1
|
Carnicer-Lombarte A, Malliaras GG, Barone DG. The Future of Biohybrid Regenerative Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2408308. [PMID: 39564751 DOI: 10.1002/adma.202408308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/13/2024] [Indexed: 11/21/2024]
Abstract
Biohybrid regenerative bioelectronics are an emerging technology combining implantable devices with cell transplantation. Once implanted, biohybrid regenerative devices integrate with host tissue. The combination of transplant and device provides an avenue to both replace damaged or dysfunctional tissue, and monitor or control its function with high precision. While early challenges in the fusion of the biological and technological components limited development of biohybrid regenerative technologies, progress in the field has resulted in a rapidly increasing number of applications. In this perspective the great potential of this emerging technology for the delivery of therapy is discussed, including both recent research progress and potential new directions. Then the technology barriers are discussed that will need to be addressed to unlock the full potential of biohybrid regenerative devices.
Collapse
Affiliation(s)
| | - George G Malliaras
- Department of Engineering, Electrical Engineering Division, University of Cambridge, Cambridge, CB3 0FA, UK
| | - Damiano G Barone
- Department of Engineering, Electrical Engineering Division, University of Cambridge, Cambridge, CB3 0FA, UK
- Department of Neurosurgery, Houston Methodist, Houston, 77030, USA
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, CB2 0QQ, UK
| |
Collapse
|
2
|
Christensen MB, Cui XT, Rieth L, Warren DJ. Editorial: Biocompatibility of implanted devices, modulation, and repair in the nervous system. Front Neurosci 2024; 18:1505912. [PMID: 39529699 PMCID: PMC11551927 DOI: 10.3389/fnins.2024.1505912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Michael B. Christensen
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
- Department of Otolaryngology—Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Xinyan T. Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Loren Rieth
- Department of Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, United States
- Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - David J. Warren
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
3
|
Boddeti U, Langbein J, McAfee D, Altshuler M, Bachani M, Zaveri HP, Spencer D, Zaghloul KA, Ksendzovsky A. Modeling seizure networks in neuron-glia cultures using microelectrode arrays. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1441345. [PMID: 39290793 PMCID: PMC11405204 DOI: 10.3389/fnetp.2024.1441345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024]
Abstract
Epilepsy is a common neurological disorder, affecting over 65 million people worldwide. Unfortunately, despite resective surgery, over 30 % of patients with drug-resistant epilepsy continue to experience seizures. Retrospective studies considering connectivity using intracranial electrocorticography (ECoG) obtained during neuromonitoring have shown that treatment failure is likely driven by failure to consider critical components of the seizure network, an idea first formally introduced in 2002. However, current studies only capture snapshots in time, precluding the ability to consider seizure network development. Over the past few years, multiwell microelectrode arrays have been increasingly used to study neuronal networks in vitro. As such, we sought to develop a novel in vitro MEA seizure model to allow for study of seizure networks. Specifically, we used 4-aminopyridine (4-AP) to capture hyperexcitable activity, and then show increased network changes after 2 days of chronic treatment. We characterize network changes using functional connectivity measures and a novel technique using dimensionality reduction. We find that 4-AP successfully captures persistently elevated mean firing rate and significant changes in underlying connectivity patterns. We believe this affords a robust in vitro seizure model from which longitudinal network changes can be studied, laying groundwork for future studies exploring seizure network development.
Collapse
Affiliation(s)
- Ujwal Boddeti
- Surgical Neurology Branch, NINDS, National Institutes of Health, Baltimore, MD, United States
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jenna Langbein
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Darrian McAfee
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelle Altshuler
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Muzna Bachani
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Hitten P Zaveri
- Department of Neurology, Yale University, New Haven, CT, United States
| | - Dennis Spencer
- Department of Neurosurgery, Yale University, New Haven, CT, United States
| | - Kareem A Zaghloul
- Surgical Neurology Branch, NINDS, National Institutes of Health, Baltimore, MD, United States
| | - Alexander Ksendzovsky
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Cao B, Huang Y, Chen L, Jia W, Li D, Jiang Y. Soft bioelectronics for diagnostic and therapeutic applications in neurological diseases. Biosens Bioelectron 2024; 259:116378. [PMID: 38759308 DOI: 10.1016/j.bios.2024.116378] [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: 02/18/2024] [Revised: 04/13/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024]
Abstract
Physical and chemical signals in the central nervous system yield crucial information that is clinically relevant under both physiological and pathological conditions. The emerging field of bioelectronics focuses on the monitoring and manipulation of neurophysiological signals with high spatiotemporal resolution and minimal invasiveness. Significant advances have been realized through innovations in materials and structural design, which have markedly enhanced mechanical and electrical properties, biocompatibility, and overall device performance. The diagnostic and therapeutic potential of soft bioelectronics has been corroborated across a diverse array of pre-clinical settings. This review summarizes recent studies that underscore the developments and applications of soft bioelectronics in neurological disorders, including neuromonitoring, neuromodulation, tumor treatment, and biosensing. Limitations and outlooks of soft devices are also discussed in terms of power supply, wireless control, biocompatibility, and the integration of artificial intelligence. This review highlights the potential of soft bioelectronics as a future platform to promote deciphering brain functions and clinical outcomes of neurological diseases.
Collapse
Affiliation(s)
- Bowen Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China; Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, United States
| | - Yewei Huang
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, United States
| | - Liangpeng Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China.
| | - Deling Li
- Department of Neurosurgery, Beijing Tiantan Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China; Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China.
| | - Yuanwen Jiang
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, United States.
| |
Collapse
|
5
|
O'Sullivan KP, Coats B. Coupled Eulerian-Lagrangian model prediction of neural tissue strain during microelectrode insertion. J Neural Eng 2024; 21:046055. [PMID: 39074496 DOI: 10.1088/1741-2552/ad68a6] [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: 02/19/2024] [Accepted: 07/29/2024] [Indexed: 07/31/2024]
Abstract
Objective.Implanted neural microelectrodes are an important tool for recording from and stimulating the cerebral cortex. The performance of chronically implanted devices, however, is often hindered by the development of a reactive tissue response. Previous computational models have investigated brain strain from micromotions of neural electrodes after they have been inserted, to investigate design parameters that might minimize triggers to the reactive tissue response. However, these models ignore tissue damage created during device insertion, an important contributing factor to the severity of inflammation. The objective of this study was to evaluate the effect of electrode geometry, insertion speed, and surface friction on brain tissue strain during insertion.Approach. Using a coupled Eulerian-Lagrangian approach, we developed a 3D finite element model (FEM) that simulates the dynamic insertion of a neural microelectrode in brain tissue. Geometry was varied to investigate tip bluntness, cross-sectional shape, and shank thickness. Insertion velocities were varied from 1 to 8 m s-1. Friction was varied from frictionless to 0.4. Tissue strain and potential microvasculature hemorrhage radius were evaluated for brain regions along the electrode shank and near its tip.Main results. Sharper tips resulted in higher mean max principal strains near the tip except for the bluntest tip on the square cross-section electrode, which exhibited high compressive strain values due to stress concentrations at the corners. The potential vascular damage radius around the electrode was primarily a function of the shank diameter, with smaller shank diameters resulting in smaller distributions of radial strain around the electrode. However, the square shank interaction with the tip taper length caused unique strain distributions that increased the damage radius in some cases. Faster insertion velocities created more strain near the tip but less strain along the shank. Increased friction between the brain and electrode created more strain near the electrode tip and along the shank, but frictionless interactions resulted in increased tearing of brain tissue near the tip.Significance. These results demonstrate the first dynamic FEM study of neural electrode insertion, identifying design factors that can reduce tissue strain and potentially mitigate initial reactive tissue responses due to traumatic microelectrode array insertion.
Collapse
Affiliation(s)
- K P O'Sullivan
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States of America
| | - B Coats
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
6
|
Song SS, Druschel LN, Kasthuri NM, Wang JJ, Conard JH, Chan ER, Acharya AP, Capadona JR. Comprehensive proteomic analysis of the differential expression of 62 proteins following intracortical microelectrode implantation. Sci Rep 2024; 14:17596. [PMID: 39080300 PMCID: PMC11289480 DOI: 10.1038/s41598-024-68017-2] [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: 02/20/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Intracortical microelectrodes (IMEs) are devices designed to be implanted into the cerebral cortex for various neuroscience and neuro-engineering applications. A critical feature of IMEs is their ability to detect neural activity from individual neurons. Currently, IMEs are limited by chronic failure, largely considered to be caused by the prolonged neuroinflammatory response to the implanted devices. Over the past few years, the characterization of the neuroinflammatory response has grown in sophistication, with the most recent advances focusing on mRNA expression following IME implantation. While gene expression studies increase our broad understanding of the relationship between IMEs and cortical tissue, advanced proteomic techniques have not been reported. Proteomic evaluation is necessary to describe the diverse changes in protein expression specific to neuroinflammation, neurodegeneration, or tissue and cellular viability, which could lead to the further development of targeted intervention strategies designed to improve IME functionality. In this study, we have characterized the expression of 62 proteins within 180 μm of the IME implant site at 4-, 8-, and 16-weeks post-implantation. We identified potential targets for immunotherapies, as well as key pathways that contribute to neuronal dieback around the IME implant.
Collapse
Affiliation(s)
- Sydney S Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Niveda M Kasthuri
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Jaime J Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Jacob H Conard
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Abhinav P Acharya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
7
|
Abbott JR, Jeakle EN, Haghighi P, Usoro JO, Sturgill BS, Wu Y, Geramifard N, Radhakrishna R, Patnaik S, Nakajima S, Hess J, Mehmood Y, Devata V, Vijayakumar G, Sood A, Doan Thai TT, Dogra K, Hernandez-Reynoso AG, Pancrazio JJ, Cogan SF. Planar amorphous silicon carbide microelectrode arrays for chronic recording in rat motor cortex. Biomaterials 2024; 308:122543. [PMID: 38547834 PMCID: PMC11065583 DOI: 10.1016/j.biomaterials.2024.122543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/21/2024]
Abstract
Chronic implantation of intracortical microelectrode arrays (MEAs) capable of recording from individual neurons can be used for the development of brain-machine interfaces. However, these devices show reduced recording capabilities under chronic conditions due, at least in part, to the brain's foreign body response (FBR). This creates a need for MEAs that can minimize the FBR to possibly enable long-term recording. A potential approach to reduce the FBR is the use of MEAs with reduced cross-sectional geometries. Here, we fabricated 4-shank amorphous silicon carbide (a-SiC) MEAs and implanted them into the motor cortex of seven female Sprague-Dawley rats. Each a-SiC MEA shank was 8 μm thick by 20 μm wide and had sixteen sputtered iridium oxide film (SIROF) electrodes (4 per shank). A-SiC was chosen as the fabrication base for its high chemical stability, good electrical insulation properties, and amenability to thin film fabrication. Electrochemical analysis and neural recordings were performed weekly for 4 months. MEAs were characterized pre-implantation in buffered saline and in vivo using electrochemical impedance spectroscopy and cyclic voltammetry at 50 mV/s and 50,000 mV/s. Neural recordings were analyzed for single unit activity. At the end of the study, animals were sacrificed for immunohistochemical analysis. We observed statistically significant, but small, increases in 1 and 30 kHz impedance values and 50,000 mV/s charge storage capacity over the 16-week implantation period. Slow sweep 50 mV/s CV and 1 Hz impedance did not significantly change over time. Impedance values increased from 11.6 MΩ to 13.5 MΩ at 1 Hz, 1.2 MΩ-2.9 MΩ at 1 kHz, and 0.11 MΩ-0.13 MΩ at 30 kHz over 16 weeks. The median charge storage capacity of the implanted electrodes at 50 mV/s was 58.1 mC/cm2 on week 1 and 55.9 mC/cm2 on week 16, and at 50,000 mV/s, 4.27 mC/cm2 on week 1 and 5.93 mC/cm2 on week 16. Devices were able to record neural activity from 92% of all active channels at the beginning of the study, At the study endpoint, a-SiC devices were still recording single-unit activity on 51% of electrochemically active electrode channels. In addition, we observed that the signal-to-noise ratio experienced a small decline of -0.19 per week. We also classified observed units as fast and slow repolarizing based on the trough-to-peak time. Although the overall presence of single units declined, fast and slow repolarizing units declined at a similar rate. At recording electrode depth, immunohistochemistry showed minimal tissue response to the a-SiC devices, as indicated by statistically insignificant differences in activated glial cell response between implanted brains slices and contralateral sham slices at 150 μm away from the implant location, as evidenced by GFAP staining. NeuN staining revealed the presence of neuronal cell bodies close to the implantation site, again statistically not different from a contralateral sham slice. These results warrant further investigation of a-SiC MEAs for future long-term implantation neural recording studies.
Collapse
Affiliation(s)
- Justin R Abbott
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Eleanor N Jeakle
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Pegah Haghighi
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Joshua O Usoro
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Brandon S Sturgill
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Yupeng Wu
- Department of Materials Science and Engineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Negar Geramifard
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Rahul Radhakrishna
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Sourav Patnaik
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Shido Nakajima
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Jordan Hess
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Yusef Mehmood
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Veda Devata
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, United States
| | - Gayathri Vijayakumar
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Armaan Sood
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Teresa Thuc Doan Thai
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Komal Dogra
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Ana G Hernandez-Reynoso
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Joseph J Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Stuart F Cogan
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States.
| |
Collapse
|
8
|
Jung T, Zeng N, Fabbri JD, Eichler G, Li Z, Willeke K, Wingel KE, Dubey A, Huq R, Sharma M, Hu Y, Ramakrishnan G, Tien K, Mantovani P, Parihar A, Yin H, Oswalt D, Misdorp A, Uguz I, Shinn T, Rodriguez GJ, Nealley C, Gonzales I, Roukes M, Knecht J, Yoshor D, Canoll P, Spinazzi E, Carloni LP, Pesaran B, Patel S, Youngerman B, Cotton RJ, Tolias A, Shepard KL. Stable, chronic in-vivo recordings from a fully wireless subdural-contained 65,536-electrode brain-computer interface device. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594333. [PMID: 38798494 PMCID: PMC11118429 DOI: 10.1101/2024.05.17.594333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Minimally invasive, high-bandwidth brain-computer-interface (BCI) devices can revolutionize human applications. With orders-of-magnitude improvements in volumetric efficiency over other BCI technologies, we developed a 50-μm-thick, mechanically flexible micro-electrocorticography (μECoG) BCI, integrating 256×256 electrodes, signal processing, data telemetry, and wireless powering on a single complementary metal-oxide-semiconductor (CMOS) substrate containing 65,536 recording and 16,384 stimulation channels, from which we can simultaneously record up to 1024 channels at a given time. Fully implanted below the dura, our chip is wirelessly powered, communicating bi-directionally with an external relay station outside the body. We demonstrated chronic, reliable recordings for up to two weeks in pigs and up to two months in behaving non-human primates from somatosensory, motor, and visual cortices, decoding brain signals at high spatiotemporal resolution.
Collapse
|
9
|
Nolta NF, Christensen MB, Tresco PA. Advanced age is not a barrier to chronic intracortical single-unit recording in rat cortex. Front Neurosci 2024; 18:1389556. [PMID: 38817909 PMCID: PMC11138162 DOI: 10.3389/fnins.2024.1389556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Available evidence suggests that as we age, our brain and immune system undergo changes that increase our susceptibility to injury, inflammation, and neurodegeneration. Since a significant portion of the potential patients treated with a microelectrode-based implant may be older, it is important to understand the recording performance of such devices in an aged population. Methods We studied the chronic recording performance and the foreign body response (FBR) to a clinically used microelectrode array implanted in the cortex of 18-month-old Sprague Dawley rats. Results and discussion To the best of our knowledge, this is the first preclinical study of its type in the older mammalian brain. Here, we show that single-unit recording performance was initially robust then gradually declined over a 12-week period, similar to what has been previously reported using younger adult rats and in clinical trials. In addition, we show that FBR biomarker distribution was similar to what has been previously described for younger adult rats implanted with multi-shank recording arrays in the motor cortex. Using a quantitative immunohistochemcal approach, we observed that the extent of astrogliosis and tissue loss near the recording zone was inversely related to recording performance. A comparison of recording performance with a younger cohort supports the notion that aging, in and of itself, is not a limiting factor for the clinical use of penetrating microelectrode recording arrays for the treatment of certain CNS disorders.
Collapse
Affiliation(s)
- Nicholas F. Nolta
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Michael B. Christensen
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
- Department of Otolaryngology – Head & Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Patrick A. Tresco
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
10
|
Song SS, Druschel LN, Conard JH, Wang JJ, Kasthuri NM, Ricky Chan E, Capadona JR. Depletion of complement factor 3 delays the neuroinflammatory response to intracortical microelectrodes. Brain Behav Immun 2024; 118:221-235. [PMID: 38458498 DOI: 10.1016/j.bbi.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/26/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
The neuroinflammatory response to intracortical microelectrodes (IMEs) used with brain-machine interfacing (BMI) applications is regarded as the primary contributor to poor chronic performance. Recent developments in high-plex gene expression technologies have allowed for an evolution in the investigation of individual proteins or genes to be able to identify specific pathways of upregulated genes that may contribute to the neuroinflammatory response. Several key pathways that are upregulated following IME implantation are involved with the complement system. The complement system is part of the innate immune system involved in recognizing and eliminating pathogens - a significant contributor to the foreign body response against biomaterials. Specifically, we have identified Complement 3 (C3) as a gene of interest because it is the intersection of several key complement pathways. In this study, we investigated the role of C3 in the IME inflammatory response by comparing the neuroinflammatory gene expression at the microelectrode implant site between C3 knockout (C3-/-) and wild-type (WT) mice. We have found that, like in WT mice, implantation of intracortical microelectrodes in C3-/- mice yields a dramatic increase in the neuroinflammatory gene expression at all post-surgery time points investigated. However, compared to WT mice, C3 depletion showed reduced expression of many neuroinflammatory genes pre-surgery and 4 weeks post-surgery. Conversely, depletion of C3 increased the expression of many neuroinflammatory genes at 8 weeks and 16 weeks post-surgery, compared to WT mice. Our results suggest that C3 depletion may be a promising therapeutic target for acute, but not chronic, relief of the neuroinflammatory response to IME implantation. Additional compensatory targets may also be required for comprehensive long-term reduction of the neuroinflammatory response for improved intracortical microelectrode performance.
Collapse
Affiliation(s)
- Sydney S Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - Jacob H Conard
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States.
| | - Jaime J Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - Niveda M Kasthuri
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States.
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
11
|
Yang Q, Vazquez AL, Cui XT. Revealing in vivo cellular mechanisms of cerebral microbleeds on neurons and microglia across cortical layers. iScience 2024; 27:109371. [PMID: 38510113 PMCID: PMC10951986 DOI: 10.1016/j.isci.2024.109371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/28/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Cerebral microbleeds (CMBs) are associated with higher risk for various neurological diseases including stroke, dementia, and Alzheimer's disease. However, the understanding of cellular pathology of CMBs, particularly in deep brain regions, remains limited. Utilizing two-photon microscopy and microprism implantation, we longitudinally imaged the impact of CMBs on neuronal and microglial activities across cortical depths in awake mice. A temporary decline in spontaneous neuronal activity occurred throughout cortical layers, followed by recovery within a week. However, significant changes of neuron-neuron activity correlations persisted for weeks. Moreover, microglial contact with neuron soma significantly increased post-microbleeds, indicating an important modulatory role of microglia. Notably, microglial contact, negatively correlated with neuronal firing rate in normal conditions, became uncorrelated after microbleeds, suggesting a decreased neuron-microglia inhibition. These findings reveal chronic alterations in cortical neuronal networks and microglial-neuronal interactions across cortical depths, shedding light on the pathology of CMBs.
Collapse
Affiliation(s)
- Qianru Yang
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alberto L. Vazquez
- Center for Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - X. Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
McNamara IN, Wellman SM, Li L, Eles JR, Savya S, Sohal HS, Angle MR, Kozai TDY. Electrode sharpness and insertion speed reduce tissue damage near high-density penetrating arrays. J Neural Eng 2024; 21:026030. [PMID: 38518365 DOI: 10.1088/1741-2552/ad36e1] [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: 11/22/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
Objective. Over the past decade, neural electrodes have played a crucial role in bridging biological tissues with electronic and robotic devices. This study focuses on evaluating the optimal tip profile and insertion speed for effectively implanting Paradromics' high-density fine microwire arrays (FμA) prototypes into the primary visual cortex (V1) of mice and rats, addressing the challenges associated with the 'bed-of-nails' effect and tissue dimpling.Approach. Tissue response was assessed by investigating the impact of electrodes on the blood-brain barrier (BBB) and cellular damage, with a specific emphasis on tailored insertion strategies to minimize tissue disruption during electrode implantation.Main results.Electro-sharpened arrays demonstrated a marked reduction in cellular damage within 50μm of the electrode tip compared to blunt and angled arrays. Histological analysis revealed that slow insertion speeds led to greater BBB compromise than fast and pneumatic methods. Successful single-unit recordings validated the efficacy of the optimized electro-sharpened arrays in capturing neural activity.Significance.These findings underscore the critical role of tailored insertion strategies in minimizing tissue damage during electrode implantation, highlighting the suitability of electro-sharpened arrays for long-term implant applications. This research contributes to a deeper understanding of the complexities associated with high-channel-count microelectrode array implantation, emphasizing the importance of meticulous assessment and optimization of key parameters for effective integration and minimal tissue disruption. By elucidating the interplay between insertion parameters and tissue response, our study lays a strong foundation for the development of advanced implantable devices with a reduction in reactive gliosis and improved performance in neural recording applications.
Collapse
Affiliation(s)
- Ingrid N McNamara
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Lehong Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sajishnu Savya
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | | | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center of the Basis of Neural Cognition, Pittsburgh, PA, United States of America
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
13
|
Song S, Druschel L, Kasthuri N, Wang J, Conard J, Chan E, Acharya A, Capadona J. Comprehensive Proteomic Analysis of the Differential Expression of 83 Proteins Following Intracortical Microelectrode Implantation. RESEARCH SQUARE 2024:rs.3.rs-4039586. [PMID: 38559066 PMCID: PMC10980140 DOI: 10.21203/rs.3.rs-4039586/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Intracortical microelectrodes (IMEs) are devices designed to be implanted into the cerebral cortex for various neuroscience and neuro-engineering applications. A critical feature of these devices is their ability to detect neural activity from individual neurons. Currently, IMEs are limited by chronic failure, largely considered to be caused by the prolonged neuroinflammatory response to the implanted devices. Over the decades, characterization of the neuroinflammatory response has grown in sophistication, with the most recent advances including advanced genomics and spatially resolved transcriptomics. While gene expression studies increase our broad understanding of the relationship between IMEs and cortical tissue, advanced proteomic techniques have not been reported. Proteomic evaluation is necessary to describe the diverse changes in protein expression specific to neuroinflammation, neurodegeneration, or tissue and cellular viability, which could lead to the development of more targeted intervention strategies designed to improve IME function. In this study, we have characterized the expression of 83 proteins within 180 μm of the IME implant site at 4-, 8-, and 16-weeks post-implantation. We identified potential targets for immunotherapies, as well as key pathways and functions that contribute to neuronal dieback around the IME implant.
Collapse
|
14
|
Capadona J, Hoeferlin G, Grabinski S, Druschel L, Duncan J, Burkhart G, Weagraff G, Lee A, Hong C, Bambroo M, Olivares H, Bajwa T, Memberg W, Sweet J, Hamedani HA, Acharya A, Hernandez-Reynoso A, Donskey C, Jaskiw G, Chan R, Ajiboye A, von Recum H, Zhang L. Bacteria Invade the Brain Following Sterile Intracortical Microelectrode Implantation. RESEARCH SQUARE 2024:rs.3.rs-3980065. [PMID: 38496527 PMCID: PMC10942555 DOI: 10.21203/rs.3.rs-3980065/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Brain-machine interface performance is largely affected by the neuroinflammatory responses resulting in large part from blood-brain barrier (BBB) damage following intracortical microelectrode implantation. Recent findings strongly suggest that certain gut bacterial constituents penetrate the BBB and are resident in various brain regions of rodents and humans, both in health and disease. Therefore, we hypothesized that damage to the BBB caused by microelectrode implantation could amplify dysregulation of the microbiome-gut-brain axis. Here, we report that bacteria, including those commonly found in the gut, enter the brain following intracortical microelectrode implantation in mice implanted with single-shank silicon microelectrodes. Systemic antibiotic treatment of mice implanted with microelectrodes to suppress bacteria resulted in differential expression of bacteria in the brain tissue and a reduced acute inflammatory response compared to untreated controls, correlating with temporary improvements in microelectrode recording performance. Long-term antibiotic treatment resulted in worsening microelectrode recording performance and dysregulation of neurodegenerative pathways. Fecal microbiome composition was similar between implanted mice and an implanted human, suggesting translational findings. However, a significant portion of invading bacteria was not resident in the brain or gut. Together, the current study established a paradigm-shifting mechanism that may contribute to chronic intracortical microelectrode recording performance and affect overall brain health following intracortical microelectrode implantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ricky Chan
- Institute for Computational Biology, Case Western Reserve University
| | | | | | | |
Collapse
|
15
|
Schlotterose L, Cossais F, Lucius R, Hattermann K. Resveratrol Alleviates the Early Challenges of Implant-Based Drug Delivery in a Human Glial Cell Model. Int J Mol Sci 2024; 25:2078. [PMID: 38396755 PMCID: PMC10889494 DOI: 10.3390/ijms25042078] [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: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Brain diseases are oftentimes life-threatening and difficult to treat. The local administration of drug substances using brain implants can increase on-site concentrations and decrease systemic side effects. However, the biocompatibility of potential brain implant materials needs to be evaluated carefully as implants can trigger foreign body reactions, particularly by increasing the microglia and astrocyte reactivity. To date, these tests have been frequently conducted in very simple in vitro models, in particular not respecting the key players in glial cell reactions and the challenges of surgical implantation characterized by the disruption of oxygen and nutrient supply. Thus, we established an in vitro model in which we treated human glial cell lines with reduced oxygen and glucose levels. The model displayed cytokine and reactive oxygen species release from reactive microglia and an increase in a marker of reactive astrocytes, galectin-3. Moreover, the treatment caused changes in the cell survival and triggered the production of hypoxia-inducible factor 1α. In this comprehensive platform, we demonstrated the protective effect of the natural polyphenol resveratrol as a model substance, which might be included in brain implants to ease the undesired glial cell response. Overall, a glial-cell-based in vitro model of the initial challenges of local brain disease treatment may prove useful for investigating new therapy options.
Collapse
Affiliation(s)
| | | | | | - Kirsten Hattermann
- Institute of Anatomy, Kiel University, 24118 Kiel, Germany; (L.S.); (R.L.)
| |
Collapse
|
16
|
Ma L, Wang W, Zhao Y, Liu M, Ye W, Li X. Application of LRG mechanism in normal pressure hydrocephalus. Heliyon 2024; 10:e23940. [PMID: 38223707 PMCID: PMC10784321 DOI: 10.1016/j.heliyon.2023.e23940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 11/02/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024] Open
Abstract
Normal pressure hydrocephalus (NPH) is a prevalent type of hydrocephalus, including secondary normal pressure hydrocephalus (SNPH) and idiopathic normal pressure hydrocephalus (INPH). However, its clinical diagnosis and pathological mechanism are still unclear. Leucine-rich α-2 glycoprotein (LRG) is involved in various human diseases, including cancer, diabetes, cardiovascular disease, and nervous system diseases. Now the physiological mechanism of LRG is still being explored. According to the current research results on LRG, we found that the agency of LRG has much to do with the known pathological process of NPH. This review focuses on analyzing the LRG signaling pathways and the pathological mechanism of NPH. According to the collected literature evidence, we speculated that LRG probably be involved in the pathological process of NPH. Finally, based on the mechanism of LRG and NPH, we also summarized the evidence of molecular targeted therapies for future research and clinical application.
Collapse
Affiliation(s)
| | | | - Yongqiang Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Menghao Liu
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Wei Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xianfeng Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
17
|
Hernandez-Reynoso AG, Sturgill BS, Hoeferlin GF, Druschel LN, Krebs OK, Menendez DM, Thai TTD, Smith TJ, Duncan J, Zhang J, Mittal G, Radhakrishna R, Desai MS, Cogan SF, Pancrazio JJ, Capadona JR. The effect of a Mn(III)tetrakis(4-benzoic acid)porphyrin (MnTBAP) coating on the chronic recording performance of planar silicon intracortical microelectrode arrays. Biomaterials 2023; 303:122351. [PMID: 37931456 PMCID: PMC10842897 DOI: 10.1016/j.biomaterials.2023.122351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
Intracortical microelectrode arrays (MEAs) are used to record neural activity. However, their implantation initiates a neuroinflammatory cascade, involving the accumulation of reactive oxygen species, leading to interface failure. Here, we coated commercially-available MEAs with Mn(III)tetrakis(4-benzoic acid)porphyrin (MnTBAP), to mitigate oxidative stress. First, we assessed the in vitro cytotoxicity of modified sample substrates. Then, we implanted 36 rats with uncoated, MnTBAP-coated ("Coated"), or (3-Aminopropyl)triethoxysilane (APTES)-coated devices - an intermediate step in the coating process. We assessed electrode performance during the acute (1-5 weeks), sub-chronic (6-11 weeks), and chronic (12-16 weeks) phases after implantation. Three subsets of animals were euthanized at different time points to assess the acute, sub-chronic and chronic immunohistological responses. Results showed that MnTBAP coatings were not cytotoxic in vitro, and their implantation in vivo improved the proportion of electrodes during the sub-chronic and chronic phases; APTES coatings resulted in failure of the neural interface during the chronic phase. In addition, MnTBAP coatings improved the quality of the signal throughout the study and reduced the neuroinflammatory response around the implant as early as two weeks, an effect that remained consistent for months post-implantation. Together, these results suggest that MnTBAP coatings are a potentially useful modification to improve MEA reliability.
Collapse
Affiliation(s)
- Ana G Hernandez-Reynoso
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Brandon S Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - George F Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Olivia K Krebs
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Teresa T D Thai
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Thomas J Smith
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Jonathan Duncan
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Jichu Zhang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Gaurav Mittal
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Rahul Radhakrishna
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Mrudang Spandan Desai
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Stuart F Cogan
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Joseph J Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| |
Collapse
|
18
|
Chen K, Garcia Padilla C, Kiselyov K, Kozai TDY. Cell-specific alterations in autophagy-lysosomal activity near the chronically implanted microelectrodes. Biomaterials 2023; 302:122316. [PMID: 37738741 PMCID: PMC10897938 DOI: 10.1016/j.biomaterials.2023.122316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/22/2023] [Accepted: 09/02/2023] [Indexed: 09/24/2023]
Abstract
Intracortical microelectrodes that can record and stimulate brain activity have become a valuable technique for basic science research and clinical applications. However, long-term implantation of these microelectrodes can lead to progressive neurodegeneration in the surrounding microenvironment, characterized by elevation in disease-associated markers. Dysregulation of autophagy-lysosomal degradation, a major intracellular waste removal process, is considered a key factor in the onset and progression of neurodegenerative diseases. It is plausible that similar dysfunctions in autophagy-lysosomal degradation contribute to tissue degeneration following implantation-induced focal brain injury, ultimately impacting recording performance. To understand how the focal, persistent brain injury caused by long-term microelectrode implantation impairs autophagy-lysosomal pathway, we employed two-photon microscopy and immunohistology. This investigation focused on the spatiotemporal characterization of autophagy-lysosomal activity near the chronically implanted microelectrode. We observed an aberrant accumulation of immature autophagy vesicles near the microelectrode over the chronic implantation period. Additionally, we found deficits in autophagy-lysosomal clearance proximal to the chronic implant, which was associated with an accumulation of autophagy cargo and a reduction in lysosomal protease level during the chronic period. Furthermore, our evidence demonstrates reactive astrocytes have myelin-containing lysosomes near the microelectrode, suggesting its role of myelin engulfment during acute implantation period. Together, this study sheds light on the process of brain tissue degeneration caused by long-term microelectrode implantation, with a specific focus on impaired intracellular waste degradation.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Camila Garcia Padilla
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, 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
|
19
|
Hoeferlin GF, Bajwa T, Olivares H, Zhang J, Druschel LN, Sturgill BS, Sobota M, Boucher P, Duncan J, Hernandez-Reynoso AG, Cogan SF, Pancrazio JJ, Capadona JR. Antioxidant Dimethyl Fumarate Temporarily but Not Chronically Improves Intracortical Microelectrode Performance. MICROMACHINES 2023; 14:1902. [PMID: 37893339 PMCID: PMC10609067 DOI: 10.3390/mi14101902] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023]
Abstract
Intracortical microelectrode arrays (MEAs) can be used in a range of applications, from basic neuroscience research to providing an intimate interface with the brain as part of a brain-computer interface (BCI) system aimed at restoring function for people living with neurological disorders or injuries. Unfortunately, MEAs tend to fail prematurely, leading to a loss in functionality for many applications. An important contributing factor in MEA failure is oxidative stress resulting from chronically inflammatory-activated microglia and macrophages releasing reactive oxygen species (ROS) around the implant site. Antioxidants offer a means for mitigating oxidative stress and improving tissue health and MEA performance. Here, we investigate using the clinically available antioxidant dimethyl fumarate (DMF) to reduce the neuroinflammatory response and improve MEA performance in a rat MEA model. Daily treatment of DMF for 16 weeks resulted in a significant improvement in the recording capabilities of MEA devices during the sub-chronic (Weeks 5-11) phase (42% active electrode yield vs. 35% for control). However, these sub-chronic improvements were lost in the chronic implantation phase, as a more exacerbated neuroinflammatory response occurs in DMF-treated animals by 16 weeks post-implantation. Yet, neuroinflammation was indiscriminate between treatment and control groups during the sub-chronic phase. Although worse for chronic use, a temporary improvement (<12 weeks) in MEA performance is meaningful. Providing short-term improvement to MEA devices using DMF can allow for improved use for limited-duration studies. Further efforts should be taken to explore the mechanism behind a worsened neuroinflammatory response at the 16-week time point for DMF-treated animals and assess its usefulness for specific applications.
Collapse
Affiliation(s)
- George F. Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Tejas Bajwa
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Hannah Olivares
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jichu Zhang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Lindsey N. Druschel
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Brandon S. Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Michael Sobota
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Pierce Boucher
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jonathan Duncan
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Ana G. Hernandez-Reynoso
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Stuart F. Cogan
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Joseph J. Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA (J.J.P.)
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA (H.O.); (J.D.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| |
Collapse
|
20
|
Song S, Druschel LN, Chan ER, Capadona JR. Differential expression of genes involved in the chronic response to intracortical microelectrodes. Acta Biomater 2023; 169:348-362. [PMID: 37507031 PMCID: PMC10528922 DOI: 10.1016/j.actbio.2023.07.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Brain-Machine Interface systems (BMIs) are clinically valuable devices that can provide functional restoration for patients with spinal cord injury or improved integration for patients requiring prostheses. Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for precisely controlling BMIs. However, intracortical microelectrodes have a demonstrated history of progressive decline in the recording performance with time, inhibiting their usefulness. One major contributor to decreased performance is the neuroinflammatory response to the implanted microelectrodes. The neuroinflammatory response can lead to neurodegeneration and the formation of a glial scar at the implant site. Historically, histological imaging of relatively few known cellular and protein markers has characterized the neuroinflammatory response to implanted microelectrode arrays. However, neuroinflammation requires many molecular players to coordinate the response - meaning traditional methods could result in an incomplete understanding. Taking advantage of recent advancements in tools to characterize the relative or absolute DNA/RNA expression levels, a few groups have begun to explore gene expression at the microelectrode-tissue interface. We have utilized a custom panel of ∼813 neuroinflammatory-specific genes developed with NanoString for bulk tissue analysis at the microelectrode-tissue interface. Our previous studies characterized the acute innate immune response to intracortical microelectrodes. Here we investigated the gene expression at the microelectrode-tissue interface in wild-type (WT) mice chronically implanted with nonfunctioning probes. We found 28 differentially expressed genes at chronic time points (4WK, 8WK, and 16WK), many in the complement and extracellular matrix system. Further, the expression levels were relatively stable over time. Genes identified here represent chronic molecular players at the microelectrode implant sites and potential therapeutic targets for the long-term integration of microelectrodes. STATEMENT OF SIGNIFICANCE: Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for the precise control of Brain-Machine Interface systems (BMIs). However, intracortical microelectrodes have a demonstrated history of progressive declines in the recording performance with time, inhibiting their usefulness. One major contributor to the decline in these devices is the neuroinflammatory response against the implanted microelectrodes. Historically, neuroinflammation to implanted microelectrode arrays has been characterized by histological imaging of relatively few known cellular and protein markers. Few studies have begun to develop a more in-depth understanding of the molecular pathways facilitating device-mediated neuroinflammation. Here, we are among the first to identify genetic pathways that could represent targets to improve the host response to intracortical microelectrodes, and ultimately device performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
21
|
Andersen MA, Schouenborg J. Polydimethylsiloxane as a more biocompatible alternative to glass in optogenetics. Sci Rep 2023; 13:16090. [PMID: 37752160 PMCID: PMC10522705 DOI: 10.1038/s41598-023-43297-2] [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: 05/12/2023] [Accepted: 09/21/2023] [Indexed: 09/28/2023] Open
Abstract
Optogenetics is highly useful to stimulate or inhibit defined neuronal populations and is often used together with electrophysiological recordings. Due to poor penetration of light in tissue, there is a need for biocompatible wave guides. Glass wave guides are relatively stiff and known to cause glia reaction that likely influence the activity in the remaining neurons. We developed highly flexible micro wave guides for optogenetics that can be used in combination with long-lasting electrophysiological recordings. We designed and evaluated polydimethylsiloxane (PDMS) mono-fibers, which use the tissue as cladding, with a diameter of 71 ± 10 µm and 126 ± 5 µm. We showed that micro PDMS fibers transmitted 9-33 mW/mm2 light energy enough to activate channelrhodopsin. This was confirmed in acute extracellular recordings in vivo in which optogenetic stimulation through the PDMS fibers generated action potentials in rat hippocampus with a short onset latency. PDMS fibers had significantly less microglia and astrocytic activation in the zone nearest to the implant as compared to glass. There was no obvious difference in number of adjacent neurons between size matched wave guides. Micro PDMS wave guide demonstrates in vivo functionality and improved biocompatibility as compared to glass. This enables the delivery of light with less tissue damage.
Collapse
Affiliation(s)
- Michael Aagaard Andersen
- Neuronano Research Center, Department of Experimental Medicine, Lund University, Lund, Sweden.
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jens Schouenborg
- Neuronano Research Center, Department of Experimental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Tian G, Yang D, Chen C, Duan X, Kim DH, Chen H. Simultaneous Presentation of Dexamethasone and Nerve Growth Factor via Layered Carbon Nanotubes and Polypyrrole to Interface Neural Cells. ACS Biomater Sci Eng 2023; 9:5015-5027. [PMID: 37489848 DOI: 10.1021/acsbiomaterials.3c00593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
The implantation of neural electrodes usually induces acute and chronic inflammation, which can result in the formation of glial scars encapsulating the implanted electrodes and the loss of neurons near the active electrode sites. Local presentation of anti-inflammatory drugs or neural protective factors has been evidenced as an effective strategy to modulate inflammatory responses and promote electrode-neuron integration. Here, a novel delivery system for the simultaneous presentation of both anti-inflammatory drugs (dexamethasone, Dex) and nerve-growth-promoting factors (nerve growth factor, NGF) from the electrode sites was developed via layer-structured carbon nanotubes and conductive polymers. The modified electrodes exhibited higher charge storage capacitance and lower electrochemical impedance compared to unmodified electrodes and electrodes coated with polypyrrole/Dex. Dex released from the functional coating under controlled electrochemical stimulation was able to inhibit the lipopolysaccharide-induced secretion or mRNA transcription of inflammatory cytokines, including nitric oxide, TNF-α, and IL-6 in RAW264.7 cells, and control the activation of cultured astrocytes. In addition, the functional coatings did not show a toxic effect on PC12 cells and primary neural cells but exhibited promoted activities on the adhesion, growth, and neurite extension of neural cells.
Collapse
Affiliation(s)
- Guangzhao Tian
- School of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Dan Yang
- School of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Chunrong Chen
- School of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Xiaoge Duan
- School of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Dong-Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Hailan Chen
- School of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| |
Collapse
|
23
|
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
|
24
|
Böhler C, Vomero M, Soula M, Vöröslakos M, Porto Cruz M, Liljemalm R, Buzsaki G, Stieglitz T, Asplund M. Multilayer Arrays for Neurotechnology Applications (MANTA): Chronically Stable Thin-Film Intracortical Implants. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207576. [PMID: 36935361 DOI: 10.1002/advs.202207576] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/07/2023] [Indexed: 05/18/2023]
Abstract
Flexible implantable neurointerfaces show great promise in addressing one of the major challenges of implantable neurotechnology, namely the loss of signal connected to unfavorable probe tissue interaction. The authors here show how multilayer polyimide probes allow high-density intracortical recordings to be combined with a reliable long-term stable tissue interface, thereby progressing toward chronic stability of implantable neurotechnology. The probes could record 10-60 single units over 5 months with a consistent peak-to-peak voltage at dimensions that ensure robust handling and insulation longevity. Probes that remain in intimate contact with the signaling tissue over months to years are a game changer for neuroscience and, importantly, open up for broader clinical translation of systems relying on neurotechnology to interface the human brain.
Collapse
Affiliation(s)
- Christian Böhler
- Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, 79110, Freiburg, Germany
| | - Maria Vomero
- Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, 79110, Freiburg, Germany
| | - Marisol Soula
- Neuroscience Institute, Langone Medical Center, New York University, New York, 10016, USA
| | - Mihály Vöröslakos
- Neuroscience Institute, Langone Medical Center, New York University, New York, 10016, USA
| | - Maria Porto Cruz
- Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, 79110, Freiburg, Germany
| | - Rickard Liljemalm
- Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110, Freiburg, Germany
| | - György Buzsaki
- Neuroscience Institute, Langone Medical Center, New York University, New York, 10016, USA
- Department of Physiology and Neuroscience, Langone Medical Center, New York University, New York, 10016, USA
- Department of Neurology, Langone Medical Center, New York University, New York, 10016, USA
| | - Thomas Stieglitz
- Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, 79110, Freiburg, Germany
- Bernstein Center Freiburg, University of Freiburg, 79110, Freiburg, Germany
| | - Maria Asplund
- Department of Microsystems Engineering (IMTEK), University of Freiburg, 79110, Freiburg, Germany
- BrainLinks-BrainTools Center, University of Freiburg, 79110, Freiburg, Germany
- Department of Microtechnology and Nanoscience, Chalmers University of Technology, Gothenburg, SE-41296, Sweden
- Division of Nursing and Medical Technology, Luleå University of Technology, Luleå, 97187, Sweden
- Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, 79110, Freiburg, Germany
| |
Collapse
|
25
|
Thompson C, Evans B, Zhao D, Purcell E. Spatiotemporal Expression of RNA-Seq Identified Proteins at the Electrode Interface. Acta Biomater 2023; 164:209-222. [PMID: 37116634 DOI: 10.1016/j.actbio.2023.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Implantation of electrodes in the brain can be used to record from or stimulate neural tissues to treat neurological disease and injury. However, the tissue response to implanted devices can limit their functional longevity. Recent RNA-seq datasets identify hundreds of genes associated with gliosis, neuronal function, myelination, and cellular metabolism that are spatiotemporally expressed in neural tissues following the insertion of microelectrodes. To validate mRNA as a predictor of protein expression, this study evaluates a sub-set of RNA-seq identified proteins (RSIP) at 24-hours, 1-week, and 6-weeks post-implantation using quantitative immunofluorescence methods. This study found that expression of RSIPs associated with glial activation (Glial fibrillary acidic protein (GFAP), Polypyrmidine tract binding protein-1 (Ptbp1)), neuronal structure (Neurofilament heavy chain (Nefh), Proteolipid protein-1 (Plp1), Myelin Basic Protein (MBP)), and iron metabolism (Transferrin (TF), Ferritin heavy chain-1 (Fth1)) reinforce transcriptional data. This study also provides additional context to the cellular distribution of RSIPs using a MATLAB-based approach to quantify immunofluorescence intensity within specific cell types. Ptbp1, TF, and Fth1 were found to be spatiotemporally distributed within neurons, astrocytes, microglia, and oligodendrocytes at the device interface relative to distal and contralateral tissues. The altered distribution of RSIPs relative to distal tissue is largely localized within 100µm of the device injury, which approaches the functional recording range of implanted electrodes. This study provides evidence that RNA-sequencing can be used to predict protein-level changes in cortical tissues and that RSIPs can be further investigated to identify new biomarkers of the tissue response that influence signal quality. STATEMENT OF SIGNIFICANCE: : Microelectrode arrays implanted into the brain are useful tools that can be used to study neuroscience and to treat pathological conditions in a clinical setting. The tissue response to these devices, however, can severely limit their functional longevity. Transcriptomics has deepened the understandings of the tissue response by revealing numerous genes which are differentially expressed following device insertion. This manuscript provides validation for the use of transcriptomics to characterize the tissue response by evaluating a subset of known differentially expressed genes at the protein level around implanted electrodes over time. In additional to validating mRNA-to-protein relationships at the device interface, this study has identified emerging trends in the spatiotemporal distribution of proteins involved with glial activation, neuronal remodeling, and essential iron binding proteins around implanted silicon devices. This study additionally provides a new MATLAB based methodology to quantify protein distribution within discrete cell types at the device interface which may be used as biomarkers for further study or therapeutic intervention in the future.
Collapse
Affiliation(s)
- Cort Thompson
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Blake Evans
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Dorothy Zhao
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Erin Purcell
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America.
| |
Collapse
|
26
|
Urdaneta ME, Kunigk NG, Peñaloza-Aponte JD, Currlin S, Malone IG, Fried SI, Otto KJ. Layer-dependent stability of intracortical recordings and neuronal cell loss. Front Neurosci 2023; 17:1096097. [PMID: 37090803 PMCID: PMC10113640 DOI: 10.3389/fnins.2023.1096097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/13/2023] [Indexed: 04/08/2023] Open
Abstract
Intracortical recordings can be used to voluntarily control external devices via brain-machine interfaces (BMI). Multiple factors, including the foreign body response (FBR), limit the stability of these neural signals over time. Current clinically approved devices consist of multi-electrode arrays with a single electrode site at the tip of each shank, confining the recording interface to a single layer of the cortex. Advancements in manufacturing technology have led to the development of high-density electrodes that can record from multiple layers. However, the long-term stability of neural recordings and the extent of neuronal cell loss around the electrode across different cortical depths have yet to be explored. To answer these questions, we recorded neural signals from rats chronically implanted with a silicon-substrate microelectrode array spanning the layers of the cortex. Our results show the long-term stability of intracortical recordings varies across cortical depth, with electrode sites around L4-L5 having the highest stability. Using machine learning guided segmentation, our novel histological technique, DeepHisto, revealed that the extent of neuronal cell loss varies across cortical layers, with L2/3 and L4 electrodes having the largest area of neuronal cell loss. These findings suggest that interfacing depth plays a major role in the FBR and long-term performance of intracortical neuroprostheses.
Collapse
Affiliation(s)
- Morgan E. Urdaneta
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Nicolas G. Kunigk
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Jesus D. Peñaloza-Aponte
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Seth Currlin
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Ian G. Malone
- Department of Electrical & Computer Engineering, University of Florida, Gainesville, FL, United States
| | - Shelley I. Fried
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Boston Veterans Affairs Healthcare System, Boston, MA, United States
| | - Kevin J. Otto
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Department of Electrical & Computer Engineering, University of Florida, Gainesville, FL, United States
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, United States
- Department of Neurology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
27
|
Letner JG, Patel PR, Hsieh JC, Smith Flores IM, della Valle E, Walker LA, Weiland JD, Chestek CA, Cai D. Post-explant profiling of subcellular-scale carbon fiber intracortical electrodes and surrounding neurons enables modeling of recorded electrophysiology. J Neural Eng 2023; 20:026019. [PMID: 36848679 PMCID: PMC10022369 DOI: 10.1088/1741-2552/acbf78] [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: 10/18/2022] [Revised: 01/12/2023] [Accepted: 02/27/2023] [Indexed: 03/01/2023]
Abstract
Objective.Characterizing the relationship between neuron spiking and the signals that electrodes record is vital to defining the neural circuits driving brain function and informing clinical brain-machine interface design. However, high electrode biocompatibility and precisely localizing neurons around the electrodes are critical to defining this relationship.Approach.Here, we demonstrate consistent localization of the recording site tips of subcellular-scale (6.8µm diameter) carbon fiber electrodes and the positions of surrounding neurons. We implanted male rats with carbon fiber electrode arrays for 6 or 12+ weeks targeting layer V motor cortex. After explanting the arrays, we immunostained the implant site and localized putative recording site tips with subcellular-cellular resolution. We then 3D segmented neuron somata within a 50µm radius from implanted tips to measure neuron positions and health and compare to healthy cortex with symmetric stereotaxic coordinates.Main results.Immunostaining of astrocyte, microglia, and neuron markers confirmed that overall tissue health was indicative of high biocompatibility near the tips. While neurons near implanted carbon fibers were stretched, their number and distribution were similar to hypothetical fibers placed in healthy contralateral brain. Such similar neuron distributions suggest that these minimally invasive electrodes demonstrate the potential to sample naturalistic neural populations. This motivated the prediction of spikes produced by nearby neurons using a simple point source model fit using recorded electrophysiology and the mean positions of the nearest neurons observed in histology. Comparing spike amplitudes suggests that the radius at which single units can be distinguished from others is near the fourth closest neuron (30.7 ± 4.6µm,X-± S) in layer V motor cortex.Significance.Collectively, these data and simulations provide the first direct evidence that neuron placement in the immediate vicinity of the recording site influences how many spike clusters can be reliably identified by spike sorting.
Collapse
Affiliation(s)
- Joseph G Letner
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Jung-Chien Hsieh
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Israel M Smith Flores
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Elena della Valle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Logan A Walker
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, Ann Arbor, MI 48109, United States of America
| | - James D Weiland
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, United States of America
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, United States of America
- Robotics Department, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Dawen Cai
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
| |
Collapse
|
28
|
Kumosa LS. Commonly Overlooked Factors in Biocompatibility Studies of Neural Implants. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205095. [PMID: 36596702 PMCID: PMC9951391 DOI: 10.1002/advs.202205095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Biocompatibility of cutting-edge neural implants, surgical tools and techniques, and therapeutic technologies is a challenging concept that can be easily misjudged. For example, neural interfaces are routinely gauged on how effectively they determine active neurons near their recording sites. Tissue integration and toxicity of neural interfaces are frequently assessed histologically in animal models to determine tissue morphological and cellular changes in response to surgical implantation and chronic presence. A disconnect between histological and efficacious biocompatibility exists, however, as neuronal numbers frequently observed near electrodes do not match recorded neuronal spiking activity. The downstream effects of the myriad surgical and experimental factors involved in such studies are rarely examined when deciding whether a technology or surgical process is biocompatible. Such surgical factors as anesthesia, temperature excursions, bleed incidence, mechanical forces generated, and metabolic conditions are known to have strong systemic and thus local cellular and extracellular consequences. Many tissue markers are extremely sensitive to the physiological state of cells and tissues, thus significantly impacting histological accuracy. This review aims to shed light on commonly overlooked factors that can have a strong impact on the assessment of neural biocompatibility and to address the mismatch between results stemming from functional and histological methods.
Collapse
Affiliation(s)
- Lucas S. Kumosa
- Neuronano Research CenterDepartment of Experimental Medical ScienceMedical FacultyLund UniversityMedicon Village, Byggnad 404 A2, Scheelevägen 8Lund223 81Sweden
| |
Collapse
|
29
|
Patel PR, Welle EJ, Letner JG, Shen H, Bullard AJ, Caldwell CM, Vega-Medina A, Richie JM, Thayer HE, Patil PG, Cai D, Chestek CA. Utah array characterization and histological analysis of a multi-year implant in non-human primate motor and sensory cortices. J Neural Eng 2023; 20:10.1088/1741-2552/acab86. [PMID: 36595323 PMCID: PMC9954796 DOI: 10.1088/1741-2552/acab86] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/14/2022] [Indexed: 12/15/2022]
Abstract
Objective.The Utah array is widely used in both clinical studies and neuroscience. It has a strong track record of safety. However, it is also known that implanted electrodes promote the formation of scar tissue in the immediate vicinity of the electrodes, which may negatively impact the ability to record neural waveforms. This scarring response has been primarily studied in rodents, which may have a very different response than primate brain.Approach.Here, we present a rare nonhuman primate histological dataset (n= 1 rhesus macaque) obtained 848 and 590 d after implantation in two brain hemispheres. For 2 of 4 arrays that remained within the cortex, NeuN was used to stain for neuron somata at three different depths along the shanks. Images were filtered and denoised, with neurons then counted in the vicinity of the arrays as well as a nearby section of control tissue. Additionally, 3 of 4 arrays were imaged with a scanning electrode microscope to evaluate any materials damage that might be present.Main results.Overall, we found a 63% percent reduction in the number of neurons surrounding the electrode shanks compared to control areas. In terms of materials, the arrays remained largely intact with metal and Parylene C present, though tip breakage and cracks were observed on many electrodes.Significance.Overall, these results suggest that the tissue response in the nonhuman primate brain shows similar neuron loss to previous studies using rodents. Electrode improvements, for example using smaller or softer probes, may therefore substantially improve the tissue response and potentially improve the neuronal recording yield in primate cortex.
Collapse
Affiliation(s)
- Paras R. Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Elissa J. Welle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Joseph G. Letner
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Hao Shen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Autumn J. Bullard
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Ciara M. Caldwell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Alexis Vega-Medina
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48019, United States of America
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
| | - Julianna M. Richie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Hope E. Thayer
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Parag G. Patil
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
| | - Dawen Cai
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48019, United States of America
| | - Cynthia A. Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, United States of America
- Robotics Program, University of Michigan, Ann Arbor, MI 48109, United States of America
| |
Collapse
|
30
|
Merken L, Schelles M, Ceyssens F, Kraft M, Janssen P. Thin flexible arrays for long-term multi-electrode recordings in macaque primary visual cortex. J Neural Eng 2022; 19. [PMID: 36215972 DOI: 10.1088/1741-2552/ac98e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/10/2022] [Indexed: 01/11/2023]
Abstract
Objective.Basic, translational and clinical neuroscience are increasingly focusing on large-scale invasive recordings of neuronal activity. However, in large animals such as nonhuman primates and humans-in which the larger brain size with sulci and gyri imposes additional challenges compared to rodents, there is a huge unmet need to record from hundreds of neurons simultaneously anywhere in the brain for long periods of time. Here, we tested the electrical and mechanical properties of thin, flexible multi-electrode arrays (MEAs) inserted into the primary visual cortex of two macaque monkeys, and assessed their magnetic resonance imaging (MRI) compatibility and their capacity to record extracellular activity over a period of 1 year.Approach.To allow insertion of the floating arrays into the visual cortex, the 20 by 100µm2shafts were temporarily strengthened by means of a resorbable poly(lactic-co-glycolic acid) coating.Main results. After manual insertion of the arrays, theex vivoandin vivoMRI compatibility of the arrays proved to be excellent. We recorded clear single-unit activity from up to 50% of the electrodes, and multi-unit activity (MUA) on 60%-100% of the electrodes, which allowed detailed measurements of the receptive fields and the orientation selectivity of the neurons. Even 1 year after insertion, we obtained significant MUA responses on 70%-100% of the electrodes, while the receptive fields remained remarkably stable over the entire recording period.Significance.Thus, the thin and flexible MEAs we tested offer several crucial advantages compared to existing arrays, most notably in terms of brain tissue compliance, scalability, and brain coverage. Future brain-machine interface applications in humans may strongly benefit from this new generation of chronically implanted MEAs.
Collapse
Affiliation(s)
- Lara Merken
- Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven 3000, Belgium.,Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| | - Maarten Schelles
- Micro- and Nanosystems (MNS), Electrical Engineering Department (ESAT), KU Leuven, Leuven 3000, Belgium.,ReVision Implant NV, Haasrode 3053, Belgium
| | | | - Michael Kraft
- Micro- and Nanosystems (MNS), Electrical Engineering Department (ESAT), KU Leuven, Leuven 3000, Belgium.,Leuven Institute for Micro- and Nanotechnology (LIMNI), Leuven 3000, Belgium
| | - Peter Janssen
- Laboratory for Neuro- and Psychophysiology, KU Leuven, Leuven 3000, Belgium.,Leuven Brain Institute, KU Leuven, Leuven 3000, Belgium
| |
Collapse
|
31
|
Veletić M, Apu EH, Simić M, Bergsland J, Balasingham I, Contag CH, Ashammakhi N. Implants with Sensing Capabilities. Chem Rev 2022; 122:16329-16363. [PMID: 35981266 DOI: 10.1021/acs.chemrev.2c00005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Because of the aging human population and increased numbers of surgical procedures being performed, there is a growing number of biomedical devices being implanted each year. Although the benefits of implants are significant, there are risks to having foreign materials in the body that may lead to complications that may remain undetectable until a time at which the damage done becomes irreversible. To address this challenge, advances in implantable sensors may enable early detection of even minor changes in the implants or the surrounding tissues and provide early cues for intervention. Therefore, integrating sensors with implants will enable real-time monitoring and lead to improvements in implant function. Sensor integration has been mostly applied to cardiovascular, neural, and orthopedic implants, and advances in combined implant-sensor devices have been significant, yet there are needs still to be addressed. Sensor-integrating implants are still in their infancy; however, some have already made it to the clinic. With an interdisciplinary approach, these sensor-integrating devices will become more efficient, providing clear paths to clinical translation in the future.
Collapse
Affiliation(s)
- Mladen Veletić
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ehsanul Hoque Apu
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Mitar Simić
- Faculty of Electrical Engineering, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Jacob Bergsland
- The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ilangko Balasingham
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
32
|
Abstract
Neuroprosthetic devices that record and modulate neural activities have demonstrated immense potential for bypassing or restoring lost neurological functions due to neural injuries and disorders. However, implantable electrical devices interfacing with brain tissue are susceptible to a series of inflammatory tissue responses along with mechanical or electrical failures which can affect the device performance over time. Several biomaterial strategies have been implemented to improve device-tissue integration for high quality and stable performance. Ranging from developing smaller, softer, and more flexible electrode designs to introducing bioactive coatings and drug-eluting layers on the electrode surface, such strategies have shown different degrees of success but with limitations. With their hydrophilic properties and specific bioactivities, carbohydrates offer a potential solution for addressing some of the limitations of the existing biomolecular approaches. In this review, we summarize the role of polysaccharides in the central nervous system, with a primary focus on glycoproteins and proteoglycans, to shed light on their untapped potential as biomaterials for neural implants. Utilization of glycosaminoglycans for neural interface and tissue regeneration applications is comprehensively reviewed to provide the current state of carbohydrate-based biomaterials for neural implants. Finally, we will discuss the challenges and opportunities of applying carbohydrate-based biomaterials for neural tissue interfaces.
Collapse
Affiliation(s)
- Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Urdaneta ME, Kunigk NG, Currlin S, Delgado F, Fried SI, Otto KJ. The Long-Term Stability of Intracortical Microstimulation and the Foreign Body Response Are Layer Dependent. Front Neurosci 2022; 16:908858. [PMID: 35769707 PMCID: PMC9234554 DOI: 10.3389/fnins.2022.908858] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Intracortical microstimulation (ICMS) of the somatosensory cortex (S1) can restore sensory function in patients with paralysis. Studies assessing the stability of ICMS have reported heterogeneous responses across electrodes and over time, potentially hindering the implementation and translatability of these technologies. The foreign body response (FBR) and the encapsulating glial scar have been associated with a decay in chronic performance of implanted electrodes. Moreover, the morphology, intrinsic properties, and function of cells vary across cortical layers, each potentially affecting the sensitivity to ICMS as well as the degree of the FBR across cortical depth. However, layer-by-layer comparisons of the long-term stability of ICMS as well as the extent of the astrocytic glial scar change across cortical layers have not been well explored. Here, we implanted silicon microelectrodes with electrode sites spanning all the layers of S1 in rats. Using a behavioral paradigm, we obtained ICMS detection thresholds from all cortical layers for up to 40 weeks. Our results showed that the sensitivity and long-term performance of ICMS is indeed layer dependent. Overall, detection thresholds decreased during the first 7 weeks post-implantation (WPI). This was followed by a period in which thresholds remained stable or increased depending on the interfacing layer: thresholds in L1 and L6 exhibited the most consistent increases over time, while those in L4 and L5 remained the most stable. Furthermore, histological investigation of the tissue surrounding the electrode showed a biological response of microglia and macrophages which peaked at L1, while the area of the astrocytic glial scar peaked at L2/3. Interestingly, the biological response of these FBR markers is less exacerbated at L4 and L5, suggesting a potential link between the FBR and the long-term stability of ICMS. These findings suggest that interfacing depth can play an important role in the design of chronically stable implantable microelectrodes.
Collapse
Affiliation(s)
- Morgan E. Urdaneta
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- *Correspondence: Morgan E. Urdaneta,
| | - Nicolas G. Kunigk
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Seth Currlin
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Francisco Delgado
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Shelley I. Fried
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Boston Veterans Affairs Healthcare System, Boston, MA, United States
| | - Kevin J. Otto
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, United States
- Department of Neurology, University of Florida, Gainesville, FL, United States
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, United States
- Kevin J. Otto,
| |
Collapse
|
34
|
Lu B, Fan P, Wang Y, Dai Y, Xie J, Yang G, Mo F, Xu Z, Song Y, Liu J, Cai X. Neuronal Electrophysiological Activities Detection of Defense Behaviors Using an Implantable Microelectrode Array in the Dorsal Periaqueductal Gray. BIOSENSORS 2022; 12:bios12040193. [PMID: 35448253 PMCID: PMC9032743 DOI: 10.3390/bios12040193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 06/07/2023]
Abstract
Defense is the basic survival mechanism of animals when facing dangers. Previous studies have shown that the midbrain periaqueduct gray (PAG) was essential for the production of defense responses. However, the correlation between the endogenous neuronal activities of the dorsal PAG (dPAG) and different defense behaviors was still unclear. In this article, we designed and manufactured microelectrode arrays (MEAs) whose detection sites were arranged to match the shape and position of dPAG in rats, and modified it with platinum-black nanoparticles to improve the detection performance. Subsequently, we successfully recorded the electrophysiological activities of dPAG neurons via designed MEAs in freely behaving rats before and after exposure to the potent analog of predator odor 2-methyl-2-thiazoline (2-MT). Results demonstrated that 2-MT could cause strong innate fear and a series of defensive behaviors, accompanied by the significantly increased average firing rate and local field potential (LFP) power of neurons in dPAG. We also observed that dPAG participated in different defense behaviors with different degrees of activation, which was significantly stronger in the flight stage. Further analysis showed that the neuronal activities of dPAG neurons were earlier than flight, and the intensity of activation was inversely proportional to the distance from predator odor. Overall, our results indicate that dPAG neuronal activities play a crucial role in controlling different types of predator odor-evoked innate fear/defensive behaviors, and provide some guidance for the prediction of defense behavior.
Collapse
Affiliation(s)
- Botao Lu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Penghui Fan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yiding Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuchuan Dai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingyu Xie
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gucheng Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fan Mo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaojie Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China; (B.L.); (P.F.); (Y.W.); (Y.D.); (J.X.); (G.Y.); (F.M.); (Z.X.); (Y.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
35
|
Otte E, Vlachos A, Asplund M. Engineering strategies towards overcoming bleeding and glial scar formation around neural probes. Cell Tissue Res 2022; 387:461-477. [PMID: 35029757 PMCID: PMC8975777 DOI: 10.1007/s00441-021-03567-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022]
Abstract
Neural probes are sophisticated electrophysiological tools used for intra-cortical recording and stimulation. These microelectrode arrays, designed to penetrate and interface the brain from within, contribute at the forefront of basic and clinical neuroscience. However, one of the challenges and currently most significant limitations is their ‘seamless’ long-term integration into the surrounding brain tissue. Following implantation, which is typically accompanied by bleeding, the tissue responds with a scarring process, resulting in a gliotic region closest to the probe. This glial scarring is often associated with neuroinflammation, neurodegeneration, and a leaky blood–brain interface (BBI). The engineering progress on minimizing this reaction in the form of improved materials, microfabrication, and surgical techniques is summarized in this review. As research over the past decade has progressed towards a more detailed understanding of the nature of this biological response, it is time to pose the question: Are penetrating probes completely free from glial scarring at all possible?
Collapse
|
36
|
Jang JW, Kang YN, Seo HW, Kim B, Choe HK, Park SH, Lee MG, Kim S. Long-term in-vivorecording performance of flexible penetrating microelectrode arrays. J Neural Eng 2021; 18. [PMID: 34795067 DOI: 10.1088/1741-2552/ac3656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/02/2021] [Indexed: 11/12/2022]
Abstract
Objective. Neural interfaces are an essential tool to enable the human body to directly communicate with machines such as computers or prosthetic robotic arms. Since invasive electrodes can be located closer to target neurons, they have advantages such as precision in stimulation and high signal-to-noise ratio (SNR) in recording, while they often exhibit unstable performance in long-termin-vivoimplantation because of the tissue damage caused by the electrodes insertion. In the present study, we investigated the electrical functionality of flexible penetrating microelectrode arrays (FPMAs) up to 3 months inin-vivoconditions.Approach. Thein-vivoexperiment was performed by implanting FPMAs in five rats. Thein-vivoimpedance as well as the action potential (AP) amplitude and SNR were analyzed over weeks. Additionally, APs were tracked over time to investigate the possibility of single neuron recording.Main results. It was observed that the FPMAs exhibited dramatic increases in impedance for the first 4 weeks after implantation, accompanied by decreases in AP amplitude. However, the increase/decrease in AP amplitude was always accompanied by the increase/decrease in background noise, resulting in quite consistently maintained SNRs. After 4 weeks of implantation, we observed two distinctive issues regarding long-term implantation, each caused by chronic tissue responses or by the delamination of insulation layer. The results demonstrate that the FPMAs successfully recorded neuronal signals up to 12 weeks, with very stably maintained SNRs, reduced by only 16.1% on average compared to the first recordings, although biological tissue reactions or physical degradation of the FPMA were present.Significance. The fabricated FPMAs successfully recorded intracortical signals for 3 months. The SNR was maintained up to 3 months and the chronic function of FPMA was comparable with other silicon based implantable electrodes.
Collapse
Affiliation(s)
- Jae-Won Jang
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yoo Na Kang
- Department of Medical Assistant Robot, Korea Institute of Machinery and Materials (KIMM), Daegu, Republic of Korea
| | - Hee Won Seo
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Boil Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Han Kyoung Choe
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sang Hyun Park
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Maan-Gee Lee
- Department of Pharmacology, School of MedicineKyungpook National University, Daegu, Republic of Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Sohee Kim
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| |
Collapse
|
37
|
Kumosa LS, Schouenborg J. Profound alterations in brain tissue linked to hypoxic episode after device implantation. Biomaterials 2021; 278:121143. [PMID: 34653937 DOI: 10.1016/j.biomaterials.2021.121143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022]
Abstract
To enable authentic interfacing with neuronal structures in the brain, preventing alterations of tissue during implantation of devices is critical. By transiently implanting oxygen microsensors into rat cortex cerebri for 2 h, substantial and long lasting (>1 h) hypoxia is routinely generated in surrounding tissues; this hypoxia is linked to implantation generated compressive forces. Preferential loss of larger neurons and reduced metabolic components in surviving neurons indicates decreased viability one week after such hypoxic, compressive implantations. By devising an implantation method that relaxes compressive forces; magnitude and duration of hypoxia generated following such an implantation are ameliorated and neurons appear similar to naïve tissues. In line with these observations, astrocyte proliferation was significantly more pronounced for more hypoxic, compressive implantations. Surprisingly, astrocyte processes were frequently found to traverse cellular boundaries into nearby neuronal nuclei, indicating injury induction of a previously not described astrocyte-neuron interaction. Found more frequently in less hypoxic, force-relaxed insertions and thus correlating to a more beneficial outcome, this finding may suggest a novel protective mechanism. In conclusion, substantial and long lasting insertion induced hypoxia around brain implants, a previously overlooked factor, is linked to significant adverse alterations in nervous tissue.
Collapse
Affiliation(s)
- Lucas S Kumosa
- Neuronano Research Center, Faculty of Medicine, Lund University, Scheelevägen 2, Medicon Village 404A2, 223 81, Lund, Sweden.
| | - Jens Schouenborg
- Neuronano Research Center, Faculty of Medicine, Lund University, Scheelevägen 2, Medicon Village 404A2, 223 81, Lund, Sweden; NanoLund, Lund University, Professorsgatan 1, 223 63, Lund, Sweden.
| |
Collapse
|
38
|
Saldanha RL, Urdaneta ME, Otto KJ. The Role of Electrode-Site Placement in the Long-Term Stability of Intracortical Microstimulation. Front Neurosci 2021; 15:712578. [PMID: 34566563 PMCID: PMC8455844 DOI: 10.3389/fnins.2021.712578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/19/2021] [Indexed: 11/13/2022] Open
Abstract
Intracortical microelectrodes are neuroprosthetic devices used in brain-machine interfaces to both record and stimulate neural activity in the brain. These technologies have been improved by advances in microfabrication, which have led to the creation of subcellular and high-density microelectrodes. The greater number of independent stimulation channels in these devices allows for improved neuromodulation selectivity, compared to single-site microelectrodes. Elements of electrode design such as electrode-site placement can influence the long-term performance of neuroprostheses. Previous studies have shown that electrode-sites placed on the edge of a planar microelectrode have greater chronic recording functionality than sites placed in the center. However, the effect of electrode-site placement on long-term intracortical microstimulation (ICMS) is still unknown. Here, we show that, in rats chronically implanted with custom-made planar silicon microelectrodes, electrode-sites on the tip of the device outperformed those on both the edge and center in terms of the effect per charge delivered, though there is still a slight advantage to using edge sites over center sites for ICMS. Longitudinal analysis of ICMS detection thresholds over a 16-week period revealed that while all sites followed a similar trend over time, the tip and edge sites consistently elicited the behavioral response with less charge compared to center sites. Furthermore, we quantified channel activity over time and found that edge sites remained more active than center sites over time, though the rate of decay of active sites for center and edge sites was comparable. Our results demonstrate that electrode-site placement plays an important role in the long-term stability of intracortical microstimulation and could be a potential factor to consider in the design of future intracortical electrodes.
Collapse
Affiliation(s)
- Ramya L Saldanha
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Morgan E Urdaneta
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States.,Department of Neuroscience, University of Florida, Gainesville, FL, United States.,Department of Materials Science and Engineering, University of Florida, Gainesville, FL, United States.,Department of Neurology, University of Florida, Gainesville, FL, United States.,Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, United States
| |
Collapse
|
39
|
Szabo E, Hess-Dunning A. Irreversible, Self-Aligned Microfluidic Packaging for Chronic Implant Applications. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2021; 31:1-10. [PMID: 35431469 PMCID: PMC9009276 DOI: 10.1088/1361-6439/ac1994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Packaging is an often overlooked component in microfluidic devices for biomedical implant applications. Robust and reliable connectors to interface microscale and macroscale features are especially critical for chronic implant applications. Existing microfluidic packaging methods are incompatible with emerging polymeric materials designed to enhance device integration with the surrounding tissue. A microfluidic connector scheme was developed to promote compatibility with novel materials and implant applications. The connectors and an adhesive wax were printed on a scaffold via additive manufacturing processes. The low-temperature packaging process entailed bonding the connector to a polymer nanocomposite-based intracortical microfluidic probe using an adhesive wax. The robustness of the packaging was assessed by measuring the tensile and shear bond strengths of the connector-adhesive wax-polymer film interface. After soak testing for 4 weeks, the bond strength continued to exceed the force required to infuse fluids through the microfluidic channel. Further, the shear bond strength exceeded typical probe insertion forces by at least 10-fold. These results support the use of the connector and thermal bonding method as a viable option for chronic implant applications.
Collapse
Affiliation(s)
- Emily Szabo
- Case Western Reserve University, Cleveland, OH 44106, USA
| | - Allison Hess-Dunning
- Case Western Reserve University, Cleveland, OH 44106, USA
- Rehabilitation Research and Development, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Cleveland, OH 44106, USA
| |
Collapse
|
40
|
Yin P, Liu Y, Xiao L, Zhang C. Advanced Metallic and Polymeric Coatings for Neural Interfacing: Structures, Properties and Tissue Responses. Polymers (Basel) 2021; 13:2834. [PMID: 34451372 PMCID: PMC8401399 DOI: 10.3390/polym13162834] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Neural electrodes are essential for nerve signal recording, neurostimulation, neuroprosthetics and neuroregeneration, which are critical for the advancement of brain science and the establishment of the next-generation brain-electronic interface, central nerve system therapeutics and artificial intelligence. However, the existing neural electrodes suffer from drawbacks such as foreign body responses, low sensitivity and limited functionalities. In order to overcome the drawbacks, efforts have been made to create new constructions and configurations of neural electrodes from soft materials, but it is also more practical and economic to improve the functionalities of the existing neural electrodes via surface coatings. In this article, recently reported surface coatings for neural electrodes are carefully categorized and analyzed. The coatings are classified into different categories based on their chemical compositions, i.e., metals, metal oxides, carbons, conducting polymers and hydrogels. The characteristic microstructures, electrochemical properties and fabrication methods of the coatings are comprehensively presented, and their structure-property correlations are discussed. Special focus is given to the biocompatibilities of the coatings, including their foreign-body response, cell affinity, and long-term stability during implantation. This review article can provide useful and sophisticated insights into the functional design, material selection and structural configuration for the next-generation multifunctional coatings of neural electrodes.
Collapse
Affiliation(s)
| | - Yang Liu
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China; (P.Y.); (L.X.)
| | | | - Chao Zhang
- Department of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China; (P.Y.); (L.X.)
| |
Collapse
|
41
|
Krishnan A, Forssell M, Du Z, Cui XT, Fedder GK, Kelly SK. Residual voltage as an ad-hoc indicator of electrode damage in biphasic electrical stimulation. J Neural Eng 2021; 18. [PMID: 34400592 DOI: 10.1088/1741-2552/ac028a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/18/2021] [Indexed: 11/12/2022]
Abstract
Objective.We derive and demonstrate how residual voltage (RV) from a biphasic electrical stimulation pulse can be used to recognize degradation at the electrode-tissue interface.Approach.Using a first order model of the electrode-tissue interface and a rectangular biphasic stimulation current waveform, we derive the equations for RV as well as RV growth over several stimulation pulses. To demonstrate the use of RV for damage detection, we simulate accelerated damage on sputtered iridium oxide film (SIROF) electrodes using potential cycling. RV measurements of the degraded electrodes are compared against standard characterization methods of cyclic voltammetry and electrochemical impedance spectroscopy.Main results.Our theoretical discussion illustrates how an intrinsic RV arises even from perfectly balanced biphasic pulses due to leakage via the charge-transfer resistance. Preliminary data inin-vivorat experiments follow the derived model of RV growth, thereby validating our hypothesis that RV is a characteristic of the electrode-tissue interface. RV can therefore be utilized for detecting damage at the electrode. Our experimental results for damage detection show that delamination of SIROF electrodes causes a reduction in charge storage capacity, which in turn reflects a measurable increase in RV.Significance.Chronically implanted electrical stimulation systems with multi-electrode arrays have been the focus of physiological engineering research for the last decade. Changes in RV over time can be a quick and effective method to identify and disconnect faulty electrodes in large arrays. Timely diagnoses of electrode status can ensure optimal long term operation, and prevent further damage to the tissue near these electrodes.
Collapse
Affiliation(s)
- Ashwati Krishnan
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Mats Forssell
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Zhanhong Du
- Shenzhen Institute of Advanced Technology, Shenzhen City, Guangdong, People's Republic of China
| | - X Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Gary K Fedder
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Shawn K Kelly
- VA, Pittsburgh, PA 15240, United States of America.,Institute of Complex Engineered Systems, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| |
Collapse
|
42
|
Francoeur MJ, Tang T, Fakhraei L, Wu X, Hulyalkar S, Cramer J, Buscher N, Ramanathan DR. Chronic, Multi-Site Recordings Supported by Two Low-Cost, Stationary Probe Designs Optimized to Capture Either Single Unit or Local Field Potential Activity in Behaving Rats. Front Psychiatry 2021; 12:678103. [PMID: 34421671 PMCID: PMC8374626 DOI: 10.3389/fpsyt.2021.678103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Rodent models of cognitive behavior have greatly contributed to our understanding of human neuropsychiatric disorders. However, to elucidate the neurobiological underpinnings of such disorders or impairments, animal models are more useful when paired with methods for measuring brain function in awake, behaving animals. Standard tools used for systems-neuroscience level investigations are not optimized for large-scale and high-throughput behavioral battery testing due to various factors including cost, time, poor longevity, and selective targeting limited to measuring only a few brain regions at a time. Here we describe two different "user-friendly" methods for building extracellular electrophysiological probes that can be used to measure either single units or local field potentials in rats performing cognitive tasks. Both probe designs leverage several readily available, yet affordable, commercial products to facilitate ease of production and offer maximum flexibility in terms of brain-target locations that can be scalable (32-64 channels) based on experimental needs. Our approach allows neural activity to be recorded simultaneously with behavior and compared between micro (single unit) and more macro (local field potentials) levels of brain activity in order to gain a better understanding of how local brain regions and their connected networks support cognitive functions in rats. We believe our novel probe designs make collecting electrophysiology data easier and will begin to fill the gap in knowledge between basic and clinical research.
Collapse
Affiliation(s)
- Miranda J. Francoeur
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Tianzhi Tang
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Leila Fakhraei
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Xuanyu Wu
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Sidharth Hulyalkar
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Jessica Cramer
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Nathalie Buscher
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Dhakshin R. Ramanathan
- Mental Health Service, VA San Diego Healthcare System, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
43
|
Lee Y, Shin H, Lee D, Choi S, Cho I, Seo J. A Lubricated Nonimmunogenic Neural Probe for Acute Insertion Trauma Minimization and Long-Term Signal Recording. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100231. [PMID: 34085402 PMCID: PMC8336494 DOI: 10.1002/advs.202100231] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/29/2021] [Indexed: 05/06/2023]
Abstract
Brain-machine interfaces (BMIs) that link the brain to a machine are promising for the treatment of neurological disorders through the bi-directional translation of neural information over extended periods. However, the longevity of such implanted devices remains limited by the deterioration of their signal sensitivity over time due to acute inflammation from insertion trauma and chronic inflammation caused by the foreign body reaction. To address this challenge, a lubricated surface is fabricated to minimize friction during insertion and avoid immunogenicity during neural signal recording. Reduced friction force leads to 86% less impulse on the brain tissue, and thus immediately increases the number of measured signal electrodes by 102% upon insertion. Furthermore, the signal measurable period increases from 8 to 16 weeks due to the prevention of gliosis. By significantly reducing insertion damage and the foreign body reaction, the lubricated immune-stealthy probe surface (LIPS) can maximize the longevity of implantable BMIs.
Collapse
Affiliation(s)
- Yeontaek Lee
- School of Electrical and Electronic EngineeringYonsei UniversitySeoul03722Republic of Korea
| | - Hyogeun Shin
- Center for BioMicrosystemsBrain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Dongwon Lee
- School of Electrical and Electronic EngineeringYonsei UniversitySeoul03722Republic of Korea
| | - Sungah Choi
- School of Electrical and Electronic EngineeringYonsei UniversitySeoul03722Republic of Korea
| | - Il‐Joo Cho
- School of Electrical and Electronic EngineeringYonsei UniversitySeoul03722Republic of Korea
- Center for BioMicrosystemsBrain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & Technology, KIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| | - Jungmok Seo
- School of Electrical and Electronic EngineeringYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
44
|
Szymanski LJ, Kellis S, Liu CY, Jones KT, Andersen RA, Commins D, Lee B, McCreery DB, Miller CA. Neuropathological effects of chronically implanted, intracortical microelectrodes in a tetraplegic patient. J Neural Eng 2021; 18. [PMID: 34314384 DOI: 10.1088/1741-2552/ac127e] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/08/2021] [Indexed: 11/12/2022]
Abstract
Objective.Intracortical microelectrode arrays (MEA) can be used as part of a brain-machine interface system to provide sensory feedback control of an artificial limb to assist persons with tetraplegia. Variability in functionality of electrodes has been reported but few studies in humans have examined the impact of chronic brain tissue responses revealed postmortem on electrode performancein vivo. Approach.In a tetraplegic man, recording MEAs were implanted into the anterior intraparietal area and Brodmann's area 5 (BA5) of the posterior parietal cortex and a recording and stimulation array was implanted in BA1 of the primary somatosensory cortex (S1). The participant expired from unrelated causes seven months after MEA implantation. The underlying tissue of two of the three devices was processed for histology and electrophysiological recordings were assessed.Main results.Recordings of neuronal activity were obtained from all three MEAs despite meningeal encapsulation. However, the S1 array had a greater encapsulation, yielded lower signal quality than the other arrays and failed to elicit somatosensory percepts with electrical stimulation. Histological examination of tissues underlying S1 and BA5 implant sites revealed localized leptomeningeal proliferation and fibrosis, lymphocytic infiltrates, astrogliosis, and foreign body reaction around the electrodes. The BA5 recording site showed focal cerebral microhemorrhages and leptomeningeal vascular ectasia. The S1 site showed focal tissue damage including vascular recanalization, neuronal loss, and extensive subcortical white matter necrosis. The tissue response at the S1 site included hemorrhagic-induced injury suggesting a likely mechanism for reduced function of the S1 implant.Significance.Our findings are similar to those from animal studies with chronic intracortical implants and suggest that vascular disruption and microhemorrhage during device implantation are important contributors to overall array and individual electrode performance and should be a topic for future device development to mitigate tissue responses. Neurosurgical considerations are also discussed.
Collapse
Affiliation(s)
- Linda J Szymanski
- Department of Pathology, Keck USC School of Medicine, Los Angeles, CA, United States of America.,Department of Pathology and Laboratory Medicine, Children's Hospital of Los Angeles, Los Angeles, CA, United States of America
| | - Spencer Kellis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States of America.,Tianqiao and Chrissy Chen Brain-Machine Interface Center, Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, United States of America.,Department of Neurosurgery, Keck USC School of Medicine, Los Angeles, CA, United States of America.,USC Neurorestoration Center, Keck USC School of Medicine, Los Angeles, CA, United States of America
| | - Charles Y Liu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States of America.,Department of Neurosurgery, Keck USC School of Medicine, Los Angeles, CA, United States of America.,USC Neurorestoration Center, Keck USC School of Medicine, Los Angeles, CA, United States of America
| | - Kymry T Jones
- Department of Pathology, Keck USC School of Medicine, Los Angeles, CA, United States of America
| | - Richard A Andersen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States of America.,Tianqiao and Chrissy Chen Brain-Machine Interface Center, Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, United States of America
| | - Deborah Commins
- Department of Pathology, Keck USC School of Medicine, Los Angeles, CA, United States of America
| | - Brian Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States of America.,Department of Neurosurgery, Keck USC School of Medicine, Los Angeles, CA, United States of America.,USC Neurorestoration Center, Keck USC School of Medicine, Los Angeles, CA, United States of America
| | - Douglas B McCreery
- Huntington Medical Research Institute, Pasadena, CA, United States of America
| | - Carol A Miller
- Department of Pathology, Keck USC School of Medicine, Los Angeles, CA, United States of America
| |
Collapse
|
45
|
Sridharan A, Muthuswamy J. Soft, Conductive, Brain-Like, Coatings at Tips of Microelectrodes Improve Electrical Stability under Chronic, In Vivo Conditions. MICROMACHINES 2021; 12:761. [PMID: 34203234 PMCID: PMC8306035 DOI: 10.3390/mi12070761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 01/27/2023]
Abstract
Several recent studies have reported improved histological and electrophysiological outcomes with soft neural interfaces that have elastic moduli ranging from 10 s of kPa to hundreds of MPa. However, many of these soft interfaces use custom fabrication processes. We test the hypothesis that a readily adoptable fabrication process for only coating the tips of microelectrodes with soft brain-like (elastic modulus of ~5 kPa) material improves the long-term electrical performance of neural interfaces. Conventional tungsten microelectrodes (n = 9 with soft coatings and n = 6 uncoated controls) and Pt/Ir microelectrodes (n = 16 with soft coatings) were implanted in six animals for durations ranging from 5 weeks to over 1 year in a subset of rats. Electrochemical impedance spectroscopy was used to assess the quality of the brain tissue-electrode interface under chronic conditions. Neural recordings were assessed for unit activity and signal quality. Electrodes with soft, silicone coatings showed relatively stable electrical impedance characteristics over 6 weeks to >1 year compared to the uncoated control electrodes. Single unit activity recorded by coated electrodes showed larger peak-to-peak amplitudes and increased number of detectable neurons compared to uncoated controls over 6-7 weeks. We demonstrate the feasibility of using a readily translatable process to create brain-like soft interfaces that can potentially overcome variable performance associated with chronic rigid neural interfaces.
Collapse
Affiliation(s)
| | - Jit Muthuswamy
- School of Biological and Health Systems Engineering, Ira A. Fulton School of Engineering, Arizona State University, Tempe, AZ 85287-9709, USA;
| |
Collapse
|
46
|
Boergens KM, Tadić A, Hopper MS, McNamara I, Fell D, Sahasrabuddhe K, Kong Y, Straka M, Sohal HS, Angle MR. Laser ablation of the pia mater for insertion of high-density microelectrode arrays in a translational sheep model. J Neural Eng 2021; 18. [PMID: 34038875 DOI: 10.1088/1741-2552/ac0585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 05/26/2021] [Indexed: 01/03/2023]
Abstract
Objective. The safe insertion of high density intracortical electrode arrays has been a long-standing practical challenge for neural interface engineering and applications such as brain-computer interfaces (BCIs). However, the pia mater can be difficult to penetrate and causes deformation of underlying cortical tissue during insertion of high-density intracortical arrays. This can lead to neuron damage or failed insertions. The development of a method to ease insertion through the pia mater would represent a significant step toward inserting high density intracortical arrays.Approach. Here we describe a surgical procedure, inspired by laser corneal ablation, that can be used in translational models to thin the pia mater.Main results. We demonstrate that controlled pia removal with laser ablation over a small area of cortex allows for microelectrode arrays to be inserted into the cortex with less force, thus reducing deformation of underlying tissue during placement of the microelectrodes. This procedure allows for insertion of high-density electrode arrays and subsequent acute recordings of spiking neuron activity in sheep cortex. We also show histological and electrophysiological evidence that laser removal of the pia does not acutely affect neuronal viability in the region.Significance. Laser ablation of the pia reduces insertion forces of high-density arrays with minimal to no acute damage to cortical neurons. This approach suggests a promising new path for clinical BCI with high-density microelectrode arrays.
Collapse
Affiliation(s)
| | | | | | | | - Devin Fell
- Paradromics, Inc., Austin, TX, United States of America
| | | | - Yifan Kong
- Paradromics, Inc., Austin, TX, United States of America
| | | | | | | |
Collapse
|
47
|
Bouadi O, Tay TL. More Than Cell Markers: Understanding Heterogeneous Glial Responses to Implantable Neural Devices. Front Cell Neurosci 2021; 15:658992. [PMID: 33912015 PMCID: PMC8071943 DOI: 10.3389/fncel.2021.658992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/17/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ouzéna Bouadi
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Faculty of Life Sciences, University of Strasbourg, Strasbourg, France
| | - Tuan Leng Tay
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools Centre, University of Freiburg, Freiburg, Germany.,Freiburg Institute of Advanced Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Dryg I, Xie Y, Bergmann M, Urban G, Shain W, Hofmann UG. Long-term in vivomonitoring of gliotic sheathing of ultrathin entropic coated brain microprobes with fiber-based optical coherence tomography. J Neural Eng 2021; 18. [PMID: 33657543 DOI: 10.1088/1741-2552/abebc2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/03/2021] [Indexed: 11/11/2022]
Abstract
Objective.Microfabricated neuroprosthetic devices have made possible important observations on neuron activity; however, long-term high-fidelity recording performance of these devices has yet to be realized. Tissue-device interactions appear to be a primary source of lost recording performance. The current state of the art for visualizing the tissue response surrounding brain implants in animals is immunohistochemistry + confocal microscopy, which is mainly performed after sacrificing the animal. Monitoring the tissue response as it develops could reveal important features of the response which may inform improvements in electrode design.Approach.Optical coherence tomography (OCT), an imaging technique commonly used in ophthalmology, has already been adapted for imaging of brain tissue. Here, we use OCT to achieve real-time,in vivomonitoring of the tissue response surrounding chronically implanted neural devices. The employed tissue-response-provoking implants are coated with a plasma-deposited nanofilm, which has been demonstrated as a biocompatible and anti-inflammatory interface for indwelling devices. We evaluate the method by comparing the OCT results to traditional histology qualitatively and quantitatively.Main results.The differences in OCT signal across the implantation period between the plasma group and the control reveal that the plasma-type coating of otherwise rigid brain probes (glass) only slightly improve the glial encapsulation in the brain parenchyma indicating that geometrical or mechanical influences are dominating the encapsulation process.Significance.Our approach can long-term monitor and compare the tissue-response to chronically-implanted neural probes with and withour plasma coating in living animal models. Our findings provide valuable insigh to the well acknowledged yet not solved challenge.
Collapse
Affiliation(s)
- Ian Dryg
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Yijing Xie
- Section for Neuroelectronic Systems, Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
| | - Michael Bergmann
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
| | - Gerald Urban
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
| | - William Shain
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Ulrich G Hofmann
- Section for Neuroelectronic Systems, Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
49
|
Atkinson D, D'Souza T, Rajput JS, Tasnim N, Muthuswamy J, Marvi H, Pancrazio JJ. Advances in Implantable Microelectrode Array Insertion and Positioning. Neuromodulation 2021; 25:789-795. [PMID: 33438369 DOI: 10.1111/ner.13355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Microelectrode arrays offer a means to probe the functional circuitry of the brain and the promise of cortical neuroprosthesis for individuals suffering from paralysis or limb loss. These devices are typically comprised of one or more shanks incorporating microelectrode sites, where the shanks are positioned by inserting the devices along a straight path that is normal to the brain surface. The lack of consistent long-term chronic recording technology has driven interest in novel probe design and approaches that go beyond the standard insertion approach that is limited to a single velocity or axis. This review offers a description of typical approaches and associated limitations and surveys emergent methods for implantation of microelectrode arrays, in particular those new approaches that leverage embedded microactuators and extend the insertion direction beyond a single axis. MATERIALS AND METHODS This review paper surveys the current technologies that enable probe implantation, repositioning, and the capability to record/stimulate from a tissue volume. A comprehensive literature search was performed using PubMed, Web of Science, and Google Scholar. RESULTS There has been substantial innovation in the development of microscale and embedded technology that enables probe repositioning to maintain quality recordings in the brain. Innovations in material science have resulted in novel strategies for deployable structures that can record from or stimulate a tissue volume. Moreover, new developments involving magnetically steerable catheters and needles offer an alternative approach to "pull" rather than "push" a probe into the tissue. CONCLUSION We envision the emergence of a new generation of probes and insertion methodologies for neuromodulation applications that enable reliable chronic performance from devices that can be positioned virtually anywhere in the brain.
Collapse
Affiliation(s)
- David Atkinson
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, USA
| | - Tania D'Souza
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, USA
| | - Jai Singh Rajput
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, USA
| | - Nishat Tasnim
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, USA
| | - Jit Muthuswamy
- Department of Biomedical Engineering, School of Biological and Health Systems, Engineering, Arizona State University, Tempe, AZ, USA
| | - Hamid Marvi
- School for Engineering of Matter Transport and Energy, Ira A. Fulton Schools of Engineering, Arizona State University, Tempe, AZ, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
50
|
Harris A. Understanding Charge Transfer on the Clinically Used Conical Utah Electrode Array: Charge Storage Capacity, Electrochemical Impedance Spectroscopy and Effective Electrode Area. J Neural Eng 2021; 18. [PMID: 33401255 DOI: 10.1088/1741-2552/abd897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/05/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The Utah electrode is used for pre/clinical studies on neural recording and stimulation. Anecdotal and empirical reports on their performance have been made, resulting in variable testing methods. An in depth investigation was performed to understand the electrochemical behaviour and charge transfer mechanisms occurring on these clinically important electrodes. APPROACH Platinum and iridium electrodes were assessed by cyclic voltammetry and electrochemical impedance spectroscopy. The effective electrode area was measured by reduction of Ru(NH3)63+. MAIN RESULTS Pristine Utah electrodes have little to no oxide present and the surface roughness is very low. Pristine iridium electrodes pass charge through capacitance and oxide formation. Hydride and anion adsorption occurs on the platinum electrode. Anodic current oxidises both metal surfaces, altering the charge transfer mechanisms at the electrode-solution interface. The charge storage capacity depends on measurement technique and electrode structure, providing no information on charge transfer mechanisms. Electrode oxidation increases pseudocapacitance, reducing impedance. Charge transfer was non-homogeneous, most likely due to the electrode geometry enhancing charge density at the electrode tip and base. Oxidation of the electrode surface enhanced charge transfer inhomogeneity. The effective electrode area could be measured by reduction of Ru(NH3)63+ and calculated with a finite cone geometry. SIGNIFICANCE Increasing electrode pseudocapacitance, demonstrated by metal oxidation, reduces impedance. Increasing electrode capacitance offers a potential route to reducing thermal noise and increasing signal-to-noise ratio of neural recording. The effective electrode area of conical electrodes can be measured. The charge density of the conical electrode was greater than expected on a planar disc electrode, indicating modification of electrode geometry can increase an electrodes safe charge injection capacity. In vivo electrochemical measurements often don't include sufficient details to understand the electrode behaviour. Electrode oxidation most likely accounts for a significant amount of variation in previously published Utah electrode impedance data.
Collapse
Affiliation(s)
- Alex Harris
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Aikenhead Centre for Medical Discovery, Melbourne, Victoria, 3065, AUSTRALIA
| |
Collapse
|