep brain stimulation electrodes. J. Neural Eng. 6, 46001 (2009).

    Article  Google Scholar 

  40. Purcell, E. K., Thompson, D. E., Ludwig, K. A. & Kipke, D. R. Flavopiridol reduces the impedance of neural prostheses in vivo without affecting recording quality. J. Neurosci. Methods 183, 149–157 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Johnson, M. D., Otto, K. J. & Kipke, D. R. Repeated voltage biasing improves unit recordings by reducing resistive tissue impedances. IEEE Trans. Neural Syst. Rehabil. Eng. 13, 160–165 (2005).

    Article  PubMed  Google Scholar 

  42. Ludwig, K. A. et al. Chronic neural recordings using silicon microelectrode arrays electrochemically deposited with a poly(3,4-ethylenedioxythiophene) (PEDOT) film. J. Neural Eng. 3, 59–70 (2006).

    Article  PubMed  Google Scholar 

  43. Prasad, A. et al. Quantifying long-term microelectrode array functionality using chronic in vivo impedance testing. J. Neural Eng. 9, 26028 (2012).

    Article  Google Scholar 

  44. McConnell, G. C. et al. Implanted neural electrodes cause chronic, local inflammation that is correlated with local neurodegeneration. J. Neural Eng. 6, 56003 (2009).

    Article  Google Scholar 

  45. Tawfik, V. L. et al. Deep brain stimulation results in local glutamate and adenosine release: investigation into the role of astrocytes. Neurosurgery 67, 367–375 (2010).

    Article  PubMed Central  PubMed  Google Scholar 

  46. McAdams, E. T., Lackermeier, A., McLaughlin, J. A., Macken, D. & Jossinet, J. The linear and non-linear electrical properties of the electrode–electrolyte interface. Biosens. Bioelectron. 10, 67–74 (1995).

    Article  CAS  Google Scholar 

  47. Mercanzini, A., Colin, P., Bensadoun, J.-C., Bertsch, A. & Renaud, P. In vivo electrical impedance spectroscopy of tissue reaction to microelectrode arrays. IEEE Trans. Biomed. Eng. 56, 1909–1918 (2009).

    Article  PubMed  Google Scholar 

  48. Kozai, T. D. Y. et al. Ultrasmall implantable composite microelectrodes with bioactive surfaces for chronic neural interfaces. Nat. Mater. 11, 1065–1073 (2012).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Robblee, L. S., McHardy, J., Agnew, W. F. & Bullara, L. A. Electrical stimulation with Pt electrodes. VII. Dissolution of Pt electrodes during electrical stimulation of the cat cerebral cortex. J. Neurosci. Methods 9, 301–308 (1983).

    Article  CAS  PubMed  Google Scholar 

  50. Wei, X. F. et al. Impedance characteristics of deep brain stimulation electrodes in vitro and in vivo. J. Neural Eng. 6, 46008 (2009).

    Article  Google Scholar 

  51. Merrill, D. R., Bikson, M. & Jefferys, J. G. R. Electrical stimulation of excitable tissue: design of efficacious and safe protocols. J. Neurosci. Methods 141, 171–198 (2005).

    Article  PubMed  Google Scholar 

  52. Otto, K. J., Johnson, M. D. & Kipke, D. R. Voltage pulses change neural interface properties and improve unit recordings with chronically implanted microelectrodes. IEEE Trans. Biomed. Eng. 53, 333–340 (2006).

    Article  PubMed  Google Scholar 

  53. Keese, C. R., Wegener, J., Walker, S. R. & Giaever, I. Electrical wound-healing assay for cells in vitro. Proc. Natl Acad. Sci. USA 101, 1554–1559 (2004).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Kilgore, K. L. & Bhadra, N. Nerve conduction block utilising high-frequency alternating current. Med. Biol. Eng. Comput. 42, 394–406 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. McIntyre, C. C. et al. Extracellular stimulation of central neurons: influence of stimulus waveform and frequency on neuronal output. J. Neurophysiol. 88, 1592–604 (2002).

    PubMed  Google Scholar 

  56. Nowak, L. G. & Bullier, J. Axons, but not cell bodies, are activated by electrical stimulation in cortical gray matter. Exp. Brain Res. 118, 477–488 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Williams, J. C., Rennaker, R. L. & Kipke, D. R. Long-term neural recording characteristics of wire microelectrode arrays implanted in cerebral cortex. Brain Res. Protoc. 4, 303–313 (1999).

    Article  CAS  Google Scholar 

  58. Preda, F. et al. Switching from constant voltage to constant current in deep brain stimulation: a multicenter experience of mixed implants for movement disorders. Eur. J. Neurol. 23, 190–195 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Timmermann, L. et al. Multiple-source current steering in subthalamic nucleus deep brain stimulation for Parkinson’s disease (the VANTAGE study): a non-randomised, prospective, multicentre, open-label study. Lancet Neurol. 14, 693–701 (2015).

    Article  PubMed  Google Scholar 

  60. McCreery, D. et al. Correlations between histology and neuronal activity recorded by microelectrodes implanted chronically in the cerebral cortex. J. Neural Eng. 13, 36012 (2016).

    Article  Google Scholar 

  61. Williams, J. C., Hippensteel, J. A., Dilgen, J., Shain, W. & Kipke, D. R. Complex impedance spectroscopy for monitoring tissue responses to inserted neural implants. J. Neural Eng. 4, 410–423 (2007).

    Article  PubMed  Google Scholar 

  62. Suner, S., Fellows, M. R., Vargas-Irwin, C., Nakata, G. K. & Donoghue, J. P. Reliability of signals from a chronically implanted, silicon-based electrode array in non-human primate primary motor cortex. IEEE Trans. Neural Syst. Rehabil. Eng. 13, 524–541 (2005).

    Article  PubMed  Google Scholar 

  63. Jiang, J., Willett, F. R. & Taylor, D. M. Relationship between microelectrode array impedance and chronic recording quality of single units and local field potentials. In 2014 36th Annual International Conference of the IEEE Engineering in Medicine and Biology Society 3045–3048 (IEEE, 2014).

  64. Kozai, T. D. Y. et al. Mechanical failure modes of chronically implanted planar silicon-based neural probes for laminar recording. Biomaterials 37, 25–39 (2015).

    Article  CAS  PubMed  Google Scholar 

  65. Malaga, K. A. et al. Data-driven model comparing the effects of glial scarring and interface interactions on chronic neural recordings in non-human primates. J. Neural Eng. 13, 16010 (2016).

    Article  Google Scholar 

  66. Humphrey, D. R. & Schmidt, E. M. in Neurophysiological Techniques Vol. II (eds Boulton, A. A., Baker, G. B. & Vanderwolf, C. H.) 1–64 (Humana Press, New York, 1990).

  67. Shoham, S. & Nagarajan, S. in Neuroprosthetics: Theory and Practice 448–465 (World Scientific, 2003).

  68. Alivisatos, A. P. et al. Nanotools for neuroscience and brain activity mapping. ACS Nano 7, 1850–1866 (2013).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Clark, J. J. et al. Chronic microsensors for longitudinal, subsecond dopamine detection in behaving animals. Nat. Methods 7, 126–129 (2010).

    Article  CAS  PubMed  Google Scholar 

  70. Grahn, P. J. et al. A neurochemical closed-loop controller for deep brain stimulation: toward individualized smart neuromodulation therapies. Front. Neurosci. 8, 169 (2014).

    PubMed Central  PubMed  Google Scholar 

  71. Hascup, E. R. et al. Histological studies of the effects of chronic implantation of ceramic-based microelectrode arrays and microdialysis probes in rat prefrontal cortex. Brain Res. 1291, 12–20 (2009).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  72. Phillips, P. E. M. & Wightman, R. M. Critical guidelines for validation of the selectivity of in-vivo chemical microsensors. Trends Anal. Chem. 22, 509–514 (2003).

    Article  CAS  Google Scholar 

  73. Roitbak, T. & Sykov, E. Diffusion barriers evoked in the rat cortex by reactive astrogliosis. Glia 28, 40–48 (1999).

    Article  CAS  PubMed  Google Scholar 

  74. Hascup, E. R. et al. Rapid microelectrode measurements and the origin and regulation of extracellular glutamate in rat prefrontal cortex. J. Neurochem. 115, 1608–1620 (2010).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  75. Melendez, R. I., Vuthiganon, J. & Kalivas, P. W. Regulation of extracellular glutamate in the prefrontal cortex: focus on the cystine glutamate exchanger and group I metabotropic glutamate receptors. J. Pharmacol. Exp. Ther. 314, 139–147 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Hille, B. Ion Channels of Excitable Membranes (Sinauer, Sunderland, MA, 2001).

    Google Scholar 

  77. Pietrobon, D. & Moskowitz, M. A. Chaos and commotion in the wake of cortical spreading depression and spreading depolarizations. Nat. Rev. Neurosci. 15, 379–393 (2014).

    Article  CAS  PubMed  Google Scholar 

  78. Haupt, C., Witte, O. W. & Frahm, C. Up-regulation of connexin43 in the glial scar following photothrombotic ischemic injury. Mol. Cell. Neurosci. 35, 89–99 (2007).

    Article  CAS  PubMed  Google Scholar 

  79. Karumbaiah, L. et al. Relationship between intracortical electrode design and chronic recording function. Biomaterials 34, 8061–8074 (2013).

    Article  CAS  PubMed  Google Scholar 

  80. Biran, R., Martin, D. C. & Tresco, P. A. Neuronal cell loss accompanies the brain tissue response to chronically implanted silicon microelectrode arrays. Exp. Neurol. 195, 115–126 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Karumbaiah, L. et al. The upregulation of specific interleukin (IL) receptor antagonists and paradoxical enhancement of neuronal apoptosis due to electrode induced strain and brain micromotion. Biomaterials 33, 5983–5996 (2012).

    Article  CAS  PubMed  Google Scholar 

  82. Kozai, T. D. Y. et al. Effects of caspase-1 knockout on chronic neural recording quality and longevity: insight into cellular and molecular mechanisms of the reactive tissue response. Biomaterials 35, 9620–9634 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  83. Vezzani, A., French, J., Bartfai, T. & Baram, T. Z. The role of inflammation in epilepsy. Nat. Rev. Neurol. 7, 31–40 (2011).

    Article  CAS  PubMed  Google Scholar 

  84. Vezzani, A. & Viviani, B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology 96, 70–82 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Zhou, C. et al. Interleukin-1β inhibits voltage-gated sodium currents in a time- and dose-dependent manner in cortical neurons. Neurochem. Res. 36, 1116–1123 (2011).

    Article  CAS  PubMed  Google Scholar 

  86. Li, X., Chen, W., Sheng, J., Cao, D. & Wang, W. Interleukin-6 inhibits voltage-gated sodium channel activity of cultured rat spinal cord neurons. Acta Neuropsychiatr. 26, 170–177 (2014).

    Article  PubMed  Google Scholar 

  87. Zhou, C., Ye, H.-H., Wang, S.-Q. & Chai, Z. Interleukin-1β regulation of N-type Ca2+ channels in cortical neurons. Neurosci. Lett. 403, 181–185 (2006).

  88. Ma, S.-H., Li, B., Huang, H.-W., Peng, Y.-P. & Qiu, Y.-H. Interleukin-6 inhibits L-type calcium channel activity of cultured cerebellar granule neurons. J. Physiol. Sci. 62, 385–392 (2012).

    Article  CAS  PubMed  Google Scholar 

  89. Liu, Z., Qiu, Y.-H., Li, B., Ma, S.-H. & Peng, Y.-P. Neuroprotection of interleukin-6 against NMDA-induced apoptosis and its signal-transduction mechanisms. Neurotox. Res. 19, 484–495 (2011).

    Article  CAS  PubMed  Google Scholar 

  90. Baldwin, K. T. & Eroglu, C. Molecular mechanisms of astrocyte-induced synaptogenesis. Curr. Opin. Neurobiol. 45, 113–120 (2017).

    Article  CAS  PubMed  Google Scholar 

  91. Salatino, J. W., Winter, B. M., Drazin, M. H. & Purcell, E. K. Functional remodeling of subtype-specific markers surrounding implanted neuroprostheses. J. Neurophysiol. 118, 194–202 (2017).

  92. Weissberg, I. et al. Albumin induces excitatory synaptogenesis through astrocytic TGF-β/ALK5 signaling in a model of acquired epilepsy following blood-brain barrier dysfunction. Neurobiol. Dis. 78, 115–125 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  93. Diniz, L. P. et al. Astrocyte transforming growth factor beta 1 promotes inhibitory synapse formation via CaM kinase II signaling. Glia 62, 1917–1931 (2014).

    Article  PubMed  Google Scholar 

  94. Lin, T.-n et al. Differential regulation of thrombospondin-1 and thrombospondin-2 after focal cerebral ischemia/reperfusion. Stroke 34, 177–186 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Carsten Möller, J. et al. Regulation of thrombospondin in the regenerating mouse facial motor nucleus. Glia 17, 121–132 (1996).

    Article  Google Scholar 

  96. Tran, M. D. & Neary, J. T. Purinergic signaling induces thrombospondin-1 expression in astrocytes. Proc. Natl Acad. Sci. USA 103, 9321–9326 (2006).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  97. Christopherson, K. S. et al. Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120, 421–433 (2005).

    Article  CAS  PubMed  Google Scholar 

  98. Kerchner, G. A. & Nicoll, R. A. Silent synapses and the emergence of a postsynaptic mechanism for LTP. Nat. Rev. Neurosci. 9, 813–825 (2008).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  99. Stellwagen, D. & Malenka, R. C. Synaptic scaling mediated by glial TNF-α. Nature 440, 1054–1059 (2006).

    Article  CAS  PubMed  Google Scholar 

  100. Beattie, E. C. et al. Control of synaptic strength by glial TNFalpha. Science 295, 2282–2285 (2002).

    Article  CAS  PubMed  Google Scholar 

  101. Chang, S.-Y., Shon, Y. M., Agnesi, F. & Lee, K. H. Microthalamotomy effect during deep brain stimulation: potential involvement of adenosine and glutamate efflux. In 2009 Annual International Conference of the IEEE Engineering in Medicine and Biology Society 3294–3297 (IEEE, 2009).

  102. Dunwiddie, T. V., Diao, L. & Proctor, W. R. Adenine nucleotides undergo rapid, quantitative conversion to adenosine in the extracellular space in rat hippocampus. J. Neurosci. 17, 7673–7682 (1997).

    CAS  PubMed  Google Scholar 

  103. Fredholm, B. & Dunwiddie, T. How does adenosine inhibit transmitter release? Trends Pharmacol. Sci. 9, 130–134 (1988).

    Article  CAS  PubMed  Google Scholar 

  104. Dunwiddie, T. V. & Fredholm, B. B. Adenosine A1 receptors inhibit adenylate cyclase activity and neurotransmitter release and hyperpolarize pyramidal neurons in rat hippocampus. J. Pharmacol. Exp. Ther. 249, 31–37 (1989).

    CAS  PubMed  Google Scholar 

  105. Stone, T. W., Ceruti, S. & Abbracchio, M. P. in Adenosine Receptors in Health and Disease. Handbook of Experimental Pharmacology Vol. 193 (eds Wilson, C. & Mustafa, S.) 535–587 (Springer, Berlin, Heidelberg, 2009).

    Chapter  Google Scholar 

  106. Ben Achour, S. & Pascual, O. Glia: the many ways to modulate synaptic plasticity. Neurochem. Int. 57, 440–445 (2010).

    Article  CAS  PubMed  Google Scholar 

  107. Gradinaru, V., Mogri, M., Thompson, K. R., Henderson, J. M. & Deisseroth, K. Optical deconstruction of parkinsonian neural circuitry. Science 324, 354–359 (2009).

    Article  CAS  PubMed  Google Scholar 

  108. Amiri, M., Montaseri, G. & Bahrami, F. On the role of astrocytes in synchronization of two coupled neurons: a mathematical perspective. Biol. Cybern. 105, 153–166 (2011).

    Article  PubMed  Google Scholar 

  109. Agulhon, C., Fiacco, T. A. & McCarthy, K. D. Hippocampal short- and long-term plasticity are not modulated by astrocyte Ca2+ signaling. Science 327, 1250–1254 (2010).

    Article  CAS  PubMed  Google Scholar 

  110. Strogatz, S. H. Exploring complex networks. Nature 410, 268–276 (2001).

    Article  CAS  PubMed  Google Scholar 

  111. Perea, G. & Araque, A. Astrocytes potentiate transmitter release at single hippocampal synapses. Science 317, 1083–1086 (2007).

    Article  CAS  PubMed  Google Scholar 

  112. Newman, E. A. New roles for astrocytes: regulation of synaptic transmission. Trends Neurosci. 26, 536–542 (2003).

    Article  CAS  PubMed  Google Scholar 

  113. Wetherington, J., Serrano, G. & Dingledine, R. Astrocytes in the epileptic brain. Neuron 58, 168–178 (2008).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  114. Fellin, T. et al. Neuronal synchrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors. Neuron 43, 729–743 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Silchenko, A. N. & Tass, P. A. Computational modeling of paroxysmal depolarization shifts in neurons induced by the glutamate release from astrocytes. Biol. Cybern. 98, 61–74 (2008).

    Article  PubMed  Google Scholar 

  116. Uhlhaas, P. J. & Singer, W. Neural synchrony in brain disorders: relevance for cognitive dysfunctions and pathophysiology. Neuron 52, 155–168 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Norden, D. M., Muccigrosso, M. M. & Godbout, J. P. Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease. Neuropharmacology 96, 29–41 (2015).

    Article  CAS  PubMed  Google Scholar 

  118. Vedam-Mai, V. et al. Deep brain stimulation and the role of astrocytes. Mol. Psychiatry 17, 124–31 (2012).

    Article  CAS  PubMed  Google Scholar 

  119. McIntyre, C. C. & Anderson, R. W. Deep brain stimulation mechanisms: the control of network activity via neurochemistry modulation. J. Neurochem. 139, 338–345 (2016).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  120. Fenoy, A. J., Goetz, L., Chabardès, S. & Xia, Y. Deep brain stimulation: are astrocytes a key driver behind the scene? CNS Neurosci. Ther. 20, 191–201 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  121. Vedam-Mai, V. et al. The national DBS brain tissue network pilot study: need for more tissue and more standardization. Cell Tissue Bank 12, 219–231 (2011).

    Article  CAS  PubMed  Google Scholar 

  122. Sun, D. A. et al. Postmortem analysis following 71 months of deep brain stimulation of the subthalamic nucleus for Parkinson disease. J. Neurosurg. 109, 325–329 (2008).

    Article  PubMed  Google Scholar 

  123. van Kuyck, K., Welkenhuysen, M., Arckens, L., Sciot, R. & Nuttin, B. Histological alterations induced by electrode implantation and electrical stimulation in the human brain: a review. Neuromodulation 10, 244–261 (2007).

    Article  PubMed  Google Scholar 

  124. Chang, S.-Y. et al. Wireless fast-scan cyclic voltammetry to monitor adenosine in patients with essential tremor during deep brain stimulation. Mayo Clin. Proc. 87, 760–765 (2012).

    Article  PubMed Central  PubMed  Google Scholar 

  125. Van Gompel, J. J. et al. Increased cortical extracellular adenosine correlates with seizure termination. Epilepsia 55, 233–244 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  126. Santello, M. & Volterra, A. TNFα in synaptic function: switching gears. Trends Neurosci. 35, 638–647 (2012).

    Article  CAS  PubMed  Google Scholar 

  127. Gellner, A.-K., Reis, J. & Fritsch, B. Glia: a neglected player in non-invasive direct current brain stimulation. Front. Cell. Neurosci. 10, 188 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  128. Monai, H. et al. Calcium imaging reveals glial involvement in transcranial direct current stimulation-induced plasticity in mouse brain. Nat. Commun. 7, 11100 (2016).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  129. Sasaki, T. et al. Application of an optogenetic byway for perturbing neuronal activity via glial photostimulation. Proc. Natl Acad. Sci. USA 109, 20720–20725 (2012).

    Article  PubMed Central  PubMed  Google Scholar 

  130. Braun, R. et al. Transcranial direct current stimulation accelerates recovery of function, induces neurogenesis and recruits oligodendrocyte precursors in a rat model of stroke. Exp. Neurol. 279, 127–136 (2016).

    Article  PubMed  Google Scholar 

  131. Arsenault, D. et al. A novel combinational approach of microstimulation and bioluminescence imaging to study the mechanisms of action of cerebral electrical stimulation in mice. J. Physiol. 593, 2257–2278 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  132. Matsuo, H. et al. Early transcutaneous electrical nerve stimulation reduces hyperalgesia and decreases activation of spinal glial cells in mice with neuropathic pain. Pain 155, 1888–1901 (2014).

    Article  CAS  PubMed  Google Scholar 

  133. Peruzzotti-Jametti, L. et al. Safety and efficacy of transcranial direct current stimulation in acute experimental ischemic stroke. Stroke 44, 3166–3174 (2013).

    Article  PubMed  Google Scholar 

  134. Iwasa, S. N., Babona-Pilipos, R. & Morshead, C. M. environmental factors that influence stem cell migration: an “electric field”. Stem Cells Int. 2017, 4276927 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  135. Vedam-Mai, V. et al. Increased precursor cell proliferation after deep brain stimulation for Parkinson’s disease: a human study. PLoS ONE 9, e88770 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  136. Toda, H., Hamani, C., Fawcett, A. P., Hutchison, W. D. & Lozano, A. M. The regulation of adult rodent hippocampal neurogenesis by deep brain stimulation. J. Neurosurg. 108, 132–138 (2008).

    Article  PubMed  Google Scholar 

  137. Encinas, J. M., Hamani, C., Lozano, A. M. & Enikolopov, G. Neurogenic hippocampal targets of deep brain stimulation. J. Comp. Neurol. 519, 6–20 (2011).

    Article  PubMed Central  PubMed  Google Scholar 

  138. Babona-Pilipos, R., Pritchard-Oh, A., Popovic, M. R. & Morshead, C. M. Biphasic monopolar electrical stimulation induces rapid and directed galvanotaxis in adult subependymal neural precursors. Stem Cell Res. Ther. 6, 67 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  139. Rueger, M. A. et al. Multi-session transcranial direct current stimulation (tDCS) elicits inflammatory and regenerative processes in the rat brain. PLoS ONE 7, e43776 (2012).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  140. Zhao, H., Steiger, A., Nohner, M. & Ye, H. specific intensity direct current (DC) electric field improves neural stem cell migration and enhances differentiation towards βIII tubulin+ neurons. PLoS ONE 10, e0129625 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  141. Khaindrava, V. et al. High frequency stimulation of the subthalamic nucleus impacts adult neurogenesis in a rat model of Parkinson’s disease. Neurobiol. Dis. 42, 284–291 (2011).

    Article  PubMed  Google Scholar 

  142. Attwell, D. et al. Glial and neuronal control of brain blood flow. Nature 468, 232–243 (2010).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  143. Zonta, M. et al. Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation. Nat. Neurosci. 6, 43–50 (2003).

    Article  CAS  PubMed  Google Scholar 

  144. Filosa, J. A., Bonev, A. D. & Nelson, M. T. Calcium dynamics in cortical astrocytes and arterioles during neurovascular coupling. Circ. Res. 95, e73–e81 (2004).

  145. Takano, T. et al. Astrocyte-mediated control of cerebral blood flow. Nat. Neurosci. 9, 260–267 (2006).

    Article  CAS  PubMed  Google Scholar 

  146. Ke, B., Liu, T.-T., Liu, C., Xiang, H. & Xiong, J. Dorsal subthalamic nucleus electrical stimulation for drug/treatment-refractory epilepsy may modulate melanocortinergic signaling in astrocytes. Epilepsy Behav. 36, 6–8 (2014).

    Article  PubMed  Google Scholar 

  147. Seymour, J. P., Wu, F., Wise, K. D. & Yoon, E. State-of-the-art MEMS and microsystem tools for brain research. Microsyst. Nanoeng. 3, 16066 (2017).

    Article  Google Scholar 

  148. Seymour, J. P. & Kipke, D. R. Neural probe design for reduced tissue encapsulation in CNS. Biomaterials 28, 3594–607 (2007).

    Article  CAS  PubMed  Google Scholar 

  149. Loane, D. J. et al. Progressive neurodegeneration after experimental brain trauma: association with chronic microglial activation. J. Neuropathol. Exp. Neurol. 73, 14–29 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  150. Johnson, V. E. et al. Inflammation and white matter degeneration persist for years after a single traumatic brain injury. Brain 136, 28–42 (2013).

    Article  PubMed Central  PubMed  Google Scholar 

  151. Perry, V. H., Nicoll, J. A. R. & Holmes, C. Microglia in neurodegenerative disease. Nat. Rev. Neurol. 6, 193–201 (2010).

    Article  PubMed  Google Scholar 

  152. Glass, C. K., Saijo, K., Winner, B., Marchetto, M. C. & Gage, F. H. Mechanisms underlying inflammation in neurodegeneration. Cell 140, 918–934 (2010).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  153. Henry, C. J., Huang, Y., Wynne, A. M. & Godbout, J. P. Peripheral lipopolysaccharide (LPS) challenge promotes microglial hyperactivity in aged mice that is associated with exaggerated induction of both pro-inflammatory IL-1β and anti-inflammatory IL-10 cytokines. Brain. Behav. Immun. 23, 309–317 (2009).

    Article  CAS  PubMed  Google Scholar 

  154. Frank, M. G. et al. mRNA up-regulation of MHC II and pivotal pro-inflammatory genes in normal brain aging. Neurobiol. Aging 27, 717–722 (2006).

    Article  CAS  PubMed  Google Scholar 

  155. Cunningham, C., Wilcockson, D. C., Campion, S., Lunnon, K. & Perry, V. H. Central and systemic endotoxin challenges exacerbate the local inflammatory response and increase neuronal death during chronic neurodegeneration. J. Neurosci. 25, 9275–9284 (2005).

    Article  CAS  PubMed  Google Scholar 

  156. Lacour, S. P. et al. Materials and technologies for soft implantable neuroprostheses. Nat. Rev. Mater. 1, 16063 (2016).

    Article  CAS  Google Scholar 

  157. Wellman, S. M. et al. A materials roadmap to functional neural interface design. Adv. Funct. Mater. https://doi.org/10.1002/adfm.201701269 (2017).

    Google Scholar 

  158. Moshayedi, P. et al. The relationship between glial cell mechanosensitivity and foreign body reactions in the central nervous system. Biomaterials 35, 3919–3925 (2014).

    Article  CAS  PubMed  Google Scholar 

  159. Velliste, M., Perel, S., Spalding, M. C., Whitford, A. S. & Schwartz, A. B. Cortical control of a prosthetic arm for self-feeding. Nature 453, 1098–1101 (2008).

    Article  CAS  PubMed  Google Scholar 

  160. Collinger, J. L. et al. High-performance neuroprosthetic control by an individual with tetraplegia. Lancet 381, 557–564 (2013).

    Article  PubMed Central  PubMed  Google Scholar 

  161. Nguyen, J. K. et al. Mechanically-compliant intracortical implants reduce the neuroinflammatory response. J. Neural Eng. 11, 56014 (2014).

    Article  Google Scholar 

  162. Kolarcik, C. L. et al. Elastomeric and soft conducting microwires for implantable neural interfaces. Soft Matter 11, 4847–4861 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  163. Hess, A. E. et al. Development of a stimuli-responsive polymer nanocomposite toward biologically optimized, MEMS-based neural probes. J. Micromech. Microeng. 21, 54009 (2011).

    Article  Google Scholar 

  164. Ware, T. et al. Thiol-ene/acrylate substrates for softening intracortical electrodes. J. Biomed. Mater. Res. Part B Appl. Biomater. 102, 1–11 (2014).

    Article  CAS  PubMed  Google Scholar 

  165. Ware, T. et al. Fabrication of responsive, softening neural interfaces. Adv. Funct. Mater. 22, 3470–3479 (2012).

    Article  CAS  Google Scholar 

  166. Capadona, J. R. et al. Mechanically adaptive nanocomposites for neural interfacing. MRS Bull. 37, 581–589 (2012).

    Article  CAS  Google Scholar 

  167. Capadona, J. R., Shanmuganathan, K., Tyler, D. J., Rowan, S. J. & Weder, C. Stimuli-responsive polymer nanocomposites inspired by the sea cucumber dermis. Science 319, 1370–1374 (2008).

    Article  CAS  PubMed  Google Scholar 

  168. Harris, J. P. et al. Mechanically adaptive intracortical implants improve the proximity of neuronal cell bodies. J. Neural Eng. 8, 66011 (2011).

    Article  CAS  Google Scholar 

  169. Ware, T. et al. Three-dimensional flexible electronics enabled by shape memory polymer substrates for responsive neural interfaces. Macromol. Mater. Eng. 297, 1193–1202 (2012).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  170. Chen, R., Canales, A. & Anikeeva, P. Neural recording and modulation technologies. Nat. Rev. Mater. 2, 16093 (2017).

    Article  CAS  Google Scholar 

  171. Fu, T.-M. et al. Stable long-term chronic brain mapping at the single-neuron level. Nat. Methods 13, 875–882 (2016).

    Article  CAS  PubMed  Google Scholar 

  172. Hong, G. et al. Syringe Injectable electronics: precise targeted delivery with quantitative input/output connectivity. Nano Lett. 15, 6979–6984 (2015).

    Article  CAS  PubMed  Google Scholar 

  173. Luan, L. et al. Ultraflexible nanoelectronic probes form reliable, glial scar–free neural integration. Sci. Adv. 3, e1601966 (2017).

  174. Kim, B. J. et al. 3D parylene sheath neural probe for chronic recordings. J. Neural Eng. 10, 045002 (2013).

    Article  CAS  Google Scholar 

  175. Kim, T. et al. Injectable, cellular-scale optoelectronics with applications for wireless optogenetics. Science 340, 211–216 (2013).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  176. Park, S. II et al. Soft, stretchable, fully implantable miniaturized optoelectronic systems for wireless optogenetics. Nat. Biotechnol. 33, 1280–1286 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  177. Metallo, C. & Trimmer, B. A. Silk coating as a novel delivery system and reversible adhesive for stiffening and shaping flexible probes. J. Biol. Methods 2, e13 (2015).

    Article  Google Scholar 

  178. Kozai, T. D. Y. et al. Chronic tissue response to carboxymethyl cellulose based dissolvable insertion needle for ultra-small neural probes. Biomaterials 35, 9255–9268 (2014).

    Article  CAS  PubMed  Google Scholar 

  179. Seymour, J. P., Langhals, N. B., Anderson, D. J. & Kipke, D. R. Novel multi-sided, microelectrode arrays for implantable neural applications. Biomed. Microdevices 13, 441–451 (2011).

    Article  PubMed Central  PubMed  Google Scholar 

  180. Skousen, J. L. et al. Reducing surface area while maintaining implant penetrating profile lowers the brain foreign body response to chronically implanted planar silicon microelectrode arrays. Prog. Brain Res. 194, 167–80 (2011).

    Article  PubMed  Google Scholar 

  181. Chen, C. S., Mrksich, M., Huang, S., Whitesides, G. M. & Ingber, D. E. Geometric control of cell life and death. Science 276, 1425–1428 (1997).

  182. Chen, C. S., Tan, J. & Tien, J. Mechanotransduction at cell-matrix and cell-cell contacts. Annu. Rev. Biomed. Eng. 6, 275–302 (2004).

    Article  CAS  PubMed  Google Scholar 

  183. Turner, A. M. P. et al. Attachment of astroglial cells to microfabricated pillar arrays of different geometries. J. Biomed. Mater. Res. 51, 430–441 (2000).

    Article  CAS  PubMed  Google Scholar 

  184. Sanders, J. E., Stiles, C. E. & Hayes, C. L. Tissue response to single-polymer fibers of varying diameters: evaluation of fibrous encapsulation and macrophage density. J. Biomed. Mater. Res. 52, 231–237 (2000).

    Article  CAS  PubMed  Google Scholar 

  185. Bernatchez, S., Parks, P. J. & Gibbons, D. F. Interaction of macrophages with fibrous materials in vitro. Biomaterials 17, 2077–2086 (1996).

    Article  CAS  PubMed  Google Scholar 

  186. Gällentoft, L. et al. Size-dependent long-term tissue response to biostable nanowires in the brain. Biomaterials 42, 172–183 (2015).

    Article  CAS  PubMed  Google Scholar 

  187. Skousen, J. L., Bridge, M. J. & Tresco, P. A. A strategy to passively reduce neuroinflammation surrounding devices implanted chronically in brain tissue by manipulating device surface permeability. Biomaterials 36, 33–43 (2015).

    Article  CAS  PubMed  Google Scholar 

  188. Saxena, T. & Bellamkonda, R. V. Implantable electronics: a sensor web for neurons. Nat. Mater. 14, 1190–1191 (2015).

    Article  CAS  PubMed  Google Scholar 

  189. Xie, C. et al. Three-dimensional macroporous nanoelectronic networks as minimally invasive brain probes. Nat. Mater. 14, 1286–1292 (2015).

    Article  CAS  PubMed  Google Scholar 

  190. Liu, J. et al. Syringe-injectable electronics. Nat. Nanotech. 10, 629–636 (2015).

    Article  CAS  Google Scholar 

  191. Fattahi, P., Yang, G., Kim, G. & Abidian, M. R. A review of organic and inorganic biomaterials for neural interfaces. Adv. Mater. 26, 1846–1885 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  192. Green, R. & Abidian, M. R. Conducting polymers for neural prosthetic and neural interface applications. Adv. Mater. 27, 7620–7637 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  193. Kolarcik, C. L. et al. In vivo effects of L1 coating on inflammation and neuronal health at the electrode–tissue interface in rat spinal cord and dorsal root ganglion. Acta Biomater. 8, 3561–3575 (2012).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  194. Rao, L., Zhou, H., Li, T., Li, C. & Duan, Y. Y. Polyethylene glycol-containing polyurethane hydrogel coatings for improving the biocompatibility of neural electrodes. Acta Biomater. 8, 2233–2242 (2012).

    Article  CAS  PubMed  Google Scholar 

  195. Kim, D.-H. et al. Dissolvable films of silk fibroin for ultrathin conformal bio-integrated electronics. Nat. Mater. 9, 511–517 (2010).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  196. Tien, L. W. et al. Silk as a multifunctional biomaterial substrate for reduced glial scarring around brain-penetrating electrodes. Adv. Funct. Mater. 23, 3185–3193 (2013).

    Article  CAS  Google Scholar 

  197. Zhong, Y. & Bellamkonda, R. V. Dexamethasone-coated neural probes elicit attenuated inflammatory response and neuronal loss compared to uncoated neural probes. Brain Res. 1148, 15–27 (2007).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  198. He, W., McConnell, G. C., Schneider, T. M. & Bellamkonda, R. V. A novel anti-inflammatory surface for neural electrodes. Adv. Mater. 19, 3529–3533 (2007).

    Article  CAS  Google Scholar 

  199. Mercanzini, A. et al. Controlled release nanoparticle-embedded coatings reduce the tissue reaction to neuroprostheses. J. Control. Release 145, 196–202 (2010).

    Article  CAS  PubMed  Google Scholar 

  200. Abidian, M. R. & Martin, D. C. Multifunctional nanobiomaterials for neural interfaces. Adv. Funct. Mater. 19, 573–585 (2009).

    Article  CAS  Google Scholar 

  201. Bezuidenhout, D. et al. Covalent incorporation and controlled release of active dexamethasone from injectable polyethylene glycol hydrogels. J. Biomed. Mater. Res. Part A 101A, 1311–1318 (2013).

    Article  CAS  Google Scholar 

  202. Gutowski, S. M. et al. Protease-degradable PEG-maleimide coating with on-demand release of IL-1Ra to improve tissue response to neural electrodes. Biomaterials 44, 55–70 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  203. Wadhwa, R., Lagenaur, C. F. & Cui, X. T. Electrochemically controlled release of dexamethasone from conducting polymer polypyrrole coated electrode. J. Control. Release 110, 531–541 (2006).

    Article  CAS  PubMed  Google Scholar 

  204. Kim, Y. et al. Stretchable nanoparticle conductors with self-organized conductive pathways. Nature 500, 59–63 (2013).

    Article  CAS  PubMed  Google Scholar 

  205. Abidian, M. R., Ludwig, K. A., Marzullo, T. C., Martin, D. C. & Kipke, D. R. Interfacing conducting polymer nanotubes with the central nervous system: chronic neural recording using poly(3,4-ethylenedioxythiophene) nanotubes. Adv. Mater. 21, 3764–3770 (2009).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  206. Keefer, E. W., Botterman, B. R., Romero, M. I., Rossi, A. F. & Gross, G. W. Carbon nanotube coating improves neuronal recordings. Nat. Nanotech. 3, 434–439 (2008).

    Article  CAS  Google Scholar 

  207. Abidian, M. R., Corey, J. M., Kipke, D. R. & Martin, D. C. Conducting-polymer nanotubes improve electrical properties, mechanical adhesion, neural attachment, and neurite outgrowth of neural electrodes. Small 6, 421–429 (2010).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  208. Eles, J. R. et al. Neuroadhesive L1 coating attenuates acute microglial encapsulation of neural electrodes as revealed by live two-photon microscopy. Biomaterials 113, 279–292 (2017).

    Article  CAS  PubMed  Google Scholar 

  209. Singh, A. V. et al. Astrocytes increase ATP exocytosis mediated calcium signaling in response to microgroove structures. Sci. Rep. 5, 7847 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  210. Min, S. K. et al. Effect of topography of an electrospun nanofiber on modulation of activity of primary rat astrocytes. Neurosci. Lett. 534, 80–84 (2013).

    Article  CAS  PubMed  Google Scholar 

  211. Zuidema, J. M. et al. Enhanced GLT-1 mediated glutamate uptake and migration of primary astrocytes directed by fibronectin-coated electrospun poly-l-lactic acid fibers. Biomaterials 35, 1439–1449 (2014).

    Article  CAS  PubMed  Google Scholar 

  212. Vishwakarma, A. et al. Engineering immunomodulatory biomaterials to tune the inflammatory response. Trends Biotechnol. 34, 470–482 (2016).

    Article  CAS  PubMed  Google Scholar 

  213. Webber, M. J., Appel, E. A., Meijer, E. W. & Langer, R. Supramolecular biomaterials. Nat. Mater. 15, 13–26 (2016).

    Article  CAS  PubMed  Google Scholar 

  214. Lu, Y., Aimetti, A. A., Langer, R. & Gu, Z. Bioresponsive materials. Nat. Rev. Mater. 2, 16075 (2016).

    Article  CAS  Google Scholar 

  215. Jackson, A., Mavoori, J. & Fetz, E. E. Long-term motor cortex plasticity induced by an electronic neural implant. Nature 444, 56–60 (2006).

  216. Hampson, R. E. et al. Facilitation and restoration of cognitive function in primate prefrontal cortex by a neuroprosthesis that utilizes minicolumn-specific neural firing. J. Neural Eng. 9, 056012 (2012).

    Google Scholar 

  217. Kipke, D. R., Vetter, R. J., Williams, J. C. & Hetke, J. F. Silicon-substrate intracortical microelectrode arrays for long-term recording of neuronal spike activity in cerebral cortex. IEEE Trans. Neural Syst. Rehabil. Eng. 11, 151–155 (2003).

    Article  PubMed  Google Scholar 

  218. Kindlmann, G. et al. Imaging of Utah electrode array, implanted in cochlear nerve. Digit. Biol. Emerg. Paradig. 6–7 (2003).

  219. Moss, J., Ryder, T., Aziz, T. Z., Graeber, M. B. & Bain, P. G. Electron microscopy of tissue adherent to explanted electrodes in dystonia and Parkinson’s disease. Brain 127, 2755–2763 (2004).

    Article  CAS  PubMed  Google Scholar 

  220. Kozai, T. D. Y., Vazquez, A. L., Weaver, C. L., Kim, S.-G. & Cui, X. T. In vivo two-photon microscopy reveals immediate microglial reaction to implantation of microelectrode through extension of processes. J. Neural Eng. 9, 066001 (2012).

    Article  PubMed Central  PubMed  Google Scholar 

  221. Ludwig, K. A. et al. Using a common average reference to improve cortical neuron recordings from microelectrode arrays. J. Neurophysiol. 101, 1679–1689 (2009).

  222. Nishiyama, A., Komitova, M., Suzuki, R. & Zhu, X. Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nat. Rev. Neurosci. 10, 9–22 (2009).

    Article  CAS  PubMed  Google Scholar 

  223. Richardson, W. D., Young, K. M., Tripathi, R. B. & McKenzie, I. NG2-glia as multipotent neural stem cells: fact or fantasy? Neuron 70, 661–73 (2011).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  224. Dimou, L. & Gallo, V. NG2-glia and their functions in the central nervous system. Glia 63, 1429–1451 (2015).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  225. Crisan, M. et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 3, 301–313 (2008).

    Article  CAS  PubMed  Google Scholar 

  226. Scadden, D. T. The stem-cell niche as an entity of action. Nature 441, 1075–1079 (2006).

    Article  CAS  PubMed  Google Scholar 

  227. Hall, C. N. et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature 508, 55–60 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  228. Fidler, P. S. et al. Comparing astrocytic cell lines that are inhibitory or permissive for axon growth: the major axon-inhibitory proteoglycan is NG2. J. Neurosci. 19, 8778–8788 (1999).

    CAS  PubMed  Google Scholar 

  229. Rivers, L. E. et al. PDGFRA/NG2 glia generate myelinating oligodendrocytes and piriform projection neurons in adult mice. Nat. Neurosci. 11, 1392–1401 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

J.W.S. was supported by National Institutes of Health (NIH) 1R21NS094900, T.D.Y.K. was supported by NIH 1R01NS094396, K.A.L. was supported by The Grainger Foundation, and E.K.P. was supported by NIH 1R21NS094900 and 5R03NS095202. The authors thank J. Eles for assistance collecting in vivo imaging data (Fig. 2a), D. Thompson and S. Yandamuri for assistance collecting data presented in Fig. 3, and M.-C. Senut of Biomilab, LLC, for providing feedback.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to researching the data and discussing the content of the manuscript, and to writing, reviewing and editing it.

Corresponding author

Correspondence to Erin K. Purcell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

A correction to this article is available online at https://doi.org/10.1038/s41551-017-0177-7.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Salatino, J.W., Ludwig, K.A., Kozai, T.D.Y. et al. Glial responses to implanted electrodes in the brain. Nat Biomed Eng 1, 862–877 (2017). https://doi.org/10.1038/s41551-017-0154-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-017-0154-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research