251
|
Petrossians A, Whalen JJ, Weiland JD, Mansfeld F. Surface modification of neural stimulating/recording electrodes with high surface area platinum-iridium alloy coatings. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2011; 2011:3001-3004. [PMID: 22254972 DOI: 10.1109/iembs.2011.6090823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
High-surface area platinum-iridium alloys were electrodeposited by on Pt and Au microelectrodes using a potential sweep technique. Detailed investigations of the structure and morphology and the electrochemical properties of the electrodeposited Pt-Ir alloy coatings were performed. Scanning electron microscopy (SEM) and energy dispersive spectroscopy (EDS) were used for the determination of the surface morphology and the chemical composition of the Pt-Ir coatings, respectively. The elemental analysis by EDS showed a nearly 60-40% Pt-Ir composition of the coatings. The electrochemical properties of the Pt-Ir coatings were evaluated using cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS). CV and EIS measurements revealed that the Pt-Ir coated electrodes exhibit significantly increased charge storage capacity and real surface area compared to uncoated Pt electrodes. Charge injection experiments of the Pt-Ir coated microelectrodes revealed low potential excursions, indicating high charge injection capabilities within safe potential limits.
Collapse
Affiliation(s)
- Artin Petrossians
- Chemical Engineering and Materials Science Department, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | |
Collapse
|
252
|
Hassler C, Guy J, Nietzschmann M, Staiger JF, Stieglitz T. Chronic intracortical implantation of saccharose-coated flexible shaft electrodes into the cortex of rats. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2011; 2011:644-647. [PMID: 22254391 DOI: 10.1109/iembs.2011.6090143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Within this study, polyimide based shaft electrodes were fabricated and dip-coated in molten saccharose to stiffen them for insertion into the brain tissue. These electrodes were then implanted successfully into the cortex of whistar rats and the insertion force during implantation was recorded. Electrochemical impedance spectroscopy was performed immediately after implantation and in regular time intervals up to 201 days after implantation to monitor the tissue response to the implanted electrodes. Depending on the measured electrode pairs and the rats, the impedance spectra behaved different over time. Either they showed a constant decrease in impedance at 1 kHz, or they showed an initial decrease to increase again later. Furthermore, physiological signal recording was performed by stimulating the rats with acoustic signals and simultaneously recording the response on the different electrode sites. Multi-unit activity was detected until 37 days after implantation with an averaged signal-to-noise ratio of 2 to 4.
Collapse
Affiliation(s)
- Christina Hassler
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering, University of Freiburg, Freiburg, Germany.
| | | | | | | | | |
Collapse
|
253
|
A comparison of the tissue response to chronically implanted Parylene-C-coated and uncoated planar silicon microelectrode arrays in rat cortex. Biomaterials 2010; 31:9163-72. [DOI: 10.1016/j.biomaterials.2010.05.050] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 05/20/2010] [Indexed: 11/21/2022]
|
254
|
Lee JY, Schmidt CE. Pyrrole-hyaluronic acid conjugates for decreasing cell binding to metals and conducting polymers. Acta Biomater 2010; 6:4396-404. [PMID: 20558330 DOI: 10.1016/j.actbio.2010.06.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 06/02/2010] [Accepted: 06/10/2010] [Indexed: 01/01/2023]
Abstract
Surface modification of electrically conductive biomaterials has been studied to improve biocompatibility for a number of applications, such as implantable sensors and microelectrode arrays. In this study we electrochemically coated electrodes with biocompatible and non-cell adhesive hyaluronic acid (HA) to reduce cellular adhesion for potential use in neural prostheses. To this end, pyrrole-conjugated hyaluronic acid (PyHA) was synthesized and employed to electrochemically coat platinum, indium-tin oxide and polystyrene sulfonate-doped polypyrrole electrodes. This PyHA conjugate consisted of (1) a pyrrole moiety that allowed the compound to be electrochemically polymerized onto a conductive substrate and (2) non-adhesive HA to minimize cell adhesion and to potentially decrease inflammatory tissue responses. Our characterization results showed the presence of a hydrophilic p(PyHA) layer on the modified electrode, and impedance measurements revealed an impedance that was statistically the same as the unmodified electrode. We found that the p(PyHA)-coated electrodes minimized adhesion and migration of fibroblasts and astrocytes for a minimum of up to 3 months. Also, the coating was stable in physiological solution for 3 months and was stable against enzymatic degradation by hyaluronidase. These studies suggest that this p(PyHA) coating has the potential to be used to mask conducting electrodes from adverse glial responses that occur upon implantation. In addition, electrochemical coating with PyHA could potentially be extended for the surface modification of other metallic and conducting substances, such as stents and biosensors.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | | |
Collapse
|
255
|
Azemi E, Lagenaur CF, Cui XT. The surface immobilization of the neural adhesion molecule L1 on neural probes and its effect on neuronal density and gliosis at the probe/tissue interface. Biomaterials 2010; 32:681-92. [PMID: 20933270 DOI: 10.1016/j.biomaterials.2010.09.033] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 09/14/2010] [Indexed: 12/16/2022]
Abstract
Brain tissue inflammatory responses, including neuronal loss and gliosis at the neural electrode/tissue interface, limit the recording stability and longevity of neural probes. The neural adhesion molecule L1 specifically promotes neurite outgrowth and neuronal survival. In this study, we covalently immobilized L1 on the surface of silicon-based neural probes and compared the tissue response between L1 modified and non-modified probes implanted in the rat cortex after 1, 4, and 8 weeks. The effect of L1 on neuronal health and survival, and glial cell reactions were evaluated with immunohistochemistry and quantitative image analysis. Similar to previous findings, persistent glial activation and significant decreases of neuronal and axonal densities were found at the vicinity of the non-modified probes. In contrast, the immediate area (100 μm) around the L1 modified probe showed no loss of neuronal bodies and a significantly increased axonal density relative to background. In this same region, immunohistochemistry analyses show a significantly lower activation of microglia and reaction of astrocytes around the L1 modified probes when compared to the control probes. These improvements in tissue reaction induced by the L1 coating are likely to lead to improved functionality of the implanted neural electrodes during chronic recordings.
Collapse
Affiliation(s)
- Erdrin Azemi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | |
Collapse
|
256
|
Mercanzini A, Reddy ST, Velluto D, Colin P, Maillard A, Bensadoun JC, Hubbell JA, Renaud P. Controlled release nanoparticle-embedded coatings reduce the tissue reaction to neuroprostheses. J Control Release 2010; 145:196-202. [DOI: 10.1016/j.jconrel.2010.04.025] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 04/26/2010] [Accepted: 04/27/2010] [Indexed: 11/28/2022]
|
257
|
Kozai TDY, Marzullo TC, Hooi F, Langhals NB, Majewska AK, Brown EB, Kipke DR. Reduction of neurovascular damage resulting from microelectrode insertion into the cerebral cortex using in vivo two-photon mapping. J Neural Eng 2010; 7:046011. [PMID: 20644246 PMCID: PMC3164482 DOI: 10.1088/1741-2560/7/4/046011] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Penetrating neural probe technologies allow investigators to record electrical signals in the brain. The implantation of probes causes acute tissue damage, partially due to vasculature disruption during probe implantation. This trauma can cause abnormal electrophysiological responses and temporary increases in neurotransmitter levels, and perpetuate chronic immune responses. A significant challenge for investigators is to examine neurovascular features below the surface of the brain in vivo. The objective of this study was to investigate localized bleeding resulting from inserting microscale neural probes into the cortex using two-photon microscopy (TPM) and to explore an approach to minimize blood vessel disruption through insertion methods and probe design. 3D TPM images of cortical neurovasculature were obtained from mice and used to select preferred insertion positions for probe insertion to reduce neurovasculature damage. There was an 82.8 +/- 14.3% reduction in neurovascular damage for probes inserted in regions devoid of major (>5 microm) sub-surface vessels. Also, the deviation of surface vessels from the vector normal to the surface as a function of depth and vessel diameter was measured and characterized. 68% of the major vessels were found to deviate less than 49 microm from their surface origin up to a depth of 500 microm. Inserting probes more than 49 microm from major surface vessels can reduce the chances of severing major sub-surface neurovasculature without using TPM.
Collapse
Affiliation(s)
- T D Y Kozai
- Neural Engineering Lab, Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | | | | | |
Collapse
|
258
|
Bamford JA, Todd KG, Mushahwar VK. The effects of intraspinal microstimulation on spinal cord tissue in the rat. Biomaterials 2010; 31:5552-63. [PMID: 20430436 PMCID: PMC2875271 DOI: 10.1016/j.biomaterials.2010.03.051] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Accepted: 03/19/2010] [Indexed: 10/19/2022]
Abstract
Intraspinal microstimulation (ISMS) involves the implantation of microwires into the spinal cord below the level of an injury to excite neural networks involved in the control of locomotion in the lower limbs. The goal of this study was to examine the potential spinal cord damage that might occur with chronic ISMS. We employed functional measures of force recruitment and immunohistochemical processing of serial spinal cord sections to evaluate any damage induced by spinal transection, implantation of ISMS arrays, and electrical stimulation of 4h/day for 30 days. Functional measurements showed no change in force recruitment following transection and chronic ISMS, indicating no changes to underlying neural networks. The implantation of sham intraspinal microwires produced a spatially-limited increase in the density of microglia/macrophages and GFAP+ astrocytes adjacent to the microwire tracks, indicating a persistent immune response. Most importantly, these results were not different from those around microwires that were chronically pulsed with charge levels up to 48nC/phase. Likewise, measurements of neuronal density indicated no decrease in neuronal cell bodies in the ventral grey matter surrounding ISMS microwires (243.6/mm2+/-35.3/mm2) compared to tissue surrounding sham microwires (207.8/mm2+/-38.8/mm2). We conclude that the implantation of intraspinal microwires and chronic application of ISMS are well tolerated by spinal cord tissue.
Collapse
Affiliation(s)
- Jeremy A. Bamford
- Centre for Neuroscience, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Kathryn G. Todd
- Centre for Neuroscience, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Vivian K. Mushahwar
- Centre for Neuroscience, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| |
Collapse
|
259
|
Lind G, Linsmeier CE, Thelin J, Schouenborg J. Gelatine-embedded electrodes—a novel biocompatible vehicle allowing implantation of highly flexible microelectrodes. J Neural Eng 2010; 7:046005. [DOI: 10.1088/1741-2560/7/4/046005] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
260
|
McCreery D, Pikov V, Troyk PR. Neuronal loss due to prolonged controlled-current stimulation with chronically implanted microelectrodes in the cat cerebral cortex. J Neural Eng 2010; 7:036005. [PMID: 20460692 PMCID: PMC2921317 DOI: 10.1088/1741-2560/7/3/036005] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Activated iridium microelectrodes were implanted for 450-1282 days in the sensorimotor cortex of seven adult domestic cats and then pulsed for 240 h (8 h per day for 30 days) at 50 Hz. Continuous stimulation at 2 nC/phase and with a geometric charge density of 100 microC cm(-2) produced no detectable change in neuronal density in the tissue surrounding the microelectrode tips. However, pulsing with a continuous 100% duty cycle at 4 nC/phase and with a geometric charge density of 200 microC cm(-2) induced loss of cortical neurons over a radius of at least 150 microm from the electrode tips. The same stimulus regimen but with a duty cycle of 50% (1 s of stimulation, and then 1 s without stimulation repeated for 8 h) produced neuronal loss within a smaller radius, approximately 60 microm from the center of the electrode tips. However, there also was significant loss of neurons surrounding the unpulsed electrodes, presumably as a result of mechanical injury due to their insertion into and long-term residence in the tissue, and this was responsible for most of the neuronal loss within 150 microm of the electrodes pulsed with the 50% duty cycle.
Collapse
Affiliation(s)
- Douglas McCreery
- Neural Engineering Program, Huntington Medical Research Institutes, Pasadena, CA 91105, USA.
| | | | | |
Collapse
|
261
|
Grand L, Wittner L, Herwik S, Göthelid E, Ruther P, Oscarsson S, Neves H, Dombovári B, Csercsa R, Karmos G, Ulbert I. Short and long term biocompatibility of NeuroProbes silicon probes. J Neurosci Methods 2010; 189:216-29. [PMID: 20399227 DOI: 10.1016/j.jneumeth.2010.04.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 03/12/2010] [Accepted: 04/08/2010] [Indexed: 12/01/2022]
Affiliation(s)
- László Grand
- Institute for Psychology, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
262
|
Marin C, Fernández E. Biocompatibility of intracortical microelectrodes: current status and future prospects. FRONTIERS IN NEUROENGINEERING 2010; 3:8. [PMID: 20577634 PMCID: PMC2889721 DOI: 10.3389/fneng.2010.00008] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 05/05/2010] [Indexed: 11/17/2022]
Abstract
Rehabilitation of sensory and/or motor functions in patients with neurological diseases is more and more dealing with artificial electrical stimulation and recording from populations of neurons using biocompatible chronic implants. As more and more patients have benefited from these approaches, the interest in neural interfaces has grown significantly. However an important problem reported with all available microelectrodes to date is long-term viability and biocompatibility. Therefore it is essential to understand the signals that lead to neuroglial activation and create a targeted intervention to control the response, reduce the adverse nature of the reactions and maintain an ideal environment for the brain-electrode interface. We discuss some of the exciting opportunities and challenges that lie in this intersection of neuroscience research, bioengineering, neurology and biomaterials.
Collapse
Affiliation(s)
- Cristina Marin
- Institute of Bioengineering, University Miguel Hernández Elche, Spain
| | | |
Collapse
|
263
|
Sommakia S, Rickus JL, Otto KJ. Effects of adsorbed proteins, an antifouling agent and long-duration DC voltage pulses on the impedance of silicon-based neural microelectrodes. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2010; 2009:7139-42. [PMID: 19963693 DOI: 10.1109/iembs.2009.5332456] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The successful use of implantable neural microelectrodes as neuroprosthetic devices depends on the mitigation of the reactive tissue response of the brain. One of the factors affecting the ultimate severity of the reactive tissue response and the in vivo electrical properties of the microelectrodes is the initial adsorption of proteins onto the surface of the implanted microelectrodes. In this study we quantify the increase in microelectrode impedance magnitude at physiological frequencies following electrode immersion in a 10% bovine serum albumin (BSA) solution. We also demonstrate the efficacy of a common antifouling molecule, poly(ethylene glycol) (PEG), in preventing a significant increase in microelectrode impedance. In addition, we show the feasibility of using long-duration DC voltage pulses to remove adsorbed proteins from the microelectrode surface.
Collapse
Affiliation(s)
- Salah Sommakia
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47906, USA.
| | | | | |
Collapse
|
264
|
Winslow BD, Tresco PA. Quantitative analysis of the tissue response to chronically implanted microwire electrodes in rat cortex. Biomaterials 2010; 31:1558-67. [DOI: 10.1016/j.biomaterials.2009.11.049] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Accepted: 11/17/2009] [Indexed: 10/20/2022]
|
265
|
Leach JB, Achyuta AKH, Murthy SK. Bridging the Divide between Neuroprosthetic Design, Tissue Engineering and Neurobiology. FRONTIERS IN NEUROENGINEERING 2010; 2:18. [PMID: 20161810 PMCID: PMC2821180 DOI: 10.3389/neuro.16.018.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 12/28/2009] [Indexed: 11/19/2022]
Abstract
Neuroprosthetic devices have made a major impact in the treatment of a variety of disorders such as paralysis and stroke. However, a major impediment in the advancement of this technology is the challenge of maintaining device performance during chronic implantation (months to years) due to complex intrinsic host responses such as gliosis or glial scarring. The objective of this review is to bring together research communities in neurobiology, tissue engineering, and neuroprosthetics to address the major obstacles encountered in the translation of neuroprosthetics technology into long-term clinical use. This article draws connections between specific challenges faced by current neuroprosthetics technology and recent advances in the areas of nerve tissue engineering and neurobiology. Within the context of the device-nervous system interface and central nervous system implants, areas of synergistic opportunity are discussed, including platforms to present cells with multiple cues, controlled delivery of bioactive factors, three-dimensional constructs and in vitro models of gliosis and brain injury, nerve regeneration strategies, and neural stem/progenitor cell biology. Finally, recent insights gained from the fields of developmental neurobiology and cancer biology are discussed as examples of exciting new biological knowledge that may provide fresh inspiration toward novel technologies to address the complexities associated with long-term neuroprosthetic device performance.
Collapse
Affiliation(s)
- Jennie B. Leach
- Department of Chemical and Biochemical Engineering, University of MarylandBaltimore, MD, USA
| | | | - Shashi K. Murthy
- Department of Chemical Engineering, Northeastern UniversityBoston, MA, USA
| |
Collapse
|
266
|
Thaning EM, Asplund MLM, Nyberg TA, Inganäs OW, von Holst H. Stability of poly(3,4-ethylene dioxythiophene) materials intended for implants. J Biomed Mater Res B Appl Biomater 2010; 93:407-15. [DOI: 10.1002/jbm.b.31597] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
267
|
Frampton JP, Hynd MR, Shuler ML, Shain W. Effects of Glial Cells on Electrode Impedance Recorded from Neural Prosthetic Devices In Vitro. Ann Biomed Eng 2010; 38:1031-47. [DOI: 10.1007/s10439-010-9911-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
268
|
Minnikanti S, Pereira MGAG, Jaraiedi S, Jackson K, Costa-Neto CM, Li Q, Peixoto N. In vivo electrochemical characterization and inflammatory response of multiwalled carbon nanotube-based electrodes in rat hippocampus. J Neural Eng 2010; 7:16002. [PMID: 20054103 DOI: 10.1088/1741-2560/7/1/016002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Stimulating neural electrodes are required to deliver charge to an environment that presents itself as hostile. The electrodes need to maintain their electrical characteristics (charge and impedance) in vivo for a proper functioning of neural prostheses. Here we design implantable multi-walled carbon nanotubes coating for stainless steel substrate electrodes, targeted at wide frequency stimulation of deep brain structures. In well-controlled, low-frequency stimulation acute experiments, we show that multi-walled carbon nanotube electrodes maintain their charge storage capacity (CSC) and impedance in vivo. The difference in average CSCs (n = 4) between the in vivo (1.111 mC cm(-2)) and in vitro (1.008 mC cm(-2)) model was statistically insignificant (p > 0.05 or P-value = 0.715, two tailed). We also report on the transcription levels of the pro-inflammatory cytokine IL-1beta and TLR2 receptor as an immediate response to low-frequency stimulation using RT-PCR. We show here that the IL-1beta is part of the inflammatory response to low-frequency stimulation, but TLR2 is not significantly increased in stimulated tissue when compared to controls. The early stages of neuroinflammation due to mechanical and electrical trauma induced by implants can be better understood by detection of pro-inflammatory molecules rather than by histological studies. Tracking of such quantitative response profits from better analysis methods over several temporal and spatial scales. Our results concerning the evaluation of such inflammatory molecules revealed that transcripts for the cytokine IL-1beta are upregulated in response to low-frequency stimulation, whereas no modulation was observed for TLR2. This result indicates that the early response of the brain to mechanical trauma and low-frequency stimulation activates the IL-1beta signaling cascade but not that of TLR2.
Collapse
Affiliation(s)
- Saugandhika Minnikanti
- Department of Electrical and Computer Engineering, Neural Engineering Laboratory, George Mason University, Fairfax, VA 22030, USA
| | | | | | | | | | | | | |
Collapse
|
269
|
The Electrochemistry of Charge Injection at the Electrode/Tissue Interface. IMPLANTABLE NEURAL PROSTHESES 2 2010. [DOI: 10.1007/978-0-387-98120-8_4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
270
|
|
271
|
Intracranial electrode implantation produces regional neuroinflammation and memory deficits in rats. Exp Neurol 2009; 222:42-50. [PMID: 20026042 DOI: 10.1016/j.expneurol.2009.12.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 08/28/2009] [Accepted: 12/05/2009] [Indexed: 11/21/2022]
Abstract
Deep brain stimulation (DBS) is an established treatment for advanced Parkinson's disease (PD). The procedure entails intracranial implantation of an electrode in a specific brain structure followed by chronic stimulation. Although the beneficial effects of DBS on motor symptoms in PD are well known, it is often accompanied by cognitive impairments, the origin of which is not fully understood. To explore the possible contribution of the surgical procedure itself, we studied the effect of electrode implantation in the subthalamic nucleus (STN) on regional neuroinflammation and memory function in rats implanted bilaterally with stainless steel electrodes. Age-matched sham and intact rats were used as controls. Brains were removed 1 or 8 weeks post-implantation and processed for in vitro autoradiography with [(3)H]PK11195, an established marker of microglial activation. Memory function was assessed by the novel object recognition test (ORT) before surgery and 2 and 8 weeks after surgery. Electrode implantation produced region-dependent changes in ligand binding density in the implanted brains at 1 as well as 8 weeks post-implantation. Cortical regions showed more intense and widespread neuroinflammation than striatal or thalamic structures. Furthermore, implanted animals showed deficits in ORT performance 2 and 8 weeks post-implantation. Thus, electrode implantation resulted in a widespread and persistent neuroinflammation and sustained memory impairment. These results suggest that the insertion and continued presence of electrodes in the brain, even without stimulation, may lead to inflammation-mediated cognitive deficits in susceptible individuals, as observed in patients treated with DBS.
Collapse
|
272
|
Long-Term Recordings of Multiple, Single-Neurons for Clinical Applications: The Emerging Role of the Bioactive Microelectrode. MATERIALS 2009. [PMCID: PMC5525202 DOI: 10.3390/ma2041762] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In 1999 we reported an important demonstration of a working brain-machine interface (BMI), in which recordings from multiple, single neurons in sensorimotor cortical areas of rats were used to directly control a robotic arm to retrieve a water reward. Subsequent studies in monkeys, using a similar approach, demonstrated that primates can use a BMI device to control a cursor on a computer screen and a robotic arm. Recent studies in humans with spinal cord injuries have shown that recordings from multiple, single neurons can be used by the patient to control the cursor on a computer screen. The promise is that one day it will be possible to use these control signals from neurons to re-activate the patient’s own limbs. However, the ability to record from large populations of single neurons for long periods of time has been hampered because either the electrode itself fails or the immunological response in the tissue surrounding the microelectrode produces a glial scar, preventing single-neuron recording. While we have largely solved the problem of mechanical or electrical failure of the electrode itself, much less is known about the long term immunological response to implantation of a microelectrode, its effect on neuronal recordings and, of greatest importance, how it can be reduced to allow long term single neuron recording. This article reviews materials approaches to resolving the glial scar to improve the longevity of recordings. The work to date suggests that approaches utilizing bioactive interventions that attempt to alter the glial response and attract neurons to the recording site are likely to be the most successful. Importantly, measures of the glial scar alone are not sufficient to assess the effect of interventions. It is imperative that recordings of single neurons accompany any study of glial activation because, at this time, we do not know the precise relationship between glial activation and loss of neuronal recordings. Moreover, new approaches to immobilize bioactive molecules on microelectrode surfaces while maintaining their functionality may open new avenues for very long term single neuron recording. Finally, it is important to have quantitative measures of glial upregulation and neuronal activity in order to assess the relationship between the two. These types of studies will help rationalize the study of interventions to improve the longevity of recordings from microelectrodes.
Collapse
|
273
|
Purcell EK, Thompson DE, Ludwig KA, Kipke DR. Flavopiridol reduces the impedance of neural prostheses in vivo without affecting recording quality. J Neurosci Methods 2009; 183:149-57. [PMID: 19560490 DOI: 10.1016/j.jneumeth.2009.06.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 06/17/2009] [Accepted: 06/18/2009] [Indexed: 02/04/2023]
Abstract
We hypothesized that re-entry into the cell cycle may be associated with reactive gliosis surrounding neural prostheses, and that administration of a cell cycle inhibitor (flavopiridol) at the time of surgery would reduce this effect. We investigated the effects of flavopiridol on recording quality and impedance over a 28-day time period and conducted histology at 3 and 28 days post-implantation. Flavopiridol reduced the expression of a cell cycle protein (cyclin D1) in microglia surrounding probes at the 3-day time point. Impedance at 1 kHz was decreased by drug administration across the study period compared to vehicle controls. Correlations between recording (SNR, units) and impedance metrics revealed a small, but statistically significant, inverse relationship between these variables. However, the relationship between impedance and recording quality was not sufficiently strong for flavopiridol to result in an improvement in SNR or the number of units detected. Our data indicate that flavopiridol is an effective, easily administered treatment for reducing impedance in vivo, potentially through inhibiting microglial encapsulation of implanted devices. This strategy may be useful in stimulation applications, where reduced impedance is desirable for achieving activation thresholds and prolonging the lifetime of the implanted power supply. While improvements in recording quality were not observed, a combination of flavopiridol with a second strategy which enhances neuronal signal detection may enhance these results in future studies.
Collapse
Affiliation(s)
- Erin K Purcell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
274
|
Jaquins-Gerstl A, Michael AC. Comparison of the brain penetration injury associated with microdialysis and voltammetry. J Neurosci Methods 2009; 183:127-35. [PMID: 19559724 PMCID: PMC2743756 DOI: 10.1016/j.jneumeth.2009.06.023] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 10/20/2022]
Abstract
Emerging evidence suggests that differences between microdialysis- and voltammetry-based estimates of extracellular dopamine in the brain might originate in the different penetration injury associated with each technique. To address this issue in a direct fashion, microdialysis probes and voltammetric microelectrodes were implanted in the rat striatum for 1, 4, or 24 h. Tissues were perfused with a suspension of fluorescently labeled nanobeads to assess blood vessels near the implant. Tissue sections (30 microm) were labeled with antibodies for PECAM, an endothelial cell marker, or GFAP, a glial marker. In non-implanted control tissue, blood vessels were reliably double-labeled with nanobeads and antiPECAM. Tissue near microdialysis probe tracks exhibited ischemia in the form of PECAM immunoreactive blood vessels devoid of nanobeads. Ischemia was most apparent after the 4-h implants. Probe tracks were surrounded by endothelial cell debris, which appeared as a diffuse halo of PECAM immunoreactivity. The halo intensity decreased with implant duration, indicative of an active wound-healing process. Consistent with this, after 24-h implants, the probe tracks were surrounded by hyperplasic and hypertrophic glia and glial processes were extending towards, and engulfing, the track. Carbon fiber microelectrodes produced a diffuse disruption of nanobead labeling but no focal disruption of blood vessels, no PECAM immunoreactive halo, and no glial activation. These findings illuminate the differences between the extent and nature of the penetration injuries associated with microdialysis and voltammetry.
Collapse
Affiliation(s)
- Andrea Jaquins-Gerstl
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Adrian C. Michael
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, United States
| |
Collapse
|
275
|
Tran PA, Zhang L, Webster TJ. Carbon nanofibers and carbon nanotubes in regenerative medicine. Adv Drug Deliv Rev 2009; 61:1097-114. [PMID: 19647768 DOI: 10.1016/j.addr.2009.07.010] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Accepted: 07/16/2009] [Indexed: 12/22/2022]
Abstract
Carbon nanotubes and carbon nanofibers have long been investigated for applications in composite structural materials, semiconductor devices, and sensors. With the recent well-documented ability to chemically modify nanofibrous carbon materials to improve their solubility and biocompatibility properties: a whole new class of bioactive carbon nanostructures has been created for biological applications. This review focuses on the latest applications of carbon nanofibers and carbon nanotubes in regenerative medicine.
Collapse
Affiliation(s)
- Phong A Tran
- Physics Department, Brown University, Providence, RI 02912, USA
| | | | | |
Collapse
|
276
|
Abidian MR, Ludwig KA, Marzullo TC, Martin DC, Kipke DR. Interfacing Conducting Polymer Nanotubes with the Central Nervous System: Chronic Neural Recording using Poly(3,4-ethylenedioxythiophene) Nanotubes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2009; 21:3764-3770. [PMID: 26345408 PMCID: PMC4559350 DOI: 10.1002/adma.200900887] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Mohammad Reza Abidian
- Department of Biomedical Engineering, The University of Michigan, 1101 Beal Ave., Ann Arbor, MI 48109 (USA)
| | - Kip A. Ludwig
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI 48109 (USA)
| | | | | | - Daryl R. Kipke
- Department of Biomedical Engineering, The University of Michigan, 1101 Beal Ave., Ann Arbor, MI 48109 (USA)
| |
Collapse
|
277
|
Lo CT, Van Tassel PR, Saltzman WM. Simultaneous release of multiple molecules from poly(lactide-co-glycolide) nanoparticles assembled onto medical devices. Biomaterials 2009; 30:4889-97. [PMID: 19592089 PMCID: PMC2742909 DOI: 10.1016/j.biomaterials.2009.05.074] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 05/21/2009] [Indexed: 10/20/2022]
Abstract
Cell and tissue responses to implanted biomaterials often limit their effectiveness and lifetime. This is particularly true for materials implanted into the brain. We present here a new approach for the modification of materials to enable release of multiple agents, which might be useful in modulating tissue responses, without changing the properties of the underlying material, in this case, a silicon probe. Poly(lactide-co-glycolide) nanoparticles (NPs) were assembled onto silicon probe surfaces by electrostatic interactions. Charged NPs were fabricated by altering the properties of the surfactant. NPs formed with poly(ethylene-alt-maleic anhydride) (PEMA) were strongly negatively charged; these NPs assembled onto probes best when suspended at nearly physiological conditions (surface density approximately 83,600+/-3000 particles/mm(2)). The percentage of surface area coverage by the NPs was estimated to be approximately 13% and was maintained over two weeks during constant exposure to PBS. Multiple fluorescent NP populations were attached to the same probe to allow visualization of simultaneous delivery of multiple agents by fluorescence microscopy. Release from NP coatings was reproducible and controllable. The distinct release profiles of each agent from the coatings were preserved upon attachment to the surfaces. The unique feature of this new system is that NPs encapsulating various molecules (i.e. drugs, proteins, or DNA) can be fabricated separately, in advance, and simply mixed prior to attachment. The versatility of this delivery system, therefore, makes it suitable for many applications.
Collapse
Affiliation(s)
- Catherine T. Lo
- Department of Biomedical Engineering, Yale University, Malone Engineering Center, Room 414, 55 Prospect Street, New Haven, CT 06511, USA
| | - Paul R. Van Tassel
- Department of Chemical Engineering, Yale University, Mason Laboratory, Room 307A, 9 Hillhouse Avenue, New Haven, CT 06511, USA
| | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, Malone Engineering Center, Room 414, 55 Prospect Street, New Haven, CT 06511, USA
| |
Collapse
|
278
|
Hascup ER, af Bjerkén S, Hascup KN, Pomerleau F, Huettl P, Strömberg I, Gerhardt GA. Histological studies of the effects of chronic implantation of ceramic-based microelectrode arrays and microdialysis probes in rat prefrontal cortex. Brain Res 2009; 1291:12-20. [PMID: 19577548 PMCID: PMC2980830 DOI: 10.1016/j.brainres.2009.06.084] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 06/23/2009] [Accepted: 06/24/2009] [Indexed: 01/31/2023]
Abstract
Chronic implantation of neurotransmitter measuring devices is essential for awake, behavioral studies occurring over multiple days. Little is known regarding the effects of long term implantation on surrounding brain parenchyma and the resulting alterations in the functional properties of this tissue. We examined the extent of tissue damage produced by chronic implantation of either ceramic microelectrode arrays (MEAs) or microdialysis probes. Histological studies were carried out on fixed tissues using stains for neurons (cresyl violet), astrocytes (GFAP), microglia (Iba1), glutamatergic nerve fibers (VGLUT1), and the blood-brain barrier (SMI-71). Nissl staining showed pronounced tissue body loss with microdialysis implants compared to MEAs. The MEAs produced mild gliosis extending 50-100 microm from the tracks, with a significant change in the affected areas starting at 3 days. By contrast, the microdialysis probes produced gliosis extending 200-300 microm from the track, which was significant at 3 and 7 days. Markers for microglia and glutamatergic fibers supported that the MEAs produce minimal damage with significant changes occurring only at 3 and 7 days that return to control levels by 1 month. SMI-71 staining supported the integrity of the blood-brain barrier out to 1 week for both the microdialysis probes and the MEAs. This data support that the ceramic MEA's small size and biocompatibility are necessary to accurately measure neurotransmitter levels in the intact brain. The minimal invasiveness of the MEAs reduce tissue loss, allowing for long term (>6 month) electrochemical and electrophysiological monitoring of brain activity.
Collapse
Affiliation(s)
- Erin R Hascup
- Anatomy and Neurobiology, Morris K. Udall Parkinson's Disease Research Center of Excellence, Center for Microelectrode Technology, University of Kentucky, College of Medicine, Lexington, KY, 40536-0098 USA.
| | | | | | | | | | | | | |
Collapse
|
279
|
Foley CP, Nishimura N, Neeves KB, Schaffer CB, Olbricht WL. Flexible microfluidic devices supported by biodegradable insertion scaffolds for convection-enhanced neural drug delivery. Biomed Microdevices 2009; 11:915-24. [PMID: 19353271 DOI: 10.1007/s10544-009-9308-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Convection enhanced delivery (CED) can improve the spatial distribution of drugs delivered directly to the brain. In CED, drugs are infused locally into tissue through a needle or catheter inserted into brain parenchyma. Transport of the infused material is dominated by convection, which enhances drug penetration into tissue compared with diffusion mediated delivery. We have fabricated and characterized an implantable microfluidic device for chronic convection enhanced delivery protocols. The device consists of a flexible parylene-C microfluidic channel that is supported during its insertion into tissue by a biodegradable poly(DL-lactide-co-glycolide) scaffold. The scaffold is designed to enable tissue penetration and then erode over time, leaving only the flexible channel implanted in the tissue. The device was able to reproducibly inject fluid into neural tissue in acute experiments with final infusate distributions that closely approximate delivery from an ideal point source. This system shows promise as a tool for chronic CED protocols.
Collapse
Affiliation(s)
- Conor P Foley
- School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | |
Collapse
|
280
|
McConnell GC, Butera RJ, Bellamkonda RV. Bioimpedance modeling to monitor astrocytic response to chronically implanted electrodes. J Neural Eng 2009; 6:055005. [DOI: 10.1088/1741-2560/6/5/055005] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
281
|
McConnell GC, Rees HD, Levey AI, Gutekunst CA, Gross RE, Bellamkonda RV. Implanted neural electrodes cause chronic, local inflammation that is correlated with local neurodegeneration. J Neural Eng 2009; 6:056003. [PMID: 19700815 DOI: 10.1088/1741-2560/6/5/056003] [Citation(s) in RCA: 316] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Prosthetic devices that are controlled by intracortical electrodes recording one's 'thoughts' are a reality today, and no longer merely in the realm of science fiction. However, widespread clinical use of implanted electrodes is hampered by a lack of reliability in chronic recordings, independent of the type of electrodes used. One major hypothesis has been that astroglial scar electrically impedes the electrodes. However, there is a temporal discrepancy between stabilization of scar's electrical properties and recording failure with recording failure lagging by 1 month. In this study, we test a possible explanation for this discrepancy: the hypothesis that chronic inflammation, due to the persistent presence of the electrode, causes a local neurodegenerative state in the immediate vicinity of the electrode. Through modulation of chronic inflammation via stab wound, electrode geometry and age-matched control, we found that after 16 weeks, animals with an increased level of chronic inflammation were associated with increased neuronal and dendritic, but not axonal, loss. We observed increased neuronal and dendritic loss 16 weeks after implantation compared to 8 weeks after implantation, suggesting that the local neurodegenerative state is progressive. After 16 weeks, we observed axonal pathology in the form of hyperphosphorylation of the protein tau in the immediate vicinity of the microelectrodes (as observed in Alzheimer's disease and other tauopathies). The results of this study suggest that a local, late onset neurodegenerative disease-like state surrounds the chronic electrodes and is a potential cause for chronic recording failure. These results also inform strategies to enhance our capability to attain reliable long-term recordings from implantable electrodes in the CNS.
Collapse
Affiliation(s)
- George C McConnell
- Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, 30332, USA
| | | | | | | | | | | |
Collapse
|
282
|
Abstract
Repositioning microelectrodes post-implantation is emerging as a promising approach to achieve long-term reliability in single neuronal recordings. The main goal of this study was to (a) assess glial reaction in response to movement of microelectrodes in the brain post-implantation and (b) determine an optimal window of time post-implantation when movement of microelectrodes within the brain would result in minimal glial reaction. Eleven Sprague-Dawley rats were implanted with two microelectrodes each that could be moved in vivo post-implantation. Three cohorts were investigated: (1) microelectrode moved at day 2 (n = 4 animals), (2) microelectrode moved at day 14 (n = 5 animals) and (3) microelectrode moved at day 28 (n = 2 animals). Histological evaluation was performed in cohorts 1-3 at four-week post-movement (30 days, 42 days and 56 days post-implantation, respectively). In addition, five control animals were implanted with microelectrodes that were not moved. Control animals were implanted for (1) 30 days (n = 1), (2) 42 days (n = 2) and (3) 56 days (n = 2) prior to histological evaluation. Quantitative assessment of glial fibrillary acidic protein (GFAP) around the tip of the microelectrodes demonstrated that GFAP levels were similar around microelectrodes moved at day 2 when compared to the 30-day controls. However, GFAP expression levels around microelectrode tips that moved at day 14 and day 28 were significantly less than those around control microelectrodes implanted for 42 and 56 days, respectively. Therefore, we conclude that moving microelectrodes after implantation is a viable strategy that does not result in any additional damage to brain tissue. Further, moving the microelectrode downwards after 14 days of implantation may actually reduce the levels of GFAP expression around the tips of the microelectrodes in the long term.
Collapse
Affiliation(s)
- Paula Stice
- Department of Bioengineering, Arizona State University, Tempe, AZ 85287-9709, USA
| | | |
Collapse
|
283
|
Chen YY, Lai HY, Lin SH, Cho CW, Chao WH, Liao CH, Tsang S, Chen YF, Lin SY. Design and fabrication of a polyimide-based microelectrode array: Application in neural recording and repeatable electrolytic lesion in rat brain. J Neurosci Methods 2009; 182:6-16. [DOI: 10.1016/j.jneumeth.2009.05.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 04/24/2009] [Accepted: 05/14/2009] [Indexed: 11/29/2022]
|
284
|
Grill WM, Norman SE, Bellamkonda RV. Implanted Neural Interfaces: Biochallenges and Engineered Solutions. Annu Rev Biomed Eng 2009; 11:1-24. [DOI: 10.1146/annurev-bioeng-061008-124927] [Citation(s) in RCA: 368] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Warren M. Grill
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708-0281;
| | - Sharon E. Norman
- Bioengineering Program Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Ravi V. Bellamkonda
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, Georgia 30332
| |
Collapse
|
285
|
Polikov VS, Su EC, Ball MA, Hong JS, Reichert WM. Control protocol for robust in vitro glial scar formation around microwires: essential roles of bFGF and serum in gliosis. J Neurosci Methods 2009; 181:170-7. [PMID: 19447137 PMCID: PMC2722030 DOI: 10.1016/j.jneumeth.2009.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 04/30/2009] [Accepted: 05/03/2009] [Indexed: 10/20/2022]
Abstract
Previously, we reported an in vitro cell culture model that recreates many of the hallmarks of glial scarring around electrodes used for recording in the brain; however, the model lacked the reproducibility necessary to establish a useful characterization tool. This methods paper describes a protocol, modeled on protocols typically used to culture neural stem/precursor cells, that generates a predictable positive control of an intense scarring reaction. Six independent cell culture variables (growth media, seeding density, bFGF addition day, serum concentration in treatment media, treatment day, and duration of culture) were varied systematically and the resulting scars were quantified. The following conditions were found to give the highest level of scarring: Neurobasal medium supplemented with B27, 10% fetal bovine serum at treatment, 10 ng/ml b-FGF addition at seeding and at treatment, treatment at least 6 days after seeding and scar growth of at least 5 days. Seeding density did not affect scarring as long as at least 500,000 cells were seeded per well, but appropriate media, bFGF, and serum were essential for significant scar formation-insights that help validate the in vitro-based approach to understanding glial scarring. With the control protocol developed in this study producing a strong, reproducible glial scarring positive control with every dissection, this culture model is suitable for the in vitro study of the mechanisms behind glial scarring and neuroelectrode failure.
Collapse
Affiliation(s)
- Vadim S. Polikov
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - Eric C. Su
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - Matthew A. Ball
- Department of Biomedical Engineering, Duke University, Durham, NC 27708
| | - Jau-Shyong Hong
- Neuropharmacology Group, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, North Carolina 27709
| | - William M. Reichert
- Department of Biomedical Engineering, Box 90281, Duke University, Durham, NC 27708-0281, Office: 919-660-5151, F AX: 919-660-5362, E-mail:
| |
Collapse
|
286
|
A decrease in EEG energy accompanies anti-epileptic drug taper during intracranial monitoring. Epilepsy Res 2009; 86:153-62. [PMID: 19632096 DOI: 10.1016/j.eplepsyres.2009.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Revised: 05/29/2009] [Accepted: 06/07/2009] [Indexed: 11/22/2022]
Abstract
OBJECTIVE During intracranial EEG (icEEG) monitoring the likelihood of observing a seizure is increased by tapering anti-epileptic drugs (AEDs). Presumably AED taper results in an increase in cortical excitation which in turn promotes seizure emergence. We measured change in signal energy of icEEGs in response to AED taper to quantify changes in excitation which accompany the increased propensity for seizures. METHODS Twelve consecutive adult patients who completed intracranial monitoring were studied. Two icEEG epochs from before and after AED taper, each 1h in duration, during wake, matched by time-of-day and removed from seizures were selected for each patient. Teager energy, a frequency weighted measure of signal energy, was estimated for both the seizure onset region as well as all other brain areas monitored. RESULTS Considerable changes in Teager energy, evaluated at a 1-h time-resolution, occur during intracranial monitoring. The most dominant trend is a decrease to lower values than those when the patient is on AEDs. A decrease of 35% was observed for both all the brain areas monitored and the seizure onset region. CONCLUSIONS A decrease in signal energy occurs during intracranial EEG monitoring, possibly accompanying AED taper. If the decrease is due to AED taper this would suggest that AEDs prevent seizures in ways other than reduction of cortical excitation and seizure generation may be influenced by factors other than poorly regulated cortical excitation.
Collapse
|
287
|
Lü X, Cui W, Huang Y, Zhao Y, Wang Z. Surface modification on silicon with chitosan and biological research. Biomed Mater 2009; 4:044103. [DOI: 10.1088/1748-6041/4/4/044103] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
288
|
Mercanzini A, Colin P, Bensadoun JC, Bertsch A, Renaud P. In Vivo Electrical Impedance Spectroscopy of Tissue Reaction to Microelectrode Arrays. IEEE Trans Biomed Eng 2009; 56:1909-18. [DOI: 10.1109/tbme.2009.2018457] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
289
|
Wolf MT, Cham JG, Branchaud EA, Mulliken GH, Burdick JW, Andersen RA. A Robotic Neural Interface for Autonomous Positioning of Extracellular Recording Electrodes. Int J Rob Res 2009. [DOI: 10.1177/0278364908103788] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this paper we describe a set of algorithms and a novel miniature device that together can autonomously position electrodes in neural tissue to obtain high-quality extracellular recordings. This robotic system moves each electrode to detect the signals of individual neurons, optimize the signal quality of a target neuron, and then maintain this signal over time. Such neuronal signals provide the key inputs for emerging neuroprosthetic medical devices and serve as the foundation of basic neuroscientific and medical research. Experimental results from extensive use of the robotic electrodes in macaque parietal cortex are presented to validate the method and to quantify its effectiveness.
Collapse
Affiliation(s)
- Michael T. Wolf
- Department of Mechanical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91106, USA,
| | - Jorge G. Cham
- Department of Mechanical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91106, USA
| | - Edward A. Branchaud
- Department of Mechanical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91106, USA
| | - Grant H. Mulliken
- Department of Mechanical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91106, USA
| | - Joel W. Burdick
- Department of Mechanical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91106, USA
| | - Richard A. Andersen
- Division of Biology California Institute of Technology, 1200 East California Boulevard, Pasadena, CA 91106, USA
| |
Collapse
|
290
|
Ward MP, Rajdev P, Ellison C, Irazoqui PP. Toward a comparison of microelectrodes for acute and chronic recordings. Brain Res 2009; 1282:183-200. [PMID: 19486899 DOI: 10.1016/j.brainres.2009.05.052] [Citation(s) in RCA: 202] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2009] [Revised: 04/27/2009] [Accepted: 05/20/2009] [Indexed: 11/28/2022]
Abstract
Several variations of microelectrode arrays are used to record and stimulate intracortical neuronal activity. Bypassing the immune response to maintain a stable recording interface remains a challenge. Companies and researchers are continuously altering the material compositions and geometries of the arrays in order to discover a combination that allows for a chronic and stable electrode-tissue interface. From this interface, they wish to obtain consistent quality recordings and a stable, low impedance pathway for charge injection over extended periods of time. Despite numerous efforts, no microelectrode array design has managed to evade the host immune response and remain fully functional. This study is an initial effort comparing several microelectrode arrays with fundamentally different configurations for use in an implantable epilepsy prosthesis. Specifically, NeuroNexus (Michigan) probes, Cyberkinetics (Utah) Silicon and Iridium Oxide arrays, ceramic-based thin-film microelectrode arrays (Drexel), and Tucker-Davis Technologies (TDT) microwire arrays are evaluated over a 31-day period in an animal model. Microelectrodes are compared in implanted rats through impedance, charge capacity, signal-to-noise ratio, recording stability, and elicited immune response. Results suggest significant variability within and between microelectrode types with no clear superior array. Some applications for the microelectrode arrays are suggested based on data collected throughout the longitudinal study. Additionally, specific limitations of assaying biological phenomena and comparing fundamentally different microelectrode arrays in a highly variable system are discussed with suggestions on how to improve the reliability of observed results and steps needed to develop a more standardized microelectrode design.
Collapse
Affiliation(s)
- Matthew P Ward
- Weldon School of Biomedical Engineering, Purdue University, MJIS 2083, 206 S Martin Jischke Drive West Lafayette, IN 47907 USA.
| | | | | | | |
Collapse
|
291
|
Abstract
Neural prosthetics capable of recording or stimulating neuronal activity may restore function for patients with motor and sensory deficits resulting from injury or degenerative disease. However, overcoming inconsistent recording quality and stability in chronic applications remains a significant challenge. A likely reason for this is the reactive tissue response to the devices following implantation into the brain, which is characterized by neuronal loss and glial encapsulation. We have developed a neural stem cell-seeded probe to facilitate integration of a synthetic prosthesis with the surrounding brain tissue. We fabricated parylene devices that include an open well seeded with neural stem cells encapsulated in an alginate hydrogel scaffold. Quantitative and qualitative data describing the distribution of neuronal, glial, and progenitor cells surrounding seeded and control devices are reported over four time points spanning 3 months. Neuronal loss and glial encapsulation associated with cell-seeded probes were mitigated during the initial week of implantation and exacerbated by 6 weeks post-insertion compared to control conditions. We hypothesize that graft cells secrete neuroprotective and neurotrophic factors that effect the desired healing response early in the study, with subsequent cell death and scaffold degradation accounting for a reversal of these results later. Applications of this biohybrid technology include future long-term neural recording and sensing studies.
Collapse
Affiliation(s)
- E K Purcell
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Ave., Ann Arbor, MI 48109-2099, USA.
| | | | | | | |
Collapse
|
292
|
Cottaris NP, Elfar SD. Assessing the efficacy of visual prostheses by decoding ms-LFPs: application to retinal implants. J Neural Eng 2009; 6:026007. [DOI: 10.1088/1741-2560/6/2/026007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
293
|
Wester BA, Lee RH, LaPlaca MC. Development and characterization of in vivo flexible electrodes compatible with large tissue displacements. J Neural Eng 2009; 6:024002. [PMID: 19255461 DOI: 10.1088/1741-2560/6/2/024002] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Electrical activity is the ultimate functional measure of neuronal tissue and recording that activity remains a key technical challenge in neuroscience. The mechanical mismatch between rigid electrodes and compliant brain tissue is a critical limitation in applications where movement is an inherent component. An electrode that permits recording of neural activity, while minimizing tissue disruption, is beneficial for applications that encompass both normal physiological movements and those which require consistent recording during large tissue displacements. In order to test the extreme of this range of movement, flexible electrodes were developed to record activity during and immediately following cortical impact in the rat. Photolithography techniques were used to fabricate flexible electrodes that were readily insertable into the brain using a parylene C base and gold conduction lines and contact pads, permitting custom geometry. We found that this electrode configuration retained mechanical and electrical integrity following both durability studies and large movements within the cortex. This novel flexible electrode configuration provides a novel platform for experimentally examining neuronal activity during a range of brain movements.
Collapse
Affiliation(s)
- B A Wester
- Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr., Atlanta, GA 30332-0535, USA.
| | | | | |
Collapse
|
294
|
Ludwig KA, Miriani RM, Langhals NB, Joseph MD, Anderson DJ, Kipke DR. Using a common average reference to improve cortical neuron recordings from microelectrode arrays. J Neurophysiol 2008; 101:1679-89. [PMID: 19109453 DOI: 10.1152/jn.90989.2008] [Citation(s) in RCA: 261] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this study, we propose and evaluate a technique known as common average referencing (CAR) to generate a more ideal reference electrode for microelectrode recordings. CAR is a computationally simple technique, and therefore amenable to both on-chip and real-time applications. CAR is commonly used in EEG, where it is necessary to identify small signal sources in very noisy recordings. To study the efficacy of common average referencing, we compared CAR to both referencing with a stainless steel bone-screw and a single microelectrode site. Data consisted of in vivo chronic recordings in anesthetized Sprague-Dawley rats drawn from prior studies, as well as previously unpublished data. By combining the data from multiple studies, we generated and analyzed one of the more comprehensive chronic neural recording datasets to date. Reference types were compared in terms of noise level, signal-to-noise ratio, and number of neurons recorded across days. Common average referencing was found to drastically outperform standard types of electrical referencing, reducing noise by >30%. As a result of the reduced noise floor, arrays referenced to a CAR yielded almost 60% more discernible neural units than traditional methods of electrical referencing. CAR should impart similar benefits to other microelectrode recording technologies-for example, chemical sensing-where similar differential recording concepts apply. In addition, we provide a mathematical justification for CAR using Gauss-Markov theorem and therefore help place the application of CAR into a theoretical context.
Collapse
Affiliation(s)
- Kip A Ludwig
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | | | | | | | |
Collapse
|
295
|
Jhaveri SJ, Hynd MR, Dowell-Mesfin N, Turner JN, Shain W, Ober CK. Release of Nerve Growth Factor from HEMA Hydrogel-Coated Substrates and Its Effect on the Differentiation of Neural Cells. Biomacromolecules 2008; 10:174-83. [DOI: 10.1021/bm801101e] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Shalin J. Jhaveri
- Department of Materials Science and Engineering and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853 and Wadsworth Center, NYS Department of Health, Albany, New York 12201
| | - Matthew R. Hynd
- Department of Materials Science and Engineering and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853 and Wadsworth Center, NYS Department of Health, Albany, New York 12201
| | - Natalie Dowell-Mesfin
- Department of Materials Science and Engineering and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853 and Wadsworth Center, NYS Department of Health, Albany, New York 12201
| | - James N. Turner
- Department of Materials Science and Engineering and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853 and Wadsworth Center, NYS Department of Health, Albany, New York 12201
| | - William Shain
- Department of Materials Science and Engineering and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853 and Wadsworth Center, NYS Department of Health, Albany, New York 12201
| | - Christopher K. Ober
- Department of Materials Science and Engineering and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853 and Wadsworth Center, NYS Department of Health, Albany, New York 12201
| |
Collapse
|
296
|
Abstract
Biomaterials are widely used to help treat neurological disorders and/or improve functional recovery in the central nervous system (CNS). This article reviews the application of biomaterials in (i) shunting systems for hydrocephalus, (ii) cortical neural prosthetics, (iii) drug delivery in the CNS, (iv) hydrogel scaffolds for CNS repair, and (v) neural stem cell encapsulation for neurotrauma. The biological and material requirements for the biomaterials in these applications are discussed. The difficulties that the biomaterials might face in each application and the possible solutions are also reviewed in this article.
Collapse
Affiliation(s)
- Yinghui Zhong
- Neurological Biomaterials and Therapeutics, Laboratory for Neuroengineering, Wallace H Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA 30332, USA
| | | |
Collapse
|
297
|
Abstract
Electrical stimulation of nerve tissue and recording of neural electrical activity are the basis of emerging prostheses and treatments for spinal cord injury, stroke, sensory deficits, and neurological disorders. An understanding of the electrochemical mechanisms underlying the behavior of neural stimulation and recording electrodes is important for the development of chronically implanted devices, particularly those employing large numbers of microelectrodes. For stimulation, materials that support charge injection by capacitive and faradaic mechanisms are available. These include titanium nitride, platinum, and iridium oxide, each with certain advantages and limitations. The use of charge-balanced waveforms and maximum electrochemical potential excursions as criteria for reversible charge injection with these electrode materials are described and critiqued. Techniques for characterizing electrochemical properties relevant to stimulation and recording are described with examples of differences in the in vitro and in vivo response of electrodes.
Collapse
|
298
|
Retterer ST, Smith KL, Bjornsson CS, Turner JN, Isaacson MS, Shain W. Constant pressure fluid infusion into rat neocortex from implantable microfluidic devices. J Neural Eng 2008; 5:385-91. [PMID: 18827310 DOI: 10.1088/1741-2560/5/4/003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Implantable electrode arrays capable of recording and stimulating neural activity with high spatial and temporal resolution will provide a foundation for future brain computer interface technology. Currently, their clinical impact has been curtailed by a general lack of functional stability, which can be attributed to the acute and chronic reactive tissue responses to devices implanted in the brain. Control of the tissue environment surrounding implanted devices through local drug delivery could significantly alter both the acute and chronic reactive responses, and thus enhance device stability. Here, we characterize pressure-mediated release of test compounds into rat cortex using an implantable microfluidic platform. A fixed volume of fluorescent cell marker cocktail was delivered using constant pressure infusion at reservoir backpressures of 0, 5 and 10 psi. Affected tissue volumes were imaged and analyzed using epifluorescence and confocal microscropies and quantitative image analysis techniques. The addressable tissue volume for the 5 and 10 psi infusions, defined by fluorescent staining with Hoescht 33342 dye, was significantly larger than the tissue volume addressed by simple diffusion (0 psi) and the tissue volume exhibiting insertion-related cell damage (stained by propidium iodide). The results demonstrate the potential for using constant pressure infusion to address relevant tissue volumes with appropriate pharmacologies to alleviate reactive biological responses around inserted neuroprosthetic devices.
Collapse
Affiliation(s)
- S T Retterer
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | | | | | | | | | | |
Collapse
|
299
|
Paralikar KJ, Clement RS. Collagenase-Aided Intracortical Microelectrode Array Insertion: Effects on Insertion Force and Recording Performance. IEEE Trans Biomed Eng 2008; 55:2258-67. [DOI: 10.1109/tbme.2008.923150] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
300
|
Azemi E, Stauffer WR, Gostock MS, Lagenaur CF, Cui XT. Surface immobilization of neural adhesion molecule L1 for improving the biocompatibility of chronic neural probes: In vitro characterization. Acta Biomater 2008; 4:1208-17. [PMID: 18420473 DOI: 10.1016/j.actbio.2008.02.028] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Revised: 12/24/2007] [Accepted: 02/26/2008] [Indexed: 12/12/2022]
Abstract
Silicon-based implantable neural electrode arrays are known to experience failure during long-term recording, partially due to host tissue responses. Surface modification and immobilization of biomolecules may provide a means to improve their biocompatibility and integration within the host brain tissue. Previously, the laminin biomolecule or laminin fragments have been used to modify the neural probe's silicon surface to promote neuronal attachment and growth. Here we report the successful immobilization of the L1 biomolecule on a silicon surface. L1 is a neuronal adhesion molecule that can specifically promote neurite outgrowth and neuronal survival. Silane chemistry and the heterobifunctional coupling agent 4-maleimidobutyric acid N-hydroxysuccinimide ester (GMBS) were used to covalently bind these two biomolecules onto the surface of silicon dioxide wafers, which mimic the surface of silicon-based implantable neural probes. After covalent binding of the biomolecules, polyethylene glycol (PEG)-NH(2) was used to cap the unreacted GMBS groups. Surface immobilization was verified by goniometry, dual polarization interferometry, and immunostaining techniques. Primary murine neurons or astrocytes were used to evaluate the modified silicon surfaces. Both L1- and laminin-modified surfaces promoted neuronal attachment, while the L1-modified surface demonstrated significantly enhanced levels of neurite outgrowth (p<0.05). In addition, the laminin-modified surface promoted astrocyte attachment, while the L1-modified surface showed significantly reduced levels of astrocyte attachment relative to the laminin-modified surface and other controls (p<0.05). These results demonstrate the ability of the L1-immobilized surface to specifically promote neuronal growth and neurite extension, while inhibiting the attachment of astrocytes, one of the main cellular components of the glial sheath. Such unique properties present vast potentials to improve the biocompatibility and chronic recording performance of neural probes.
Collapse
|