1
|
Li F, Gallego J, Tirko NN, Greaser J, Bashe D, Patel R, Shaker E, Van Valkenburg GE, Alsubhi AS, Wellman S, Singh V, Padilla CG, Gheres KW, Broussard JI, Bagwell R, Mulvihill M, Kozai TDY. Low-intensity pulsed ultrasound stimulation (LIPUS) modulates microglial activation following intracortical microelectrode implantation. Nat Commun 2024; 15:5512. [PMID: 38951525 PMCID: PMC11217463 DOI: 10.1038/s41467-024-49709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
Microglia are important players in surveillance and repair of the brain. Implanting an electrode into the cortex activates microglia, produces an inflammatory cascade, triggers the foreign body response, and opens the blood-brain barrier. These changes can impede intracortical brain-computer interfaces performance. Using two-photon imaging of implanted microelectrodes, we test the hypothesis that low-intensity pulsed ultrasound stimulation can reduce microglia-mediated neuroinflammation following the implantation of microelectrodes. In the first week of treatment, we found that low-intensity pulsed ultrasound stimulation increased microglia migration speed by 128%, enhanced microglia expansion area by 109%, and a reduction in microglial activation by 17%, indicating improved tissue healing and surveillance. Microglial coverage of the microelectrode was reduced by 50% and astrocytic scarring by 36% resulting in an increase in recording performance at chronic time. The data indicate that low-intensity pulsed ultrasound stimulation helps reduce the foreign body response around chronic intracortical microelectrodes.
Collapse
Affiliation(s)
- Fan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- Computational Modeling and Simulation PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jazlyn Gallego
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Natasha N Tirko
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | | | - Derek Bashe
- Washington University in St. Louis, St. Louis, MO, USA
| | - Rudra Patel
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Shaker
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | - Vanshika Singh
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Camila Garcia Padilla
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | | | | | | | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Cui J, Mivalt F, Sladky V, Kim J, Richner TJ, Lundstrom BN, Van Gompel JJ, Wang HL, Miller KJ, Gregg N, Wu LJ, Denison T, Winter B, Brinkmann BH, Kremen V, Worrell GA. Acute to long-term characteristics of impedance recordings during neurostimulation in humans. J Neural Eng 2024; 21:10.1088/1741-2552/ad3416. [PMID: 38484397 PMCID: PMC11044203 DOI: 10.1088/1741-2552/ad3416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Objective.This study aims to characterize the time course of impedance, a crucial electrophysiological property of brain tissue, in the human thalamus (THL), amygdala-hippocampus, and posterior hippocampus over an extended period.Approach.Impedance was periodically sampled every 5-15 min over several months in five subjects with drug-resistant epilepsy using an investigational neuromodulation device. Initially, we employed descriptive piecewise and continuous mathematical models to characterize the impedance response for approximately three weeks post-electrode implantation. We then explored the temporal dynamics of impedance during periods when electrical stimulation was temporarily halted, observing a monotonic increase (rebound) in impedance before it stabilized at a higher value. Lastly, we assessed the stability of amplitude and phase over the 24 h impedance cycle throughout the multi-month recording.Main results.Immediately post-implantation, the impedance decreased, reaching a minimum value in all brain regions within approximately two days, and then increased monotonically over about 14 d to a stable value. The models accounted for the variance in short-term impedance changes. Notably, the minimum impedance of the THL in the most epileptogenic hemisphere was significantly lower than in other regions. During the gaps in electrical stimulation, the impedance rebound decreased over time and stabilized around 200 days post-implant, likely indicative of the foreign body response and fibrous tissue encapsulation around the electrodes. The amplitude and phase of the 24 h impedance oscillation remained stable throughout the multi-month recording, with circadian variation in impedance dominating the long-term measures.Significance.Our findings illustrate the complex temporal dynamics of impedance in implanted electrodes and the impact of electrical stimulation. We discuss these dynamics in the context of the known biological foreign body response of the brain to implanted electrodes. The data suggest that the temporal dynamics of impedance are dependent on the anatomical location and tissue epileptogenicity. These insights may offer additional guidance for the delivery of therapeutic stimulation at various time points post-implantation for neuromodulation therapy.
Collapse
Affiliation(s)
- Jie Cui
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Mayo College of Medicine and Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Filip Mivalt
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Vladimir Sladky
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| | - Jiwon Kim
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | - Hai-long Wang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kai J. Miller
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Nicholas Gregg
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Long Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy Denison
- Department of Engineering Science, University of Oxford; MRC Brain Network Dynamics Unit, University of Oxford, OX3 7DQ UK
| | - Bailey Winter
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo College of Medicine and Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Benjamin H. Brinkmann
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Vaclav Kremen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Czech Institute of Informatics, Robotics, and Cybernetics, Czech Technical University, Prague, Czech Republic
| | - Gregory A. Worrell
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
3
|
Cui J, Mivalt F, Sladky V, Kim J, Richner TJ, Lundstrom BN, Van Gompel JJ, Wang HL, Miller KJ, Gregg N, Wu LJ, Denison T, Winter B, Brinkmann BH, Kremen V, Worrell GA. Acute to long-term characteristics of impedance recordings during neurostimulation in humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.23.24301672. [PMID: 38343858 PMCID: PMC10854350 DOI: 10.1101/2024.01.23.24301672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Objective This study aims to characterize the time course of impedance, a crucial electrophysiological property of brain tissue, in the human thalamus (THL), amygdala-hippocampus (AMG-HPC), and posterior hippocampus (post-HPC) over an extended period. Approach Impedance was periodically sampled every 5-15 minutes over several months in five subjects with drug-resistant epilepsy using an experimental neuromodulation device. Initially, we employed descriptive piecewise and continuous mathematical models to characterize the impedance response for approximately three weeks post-electrode implantation. We then explored the temporal dynamics of impedance during periods when electrical stimulation was temporarily halted, observing a monotonic increase (rebound) in impedance before it stabilized at a higher value. Lastly, we assessed the stability of amplitude and phase over the 24-hour impedance cycle throughout the multi-month recording. Main results Immediately post-implantation, the impedance decreased, reaching a minimum value in all brain regions within approximately two days, and then increased monotonically over about 14 days to a stable value. The models accounted for the variance in short-term impedance changes. Notably, the minimum impedance of the THL in the most epileptogenic hemisphere was significantly lower than in other regions. During the gaps in electrical stimulation, the impedance rebound decreased over time and stabilized around 200 days post-implant, likely indicative of the foreign body response and fibrous tissue encapsulation around the electrodes. The amplitude and phase of the 24-hour impedance oscillation remained stable throughout the multi-month recording, with circadian variation in impedance dominating the long-term measures. Significance Our findings illustrate the complex temporal dynamics of impedance in implanted electrodes and the impact of electrical stimulation. We discuss these dynamics in the context of the known biological foreign body response of the brain to implanted electrodes. The data suggest that the temporal dynamics of impedance are dependent on the anatomical location and tissue epileptogenicity. These insights may offer additional guidance for the delivery of therapeutic stimulation at various time points post-implantation for neuromodulation therapy.
Collapse
Affiliation(s)
- Jie Cui
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Mayo College of Medicine and Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Filip Mivalt
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Vladimir Sladky
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Kladno, Czech Republic
| | - Jiwon Kim
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | - Hai-long Wang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kai J. Miller
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Nicholas Gregg
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Long Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy Denison
- Department of Engineering Science, University of Oxford; MRC Brain Network Dynamics Unit, University of Oxford, OX3 7DQ UK
| | - Bailey Winter
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Mayo College of Medicine and Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Benjamin H. Brinkmann
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Vaclav Kremen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Czech Institute of Informatics, Robotics, and Cybernetics, Czech Technical University, Prague, Czech Republic
| | - Gregory A. Worrell
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
4
|
Li F, Gallego J, Tirko NN, Greaser J, Bashe D, Patel R, Shaker E, Van Valkenburg GE, Alsubhi AS, Wellman S, Singh V, Padill CG, Gheres KW, Bagwell R, Mulvihill M, Kozai TDY. Low-intensity pulsed ultrasound stimulation (LIPUS) modulates microglial activation following intracortical microelectrode implantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570162. [PMID: 38105969 PMCID: PMC10723293 DOI: 10.1101/2023.12.05.570162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Microglia are important players in surveillance and repair of the brain. Their activation mediates neuroinflammation caused by intracortical microelectrode implantation, which impedes the application of intracortical brain-computer interfaces (BCIs). While low-intensity pulsed ultrasound stimulation (LIPUS) can attenuate microglial activation, its potential to modulate the microglia-mediated neuroinflammation and enhance the bio-integration of microelectrodes remains insufficiently explored. We found that LIPUS increased microglia migration speed from 0.59±0.04 to 1.35±0.07 µm/hr on day 1 and enhanced microglia expansion area from 44.50±6.86 to 93.15±8.77 µm 2 /min on day 7, indicating improved tissue healing and surveillance. Furthermore, LIPUS reduced microglial activation by 17% on day 6, vessel-associated microglia ratio from 70.67±6.15 to 40.43±3.87% on day 7, and vessel diameter by 20% on day 28. Additionally, microglial coverage of the microelectrode was reduced by 50% in week 1, indicating better tissue-microelectrode integration. These data reveal that LIPUS helps resolve neuroinflammation around chronic intracortical microelectrodes.
Collapse
|
5
|
Franklin ME, Bennett C, Arboite M, Alvarez-Ciara A, Corrales N, Verdelus J, Dietrich WD, Keane RW, de Rivero Vaccari JP, Prasad A. Activation of inflammasomes and their effects on neuroinflammation at the microelectrode-tissue interface in intracortical implants. Biomaterials 2023; 297:122102. [PMID: 37015177 PMCID: PMC10614166 DOI: 10.1016/j.biomaterials.2023.122102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Invasive neuroprosthetics rely on microelectrodes (MEs) to record or stimulate the activity of large neuron assemblies. However, MEs are subjected to tissue reactivity in the central nervous system (CNS) due to the foreign body response (FBR) that contribute to chronic neuroinflammation and ultimately result in ME failure. An endogenous, acute set of mechanisms responsible for the recognition and targeting of foreign objects, called the innate immune response, immediately follows the ME implant-induced trauma. Inflammasomes are multiprotein structures that play a critical role in the initiation of an innate immune response following CNS injuries. The activation of inflammasomes facilitates a range of innate immune response cascades and results in neuroinflammation and programmed cell death. Despite our current understanding of inflammasomes, their roles in the context of neural device implantation remain unknown. In this study, we implanted a non-functional Utah electrode array (UEA) into the rat somatosensory cortex and studied the inflammasome signaling and the corresponding downstream effects on inflammatory cytokine expression and the inflammasome-mediated cell death mechanism of pyroptosis. Our results not only demonstrate the continuous activation of inflammasomes and their contribution to neuroinflammation at the electrode-tissue interface but also reveal the therapeutic potential of targeting inflammasomes to attenuate the FBR in invasive neuroprosthetics.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Maelle Arboite
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jennifer Verdelus
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
| | - Robert W Keane
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA.
| |
Collapse
|
6
|
Wellens J, Deschaume O, Putzeys T, Eyley S, Thielemans W, Verhaert N, Bartic C. Sulfobetaine-based ultrathin coatings as effective antifouling layers for implantable neuroprosthetic devices. Biosens Bioelectron 2023; 226:115121. [PMID: 36774733 DOI: 10.1016/j.bios.2023.115121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Foreign body response (FBR), inflammation, and fibrotic encapsulation of neural implants remain major problems affecting the impedance of the electrode-tissue interface and altering the device performance. Adhesion of proteins and cells (e.g., pro-inflammatory macrophages, and fibroblasts) triggers the FBR cascade and can be diminished by applying antifouling coatings onto the implanted devices. In this paper, we report the deposition and characterization of a thin (±6 nm) sulfobetaine-based coating onto microfabricated platinum electrodes and cochlear implant (CI) electrode arrays. We found that this coating has stable cell and protein-repellent properties, for at least 31 days in vitro, not affected by electrical stimulation protocols. Additionally, its effect on the electrochemical properties relevant to stimulation (i.e., impedance, charge injection capacity) was negligible. When applied to clinical CI electrode arrays, the film was successful at inhibiting fibroblast adhesion on both the silicone packaging and the platinum/iridium electrodes. In vitro, in fibroblast cultures, coated CI electrode arrays maintained impedance values up to five times lower compared to non-coated devices. Our studies demonstrate that such thin sulfobetaine containing layers are stable and prevent protein and cell adhesion in vitro and are compatible for use on CI electrode arrays. Future in vivo studies should be conducted to investigate its ability to mitigate biofouling, fibrosis, and the resulting impedance changes upon long-term implantation in vivo.
Collapse
Affiliation(s)
- Jolan Wellens
- Laboratory for Soft Matter and Biophysics, Dept. Physics and Astronomy, KU Leuven, Celestijnenlaan 200D, 3001, Leuven, Belgium
| | - Olivier Deschaume
- Laboratory for Soft Matter and Biophysics, Dept. Physics and Astronomy, KU Leuven, Celestijnenlaan 200D, 3001, Leuven, Belgium
| | - Tristan Putzeys
- Laboratory for Soft Matter and Biophysics, Dept. Physics and Astronomy, KU Leuven, Celestijnenlaan 200D, 3001, Leuven, Belgium; Experimental Oto-rhino-laryngology Research Group, Dept. Neuroscience, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Samuel Eyley
- Sustainable Materials Lab, Department of Chemical Engineering, KU Leuven, Campus Kulak Kortrijk, Etienne Sabbelaan 53, 8500, Kortrijk, Belgium
| | - Wim Thielemans
- Sustainable Materials Lab, Department of Chemical Engineering, KU Leuven, Campus Kulak Kortrijk, Etienne Sabbelaan 53, 8500, Kortrijk, Belgium
| | - Nicolas Verhaert
- Experimental Oto-rhino-laryngology Research Group, Dept. Neuroscience, KU Leuven, Herestraat 49, 3000, Leuven, Belgium; Department of Otorhinolaryngology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Carmen Bartic
- Laboratory for Soft Matter and Biophysics, Dept. Physics and Astronomy, KU Leuven, Celestijnenlaan 200D, 3001, Leuven, Belgium.
| |
Collapse
|
7
|
Krukiewicz K. Electrochemical impedance spectroscopy as a versatile tool for the characterization of neural tissue: A mini review. Electrochem commun 2020. [DOI: 10.1016/j.elecom.2020.106742] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
8
|
Koss KM, Tsui C, Unsworth LD. Enzymatic Activity in Fractal Networks of Self-Assembling Peptides. Biomacromolecules 2018; 20:422-434. [DOI: 10.1021/acs.biomac.8b01496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Kyle M. Koss
- Department of Chemical and Materials Engineering, University of Alberta, 13-390 Floor - Donadeo Innovation Centre for Engineering
(ICE), 9211-116 Street NW, Edmonton, AB T6G 1H9, Canada
| | - Christopher Tsui
- Department of Chemical and Materials Engineering, University of Alberta, 13-390 Floor - Donadeo Innovation Centre for Engineering
(ICE), 9211-116 Street NW, Edmonton, AB T6G 1H9, Canada
| | - Larry D. Unsworth
- Department of Chemical and Materials Engineering, University of Alberta, 13-390 Floor - Donadeo Innovation Centre for Engineering
(ICE), 9211-116 Street NW, Edmonton, AB T6G 1H9, Canada
| |
Collapse
|
9
|
|
10
|
Lotti F, Ranieri F, Vadalà G, Zollo L, Di Pino G. Invasive Intraneural Interfaces: Foreign Body Reaction Issues. Front Neurosci 2017; 11:497. [PMID: 28932181 PMCID: PMC5592213 DOI: 10.3389/fnins.2017.00497] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
Intraneural interfaces are stimulation/registration devices designed to couple the peripheral nervous system (PNS) with the environment. Over the last years, their use has increased in a wide range of applications, such as the control of a new generation of neural-interfaced prostheses. At present, the success of this technology is limited by an electrical impedance increase, due to an inflammatory response called foreign body reaction (FBR), which leads to the formation of a fibrotic tissue around the interface, eventually causing an inefficient transduction of the electrical signal. Based on recent developments in biomaterials and inflammatory/fibrotic pathologies, we explore and select the biological solutions that might be adopted in the neural interfaces FBR context: modifications of the interface surface, such as organic and synthetic coatings; the use of specific drugs or molecular biology tools to target the microenvironment around the interface; the development of bio-engineered-scaffold to reduce immune response and promote interface-tissue integration. By linking what we believe are the major crucial steps of the FBR process with related solutions, we point out the main issues that future research has to focus on: biocompatibility without losing signal conduction properties, good reproducible in vitro/in vivo models, drugs exhaustion and undesired side effects. The underlined pros and cons of proposed solutions show clearly the importance of a better understanding of all the molecular and cellular pathways involved and the need of a multi-target action based on a bio-engineered combination approach.
Collapse
Affiliation(s)
- Fiorenza Lotti
- NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Università Campus Bio-MedicoRome, Italy.,Research Unit of Orthopaedic and Trauma Surgery, Università Campus Bio-MedicoRome, Italy
| | - Federico Ranieri
- NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Università Campus Bio-MedicoRome, Italy.,Fondazione Alberto Sordi-Research Institute for AgingRome, Italy.,Research Unit of Neurology, Neurophysiology and Neurobiology, Università Campus Bio-MedicoRome, Italy
| | - Gianluca Vadalà
- Research Unit of Orthopaedic and Trauma Surgery, Università Campus Bio-MedicoRome, Italy
| | - Loredana Zollo
- Research Unit of Biomedical Robotics and Biomicrosystems, Università Campus Bio-MedicoRome, Italy
| | - Giovanni Di Pino
- NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Università Campus Bio-MedicoRome, Italy.,Research Unit of Neurology, Neurophysiology and Neurobiology, Università Campus Bio-MedicoRome, Italy
| |
Collapse
|
11
|
Matsumura R, Yamamoto H, Niwano M, Hirano-Iwata A. An electrically resistive sheet of glial cells for amplifying signals of neuronal extracellular recordings. APPLIED PHYSICS LETTERS 2016; 108:023701. [PMID: 27703279 PMCID: PMC5035130 DOI: 10.1063/1.4939629] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/24/2015] [Indexed: 06/06/2023]
Abstract
Electrical signals of neuronal cells can be recorded non-invasively and with a high degree of temporal resolution using multielectrode arrays (MEAs). However, signals that are recorded with these devices are small, usually 0.01%-0.1% of intracellular recordings. Here, we show that the amplitude of neuronal signals recorded with MEA devices can be amplified by covering neuronal networks with an electrically resistive sheet. The resistive sheet used in this study is a monolayer of glial cells, supportive cells in the brain. The glial cells were grown on a collagen-gel film that is permeable to oxygen and other nutrients. The impedance of the glial sheet was measured by electrochemical impedance spectroscopy, and equivalent circuit simulations were performed to theoretically investigate the effect of covering the neurons with such a resistive sheet. Finally, the effect of the resistive glial sheet was confirmed experimentally, showing a 6-fold increase in neuronal signals. This technique feasibly amplifies signals of MEA recordings.
Collapse
Affiliation(s)
- R Matsumura
- Graduate School of Biomedical Engineering, Tohoku University , 6-6 Aramaki-aza Aoba, Aoba-ku, Sendai 980-8579, Japan
| | - H Yamamoto
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University , 6-3 Aramaki-aza Aoba, Aoba-ku, Sendai 980-8578, Japan
| | - M Niwano
- Research Institute of Electrical Communication, Tohoku University , 2-1-1 Katahira, Aoba-ku, Sendai 980-8577, Japan
| | - A Hirano-Iwata
- Graduate School of Biomedical Engineering, Tohoku University , 6-6 Aramaki-aza Aoba, Aoba-ku, Sendai 980-8579, Japan
| |
Collapse
|
12
|
Lee JY, Khaing ZZ, Siegel JJ, Schmidt CE. Surface modification of neural electrodes with pyrrole-hyaluronic acid conjugate to attenuate reactive astrogliosis in vivo. RSC Adv 2015; 5:39228-39231. [PMID: 35528963 PMCID: PMC9075707 DOI: 10.1039/c5ra03294f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023] Open
Abstract
Surface of neural probes were electrochemically modified with a non-cell adhesive and biocompatible conjugate, pyrrole-hyaluronic acid (PyHA), to reduce reactive astrogliosis. Poly(PyHA)-modified wire electrodes were implanted into rat motor cortices for three weeks and were found to markedly reduce the expression of glial fibrillary acidic protein compared to uncoated electrodes.
Collapse
Affiliation(s)
- J Y Lee
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- School of Materials Science and Engineering, Gwangju Institute of Science and Engineering, Gwangju, South Korea
| | - Z Z Khaing
- Department of Biomedical Engineering, The University of Florida at Gainesville, Gainesville, FL, USA
| | - J J Siegel
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX, USA
| | - C E Schmidt
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Biomedical Engineering, The University of Florida at Gainesville, Gainesville, FL, USA
| |
Collapse
|
13
|
Gutowski SM, Shoemaker JT, Templeman KL, Wei Y, Latour RA, Bellamkonda RV, LaPlaca MC, García AJ. Protease-degradable PEG-maleimide coating with on-demand release of IL-1Ra to improve tissue response to neural electrodes. Biomaterials 2015; 44:55-70. [PMID: 25617126 DOI: 10.1016/j.biomaterials.2014.12.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 12/01/2014] [Accepted: 12/16/2014] [Indexed: 01/18/2023]
Abstract
Neural electrodes are an important part of brain-machine interface devices that can restore functionality to patients with sensory and movement disorders. Chronically implanted neural electrodes induce an unfavorable tissue response which includes inflammation, scar formation, and neuronal cell death, eventually causing loss of electrode function. We developed a poly(ethylene glycol) hydrogel coating for neural electrodes with non-fouling characteristics, incorporated an anti-inflammatory agent, and engineered a stimulus-responsive degradable portion for on-demand release of the anti-inflammatory agent in response to inflammatory stimuli. This coating reduces in vitro glial cell adhesion, cell spreading, and cytokine release compared to uncoated controls. We also analyzed the in vivo tissue response using immunohistochemistry and microarray qRT-PCR. Although no differences were observed among coated and uncoated electrodes for inflammatory cell markers, lower IgG penetration into the tissue around PEG+IL-1Ra coated electrodes indicates an improvement in blood-brain barrier integrity. Gene expression analysis showed higher expression of IL-6 and MMP-2 around PEG+IL-1Ra samples, as well as an increase in CNTF expression, an important marker for neuronal survival. Importantly, increased neuronal survival around coated electrodes compared to uncoated controls was observed. Collectively, these results indicate promising findings for an engineered coating to increase neuronal survival and improve tissue response around implanted neural electrodes.
Collapse
Affiliation(s)
- Stacie M Gutowski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - James T Shoemaker
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kellie L Templeman
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yang Wei
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Robert A Latour
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Ravi V Bellamkonda
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michelle C LaPlaca
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
14
|
Accurate resistivity mouse brain mapping using microelectrode arrays. Biosens Bioelectron 2014; 60:143-53. [DOI: 10.1016/j.bios.2014.03.044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/11/2014] [Accepted: 03/21/2014] [Indexed: 12/19/2022]
|
15
|
Sommakia S, Gaire J, Rickus JL, Otto KJ. Resistive and reactive changes to the impedance of intracortical microelectrodes can be mitigated with polyethylene glycol under acute in vitro and in vivo settings. FRONTIERS IN NEUROENGINEERING 2014; 7:33. [PMID: 25136315 PMCID: PMC4120760 DOI: 10.3389/fneng.2014.00033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/11/2014] [Indexed: 01/05/2023]
Abstract
The reactive response of brain tissue to implantable intracortical microelectrodes is thought to negatively affect their recordable signal quality and impedance, resulting in unreliable longitudinal performance. The relationship between the progression of the reactive tissue into a glial scar and the decline in device performance is unclear. We show that exposure to a model protein solution in vitro and acute implantation result in both resistive and capacitive changes to electrode impedance, rather than purely resistive changes. We also show that applying 4000 MW polyethylene glycol (PEG) prevents impedance increases in vitro, and reduces the percent change in impedance in vivo following implantation. Our results highlight the importance of considering the contributions of non-cellular components to the decline in neural microelectrode performance, and present a proof of concept for using a simple dip-coated PEG film to modulate changes in microelectrode impedance.
Collapse
Affiliation(s)
- Salah Sommakia
- Weldon School of Biomedical Engineering, Purdue University West Lafayette, IN, USA ; Physiological Sensing Facility at the Bindley Bioscience Center and Birck Nanotechnology Center, Purdue University West Lafayette, IN, USA
| | - Janak Gaire
- Department of Biological Sciences, Purdue University West Lafayette, IN, USA
| | - Jenna L Rickus
- Weldon School of Biomedical Engineering, Purdue University West Lafayette, IN, USA ; Physiological Sensing Facility at the Bindley Bioscience Center and Birck Nanotechnology Center, Purdue University West Lafayette, IN, USA ; Department of Agricultural and Biological Engineering, Purdue University West Lafayette, IN, USA
| | - Kevin J Otto
- Weldon School of Biomedical Engineering, Purdue University West Lafayette, IN, USA ; Department of Biological Sciences, Purdue University West Lafayette, IN, USA
| |
Collapse
|
16
|
Cicchetti F, Barker RA. The glial response to intracerebrally delivered therapies for neurodegenerative disorders: is this a critical issue? Front Pharmacol 2014; 5:139. [PMID: 25071571 PMCID: PMC4090753 DOI: 10.3389/fphar.2014.00139] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/24/2014] [Indexed: 12/20/2022] Open
Abstract
The role of glial cells in the pathogenesis of many neurodegenerative conditions of the central nervous system (CNS) is now well established (as is discussed in other reviews in this special issue of Frontiers in Neuropharmacology). What is less clear is whether there are changes in these same cells in terms of their behavior and function in response to invasive experimental therapeutic interventions for these diseases. This has, and will continue to become more of an issue as we enter a new era of novel treatments which require the agent to be directly placed/infused into the CNS such as deep brain stimulation (DBS), cell transplants, gene therapies and growth factor infusions. To date, all of these treatments have produced variable outcomes and the reasons for this have been widely debated but the host astrocytic and/or microglial response induced by such invasively delivered agents has not been discussed in any detail. In this review, we have attempted to summarize the limited published data on this, in particular we discuss the small number of human post-mortem studies reported in this field. By so doing, we hope to provide a better description and understanding of the extent and nature of both the astrocytic and microglial response, which in turn could lead to modifications in the way these therapeutic interventions are delivered.
Collapse
Affiliation(s)
- Francesca Cicchetti
- Axe Neurosciences, Centre de Recherche du CHU de Québec Québec, QC, Canada ; Département de Psychiatrie et Neurosciences, Université Laval Québec, QC, Canada
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge Cambridge, UK
| |
Collapse
|
17
|
Potter-Baker KA, Ravikumar M, Burke AA, Meador WD, Householder KT, Buck AC, Sunil S, Stewart WG, Anna JP, Tomaszewski WH, Capadona JR. A comparison of neuroinflammation to implanted microelectrodes in rat and mouse models. Biomaterials 2014; 35:5637-46. [PMID: 24755527 PMCID: PMC4071936 DOI: 10.1016/j.biomaterials.2014.03.076] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/27/2014] [Indexed: 12/31/2022]
Abstract
Rat models have emerged as a common tool to study neuroinflammation to intracortical microelectrodes. While a number of studies have attempted to understand the factors resulting in neuroinflammation using rat models, a complete understanding of key mechanistic pathways remains elusive. Transgenic mouse models, however, could facilitate a deeper understanding of mechanistic pathways due to an ease of genetic alteration. Therefore, the goal of the present study is to compare neuroinflammation following microelectrode implantation between the rat and the mouse model. Our study suggests that subtle differences in the classic neuroinflammatory markers exist between the animal models at both two and sixteen weeks post implantation. Most notably, neuronal densities surrounding microelectrodes were significantly lower in the rat model at two weeks, while similar densities were observed between the animal models at sixteen weeks. Physiological differences between the species and slight alterations in surgical methods are likely key contributors to the observed differences. Moving forward, we propose that differences in the time course of neuroinflammation between the animal models should be considered when trying to understand and prevent intracortical microelectrode failure.
Collapse
Affiliation(s)
- Kelsey A Potter-Baker
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Madhumitha Ravikumar
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Alan A Burke
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - William D Meador
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - Kyle T Householder
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Amy C Buck
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Smrithi Sunil
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA
| | - Wade G Stewart
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - Jake P Anna
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - William H Tomaszewski
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Wickenden Bldg., Cleveland, OH 44106, USA; Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland, OH 44106-1702, USA.
| |
Collapse
|
18
|
Jeffery AF, Churchward MA, Mushahwar VK, Todd KG, Elias AL. Hyaluronic Acid-Based 3D Culture Model for In Vitro Testing of Electrode Biocompatibility. Biomacromolecules 2014; 15:2157-65. [DOI: 10.1021/bm500318d] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Andrea F. Jeffery
- Chemical
and Materials Engineering, University of Alberta, Edmonton, AB T6G 2V4, Canada
- Alberta Innovates-Health
Solutions Interdisciplinary Team in Smart Neural Prostheses (Project
SMART), University of Alberta, AB, Canada
| | - Matthew A. Churchward
- Department
of Psychiatry, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Alberta Innovates-Health
Solutions Interdisciplinary Team in Smart Neural Prostheses (Project
SMART), University of Alberta, AB, Canada
| | - Vivian K. Mushahwar
- Division
of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Centre
for Neuroscience, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Alberta Innovates-Health
Solutions Interdisciplinary Team in Smart Neural Prostheses (Project
SMART), University of Alberta, AB, Canada
| | - Kathryn G. Todd
- Department
of Psychiatry, University of Alberta, Edmonton, AB T6G 2G3, Canada
- Centre
for Neuroscience, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Alberta Innovates-Health
Solutions Interdisciplinary Team in Smart Neural Prostheses (Project
SMART), University of Alberta, AB, Canada
| | - Anastasia L. Elias
- Chemical
and Materials Engineering, University of Alberta, Edmonton, AB T6G 2V4, Canada
- Alberta Innovates-Health
Solutions Interdisciplinary Team in Smart Neural Prostheses (Project
SMART), University of Alberta, AB, Canada
| |
Collapse
|
19
|
Ouyang L, Shaw CL, Kuo CC, Griffin AL, Martin DC. In vivo polymerization of poly(3,4-ethylenedioxythiophene) in the living rat hippocampus does not cause a significant loss of performance in a delayed alternation task. J Neural Eng 2014; 11:026005. [PMID: 24503720 DOI: 10.1088/1741-2560/11/2/026005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
After extended implantation times, traditional intracortical neural probes exhibit a foreign-body reaction characterized by a reactive glial sheath that has been associated with increased system impedance and signal deterioration. Previously, we have proposed that the local in vivo polymerization of an electronically and ionically conducting polymer, poly(3,4-ethylenedioxythiophene) (PEDOT), might help to rebuild charge transport pathways across the glial scar between the device and surrounding parenchyma (Richardson-Burns et al 2007 J. Neural Eng. 4 L6-13). The EDOT monomer can be delivered via a microcannula/electrode system into the brain tissue of living animals followed by direct electrochemical polymerization, using the electrode itself as a source of oxidative current. In this study, we investigated the long-term effect of local in vivo PEDOT deposition on hippocampal neural function and histology. Rodent subjects were trained on a hippocampus-dependent task, delayed alternation (DA), and implanted with the microcannula/electrode system in the hippocampus. The animals were divided into four groups with different delay times between the initial surgery and the electrochemical polymerization: (1) control (no polymerization), (2) immediate (polymerization within 5 min of device implantation), (3) early (polymerization within 3-4 weeks after implantation) and (4) late (polymerization 7-8 weeks after polymerization). System impedance at 1 kHz was recorded and the tissue reactions were evaluated by immunohistochemistry. We found that under our deposition conditions, PEDOT typically grew at the tip of the electrode, forming an ∼500 µm cloud in the tissue. This is much larger than the typical width of the glial scar (∼150 µm). After polymerization, the impedance amplitude near the neurologically important frequency of 1 kHz dropped for all the groups; however, there was a time window of 3-4 weeks for an optimal decrease in impedance. For all surgery-polymerization time intervals, the polymerization did not cause significant deficits in performance of the DA task, suggesting that hippocampal function was not impaired by PEDOT deposition. However, GFAP+ and ED-1+ cells were also found at the deposition two weeks after the polymerization, suggesting potential secondary scarring. Therefore, less extensive deposition or milder deposition conditions may be desirable to minimize this scarring while maintaining decreased system impedance.
Collapse
Affiliation(s)
- Liangqi Ouyang
- Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA
| | | | | | | | | |
Collapse
|
20
|
Prasad A, Xue QS, Dieme R, Sankar V, Mayrand RC, Nishida T, Streit WJ, Sanchez JC. Abiotic-biotic characterization of Pt/Ir microelectrode arrays in chronic implants. FRONTIERS IN NEUROENGINEERING 2014; 7:2. [PMID: 24550823 PMCID: PMC3912984 DOI: 10.3389/fneng.2014.00002] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/14/2014] [Indexed: 11/13/2022]
Abstract
Pt/Ir electrodes have been extensively used in neurophysiology research in recent years as they provide a more inert recording surface as compared to tungsten or stainless steel. While floating microelectrode arrays (FMA) consisting of Pt/Ir electrodes are an option for neuroprosthetic applications, long-term in vivo functional performance characterization of these FMAs is lacking. In this study, we have performed comprehensive abiotic-biotic characterization of Pt/Ir arrays in 12 rats with implant periods ranging from 1 week up to 6 months. Each of the FMAs consisted of 16-channel, 1.5 mm long, and 75 μm diameter microwires with tapered tips that were implanted into the somatosensory cortex. Abiotic characterization included (1) pre-implant and post-explant scanning electron microscopy (SEM) to study recording site changes, insulation delamination and cracking, and (2) chronic in vivo electrode impedance spectroscopy. Biotic characterization included study of microglial responses using a panel of antibodies, such as Iba1, ED1, and anti-ferritin, the latter being indicative of blood-brain barrier (BBB) disruption. Significant structural variation was observed pre-implantation among the arrays in the form of irregular insulation, cracks in insulation/recording surface, and insulation delamination. We observed delamination and cracking of insulation in almost all electrodes post-implantation. These changes altered the electrochemical surface area of the electrodes and resulted in declining impedance over the long-term due to formation of electrical leakage pathways. In general, the decline in impedance corresponded with poor electrode functional performance, which was quantified via electrode yield. Our abiotic results suggest that manufacturing variability and insulation material as an important factor contributing to electrode failure. Biotic results show that electrode performance was not correlated with microglial activation (neuroinflammation) as we were able to observe poor performance in the absence of neuroinflammation, as well as good performance in the presence of neuroinflammation. One biotic change that correlated well with poor electrode performance was intraparenchymal bleeding, which was evident macroscopically in some rats and presented microscopically by intense ferritin immunoreactivity in microglia/macrophages. Thus, we currently consider intraparenchymal bleeding, suboptimal electrode fabrication, and insulation delamination as the major factors contributing toward electrode failure.
Collapse
Affiliation(s)
- Abhishek Prasad
- Department of Biomedical Engineering, University of Miami Coral Gables, FL, USA
| | - Qing-Shan Xue
- Department of Neuroscience, University of Florida Gainesville, FL, USA
| | - Robert Dieme
- Department of Electrical and Computer Engineering, University of Florida Gainesville, FL, USA
| | - Viswanath Sankar
- Department of Electrical and Computer Engineering, University of Florida Gainesville, FL, USA
| | - Roxanne C Mayrand
- Department of Neuroscience, University of Miami Coral Gables, FL, USA
| | - Toshikazu Nishida
- Department of Electrical and Computer Engineering, University of Florida Gainesville, FL, USA
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida Gainesville, FL, USA
| | - Justin C Sanchez
- Department of Biomedical Engineering, University of Miami Coral Gables, FL, USA ; Department of Neuroscience, University of Miami Coral Gables, FL, USA ; Miami Project to Cure Paralysis, University of Miami Miami, FL, USA
| |
Collapse
|
21
|
Groothuis J, Ramsey NF, Ramakers GM, van der Plasse G. Physiological Challenges for Intracortical Electrodes. Brain Stimul 2014; 7:1-6. [DOI: 10.1016/j.brs.2013.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 02/08/2023] Open
|
22
|
Ng KA, Xu YP. A compact, low input capacitance neural recording amplifier. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2013; 7:610-620. [PMID: 24144666 DOI: 10.1109/tbcas.2013.2280066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Conventional capacitively coupled neural recording amplifiers often present a large input load capacitance to the neural signal source and hence take up large circuit area. They suffer due to the unavoidable trade-off between the input capacitance and chip area versus the amplifier gain. In this work, this trade-off is relaxed by replacing the single feedback capacitor with a clamped T-capacitor network. With this simple modification, the proposed amplifier can achieve the same mid-band gain with less input capacitance, resulting in a higher input impedance and a smaller silicon area. Prototype neural recording amplifiers based on this proposal were fabricated in 0.35 μm CMOS, and their performance is reported. The amplifiers occupy smaller area and have lower input loading capacitance compared to conventional neural amplifiers. One of the proposed amplifiers occupies merely 0.056 mm(2). It achieves 38.1-dB mid-band gain with 1.6 pF input capacitance, and hence has an effective feedback capacitance of 20 fF. Consuming 6 μW, it has an input referred noise of 13.3 μVrms over 8.5 kHz bandwidth and NEF of 7.87. In-vivo recordings from animal experiments are also demonstrated.
Collapse
|
23
|
Ghane-Motlagh B, Sawan M. Design and Implementation Challenges of Microelectrode Arrays: A Review. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/msa.2013.48059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
24
|
Davis TS, Parker RA, House PA, Bagley E, Wendelken S, Normann RA, Greger B. Spatial and temporal characteristics of V1 microstimulation during chronic implantation of a microelectrode array in a behaving macaque. J Neural Eng 2012; 9:065003. [PMID: 23186948 PMCID: PMC3521049 DOI: 10.1088/1741-2560/9/6/065003] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE It has been hypothesized that a vision prosthesis capable of evoking useful visual percepts can be based upon electrically stimulating the primary visual cortex (V1) of a blind human subject via penetrating microelectrode arrays. As a continuation of earlier work, we examined several spatial and temporal characteristics of V1 microstimulation. APPROACH An array of 100 penetrating microelectrodes was chronically implanted in V1 of a behaving macaque monkey. Microstimulation thresholds were measured using a two-alternative forced choice detection task. Relative locations of electrically-evoked percepts were measured using a memory saccade-to-target task. MAIN RESULTS The principal finding was that two years after implantation we were able to evoke behavioural responses to electric stimulation across the spatial extent of the array using groups of contiguous electrodes. Consistent responses to stimulation were evoked at an average threshold current per electrode of 204 ± 49 µA (mean ± std) for groups of four electrodes and 91 ± 25 µA for groups of nine electrodes. Saccades to electrically-evoked percepts using groups of nine electrodes showed that the animal could discriminate spatially distinct percepts with groups having an average separation of 1.6 ± 0.3 mm (mean ± std) in cortex and 1.0° ± 0.2° in visual space. Significance. These results demonstrate chronic perceptual functionality and provide evidence for the feasibility of a cortically-based vision prosthesis for the blind using penetrating microelectrodes.
Collapse
Affiliation(s)
- T S Davis
- Department of Bioengineering, University of Utah, UT, USA
| | - R A Parker
- Interdepartmental Program in Neuroscience, University of Utah, UT, USA
| | - P A House
- Department of Neurosurgery, University of Utah, UT, USA
| | - E Bagley
- Department of Bioengineering, University of Utah, UT, USA
| | - S Wendelken
- Department of Bioengineering, University of Utah, UT, USA
| | - R A Normann
- Department of Bioengineering, University of Utah, UT, USA
- Department of Ophthalmology and Visual Sciences, University of Utah, UT, USA
| | - B Greger
- Department of Bioengineering, University of Utah, UT, USA
- Department of Ophthalmology and Visual Sciences, University of Utah, UT, USA
| |
Collapse
|
25
|
Prasad A, Xue QS, Sankar V, Nishida T, Shaw G, Streit WJ, Sanchez JC. Comprehensive characterization and failure modes of tungsten microwire arrays in chronic neural implants. J Neural Eng 2012; 9:056015. [DOI: 10.1088/1741-2560/9/5/056015] [Citation(s) in RCA: 218] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
26
|
Guo X, Ayala JE, Gonzalez M, Stancescu M, Lambert S, Hickman JJ. Tissue engineering the monosynaptic circuit of the stretch reflex arc with co-culture of embryonic motoneurons and proprioceptive sensory neurons. Biomaterials 2012; 33:5723-31. [PMID: 22594977 DOI: 10.1016/j.biomaterials.2012.04.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 04/16/2012] [Indexed: 01/08/2023]
Abstract
The sensory circuit of the stretch reflex arc is composed of intrafusal muscle fibers and their innervating proprioceptive neurons that convert mechanical information regarding muscle length and tension into action potentials that synapse onto the homonymous motoneurons in the ventral spinal cord which innervate the extrafusal fibers of the same muscle. To date, the in vitro synaptic connection between proprioceptive sensory neurons and spinal motoneurons has not been demonstrated. A functional in vitro system demonstrating this connection would enable the understanding of feedback by the integration of sensory input into the spinal reflex arc. Here we report a co-culture of rat embryonic motoneurons and proprioceptive sensory neurons from dorsal root ganglia (DRG) in a defined serum-free medium on a synthetic silane substrate (DETA). Furthermore, we have demonstrated functional synapse formation in the co-culture by immunocytochemistry and electrophysiological analysis. This work will be valuable for enabling in vitro model systems for the study of spinal motor control and related pathologies such as spinal cord injury, muscular dystrophy and spasticity by improving our understanding of the integration of the mechanosensitive feedback mechanism.
Collapse
Affiliation(s)
- Xiufang Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | | | | | | | | | | |
Collapse
|
27
|
Prasad A, Sanchez JC. Quantifying long-term microelectrode array functionality using chronic in vivo impedance testing. J Neural Eng 2012; 9:026028. [PMID: 22442134 DOI: 10.1088/1741-2560/9/2/026028] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Long-term acquisition of high-quality neural recordings is a cornerstone of neuroprosthetic system design. Mitigating the experimental variability of chronically implanted arrays has been a formidable task because the sensor recording sites can be influenced by biotic and abiotic responses. Several studies have implicated changes in electrical interface impedance as a preliminary marker to infer electrode viability. Microelectrode impedance plays an important role in the monitoring of low amplitude and high-resolution extracellular neural signals. In this work, we seek to quantify long-term microelectrode array functionality and derive an impedance-based predictor for electrode functionality that correlates the recording site electrical properties with the functional neuronal recordings in vivo. High temporal resolution metrics of this type would allow one to assess, predict, and improve electrode performance in the future. In a large cohort of animals, we performed daily impedance measurements and neural signal recordings over long periods (up to 21 weeks) of time in rats using tungsten microwire arrays implanted into the somatosensory cortex. This study revealed that there was a time-varying trend in the modulation of impedance that was related to electrode performance. Single units were best detected from electrodes at time points when the electrode entered into the 40-150 KΩ impedance range. This impedance trend was modeled across the full cohort of animals to predict future electrode performance. The model was tested on data from all animals and was able to provide predictions of electrode performance chronically. Insight from this study can be combined with knowledge of electrode materials and histological analysis to provide a more comprehensive predictive model of electrode failure in the future.
Collapse
Affiliation(s)
- Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA.
| | | |
Collapse
|
28
|
Reduction of autofluorescence at the microelectrode-cortical tissue interface improves antibody detection. J Neurosci Methods 2012; 203:96-105. [PMID: 21978484 DOI: 10.1016/j.jneumeth.2011.09.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 09/13/2011] [Accepted: 09/21/2011] [Indexed: 12/30/2022]
Abstract
Immunohistochemistry (IHC) remains among the most utilized methods for detection of inflammatory events occurring at the microelectrode-cortical tissue interface. It has further become a standard protocol to quantify the intensity of this resulting fluorescent signal, normalized to "background", as a measurement of the extent of inflammatory events. Unfortunately, several sources of autofluorescence could result in variations in this user-defined "background". Notably, we found that the presence of hemosiderin-laden macrophages (HLMs) at the interface resulted in a variable source of background in both green and red fluorescent channels. The HLM-derived autofluorescence prevented the reproducible detection of presumably low-level antigens at the interface. Here we show that treatment of the native cortical tissue for no less than 10 min, with a minimum of 0.5mM copper sulfate, resulted in at least a 70% reduction in native HLM autofluorescence in both green and red fluorescent channels. In the case of highly expressed antigens, such as glial fibrillar acidic protein (GFAP), treatment of immuno-labeled tissue with copper sulfate reduced tissue background, compared to standard IHC methodology, but did not result in significant differences in the quantification of normalized signal intensity. However, treatment with copper sulfate substantially enhanced the detection efficiency of weakly expressed antigens at the device-tissue interface. This study demonstrates that the inclusion of copper sulfate incubation during IHC tissue preparation significantly reduced HLM-derived autofluorescence, and allowed for more accurate detection and quantification of faintly expressed inflammatory markers at the device-tissue interface.
Collapse
|
29
|
Prasad A, Xue QS, Sankar V, Nishida T, Shaw G, Streit W, Sanchez JC. Comprehensive characterization of tungsten microwires in chronic neurocortical implants. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2012:755-758. [PMID: 23366002 DOI: 10.1109/embc.2012.6346041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The long-term performance of chronic microelectrode array implants for neural ensemble recording is affected by temporal degradation in signal quality due to several factors including structural changes in the recording surface, electrical responses, and tissue immune reactivity. This study combines the information available from the temporal aggregation of both biotic and abiotic metrics to analyze and quantify the combined effects on long-term performance. Study of a 42-day implant showed there was a functional relationship between the measured impedance and the array neuronal yield. This was correlated with structural changes in the recording sites, microglial activation/degeneration, and elevation of a blood biochemical marker for axonal injury. We seek to elucidate the mechanisms of chronic microelectrode array failure through the study of the combined effects of these biotic and abiotic factors.
Collapse
Affiliation(s)
- Abhishek Prasad
- Department of Biomedical Engineering and the Miami Project to Cure Paralysis, University of Miami, Coral Gables, FL 33146, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Hara SA, Kim BJ, Kuo JTW, Lee C, Gutierrez CA, Hoang T, Meng E. Pre-implantation electrochemical characterization of a Parylene C sheath microelectrode array probe. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2012; 2012:5126-5129. [PMID: 23367082 DOI: 10.1109/embc.2012.6347147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
We present the preliminary electrochemical characterization of 3D Parylene C sheath microelectrode array probes towards realizing reliable chronic neuroprosthetic recordings. Electrochemical techniques were used to verify electrode integrity after our novel post-fabrication thermoforming process was applied to flat surface micromachined structures to achieve a hollow sheath probe shape. Characterization of subsequent neurotrophic coatings was performed and accelerated life testing was used to simulate six months in vivo. Prior to probe implantation, crosstalk was measured and electrode surface properties were evaluated through the use of electrochemical impedance spectroscopy.
Collapse
Affiliation(s)
- Seth A Hara
- Department of Biomedical Engineering at the University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Hashemi P, Walsh PL, Guillot TS, Gras-Najjar J, Takmakov P, Crews FT, Wightman RM. Chronically Implanted, Nafion-Coated Ag/AgCl Reference Electrodes for Neurochemical Applications. ACS Chem Neurosci 2011; 2:658-666. [PMID: 22125666 DOI: 10.1021/cn2000684] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Fast-scan cyclic voltammetry (FSCV) at carbon fiber microelectrodes can be used to measure behaviorally correlated dopamine changes in the extracellular fluid of the brain of freely moving rats. These experiments employ a chronically implanted Ag/AgCl reference electrode. When dopamine measurements are taken 4 days after implantation, there is often a potential shift, typically greater than +0.2 V, in the anodic and cathodic peaks in the cyclic voltammogram for dopamine. In this work, we optimized a method to coat sintered Ag/AgCl reference electrodes with the perfluorinated polymer, Nafion, to prevent this shift. We find that we can stabilize reference electrodes for up to 28 days. Immunohistochemistry of the tissue around the implant site shows extensive glial encapsulation around both bare and Nafion-coated devices. However, the lesion around bare electrodes has a rough texture implying that these cells are strongly adsorbed onto the bare reference electrode, while the lesion around a Nafion-coated electrode shows that cells are more intact implying that they adsorb less strongly. EDS and SEM analysis of the surface of the electrodes confirms this by visualizing a heavy build up of plaques, organic in nature, only on bare electrodes. Impedance spectroscopy indicates no difference between the impedance of bare and Nafion-coated Ag/AgCl electrodes, indicating that glial encapsulation does not lead to an increase in uncompensated resistance between the working and reference electrodes. The electrochemical shift therefore must be due to the unique chemical microenvironment around the reference electrode that alters the chloride equilibrium, a process that the Nafion coating prevents.
Collapse
Affiliation(s)
- Parastoo Hashemi
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| | - Paul L. Walsh
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| | - Thomas S. Guillot
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| | - Julie Gras-Najjar
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| | - Pavel Takmakov
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| | - Fulton T. Crews
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| | - R. Mark Wightman
- Department of Chemistry and ‡Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
32
|
Harris JP, Capadona JR, Miller RH, Healy BC, Shanmuganathan K, Rowan SJ, Weder C, Tyler DJ. Mechanically adaptive intracortical implants improve the proximity of neuronal cell bodies. J Neural Eng 2011; 8:066011. [PMID: 22049097 DOI: 10.1088/1741-2560/8/6/066011] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The hypothesis is that the mechanical mismatch between brain tissue and microelectrodes influences the inflammatory response. Our unique, mechanically adaptive polymer nanocomposite enabled this study within the cerebral cortex of rats. The initial tensile storage modulus of 5 GPa decreases to 12 MPa within 15 min under physiological conditions. The response to the nanocomposite was compared to surface-matched, stiffer implants of traditional wires (411 GPa) coated with the identical polymer substrate and implanted on the contralateral side. Both implants were tethered. Fluorescent immunohistochemistry labeling examined neurons, intermediate filaments, macrophages, microglia and proteoglycans. We demonstrate, for the first time, a system that decouples the mechanical and surface chemistry components of the neural response. The neuronal nuclei density within 100 µm of the device at four weeks post-implantation was greater for the compliant nanocomposite compared to the stiff wire. At eight weeks post-implantation, the neuronal nuclei density around the nanocomposite was maintained, but the density around the wire recovered to match that of the nanocomposite. The glial scar response to the compliant nanocomposite was less vigorous than it was to the stiffer wire. The results suggest that mechanically associated factors such as proteoglycans and intermediate filaments are important modulators of the response of the compliant nanocomposite.
Collapse
Affiliation(s)
- J P Harris
- Department of Biomedical Engineering, CWRU, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Ereifej ES, Khan S, Newaz G, Zhang J, Auner GW, VandeVord PJ. Characterization of astrocyte reactivity and gene expression on biomaterials for neural electrodes. J Biomed Mater Res A 2011; 99:141-50. [PMID: 21812095 DOI: 10.1002/jbm.a.33170] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 02/15/2011] [Accepted: 04/29/2011] [Indexed: 01/06/2023]
Abstract
Neural electrode devices hold great promise to help people with the restoration of lost functions. However, research is lacking in the biomaterial design of a stable, long-term device. Glial scarring is initiated when a device is inserted into brain tissue and an inflammatory response ensues. Astrocytes become hypertrophic, hyperplastic, and upregulate glial-fibrillary acidic protein. This study was designed to investigate the astrocyte proliferation, viability, morphology, and gene expression to assess the reactive state of the cells on different material surfaces. Although platinum and silicon have been extensively characterized both in vivo and in vitro for their biocompatibility with neuronal cells, this study used the novel usage of PMMA and SU-8 in neural electrodes by comparative analysis of materials' biocompatibility. This study has shown evidence of noncytotoxicity of SU-8. We have also confirmed the biocompatibility of PMMA with astrocytes. Moreover, we have established sound guidelines of which neural implant materials should meet to be depicted biocompatible.
Collapse
Affiliation(s)
- Evon S Ereifej
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan, USA
| | | | | | | | | | | |
Collapse
|
34
|
Nowak K, Mix E, Gimsa J, Strauss U, Sriperumbudur KK, Benecke R, Gimsa U. Optimizing a rodent model of Parkinson's disease for exploring the effects and mechanisms of deep brain stimulation. PARKINSONS DISEASE 2011; 2011:414682. [PMID: 21603182 PMCID: PMC3096058 DOI: 10.4061/2011/414682] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 01/28/2011] [Indexed: 11/20/2022]
Abstract
Deep brain stimulation (DBS) has become a treatment for a growing number of neurological and psychiatric disorders, especially for therapy-refractory Parkinson's disease (PD). However, not all of the symptoms of PD are sufficiently improved in all patients, and side effects may occur. Further progress depends on a deeper insight into the mechanisms of action of DBS in the context of disturbed brain circuits. For this, optimized animal models have to be developed. We review not only charge transfer mechanisms at the electrode/tissue interface and strategies to increase the stimulation's energy-efficiency but also the electrochemical, electrophysiological, biochemical and functional effects of DBS. We introduce a hemi-Parkinsonian rat model for long-term experiments with chronically instrumented rats carrying a backpack stimulator and implanted platinum/iridium electrodes. This model is suitable for (1) elucidating the electrochemical processes at the electrode/tissue interface, (2) analyzing the molecular, cellular and behavioral stimulation effects, (3) testing new target regions for DBS, (4) screening for potential neuroprotective DBS effects, and (5) improving the efficacy and safety of the method. An outlook is given on further developments of experimental DBS, including the use of transgenic animals and the testing of closed-loop systems for the direct on-demand application of electric stimulation.
Collapse
Affiliation(s)
- Karl Nowak
- Department of Neurology, University of Rostock, Gehlsheimer Straße 20, 18147 Rostock, Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Han N, Rao SS, Johnson J, Parikh KS, Bradley PA, Lannutti JJ, Winter JO. Hydrogel-electrospun fiber mat composite coatings for neural prostheses. FRONTIERS IN NEUROENGINEERING 2011; 4:2. [PMID: 21441993 PMCID: PMC3061411 DOI: 10.3389/fneng.2011.00002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 02/21/2011] [Indexed: 01/08/2023]
Abstract
Achieving stable, long-term performance of implanted neural prosthetic devices has been challenging because of implantation related neuron loss and a foreign body response that results in encapsulating glial scar formation. To improve neuron–prosthesis integration and form chronic, stable interfaces, we investigated the potential of neurotrophin-eluting hydrogel–electrospun fiber mat (EFM) composite coatings. In particular, poly(ethylene glycol)-poly(ε-caprolactone) (PEGPCL) hydrogel–poly(ε-caprolactone) EFM composites were applied as coatings for multielectrode arrays. Coatings were stable and persisted on electrode surfaces for over 1 month under an agarose gel tissue phantom and over 9 months in a PBS immersion bath. To demonstrate drug release, a neurotrophin, nerve growth factor (NGF), was loaded in the PEGPCL hydrogel layer, and coating cytotoxicity and sustained NGF release were evaluated using a PC12 cell culture model. Quantitative MTT assays showed that these coatings had no significant toxicity toward PC12 cells, and neurite extension at day 7 and 14 confirmed sustained release of NGF at biologically significant concentrations for at least 2 weeks. Our results demonstrate that hydrogel–EFM composite materials can be applied to neural prostheses to improve neuron–electrode proximity and enhance long-term device performance and function.
Collapse
Affiliation(s)
- Ning Han
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|