1
|
Villa J, Cury J, Kessler L, Tan X, Richter CP. Enhancing biocompatibility of the brain-machine interface: A review. Bioact Mater 2024; 42:531-549. [PMID: 39308547 PMCID: PMC11416625 DOI: 10.1016/j.bioactmat.2024.08.034] [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: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
In vivo implantation of microelectrodes opens the door to studying neural circuits and restoring damaged neural pathways through direct electrical stimulation and recording. Although some neuroprostheses have achieved clinical success, electrode material properties, inflammatory response, and glial scar formation at the electrode-tissue interfaces affect performance and sustainability. Those challenges can be addressed by improving some of the materials' mechanical, physical, chemical, and electrical properties. This paper reviews materials and designs of current microelectrodes and discusses perspectives to advance neuroprosthetics performance.
Collapse
Affiliation(s)
- Jordan Villa
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Joaquin Cury
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Lexie Kessler
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Xiaodong Tan
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
| | - Claus-Peter Richter
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Biomedical Engineering, Northwestern University, USA
| |
Collapse
|
2
|
Druschel LN, Kasthuri NM, Song SS, Wang JJ, Hess-Dunning A, Chan ER, Capadona JR. Cell-specific spatial profiling of targeted protein expression to characterize the impact of intracortical microelectrode implantation on neuronal health. J Mater Chem B 2024. [PMID: 39479901 PMCID: PMC11525954 DOI: 10.1039/d4tb01628a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024]
Abstract
Intracortical microelectrode arrays (MEAs) can record neuronal activity and advance brain-computer interface (BCI) devices. Implantation of the invasive MEA kills local neurons, which has been documented using immunohistochemistry (IHC). Neuronal nuclear protein (NeuN), a protein that lines the nuclei of exclusively neuronal cells, has been used as a marker for neuronal health and survival for decades in neuroscience and neural engineering. NeuN staining is often used to describe the neuronal response to intracortical microelectrode array (MEA) implantation. However, IHC is semiquantitative, relying on intensity readings rather than directly counting expressed proteins. To supplement previous IHC studies, we evaluated the expression of proteins representing different aspects of neuronal structure or function: microtubule-associated protein 2 (MAP2), neurofilament light (NfL), synaptophysin (SYP), myelin basic protein (MBP), and oligodendrocyte transcription factor 2 (OLIG2) following a neural injury caused by intracortical MEA implantation. Together, these five proteins evaluate the cytoskeletal structure, neurotransmitter release, and myelination of neurons. To fully evaluate neuronal health in NeuN-positive (NeuN+) regions, we only quantified protein expression in NeuN+ regions, making this the first-ever cell-specific spatial profiling evaluation of targeted proteins by multiplex immunochemistry following MEA implantation. We performed our protein quantification along with NeuN IHC to compare the results of the two techniques directly. We found that NeuN immunohistochemical analysis does not show the same trends as MAP2, NfL, SYP, MBP, and OLIG2 expression. Further, we found that all five quantified proteins show a decreased expression pattern that aligns more with historic intracortical MEA recording performance.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - Allison Hess-Dunning
- 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
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, 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
|
3
|
Sands I, Demarco R, Thurber L, Esteban-Linares A, Song D, Meng E, Chen Y. Interface-Mediated Neurogenic Signaling: The Impact of Surface Geometry and Chemistry on Neural Cell Behavior for Regenerative and Brain-Machine Interfacing Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401750. [PMID: 38961531 PMCID: PMC11326983 DOI: 10.1002/adma.202401750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/17/2024] [Indexed: 07/05/2024]
Abstract
Nanomaterial advancements have driven progress in central and peripheral nervous system applications such as tissue regeneration and brain-machine interfacing. Ideally, neural interfaces with native tissue shall seamlessly integrate, a process that is often mediated by the interfacial material properties. Surface topography and material chemistry are significant extracellular stimuli that can influence neural cell behavior to facilitate tissue integration and augment therapeutic outcomes. This review characterizes topographical modifications, including micropillars, microchannels, surface roughness, and porosity, implemented on regenerative scaffolding and brain-machine interfaces. Their impact on neural cell response is summarized through neurogenic outcome and mechanistic analysis. The effects of surface chemistry on neural cell signaling with common interfacing compounds like carbon-based nanomaterials, conductive polymers, and biologically inspired matrices are also reviewed. Finally, the impact of these extracellular mediated neural cues on intracellular signaling cascades is discussed to provide perspective on the manipulation of neuron and neuroglia cell microenvironments to drive therapeutic outcomes.
Collapse
Affiliation(s)
- Ian Sands
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Ryan Demarco
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Laura Thurber
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Alberto Esteban-Linares
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Dong Song
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Ellis Meng
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| |
Collapse
|
4
|
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
|
5
|
Orlemann C, Boehler C, Kooijmans RN, Li B, Asplund M, Roelfsema PR. Flexible Polymer Electrodes for Stable Prosthetic Visual Perception in Mice. Adv Healthc Mater 2024; 13:e2304169. [PMID: 38324245 PMCID: PMC11468866 DOI: 10.1002/adhm.202304169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/01/2024] [Indexed: 02/08/2024]
Abstract
Brain interfaces that can stimulate neurons, cause minimal damage, and work for a long time will be central for future neuroprosthetics. Here, the long-term performance of highly flexible, thin polyimide shanks with several small (<15 µm) electrodes during electrical microstimulation of the visual cortex, is reported. The electrodes exhibit a remarkable stability when several billions of electrical pulses are applied in vitro. When the devices are implanted in the primary visual cortex (area V1) of mice and the animals are trained to detect electrical microstimulation, it is found that the perceptual thresholds are 2-20 microamperes (µA), which is far below the maximal currents that the electrodes can withstand. The long-term functionality of the devices in vivo is excellent, with stable performance for up to more than a year and little damage to the brain tissue. These results demonstrate the potential of thin floating electrodes for the long-term restoration of lost sensory functions.
Collapse
Affiliation(s)
- Corinne Orlemann
- Department of Vision and CognitionNetherlands Institute for NeuroscienceRoyal Netherlands Academy of Arts and SciencesAmsterdam1105 BAThe Netherlands
| | - Christian Boehler
- Department of Microsystems Engineering (IMTEK)University of Freiburg79110FreiburgGermany
- BrainLinks‐BrainTools CenterUniversity of Freiburg79110FreiburgGermany
| | - Roxana N. Kooijmans
- Department of Vision and CognitionNetherlands Institute for NeuroscienceRoyal Netherlands Academy of Arts and SciencesAmsterdam1105 BAThe Netherlands
- Institute for Neuroscience and Medicine (INM‐1)Forschungszentrum Jülich52428JülichGermany
| | - Bingshuo Li
- Department of Vision and CognitionNetherlands Institute for NeuroscienceRoyal Netherlands Academy of Arts and SciencesAmsterdam1105 BAThe Netherlands
| | - Maria Asplund
- Department of Microsystems Engineering (IMTEK)University of Freiburg79110FreiburgGermany
- BrainLinks‐BrainTools CenterUniversity of Freiburg79110FreiburgGermany
- Department of Microtechnology and NanoscienceChalmers University of TechnologyGothenburg412 96Sweden
| | - Pieter R. Roelfsema
- Department of Vision and CognitionNetherlands Institute for NeuroscienceRoyal Netherlands Academy of Arts and SciencesAmsterdam1105 BAThe Netherlands
- Laboratory of Visual Brain TherapySorbonne UniversitéInstitut National de la Santé et de la Recherche MédicaleCentre National de la Recherche ScientifiqueInstitut de la VisionParisF‐75012France
- Department of Integrative NeurophysiologyCentre for Neurogenomics and Cognitive ResearchVU UniversityAmsterdam1081 HVThe Netherlands
- Department of NeurosurgeryAmsterdam University Medical CenterUniversity of AmsterdamAmsterdam1105 AZThe Netherlands
| |
Collapse
|
6
|
Gupta B, Saxena A, Perillo ML, Wade-Kleyn LC, Thompson CH, Purcell EK. Structural, Functional, and Genetic Changes Surrounding Electrodes Implanted in the Brain. Acc Chem Res 2024; 57:1346-1359. [PMID: 38630432 PMCID: PMC11079975 DOI: 10.1021/acs.accounts.4c00057] [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: 01/25/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
Implantable neurotechnology enables monitoring and stimulating of the brain signals responsible for performing cognitive, motor, and sensory tasks. Electrode arrays implanted in the brain are increasingly used in the clinic to treat a variety of sources of neurological diseases and injuries. However, the implantation of a foreign body typically initiates a tissue response characterized by physical disruption of vasculature and the neuropil as well as the initiation of inflammation and the induction of reactive glial states. Likewise, electrical stimulation can induce damage to the surrounding tissue depending on the intensity and waveform parameters of the applied stimulus. These phenomena, in turn, are likely influenced by the surface chemistry and characteristics of the materials employed, but further information is needed to effectively link the biological responses observed to specific aspects of device design. In order to inform improved design of implantable neurotechnology, we are investigating the basic science principles governing device-tissue integration. We are employing multiple techniques to characterize the structural, functional, and genetic changes that occur in the cells surrounding implanted electrodes. First, we have developed a new "device-in-slice" technique to capture chronically implanted electrodes within thick slices of live rat brain tissue for interrogation with single-cell electrophysiology and two-photon imaging techniques. Our data revealed several new observations of tissue remodeling surrounding devices: (a) there was significant disruption of dendritic arbors in neurons near implants, where losses were driven asymmetrically on the implant-facing side. (b) There was a significant loss of dendritic spine densities in neurons near implants, with a shift toward more immature (nonfunctional) morphologies. (c) There was a reduction in excitatory neurotransmission surrounding implants, as evidenced by a reduction in the frequency of excitatory postsynaptic currents (EPSCs). Lastly, (d) there were changes in the electrophysiological underpinnings of neuronal spiking regularity. In parallel, we initiated new studies to explore changes in gene expression surrounding devices through spatial transcriptomics, which we applied to both recording and stimulating arrays. We found that (a) device implantation is associated with the induction of hundreds of genes associated with neuroinflammation, glial reactivity, oligodendrocyte function, and cellular metabolism and (b) electrical stimulation induces gene expression associated with damage or plasticity in a manner dependent upon the intensity of the applied stimulus. We are currently developing computational analysis tools to distill biomarkers of device-tissue interactions from large transcriptomics data sets. These results improve the current understanding of the biological response to electrodes implanted in the brain while producing new biomarkers for benchmarking the effects of novel electrode designs on responses. As the next generation of neurotechnology is developed, it will be increasingly important to understand the influence of novel materials, surface chemistries, and implant architectures on device performance as well as the relationship with the induction of specific cellular signaling pathways.
Collapse
Affiliation(s)
- Bhavna Gupta
- Neuroscience
Program, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
| | - Akash Saxena
- Institute
for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
| | - Mason L. Perillo
- Department
of Biomedical Engineering, Michigan State
University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
| | - Lauren C. Wade-Kleyn
- Department
of Biomedical Engineering, Michigan State
University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
| | - Cort H. Thompson
- Department
of Biomedical Engineering, Michigan State
University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
| | - Erin K. Purcell
- Department
of Biomedical Engineering, Michigan State
University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Neuroscience
Program, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, 775 Woodlot Dr., East Lansing, Michigan 48824, United States
| |
Collapse
|
7
|
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
|
8
|
Raghuram V, Datye AD, Fried SI, Timko BP. Transparent and Conformal Microcoil Arrays for Spatially Selective Neuronal Activation. DEVICE 2024; 2:100290. [PMID: 39184953 PMCID: PMC11343507 DOI: 10.1016/j.device.2024.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Micromagnetic stimulation (μMS) using small, implantable microcoils is a promising method for achieving neuronal activation with high spatial resolution and low toxicity. Herein, we report a microcoil array for localized activation of cortical neurons and retinal ganglion cells. We developed a computational model to relate the electric field gradient (activating function) to the geometry and arrangement of microcoils, and selected a design that produced an anisotropic region of activation <50 μm wide. The device was comprised of an SU-8/Cu/SU-8 tri-layer structure, which was flexible, transparent and conformal and featured four individually-addressable microcoils. Interfaced with cortex or retina explants from GCaMP6-expressing mice, we observed that individual neurons localized within 40 μm of a microcoil tip could be activated repeatedly and in a dose- (power-) dependent fashion. These results demonstrate the potential of μMS devices for brain-machine interfaces and could enable routes toward bioelectronic therapies including prosthetic vision devices.
Collapse
Affiliation(s)
- Vineeth Raghuram
- Dept. of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Boston Veterans Affairs Healthcare System, Boston, MA 02130, USA
- Dept. of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Aditya D. Datye
- Dept. of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Shelley I. Fried
- Dept. of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Boston Veterans Affairs Healthcare System, Boston, MA 02130, USA
- Dept. of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Brian P. Timko
- Dept. of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Lead Contact
| |
Collapse
|
9
|
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
|
10
|
Mueller NN, Kim Y, Ocoko MYM, Dernelle P, Kale I, Patwa S, Hermoso AC, Chirra D, Capadona JR, Hess-Dunning A. Effects of Micromachining on Anti-oxidant Elution from a Mechanically-Adaptive Polymer. JOURNAL OF MICROMECHANICS AND MICROENGINEERING : STRUCTURES, DEVICES, AND SYSTEMS 2024; 34:10.1088/1361-6439/ad27f7. [PMID: 38586082 PMCID: PMC10996452 DOI: 10.1088/1361-6439/ad27f7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Intracortical microelectrodes (IMEs) can be used to restore motor and sensory function as a part of brain-computer interfaces in individuals with neuromusculoskeletal disorders. However, the neuroinflammatory response to IMEs can result in their premature failure, leading to reduced therapeutic efficacy. Mechanically-adaptive, resveratrol-eluting (MARE) neural probes target two mechanisms believed to contribute to the neuroinflammatory response by reducing the mechanical mismatch between the brain tissue and device, as well as locally delivering an antioxidant therapeutic. To create the mechanically-adaptive substrate, a dispersion, casting, and evaporation method is used, followed by a microfabrication process to integrate functional recording electrodes on the material. Resveratrol release experiments were completed to generate a resveratrol release profile and demonstrated that the MARE probes are capable of long-term controlled release. Additionally, our results showed that resveratrol can be degraded by laser-micromachining, an important consideration for future device fabrication. Finally, the electrodes were shown to have a suitable impedance for single-unit neural recording and could record single units in vivo.
Collapse
Affiliation(s)
- Natalie N Mueller
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Youjoung Kim
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Mali Ya Mungu Ocoko
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Peter Dernelle
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Ishani Kale
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Simran Patwa
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Anna Clarissa Hermoso
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Deeksha Chirra
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Allison Hess-Dunning
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| |
Collapse
|
11
|
Sturgill B, Hernandez-Reynoso AG, Druschel LN, Smith TJ, Boucher PE, Hoeferlin GF, Thai TTD, Jiang MS, Hess JL, Alam NN, Menendez DM, Duncan JL, Cogan SF, Pancrazio JJ, Capadona JR. Reactive Amine Functionalized Microelectrode Arrays Provide Short-Term Benefit but Long-Term Detriment to In Vivo Recording Performance. ACS APPLIED BIO MATERIALS 2024; 7:1052-1063. [PMID: 38290529 PMCID: PMC10880090 DOI: 10.1021/acsabm.3c01014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Intracortical microelectrode arrays (MEAs) are used for recording neural signals. However, indwelling devices result in chronic neuroinflammation, which leads to decreased recording performance through degradation of the device and surrounding tissue. Coating the MEAs with bioactive molecules is being explored to mitigate neuroinflammation. Such approaches often require an intermediate functionalization step such as (3-aminopropyl)triethoxysilane (APTES), which serves as a linker. However, the standalone effect of this intermediate step has not been previously characterized. Here, we investigated the effect of coating MEAs with APTES by comparing APTES-coated to uncoated controls in vivo and ex vivo. First, we measured water contact angles between silicon uncoated and APTES-coated substrates to verify the hydrophilic characteristics of the APTES coating. Next, we implanted MEAs in the motor cortex (M1) of Sprague-Dawley rats with uncoated or APTES-coated devices. We assessed changes in the electrochemical impedance and neural recording performance over a chronic implantation period of 16 weeks. Additionally, histology and bulk gene expression were analyzed to understand further the reactive tissue changes arising from the coating. Results showed that APTES increased the hydrophilicity of the devices and decreased electrochemical impedance at 1 kHz. APTES coatings proved detrimental to the recording performance, as shown by a constant decay up to 16 weeks postimplantation. Bulk gene analysis showed differential changes in gene expression between groups that were inconclusive with regard to the long-term effect on neuronal tissue. Together, these results suggest that APTES coatings are ultimately detrimental to chronic neural recordings. Furthermore, interpretations of studies using APTES as a functionalization step should consider the potential consequences if the final functionalization step is incomplete.
Collapse
Affiliation(s)
- Brandon
S. Sturgill
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Ana G. Hernandez-Reynoso
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Lindsey N. Druschel
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Thomas J. Smith
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Pierce E. Boucher
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - George F. Hoeferlin
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Teresa Thuc Doan Thai
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Madison S. Jiang
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Jordan L. Hess
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Neeha N. Alam
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Dhariyat M. Menendez
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Jonathan L. Duncan
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Stuart F. Cogan
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Joseph J. Pancrazio
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Jeffrey R. Capadona
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| |
Collapse
|
12
|
Duncan JL, Wang JJ, Glusauskas G, Weagraff GR, Gao Y, Hoeferlin GF, Hunter AH, Hess-Dunning A, Ereifej ES, Capadona JR. In Vivo Characterization of Intracortical Probes with Focused Ion Beam-Etched Nanopatterned Topographies. MICROMACHINES 2024; 15:286. [PMID: 38399014 PMCID: PMC10893395 DOI: 10.3390/mi15020286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
(1) Background: Intracortical microelectrodes (IMEs) are an important part of interfacing with the central nervous system (CNS) and recording neural signals. However, recording electrodes have shown a characteristic steady decline in recording performance owing to chronic neuroinflammation. The topography of implanted devices has been explored to mimic the nanoscale three-dimensional architecture of the extracellular matrix. Our previous work used histology to study the implant sites of non-recording probes and showed that a nanoscale topography at the probe surface mitigated the neuroinflammatory response compared to probes with smooth surfaces. Here, we hypothesized that the improvement in the neuroinflammatory response for probes with nanoscale surface topography would extend to improved recording performance. (2) Methods: A novel design modification was implemented on planar silicon-based neural probes by etching nanopatterned grooves (with a 500 nm pitch) into the probe shank. To assess the hypothesis, two groups of rats were implanted with either nanopatterned (n = 6) or smooth control (n = 6) probes, and their recording performance was evaluated over 4 weeks. Postmortem gene expression analysis was performed to compare the neuroinflammatory response from the two groups. (3) Results: Nanopatterned probes demonstrated an increased impedance and noise floor compared to controls. However, the recording performances of the nanopatterned and smooth probes were similar, with active electrode yields for control probes and nanopatterned probes being approximately 50% and 45%, respectively, by 4 weeks post-implantation. Gene expression analysis showed one gene, Sirt1, differentially expressed out of 152 in the panel. (4) Conclusions: this study provides a foundation for investigating novel nanoscale topographies on neural probes.
Collapse
Affiliation(s)
- Jonathan L. Duncan
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Jaime J. Wang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Gabriele Glusauskas
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Gwendolyn R. Weagraff
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Yue Gao
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
| | - George F. Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Allen H. Hunter
- Michigan Center for Materials Characterization, University of Michigan, 500 S. State St, Ann Arbor, MI 48109, USA
| | - Allison Hess-Dunning
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| | - Evon S. Ereifej
- Department of Biomedical Engineering, University of Michigan, 500 S. State St, Ann Arbor, MI 48109, USA
- Veterans Affairs Hospital, 2215 Fuller Rd, Ann Arbor, MI 48105, USA
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH 44106, USA
| |
Collapse
|
13
|
Vėbraitė I, Bar-Haim C, David-Pur M, Hanein Y. Bi-directional electrical recording and stimulation of the intact retina with a screen-printed soft probe: a feasibility study. Front Neurosci 2024; 17:1288069. [PMID: 38264499 PMCID: PMC10804455 DOI: 10.3389/fnins.2023.1288069] [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: 09/03/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Electrophysiological investigations of intact neural circuits are challenged by the gentle and complex nature of neural tissues. Bi-directional electrophysiological interfacing with the retina, in its intact form, is particularly demanding and currently there is no feasible approach to achieve such investigations. Here we present a feasibility study of a novel soft multi-electrode array suitable for bi-directional electrophysiological study of the intact retina. Methods Screen-printed soft electrode arrays were developed and tested. The soft probes were designed to accommodate the curvature of the retina in the eye and offer an opportunity to study the retina in its intact form. Results For the first time, we show both electrical recording and stimulation capabilities from the intact retina. In particular, we demonstrate the ability to characterize retina responses to electrical stimulation and reveal stable, direct, and indirect responses compared with ex-vivo conditions. Discussion These results demonstrate the unique performances of the new probe while also suggesting that intact retinas retain better stability and robustness than ex-vivo retinas making them more suitable for characterizing retina responses to electrical stimulation.
Collapse
Affiliation(s)
- Ieva Vėbraitė
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Chen Bar-Haim
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Moshe David-Pur
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yael Hanein
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
14
|
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: 4] [Impact Index Per Article: 4.0] [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
|
15
|
Perna A, Angotzi GN, Berdondini L, Ribeiro JF. Advancing the interfacing performances of chronically implantable neural probes in the era of CMOS neuroelectronics. Front Neurosci 2023; 17:1275908. [PMID: 38027514 PMCID: PMC10644322 DOI: 10.3389/fnins.2023.1275908] [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: 08/10/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Tissue penetrating microelectrode neural probes can record electrophysiological brain signals at resolutions down to single neurons, making them invaluable tools for neuroscience research and Brain-Computer-Interfaces (BCIs). The known gradual decrease of their electrical interfacing performances in chronic settings, however, remains a major challenge. A key factor leading to such decay is Foreign Body Reaction (FBR), which is the cascade of biological responses that occurs in the brain in the presence of a tissue damaging artificial device. Interestingly, the recent adoption of Complementary Metal Oxide Semiconductor (CMOS) technology to realize implantable neural probes capable of monitoring hundreds to thousands of neurons simultaneously, may open new opportunities to face the FBR challenge. Indeed, this shift from passive Micro Electro-Mechanical Systems (MEMS) to active CMOS neural probe technologies creates important, yet unexplored, opportunities to tune probe features such as the mechanical properties of the probe, its layout, size, and surface physicochemical properties, to minimize tissue damage and consequently FBR. Here, we will first review relevant literature on FBR to provide a better understanding of the processes and sources underlying this tissue response. Methods to assess FBR will be described, including conventional approaches based on the imaging of biomarkers, and more recent transcriptomics technologies. Then, we will consider emerging opportunities offered by the features of CMOS probes. Finally, we will describe a prototypical neural probe that may meet the needs for advancing clinical BCIs, and we propose axial insertion force as a potential metric to assess the influence of probe features on acute tissue damage and to control the implantation procedure to minimize iatrogenic injury and subsequent FBR.
Collapse
Affiliation(s)
- Alberto Perna
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Istituto Italiano di Tecnologia, Genova, Italy
| | - Gian Nicola Angotzi
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| | - Luca Berdondini
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| | - João Filipe Ribeiro
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| |
Collapse
|
16
|
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: 4] [Impact Index Per Article: 4.0] [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
|
17
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. Biomaterials 2023; 301:122210. [PMID: 37413842 PMCID: PMC10528716 DOI: 10.1016/j.biomaterials.2023.122210] [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: 02/06/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over the chronic implantation period. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, 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
|
18
|
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: 7.0] [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
|
19
|
Smith TJ, Wu Y, Cheon C, Khan AA, Srinivasan H, Capadona JR, Cogan SF, Pancrazio JJ, Engineer CT, Hernandez-Reynoso AG. Behavioral paradigm for the evaluation of stimulation-evoked somatosensory perception thresholds in rats. Front Neurosci 2023; 17:1202258. [PMID: 37383105 PMCID: PMC10293669 DOI: 10.3389/fnins.2023.1202258] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Intracortical microstimulation (ICMS) of the somatosensory cortex via penetrating microelectrode arrays (MEAs) can evoke cutaneous and proprioceptive sensations for restoration of perception in individuals with spinal cord injuries. However, ICMS current amplitudes needed to evoke these sensory percepts tend to change over time following implantation. Animal models have been used to investigate the mechanisms by which these changes occur and aid in the development of new engineering strategies to mitigate such changes. Non-human primates are commonly the animal of choice for investigating ICMS, but ethical concerns exist regarding their use. Rodents are a preferred animal model due to their availability, affordability, and ease of handling, but there are limited choices of behavioral tasks for investigating ICMS. In this study, we investigated the application of an innovative behavioral go/no-go paradigm capable of estimating ICMS-evoked sensory perception thresholds in freely moving rats. We divided animals into two groups, one receiving ICMS and a control group receiving auditory tones. Then, we trained the animals to nose-poke - a well-established behavioral task for rats - following either a suprathreshold ICMS current-controlled pulse train or frequency-controlled auditory tone. Animals received a sugar pellet reward when nose-poking correctly. When nose-poking incorrectly, animals received a mild air puff. After animals became proficient in this task, as defined by accuracy, precision, and other performance metrics, they continued to the next phase for perception threshold detection, where we varied the ICMS amplitude using a modified staircase method. Finally, we used non-linear regression to estimate perception thresholds. Results indicated that our behavioral protocol could estimate ICMS perception thresholds based on ~95% accuracy of rat nose-poke responses to the conditioned stimulus. This behavioral paradigm provides a robust methodology for evaluating stimulation-evoked somatosensory percepts in rats comparable to the evaluation of auditory percepts. In future studies, this validated methodology can be used to study the performance of novel MEA device technologies on ICMS-evoked perception threshold stability using freely moving rats or to investigate information processing principles in neural circuits related to sensory perception discrimination.
Collapse
Affiliation(s)
- Thomas J. Smith
- School of Behavioral and Brain Sciences, 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
| | - Claire Cheon
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, United States
| | - Arlin A. Khan
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Hari Srinivasan
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Stuart F. Cogan
- 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
| | - Crystal T. Engineer
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, United States
- Texas Biomedical Device Center, The University of Texas at Dallas, Richardson, TX, United States
| | | |
Collapse
|
20
|
Kim Y, Mueller NN, Schwartzman WE, Sarno D, Wynder R, Hoeferlin GF, Gisser K, Capadona JR, Hess-Dunning A. Fabrication Methods and Chronic In Vivo Validation of Mechanically Adaptive Microfluidic Intracortical Devices. MICROMACHINES 2023; 14:1015. [PMID: 37241639 PMCID: PMC10223487 DOI: 10.3390/mi14051015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023]
Abstract
Intracortical neural probes are both a powerful tool in basic neuroscience studies of brain function and a critical component of brain computer interfaces (BCIs) designed to restore function to paralyzed patients. Intracortical neural probes can be used both to detect neural activity at single unit resolution and to stimulate small populations of neurons with high resolution. Unfortunately, intracortical neural probes tend to fail at chronic timepoints in large part due to the neuroinflammatory response that follows implantation and persistent dwelling in the cortex. Many promising approaches are under development to circumvent the inflammatory response, including the development of less inflammatory materials/device designs and the delivery of antioxidant or anti-inflammatory therapies. Here, we report on our recent efforts to integrate the neuroprotective effects of both a dynamically softening polymer substrate designed to minimize tissue strain and localized drug delivery at the intracortical neural probe/tissue interface through the incorporation of microfluidic channels within the probe. The fabrication process and device design were both optimized with respect to the resulting device mechanical properties, stability, and microfluidic functionality. The optimized devices were successfully able to deliver an antioxidant solution throughout a six-week in vivo rat study. Histological data indicated that a multi-outlet design was most effective at reducing markers of inflammation. The ability to reduce inflammation through a combined approach of drug delivery and soft materials as a platform technology allows future studies to explore additional therapeutics to further enhance intracortical neural probes performance and longevity for clinical applications.
Collapse
Affiliation(s)
- Youjoung Kim
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Natalie N Mueller
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - William E Schwartzman
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Danielle Sarno
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Reagan Wynder
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - George F Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Kaela Gisser
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, 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 VA Medical Center, Cleveland, OH 44106, USA
| | - Allison Hess-Dunning
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
21
|
Smith TJ, Wu Y, Cheon C, Khan AA, Srinivasan H, Capadona JR, Cogan SF, Pancrazio JJ, Engineer CT, Hernandez-Reynoso AG. Behavioral Paradigm for the Evaluation of Stimulation-Evoked Somatosensory Perception Thresholds in Rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.537848. [PMID: 37205577 PMCID: PMC10187227 DOI: 10.1101/2023.05.04.537848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Intracortical microstimulation (ICMS) of the somatosensory cortex via penetrating microelectrode arrays (MEAs) can evoke cutaneous and proprioceptive sensations for restoration of perception in individuals with spinal cord injuries. However, ICMS current amplitudes needed to evoke these sensory percepts tend to change over time following implantation. Animal models have been used to investigate the mechanisms by which these changes occur and aid in the development of new engineering strategies to mitigate such changes. Non-human primates are commonly the animal of choice for investigating ICMS, but ethical concerns exist regarding their use. Rodents are a preferred animal model due to their availability, affordability, and ease of handling, but there are limited choices of behavioral tasks for investigating ICMS. In this study, we investigated the application of an innovative behavioral go/no-go paradigm capable of estimating ICMS-evoked sensory perception thresholds in freely moving rats. We divided animals into two groups, one receiving ICMS and a control group receiving auditory tones. Then, we trained the animals to nose-poke - a well-established behavioral task for rats - following either a suprathreshold ICMS current-controlled pulse train or frequency-controlled auditory tone. Animals received a sugar pellet reward when nose-poking correctly. When nose-poking incorrectly, animals received a mild air puff. After animals became proficient in this task, as defined by accuracy, precision, and other performance metrics, they continued to the next phase for perception threshold detection, where we varied the ICMS amplitude using a modified staircase method. Finally, we used non-linear regression to estimate perception thresholds. Results indicated that our behavioral protocol could estimate ICMS perception thresholds based on ∼95% accuracy of rat nose-poke responses to the conditioned stimulus. This behavioral paradigm provides a robust methodology for evaluating stimulation-evoked somatosensory percepts in rats comparable to the evaluation of auditory percepts. In future studies, this validated methodology can be used to study the performance of novel MEA device technologies on ICMS-evoked perception threshold stability using freely moving rats or to investigate information processing principles in neural circuits related to sensory perception discrimination.
Collapse
|
22
|
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
|
23
|
Sharon A, Jankowski MM, Shmoel N, Erez H, Spira ME. Significantly reduced inflammatory foreign-body-response to neuroimplants and improved recording performance in young compared to adult rats. Acta Biomater 2023; 158:292-307. [PMID: 36632879 DOI: 10.1016/j.actbio.2023.01.002] [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: 09/22/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
The multicellular inflammatory encapsulation of implanted intracortical multielectrode arrays (MEA) is associated with severe deterioration of their field potentials' (FP) recording performance, which thus limits the use of brain implants in basic research and clinical applications. Therefore, extensive efforts have been made to identify the conditions in which the inflammatory foreign body response (FBR) is alleviated, or to develop methods to mitigate the formation of the inflammatory barrier. Here, for the first time, we show that (1) in young rats (74±8 gr, 4 weeks old at the onset of the experiments), cortical tissue recovery following MEA implantation proceeds with ameliorated inflammatory scar as compared to adult rats (242 ± 18 gr, 9 weeks old at the experimental onset); (2) in contrast to adult rats in which the Colony Stimulating factor 1 Receptor (CSF1R) antagonist chow eliminated ∼95% of the cortical microglia but not microglia adhering to the implant surfaces, in young rats the microglia adhering to the implant were eliminated along with the parenchymal microglia population. The removal of microglia adhering to the implant surfaces was correlated with improved recording performance by in-house fabricated Perforated Polyimide MEA Platforms (PPMP). These results support the hypothesis that microglia adhering to the surface of the electrodes, rather than the multicellular inflammatory scar, is the major underlying mechanism that deteriorates implant recording performance, and that young rats provide an advantageous model to study months-long, multisite electrophysiology in freely behaving rats. STATEMENT OF SIGNIFICANCE: Multisite electrophysiological recordings and stimulation devices play central roles in basic brain research and medical applications. The insertion of multielectrode-array platforms into the brain's parenchyma unavoidably injures the tissue, and initiates a multicellular inflammatory cascade culminating in the formation of an encapsulating scar tissue (the foreign body response-FBR). The dominant view, which directs most current research efforts to mitigate the FBR, holds that the FBR is the major hurdle to effective electrophysiological use of neuroprobes. By contrast, this report demonstrates that microglia adhering to the surface of a neuroimplants, rather than the multicellular FBR, underlie the performance deterioration of neuroimplants. These findings pave the way to the development of novel and focused strategies to overcome the functional deterioration of neuroimplants.
Collapse
Affiliation(s)
- Aviv Sharon
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maciej M Jankowski
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel; Edmond and Lily Safra Center for Brain Sciences, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nava Shmoel
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Erez
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Micha E Spira
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
24
|
Ziemba AM, Woodson MCC, Funnell JL, Wich D, Balouch B, Rende D, Amato DN, Bao J, Oprea I, Cao D, Bajalo N, Ereifej ES, Capadona JR, Palermo EF, Gilbert RJ. Development of a Slow-Degrading Polymerized Curcumin Coating for Intracortical Microelectrodes. ACS APPLIED BIO MATERIALS 2023; 6:806-818. [PMID: 36749645 PMCID: PMC11366415 DOI: 10.1021/acsabm.2c00969] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intracortical microelectrodes are used with brain-computer interfaces to restore lost limb function following nervous system injury. While promising, recording ability of intracortical microelectrodes diminishes over time due, in part, to neuroinflammation. As curcumin has demonstrated neuroprotection through anti-inflammatory activity, we fabricated a 300 nm-thick intracortical microelectrode coating consisting of a polyurethane copolymer of curcumin and polyethylene glycol (PEG), denoted as poly(curcumin-PEG1000 carbamate) (PCPC). The uniform PCPC coating reduced silicon wafer hardness by two orders of magnitude and readily absorbed water within minutes, demonstrating that the coating is soft and hydrophilic in nature. Using an in vitro release model, curcumin eluted from the PCPC coating into the supernatant over 1 week; the majority of the coating was intact after an 8-week incubation in buffer, demonstrating potential for longer term curcumin release and softness. Assessing the efficacy of PCPC within a rat intracortical microelectrode model in vivo, there were no significant differences in tissue inflammation, scarring, neuron viability, and myelin damage between the uncoated and PCPC-coated probes. As the first study to implant nonfunctional probes with a polymerized curcumin coating, we have demonstrated the biocompatibility of a PCPC coating and presented a starting point in the design of poly(pro-curcumin) polymers as coating materials for intracortical electrodes.
Collapse
Affiliation(s)
- Alexis M Ziemba
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Neuroscience Program, Department of Biological Sciences, Smith College, Northampton 01063, Massachusetts, United States
| | - Mary Clare Crochiere Woodson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Jessica L Funnell
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Douglas Wich
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Bailey Balouch
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Deniz Rende
- Center for Materials, Devices, and Integrated Systems, Rensselaer Polytechnic Institute, 110 8th Street, Troy 12180-3590, New York, United States
| | - Dahlia N Amato
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Jonathan Bao
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Ingrid Oprea
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Dominica Cao
- Neuroscience Program, Department of Biological Sciences, Smith College, Northampton 01063, Massachusetts, United States
| | - Neda Bajalo
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Evon S Ereifej
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor 48104, Michigan, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor 48104, Michigan, United States
- Department of Neurology, University of Michigan, Ann Arbor 48104, Michigan, United States
- United States Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland 44106, Ohio, United States
| | - Jeffrey R Capadona
- United States Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland 44106, Ohio, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland 44106, Ohio, United States
| | - Edmund F Palermo
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| |
Collapse
|
25
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating Clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526463. [PMID: 36778360 PMCID: PMC9915570 DOI: 10.1101/2023.01.31.526463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over chronic implantation. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period. Abstract Figure
Collapse
|
26
|
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: 1.0] [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
|
27
|
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: 15] [Impact Index Per Article: 15.0] [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
|
28
|
Chen R, Funnell JL, Quinones GB, Bentley M, Capadona JR, Gilbert RJ, Palermo EF. Poly(pro-curcumin) Materials Exhibit Dual Release Rates and Prolonged Antioxidant Activity as Thin Films and Self-Assembled Particles. Biomacromolecules 2023; 24:294-307. [PMID: 36512693 DOI: 10.1021/acs.biomac.2c01135] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Curcumin is a natural polyphenol that exhibits remarkable antioxidant and anti-inflammatory activities; however, its clinical application is limited in part by its physiological instability. Here, we report the synthesis of curcumin-derived polyesters that release curcumin upon hydrolytic degradation to improve curcumin stability and solubility in physiological conditions. Curcumin was incorporated in the polymer backbone by a one-pot condensation polymerization in the presence of sebacoyl chloride and polyethylene glycol (PEG, Mn = 1 kDa). The thermal and mechanical properties, surface wettability, self-assembly behavior, and drug-release kinetics all depend sensitively on the mole percentage of curcumin incorporated in these statistical copolymers. Curcumin release was triggered by the hydrolysis of phenolic esters on the polymer backbone, which was confirmed using a PEGylated curcumin model compound, which represented a putative repeating unit within the polymer. The release rate of curcumin was controlled by the hydrophilicity of the polymers. Burst release (2 days) and extended release (>8 weeks) can be achieved from the same polymer depending on curcumin content in the copolymer. The materials can quench free radicals for at least 8 weeks and protect primary neurons from oxidative stress in vitro. Further, these copolymer materials could be processed into both thin films and self-assembled particles, depending on the solvent-based casting conditions. Finally, we envision that these materials may have potential for neural tissue engineering application, where antioxidant release can mitigate oxidative stress and the inflammatory response following neural injury.
Collapse
Affiliation(s)
- Ruiwen Chen
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Jessica L Funnell
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Geraldine B Quinones
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Marvin Bentley
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States.,Advanced Platform Technology Center, L. Stokes Cleveland VA Medical Center, Cleveland, Ohio 44106, United States
| | - Ryan J Gilbert
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Albany Stratton Veteran Affairs Medical Center, Albany, New York 12208, United States
| | - Edmund F Palermo
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States.,Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| |
Collapse
|
29
|
Riggins TE, Whitsitt QA, Saxena A, Hunter E, Hunt B, Thompson CH, Moore MG, Purcell EK. Gene Expression Changes in Cultured Reactive Rat Astrocyte Models and Comparison to Device-Associated Effects in the Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.522870. [PMID: 36712012 PMCID: PMC9881929 DOI: 10.1101/2023.01.06.522870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Implanted microelectrode arrays hold immense therapeutic potential for many neurodegenerative diseases. However, a foreign body response limits long-term device performance. Recent literature supports the role of astrocytes in the response to damage to the central nervous system (CNS) and suggests that reactive astrocytes exist on a spectrum of phenotypes, from beneficial to neurotoxic. The goal of our study was to gain insight into the subtypes of reactive astrocytes responding to electrodes implanted in the brain. In this study, we tested the transcriptomic profile of two reactive astrocyte culture models (cytokine cocktail or lipopolysaccharide, LPS) utilizing RNA sequencing, which we then compared to differential gene expression surrounding devices inserted into rat motor cortex via spatial transcriptomics. We interpreted changes in the genetic expression of the culture models to that of 24 hour, 1 week and 6 week rat tissue samples at multiple distances radiating from the injury site. We found overlapping expression of up to ∼250 genes between in vitro models and in vivo effects, depending on duration of implantation. Cytokine-induced cells shared more genes in common with chronically implanted tissue (≥1 week) in comparison to LPS-exposed cells. We revealed localized expression of a subset of these intersecting genes (e.g., Serping1, Chi3l1, and Cyp7b1) in regions of device-encapsulating, glial fibrillary acidic protein (GFAP)-expressing astrocytes identified with immunohistochemistry. We applied a factorization approach to assess the strength of the relationship between reactivity markers and the spatial distribution of GFAP-expressing astrocytes in vivo . We also provide lists of hundreds of differentially expressed genes between reactive culture models and untreated controls, and we observed 311 shared genes between the cytokine induced model and the LPS-reaction induced control model. Our results show that comparisons of reactive astrocyte culture models with spatial transcriptomics data can reveal new biomarkers of the foreign body response to implantable neurotechnology. These comparisons also provide a strategy to assess the development of in vitro models of the tissue response to implanted electrodes.
Collapse
|
30
|
Williams NP, Kushwah N, Dhawan V, Zheng XS, Cui XT. Effects of central nervous system electrical stimulation on non-neuronal cells. Front Neurosci 2022; 16:967491. [PMID: 36188481 PMCID: PMC9521315 DOI: 10.3389/fnins.2022.967491] [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: 06/12/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past few decades, much progress has been made in the clinical use of electrical stimulation of the central nervous system (CNS) to treat an ever-growing number of conditions from Parkinson's disease (PD) to epilepsy as well as for sensory restoration and many other applications. However, little is known about the effects of microstimulation at the cellular level. Most of the existing research focuses on the effects of electrical stimulation on neurons. Other cells of the CNS such as microglia, astrocytes, oligodendrocytes, and vascular endothelial cells have been understudied in terms of their response to stimulation. The varied and critical functions of these cell types are now beginning to be better understood, and their vital roles in brain function in both health and disease are becoming better appreciated. To shed light on the importance of the way electrical stimulation as distinct from device implantation impacts non-neuronal cell types, this review will first summarize common stimulation modalities from the perspective of device design and stimulation parameters and how these different parameters have an impact on the physiological response. Following this, what is known about the responses of different cell types to different stimulation modalities will be summarized, drawing on findings from both clinical studies as well as clinically relevant animal models and in vitro systems.
Collapse
Affiliation(s)
- Nathaniel P. Williams
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Neetu Kushwah
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Xin Sally Zheng
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| |
Collapse
|
31
|
Kushwah N, Woeppel K, Dhawan V, Shi D, Cui XT. Effects of neuronal cell adhesion molecule L1 and nanoparticle surface modification on microglia. Acta Biomater 2022; 149:273-286. [PMID: 35764240 PMCID: PMC10018678 DOI: 10.1016/j.actbio.2022.06.038] [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: 01/08/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022]
Abstract
Microelectrode arrays for neural recording suffer from low yield and stability partly due to the inflammatory host responses. A neuronal cell adhesion molecule L1 coating has been shown to promote electrode-neuron integration, reduce microglia activation and improve recording. Coupling L1 to surface via a nanoparticle (NP) base layer further increased the protein surface density and stability. However, the exact L1-microglia interaction in these coatings has not been studied. Here we cultured primary microglia on L1 modified surfaces (with and without NP) and characterized microglia activation upon phorbol myristate acetate (PMA) and lipopolysaccharide (LPS) stimulation. Results showed L1 coatings reduced microglia's superoxide production in response to PMA and presented intrinsic antioxidant properties. Meanwhile, L1 decreased iNOS, NO, and pro-inflammatory cytokines (TNF alpha, IL-6, IL-1 beta), while increased anti-inflammatory cytokines (TGF beta 1, IL-10) in LPS stimulated microglia. Furthermore, L1 increased Arg-1 expression and phagocytosis upon LPS stimulation. Rougher NP surface showed lower number of microglia attached per area than their smooth counterpart, lower IL-6 release and superoxide production, and higher intrinsic reducing potential. Finally, we examined the effect of L1 and nanoparticle modifications on microglia response in vivo over 8 weeks with 2-photon imaging. Microglial coverage on the implant surface was found to be lower on the L1 modified substrates relative to unmodified, consistent with the in vitro observation. Our results indicate L1 significantly reduces superoxide production and inflammatory response of microglia and promotes wound healing, while L1 immobilization via a nanoparticle base layer brings added benefit without adverse effects. STATEMENT OF SIGNIFICANCE: Surface modification of microelectrode arrays with L1 has been shown to reduce microglia coverage on neural probe surface in vivo and improves neural recording, but the specific mechanism of action is not fully understood. The results in this study show that surface bound L1 reduces superoxide production from cultured microglia via direct reduction reaction and signaling pathways, increases anti-inflammatory cytokine release and phagocytosis in response to PMA or LPS stimulation. Additionally, roughening the surface with nanoparticles prior to L1 immobilization further increased the benefit of L1 in reducing microglia activation and oxidative stress. Together, our findings shed light on the mechanisms of action of nanotextured and neuroadhesive neural implant coatings and guide future development of seamless tissue interface.
Collapse
Affiliation(s)
- Neetu Kushwah
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Kevin Woeppel
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States
| | - Vaishnavi Dhawan
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States
| | - Delin Shi
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States
| | - Xinyan Tracy Cui
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States.
| |
Collapse
|
32
|
Song S, Regan B, Ereifej ES, Chan ER, Capadona JR. Neuroinflammatory Gene Expression Analysis Reveals Pathways of Interest as Potential Targets to Improve the Recording Performance of Intracortical Microelectrodes. Cells 2022; 11:2348. [PMID: 35954192 PMCID: PMC9367362 DOI: 10.3390/cells11152348] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
Intracortical microelectrodes are a critical component of brain-machine interface (BMI) systems. The recording performance of intracortical microelectrodes used for both basic neuroscience research and clinical applications of BMIs decreases over time, limiting the utility of the devices. The neuroinflammatory response to the microelectrode has been identified as a significant contributing factor to its performance. Traditionally, pathological assessment has been limited to a dozen or so known neuroinflammatory proteins, and only a few groups have begun to explore changes in gene expression following microelectrode implantation. Our initial characterization of gene expression profiles of the neuroinflammatory response to mice implanted with non-functional intracortical probes revealed many upregulated genes that could inform future therapeutic targets. Emphasis was placed on the most significant gene expression changes and genes involved in multiple innate immune sets, including Cd14, C3, Itgam, and Irak4. In previous studies, inhibition of Cluster of Differentiation 14 (Cd14) improved microelectrode performance for up to two weeks after electrode implantation, suggesting CD14 can be explored as a potential therapeutic target. However, all measures of improvements in signal quality and electrode performance lost statistical significance after two weeks. Therefore, the current study investigated the expression of genes in the neuroinflammatory pathway at the tissue-microelectrode interface in Cd14-/- mice to understand better how Cd14 inhibition was connected to temporary improvements in recording quality over the initial 2-weeks post-surgery, allowing for the identification of potential co-therapeutic targets that may work synergistically with or after CD14 inhibition to improve microelectrode performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106, USA; (S.S.); (E.S.E.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Brianna Regan
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evon S. Ereifej
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106, USA; (S.S.); (E.S.E.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA;
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - E. Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr. Drive, Cleveland, OH 44106, USA; (S.S.); (E.S.E.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
33
|
Ruhunage CK, Dhawan V, McKenzie TJ, Hoque A, Rahm CE, Nawarathne CP, Ayres N, Cui XT, Alvarez NT. Hydrophilic Micro- and Macroelectrodes with Antibiofouling Properties for Biomedical Applications. ACS Biomater Sci Eng 2022; 8:2920-2931. [PMID: 35710337 PMCID: PMC10080669 DOI: 10.1021/acsbiomaterials.2c00173] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Implantable neural electrodes are generally used to record the electrical activity of neurons and to stimulate neurons in the nervous system. Biofouling triggered by inflammatory responses can dramatically affect the performance of neural electrodes, resulting in decreased signal sensitivity and consistency over time. Thus, long-term clinical applications require electrically conducting electrode materials with reduced dimensions, high flexibility, and antibiofouling properties that can reduce the degree of inflammatory reactions and increase the lifetime of neural electrodes. Carbon nanotubes (CNTs) are well known to form flexible assemblies such as CNT fibers. Herein, we report the covalent functionalization of predefined CNT fiber and film surfaces with hydrophilic, antibiofouling phosphorylcholine (PC) molecules. The electrochemical and spectroscopic characteristics, impedance properties, hydrophilicity, and in vitro antifouling nature of the functionalized CNT surfaces were evaluated. The hydrophilicity of the functionalized CNT films was demonstrated by a decrease in the static contact angle from 134.4° ± 3.9° before to 15.7° ± 1.5° after one and fully wetting after three functionalization cycles, respectively. In addition, the extent of protein absorption on the functionalized CNT films was significantly lower than that on the nonfunctionalized CNT film. Surprisingly, the faradic charge-transfer properties and impedance of the CNT assemblies were preserved after functionalization with PC molecules. These functionalized CNT assemblies are promising for the development of low-impedance neural electrodes with higher hydrophilicity and protein-fouling resistance to inhibit inflammatory responses.
Collapse
Affiliation(s)
- Chethani K Ruhunage
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Tucker J McKenzie
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Abdul Hoque
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Connor E Rahm
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Chaminda P Nawarathne
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Neil Ayres
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Noe T Alvarez
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| |
Collapse
|
34
|
Riggins TE, Li W, Purcell EK. Atomic Force Microscope Characterization of the Bending Stiffness and Surface Topography of Silicon and Polymeric Electrodes. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:2348-2352. [PMID: 36085626 DOI: 10.1109/embc48229.2022.9871216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Implanted electrodes in the brain are increasingly used in research and clinical settings to understand and treat neurological conditions. However, a foreign body response typically occurs after implantation, and glial encapsulation of the device is a commonly observed. Multiple factors affect how gliosis surrounding the implantable electrodes evolves. Characterizing and measuring the surface features and mechanical properties of these devices may allow us to predict where gliosis will occur, and understanding how electrode design features may impact astrogliosis may give researchers a set of design guidelines to follow to maximize chronic performance. In this study, we used atomic force microscopy to measure surface roughness on parylene, polyimide, and silicon devices. Multiple features on microelectrode arrays were measured, including electrode sites, traces, and the bulk substrate. We found differences in surface roughness according to device material, but not device features. We also directly measured the bending stiffness of silicon devices, providing a more exact quantification of this property to corroborate calculated estimates.
Collapse
|
35
|
Atherton E, Hu Y, Brown S, Papiez E, Ling V, Colvin V, Borton D. A 3D in vitro model of the device-tissue interface: Functional and structural symptoms of innate neuroinflammation are mitigated by antioxidant ceria nanoparticles. J Neural Eng 2022; 19. [PMID: 35447619 DOI: 10.1088/1741-2552/ac6908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/20/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE The recording instability of neural implants due to neuroinflammation at the device-tissue interface is a primary roadblock to broad adoption of brain-machine interfaces. While a multiphasic immune response, marked by glial scaring, oxidative stress (OS), and neurodegeneration, is well-characterized, the independent contributions of systemic and local "innate" immune responses are not well-understood. We aimed to understand and mitigate the isolated the innate neuroinflammatory response to devices. APPROACH Three-dimensional primary neural cultures provide a unique environment for studying the drivers of neuroinflammation by decoupling the innate and systemic immune systems, while conserving an endogenous extracellular matrix and structural and functional network complexity. We created a three-dimensional in vitro model of the DTI by seeding primary cortical cells around microwires. Live imaging of both dye and AAV-mediated functional, structural, and lipid peroxidation fluorescence was employed to characterize the neuroinflammatory response. MAIN RESULTS Live imaging of microtissues over time revealed independent innate neuroinflammation, marked by increased OS, decreased neuronal density, and increased functional connectivity. We demonstrated the use of this model for therapeutic screening by directly applying drugs to neural tissue, bypassing low bioavailability through the in vivo blood brain barrier. As there is growing interest in long-acting antioxidant therapies, we tested efficacy of "perpetual" antioxidant ceria nanoparticles, which reduced OS, increased neuronal density, and protected functional connectivity. SIGNIFICANCE Our 3D in vitro model of the device-tissue interface exhibited symptoms of OS-mediated innate neuroinflammation, indicating a significant local immune response to devices. The dysregulation of functional connectivity of microcircuits surround implants suggests the presence of an observer effect, in which the process of recording neural activity may fundamentally change the neural signal. Finally, the demonstration of antioxidant ceria nanoparticle treatment exhibited substantial promise as a neuroprotective and anti-inflammatory treatment strategy.
Collapse
Affiliation(s)
- Elaina Atherton
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Yue Hu
- Department of Chemistry, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Sophie Brown
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Emily Papiez
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Vivian Ling
- Department of Chemistry, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - Vicki Colvin
- Department of Chemistry, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| | - David Borton
- School of Engineering, Brown University, 182 Hope Street, Providence, RI 02912, USA, Providence, Rhode Island, 02912, UNITED STATES
| |
Collapse
|
36
|
Sturgill B, Radhakrishna R, Thai TTD, Patnaik SS, Capadona JR, Pancrazio JJ. Characterization of Active Electrode Yield for Intracortical Arrays: Awake versus Anesthesia. MICROMACHINES 2022; 13:mi13030480. [PMID: 35334770 PMCID: PMC8955818 DOI: 10.3390/mi13030480] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/04/2022]
Abstract
Intracortical microelectrode arrays are used for recording neural signals at single-unit resolution and are promising tools for studying brain function and developing neuroprosthetics. Research is being done to increase the chronic performance and reliability of these probes, which tend to decrease or fail within several months of implantation. Although recording paradigms vary, studies focused on assessing the reliability and performance of these devices often perform recordings under anesthesia. However, anesthetics—such as isoflurane—are known to alter neural activity and electrophysiologic function. Therefore, we compared the neural recording performance under anesthesia (2% isoflurane) followed by awake conditions for probes implanted in the motor cortex of both male and female Sprague-Dawley rats. While the single-unit spike rate was significantly higher by almost 600% under awake compared to anesthetized conditions, we found no difference in the active electrode yield between the two conditions two weeks after surgery. Additionally, the signal-to-noise ratio was greater under anesthesia due to the noise levels being nearly 50% greater in awake recordings, even though there was a 14% increase in the peak-to-peak voltage of distinguished single units when awake. We observe that these findings are similar for chronic time points as well. Our observations indicate that either anesthetized or awake recordings are acceptable for studies assessing the chronic reliability and performance of intracortical microelectrode arrays.
Collapse
Affiliation(s)
- Brandon Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Rahul Radhakrishna
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Teresa Thuc Doan Thai
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Sourav S. Patnaik
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA;
| | - Joseph J. Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
- Correspondence:
| |
Collapse
|
37
|
Lim J, Yoon J, Shin M, Lee KB, Choi JW. Biomolecular Electron Controller Composed of Nanobiohybrid with Electrically Released Complex for Spatiotemporal Control of Neuronal Differentiation. SMALL METHODS 2022; 6:e2100912. [PMID: 35174997 DOI: 10.1002/smtd.202100912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/11/2021] [Indexed: 06/14/2023]
Abstract
In vitro spatiotemporal control of cell differentiation is a critical issue in several biomedical fields such as stem cell therapy and regenerative medicine, as it enables the generation of heterogeneous tissue structures similar to those of their native counterparts. However, the simultaneous control of both spatial and temporal cell differentiation poses important challenges, and therefore no previous studies have achieved this goal. Here, the authors develop a cell differentiation biomolecular electron controller ("Biomoletron") composed of recombinant proteins, DNA, Au nanoparticles, peptides, and an electrically released complex with retinoic acid (RA) to spatiotemporally control SH-SY5Y cell differentiation. RA is only released from the Biomoletron when the complex is electrically stimulated, thus demonstrating the temporal control of SH-SY5Y cell differentiation. Furthermore, by introducing a patterned Au substrate that allows controlling the area where the Biomoletron is immobilized, spatiotemporal differentiation of the SH-SY5Y cell is successfully achieved. Therefore, the proposed Biomoletron-mediated differentiation method provides a promising strategy for spatiotemporal cell differentiation control with applications in regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Joungpyo Lim
- Department of Chemical & Biomolecular Engineering, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Jinho Yoon
- Department of Chemical & Biomolecular Engineering, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Minkyu Shin
- Department of Chemical & Biomolecular Engineering, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Jeong-Woo Choi
- Department of Chemical & Biomolecular Engineering, Sogang University, Mapo-gu, Seoul, 04107, Republic of Korea
| |
Collapse
|
38
|
Kim Y, Ereifej ES, Schwartzman WE, Meade SM, Chen K, Rayyan J, Feng H, Aluri V, Mueller NN, Bhambra R, Bhambra S, Taylor DM, Capadona JR. Investigation of the Feasibility of Ventricular Delivery of Resveratrol to the Microelectrode Tissue Interface. MICROMACHINES 2021; 12:1446. [PMID: 34945296 PMCID: PMC8708660 DOI: 10.3390/mi12121446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/12/2021] [Accepted: 11/19/2021] [Indexed: 12/02/2022]
Abstract
(1) Background: Intracortical microelectrodes (IMEs) are essential to basic brain research and clinical brain-machine interfacing applications. However, the foreign body response to IMEs results in chronic inflammation and an increase in levels of reactive oxygen and nitrogen species (ROS/RNS). The current study builds on our previous work, by testing a new delivery method of a promising antioxidant as a means of extending intracortical microelectrodes performance. While resveratrol has shown efficacy in improving tissue response, chronic delivery has proven difficult because of its low solubility in water and low bioavailability due to extensive first pass metabolism. (2) Methods: Investigation of an intraventricular delivery of resveratrol in rats was performed herein to circumvent bioavailability hurdles of resveratrol delivery to the brain. (3) Results: Intraventricular delivery of resveratrol in rats delivered resveratrol to the electrode interface. However, intraventricular delivery did not have a significant impact on electrophysiological recordings over the six-week study. Histological findings indicated that rats receiving intraventricular delivery of resveratrol had a decrease of oxidative stress, yet other biomarkers of inflammation were found to be not significantly different from control groups. However, investigation of the bioavailability of resveratrol indicated a decrease in resveratrol accumulation in the brain with time coupled with inconsistent drug elution from the cannulas. Further inspection showed that there may be tissue or cellular debris clogging the cannulas, resulting in variable elution, which may have impacted the results of the study. (4) Conclusions: These results indicate that the intraventricular delivery approach described herein needs further optimization, or may not be well suited for this application.
Collapse
Affiliation(s)
- Youjoung Kim
- 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
| | - Evon S. Ereifej
- 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
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - William E. Schwartzman
- 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
| | - Seth M. Meade
- 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
| | - Keying Chen
- 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 Rayyan
- 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
| | - He Feng
- 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
| | - Varoon Aluri
- 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
| | - Natalie N. Mueller
- 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
| | - Raman Bhambra
- 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
| | - Sahaj Bhambra
- 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
| | - Dawn M. Taylor
- 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
- Cleveland Functional Electrical Stimulation Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Rehabilitation Research and Development, Cleveland, OH 44106, USA
- Department of Neurosciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, 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
|
39
|
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: 0.7] [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
|
40
|
McGlynn E, Nabaei V, Ren E, Galeote‐Checa G, Das R, Curia G, Heidari H. The Future of Neuroscience: Flexible and Wireless Implantable Neural Electronics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002693. [PMID: 34026431 PMCID: PMC8132070 DOI: 10.1002/advs.202002693] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/15/2021] [Indexed: 05/04/2023]
Abstract
Neurological diseases are a prevalent cause of global mortality and are of growing concern when considering an ageing global population. Traditional treatments are accompanied by serious side effects including repeated treatment sessions, invasive surgeries, or infections. For example, in the case of deep brain stimulation, large, stiff, and battery powered neural probes recruit thousands of neurons with each pulse, and can invoke a vigorous immune response. This paper presents challenges in engineering and neuroscience in developing miniaturized and biointegrated alternatives, in the form of microelectrode probes. Progress in design and topology of neural implants has shifted the goal post toward highly specific recording and stimulation, targeting small groups of neurons and reducing the foreign body response with biomimetic design principles. Implantable device design recommendations, fabrication techniques, and clinical evaluation of the impact flexible, integrated probes will have on the treatment of neurological disorders are provided in this report. The choice of biocompatible material dictates fabrication techniques as novel methods reduce the complexity of manufacture. Wireless power, the final hurdle to truly implantable neural interfaces, is discussed. These aspects are the driving force behind continued research: significant breakthroughs in any one of these areas will revolutionize the treatment of neurological disorders.
Collapse
Affiliation(s)
- Eve McGlynn
- Microelectronics LabJames Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUnited Kingdom
| | - Vahid Nabaei
- Microelectronics LabJames Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUnited Kingdom
| | - Elisa Ren
- Laboratory of Experimental Electroencephalography and NeurophysiologyDepartment of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModena41125Italy
| | - Gabriel Galeote‐Checa
- Microelectronics LabJames Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUnited Kingdom
| | - Rupam Das
- Microelectronics LabJames Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUnited Kingdom
| | - Giulia Curia
- Laboratory of Experimental Electroencephalography and NeurophysiologyDepartment of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModena41125Italy
| | - Hadi Heidari
- Microelectronics LabJames Watt School of EngineeringUniversity of GlasgowGlasgowG12 8QQUnited Kingdom
| |
Collapse
|
41
|
Thompson CH, Saxena A, Heelan N, Salatino J, Purcell EK. Spatiotemporal patterns of gene expression around implanted silicon electrode arrays. J Neural Eng 2021; 18:10.1088/1741-2552/abf2e6. [PMID: 33780909 PMCID: PMC8650570 DOI: 10.1088/1741-2552/abf2e6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/29/2021] [Indexed: 11/12/2022]
Abstract
Objective.Intracortical brain interfaces are an ever evolving technology with growing potential for clinical and research applications. The chronic tissue response to these devices traditionally has been characterized by glial scarring, inflammation, oxidative stress, neuronal loss, and blood-brain barrier disruptions. The full complexity of the tissue response to implanted devices is still under investigation.Approach.In this study, we have utilized RNA-sequencing to identify the spatiotemporal gene expression patterns in interfacial (within 100µm) and distal (500µm from implant) brain tissue around implanted silicon microelectrode arrays. Naïve, unimplanted tissue served as a control.Main results.The data revealed significant overall differential expression (DE) in contrasts comparing interfacial tissue vs naïve (157 DE genes), interfacial vs distal (94 DE genes), and distal vs naïve tissues (21 DE genes). Our results captured previously characterized mechanisms of the foreign body response, such as astroglial encapsulation, as well as novel mechanisms which have not yet been characterized in the context of indwelling neurotechnologies. In particular, we have observed perturbations in multiple neuron-associated genes which potentially impact the intrinsic function and structure of neurons at the device interface. In addition to neuron-associated genes, the results presented in this study identified significant DE in genes which are associated with oligodendrocyte, microglia, and astrocyte involvement in the chronic tissue response.Significance. The results of this study increase the fundamental understanding of the complexity of tissue response in the brain and provide an expanded toolkit for future investigation into the bio-integration of implanted electronics with tissues in the central nervous system.
Collapse
Affiliation(s)
- Cort H Thompson
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Akash Saxena
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Nicholas Heelan
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States of America
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, United States of America
| | - Joseph Salatino
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Erin K Purcell
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| |
Collapse
|
42
|
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
|
43
|
Sharon A, Jankowski MM, Shmoel N, Erez H, Spira ME. Inflammatory Foreign Body Response Induced by Neuro-Implants in Rat Cortices Depleted of Resident Microglia by a CSF1R Inhibitor and Its Implications. Front Neurosci 2021; 15:646914. [PMID: 33841088 PMCID: PMC8032961 DOI: 10.3389/fnins.2021.646914] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/25/2021] [Indexed: 12/30/2022] Open
Abstract
Inflammatory encapsulation of implanted cortical-neuro-probes [the foreign body response (FBR)] severely limits their use in basic brain research and in clinical applications. A better understanding of the inflammatory FBR is needed to effectively mitigate these critical limitations. Combining the use of the brain permeant colony stimulating factor 1 receptor inhibitor PLX5622 and a perforated polyimide-based multielectrode array platform (PPMP) that can be sectioned along with the surrounding tissue, we examined the contribution of microglia to the formation of inflammatory FBR. To that end, we imaged the inflammatory processes induced by PPMP implantations after eliminating 89-94% of the cortical microglia by PLX5622 treatment. The observations showed that: (I) inflammatory encapsulation of implanted PPMPs proceeds by astrocytes in microglia-free cortices. The activated astrocytes adhered to the PPMP's surfaces. This suggests that the roles of microglia in the FBR might be redundant. (II) PPMP implantation into control or continuously PLX5622-treated rats triggered a localized surge of microglia mitosis. The daughter cells that formed a "cloud" of short-lived (T 1 / 2 ≤ 14 days) microglia around and in contact with the implant surfaces were PLX5622 insensitive. (III) Neuron degeneration by PPMP implantation and the ensuing recovery in time, space, and density progressed in a similar manner in the cortices following 89-94% depletion of microglia. This implies that microglia do not serve a protective role with respect to the neurons. (IV) Although the overall cell composition and dimensions of the encapsulating scar in PLX5622-treated rats differed from the controls, the recorded field potential (FP) qualities and yield were undistinguishable. This is accounted for by assuming that the FP amplitudes in the control and PLX5622-treated rats were related to the seal resistance formed at the interface between the adhering microglia and/or astrocytes and the PPMP platform rather than across the scar tissue. These observations suggest that the prevention of both astrocytes and microglia adhesion to the electrodes is required to improve FP recording quality and yield.
Collapse
Affiliation(s)
- Aviv Sharon
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maciej M. Jankowski
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nava Shmoel
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Erez
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Micha E. Spira
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
44
|
Chen K, Wellman SM, Yaxiaer Y, Eles JR, Kozai TD. In vivo spatiotemporal patterns of oligodendrocyte and myelin damage at the neural electrode interface. Biomaterials 2020; 268:120526. [PMID: 33302121 DOI: 10.1016/j.biomaterials.2020.120526] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022]
Abstract
Intracortical microelectrodes with the ability to detect intrinsic electrical signals and/or deliver electrical stimulation into local brain regions have been a powerful tool to understand brain circuitry and for therapeutic applications to neurological disorders. However, the chronic stability and sensitivity of these intracortical microelectrodes are challenged by overwhelming biological responses, including severe neuronal loss and thick glial encapsulation. Unlike microglia and astrocytes whose activity have been extensively examined, oligodendrocytes and their myelin processes remain poorly studied within the neural interface field. Oligodendrocytes have been widely recognized to modulate electrical signal conductance along axons through insulating myelin segments. Emerging evidence offers an alternative perspective on neuron-oligodendrocyte coupling where oligodendrocytes provide metabolic and neurotrophic support to neurons through cytoplasmic myelin channels and monocarboxylate transporters. This study uses in vivo multi-photon microscopy to gain insights into the dynamics of oligodendrocyte soma and myelin processes in response to chronic device implantation injury over 4 weeks. We observe that implantation induces acute oligodendrocyte injury including initial deformation and substantial myelinosome formation, an early sign of myelin injury. Over chronic implantation periods, myelin and oligodendrocyte soma suffer severe degeneration proximal to the interface. Interestingly, wound healing attempts such as oligodendrogenesis are initiated over time, however they are hampered by continued degeneration near the implant. Nevertheless, this detailed characterization of oligodendrocyte spatiotemporal dynamics during microelectrode-induced inflammation may provide insights for novel intervention targets to facilitate oligodendrogenesis, enhance the integration of neural-electrode interfaces, and improve long-term functional performance.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, USA
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, USA
| | - Yalikun Yaxiaer
- Eberly College of Science, Pennsylvania State University, University Park, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, USA
| | - Takashi Dy Kozai
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, USA; Center for Neuroscience, University of Pittsburgh, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, USA; NeuroTech Center, University of Pittsburgh Brain Institute, USA.
| |
Collapse
|
45
|
Dunlap CF, Colachis SC, Meyers EC, Bockbrader MA, Friedenberg DA. Classifying Intracortical Brain-Machine Interface Signal Disruptions Based on System Performance and Applicable Compensatory Strategies: A Review. Front Neurorobot 2020; 14:558987. [PMID: 33162885 PMCID: PMC7581895 DOI: 10.3389/fnbot.2020.558987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/09/2020] [Indexed: 12/18/2022] Open
Abstract
Brain-machine interfaces (BMIs) record and translate neural activity into a control signal for assistive or other devices. Intracortical microelectrode arrays (MEAs) enable high degree-of-freedom BMI control for complex tasks by providing fine-resolution neural recording. However, chronically implanted MEAs are subject to a dynamic in vivo environment where transient or systematic disruptions can interfere with neural recording and degrade BMI performance. Typically, neural implant failure modes have been categorized as biological, material, or mechanical. While this categorization provides insight into a disruption's causal etiology, it is less helpful for understanding degree of impact on BMI function or possible strategies for compensation. Therefore, we propose a complementary classification framework for intracortical recording disruptions that is based on duration of impact on BMI performance and requirement for and responsiveness to interventions: (1) Transient disruptions interfere with recordings on the time scale of minutes to hours and can resolve spontaneously; (2) Reversible disruptions cause persistent interference in recordings but the root cause can be remedied by an appropriate intervention; (3) Irreversible compensable disruptions cause persistent or progressive decline in signal quality, but their effects on BMI performance can be mitigated algorithmically; and (4) Irreversible non-compensable disruptions cause permanent signal loss that is not amenable to remediation or compensation. This conceptualization of intracortical BMI disruption types is useful for highlighting specific areas for potential hardware improvements and also identifying opportunities for algorithmic interventions. We review recording disruptions that have been reported for MEAs and demonstrate how biological, material, and mechanical mechanisms of disruption can be further categorized according to their impact on signal characteristics. Then we discuss potential compensatory protocols for each of the proposed disruption classes. Specifically, transient disruptions may be minimized by using robust neural decoder features, data augmentation methods, adaptive machine learning models, and specialized signal referencing techniques. Statistical Process Control methods can identify reparable disruptions for rapid intervention. In-vivo diagnostics such as impedance spectroscopy can inform neural feature selection and decoding models to compensate for irreversible disruptions. Additional compensatory strategies for irreversible disruptions include information salvage techniques, data augmentation during decoder training, and adaptive decoding methods to down-weight damaged channels.
Collapse
Affiliation(s)
- Collin F. Dunlap
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
- Medical Devices and Neuromodulation, Battelle Memorial Institute, Columbus, OH, United States
| | - Samuel C. Colachis
- Medical Devices and Neuromodulation, Battelle Memorial Institute, Columbus, OH, United States
| | - Eric C. Meyers
- Medical Devices and Neuromodulation, Battelle Memorial Institute, Columbus, OH, United States
| | - Marcia A. Bockbrader
- Department of Physical Medicine and Rehabilitation, The Ohio State University, Columbus, OH, United States
| | - David A. Friedenberg
- Advanced Analytics and Health Research, Battelle Memorial Institute, Columbus, OH, United States
| |
Collapse
|
46
|
Investigating the Association between Motor Function, Neuroinflammation, and Recording Metrics in the Performance of Intracortical Microelectrode Implanted in Motor Cortex. MICROMACHINES 2020; 11:mi11090838. [PMID: 32899336 PMCID: PMC7570280 DOI: 10.3390/mi11090838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 11/26/2022]
Abstract
Long-term reliability of intracortical microelectrodes remains a challenge for increased acceptance and deployment. There are conflicting reports comparing measurements associated with recording quality with postmortem histology, in attempts to better understand failure of intracortical microelectrodes (IMEs). Our group has recently introduced the assessment of motor behavior tasks as another metric to evaluate the effects of IME implantation. We hypothesized that adding the third dimension to our analysis, functional behavior testing, could provide substantial insight on the health of the tissue, success of surgery/implantation, and the long-term performance of the implanted device. Here we present our novel analysis scheme including: (1) the use of numerical formal concept analysis (nFCA) and (2) a regression analysis utilizing modern model/variable selection. The analyses found complimentary relationships between the variables. The histological variables for glial cell activation had associations between each other, as well as the neuronal density around the electrode interface. The neuronal density had associations to the electrophysiological recordings and some of the motor behavior metrics analyzed. The novel analyses presented herein describe a valuable tool that can be utilized to assess and understand relationships between diverse variables being investigated. These models can be applied to a wide range of ongoing investigations utilizing various devices and therapeutics.
Collapse
|
47
|
Haley RM, Zuckerman ST, Dakhlallah H, Capadona JR, von Recum HA, Ereifej ES. Resveratrol Delivery from Implanted Cyclodextrin Polymers Provides Sustained Antioxidant Effect on Implanted Neural Probes. Int J Mol Sci 2020; 21:ijms21103579. [PMID: 32438593 PMCID: PMC7279014 DOI: 10.3390/ijms21103579] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/08/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Intracortical microelectrodes are valuable tools used to study and treat neurological diseases. Due in large part to the oxidative stress and inflammatory response occurring after electrode implantation, the signal quality of these electrodes decreases over time. To alleviate this response, resveratrol, a natural antioxidant which elicits neuroprotective effects through reduction of oxidative stress, was utilized. This work compares traditional systemic delivery of resveratrol to the novel cyclodextrin polymer (pCD) local delivery approach presented herein, both in vitro and in vivo. The pCD displayed an extended resveratrol release for 100 days, as well as 60 days of free radical scavenging activity in vitro. In vivo results indicated that our pCD delivery system successfully delivered resveratrol to the brain with a sustained release for the entire short-duration study (up to 7 days). Interestingly, significantly greater concentrations of resveratrol metabolites were found at the intracortical probe implantation site compared to the systemic administration of resveratrol. Together, our pilot results provide support for the possibility of improving the delivery of resveratrol in an attempt to stabilize long-term neural interfacing applications.
Collapse
Affiliation(s)
- Rebecca M. Haley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (R.M.H.); (J.R.C.)
| | - Sean T. Zuckerman
- Affinity Therapeutics, LLC, 11000 Cedar Avenue, Suite 285, Cleveland, OH 44106, USA;
| | - Hassan Dakhlallah
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA;
| | - Jeffery R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (R.M.H.); (J.R.C.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
| | - Horst A. von Recum
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (R.M.H.); (J.R.C.)
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
- Correspondence: (H.A.v.R.); (E.S.E.)
| | - Evon S. Ereifej
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA; (R.M.H.); (J.R.C.)
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA;
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: (H.A.v.R.); (E.S.E.)
| |
Collapse
|
48
|
Mahajan S, Hermann JK, Bedell HW, Sharkins JA, Chen L, Chen K, Meade SM, Smith CS, Rayyan J, Feng H, Kim Y, Schiefer MA, Taylor DM, Capadona JR, Ereifej ES. Toward Standardization of Electrophysiology and Computational Tissue Strain in Rodent Intracortical Microelectrode Models. Front Bioeng Biotechnol 2020; 8:416. [PMID: 32457888 PMCID: PMC7225268 DOI: 10.3389/fbioe.2020.00416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/14/2020] [Indexed: 12/26/2022] Open
Abstract
Progress has been made in the field of neural interfacing using both mouse and rat models, yet standardization of these models' interchangeability has yet to be established. The mouse model allows for transgenic, optogenetic, and advanced imaging modalities which can be used to examine the biological impact and failure mechanisms associated with the neural implant itself. The ability to directly compare electrophysiological data between mouse and rat models is crucial for the development and assessment of neural interfaces. The most obvious difference in the two rodent models is size, which raises concern for the role of device-induced tissue strain. Strain exerted on brain tissue by implanted microelectrode arrays is hypothesized to affect long-term recording performance. Therefore, understanding any potential differences in tissue strain caused by differences in the implant to tissue size ratio is crucial for validating the interchangeability of rat and mouse models. Hence, this study is aimed at investigating the electrophysiological variances and predictive device-induced tissue strain. Rat and mouse electrophysiological recordings were collected from implanted animals for eight weeks. A finite element model was utilized to assess the tissue strain from implanted intracortical microelectrodes, taking into account the differences in the depth within the cortex, implantation depth, and electrode geometry between the two models. The rat model demonstrated a larger percentage of channels recording single unit activity and number of units recorded per channel at acute but not chronic time points, relative to the mouse model Additionally, the finite element models also revealed no predictive differences in tissue strain between the two rodent models. Collectively our results show that these two models are comparable after taking into consideration some recommendations to maintain uniform conditions for future studies where direct comparisons of electrophysiological and tissue strain data between the two animal models will be required.
Collapse
Affiliation(s)
- Shreya Mahajan
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, United States
| | - John K. Hermann
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Hillary W. Bedell
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Jonah A. Sharkins
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Lei Chen
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Keying Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Seth M. Meade
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Cara S. Smith
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Jacob Rayyan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - He Feng
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Youjoung Kim
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Matthew A. Schiefer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Dawn M. Taylor
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Department of Neuroscience, The Cleveland Clinic, Cleveland, OH, United States
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Evon S. Ereifej
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
49
|
Fallegger F, Schiavone G, Lacour SP. Conformable Hybrid Systems for Implantable Bioelectronic Interfaces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903904. [PMID: 31608508 DOI: 10.1002/adma.201903904] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/20/2019] [Indexed: 05/27/2023]
Abstract
Conformable bioelectronic systems are promising tools that may aid the understanding of diseases, alleviate pathological symptoms such as chronic pain, heart arrhythmia, and dysfunctions, and assist in reversing conditions such as deafness, blindness, and paralysis. Combining reduced invasiveness with advanced electronic functions, hybrid bioelectronic systems have evolved tremendously in the last decade, pushed by progress in materials science, micro- and nanofabrication, system assembly and packaging, and biomedical engineering. Hybrid integration refers here to a technological approach to embed within mechanically compliant carrier substrates electronic components and circuits prepared with traditional electronic materials. This combination leverages mechanical and electronic performance of polymer substrates and device materials, respectively, and offers many opportunities for man-made systems to communicate with the body with unmet precision. However, trade-offs between materials selection, manufacturing processes, resolution, electrical function, mechanical integrity, biointegration, and reliability should be considered. Herein, prominent trends in manufacturing conformable hybrid systems are analyzed and key design, function, and validation principles are outlined together with the remaining challenges to produce reliable conformable, hybrid bioelectronic systems.
Collapse
Affiliation(s)
- Florian Fallegger
- Bertarelli Foundation Chair in Neuroprosthetic Technology, Laboratory for Soft Bioelectronic Interfaces, Institute of Microengineering, Institute of Bioengineering, Center for Neuroprosthetics, Ecole Polytechnique Fédérale de Lausanne, 1202, Geneva, Switzerland
| | - Giuseppe Schiavone
- Bertarelli Foundation Chair in Neuroprosthetic Technology, Laboratory for Soft Bioelectronic Interfaces, Institute of Microengineering, Institute of Bioengineering, Center for Neuroprosthetics, Ecole Polytechnique Fédérale de Lausanne, 1202, Geneva, Switzerland
| | - Stéphanie P Lacour
- Bertarelli Foundation Chair in Neuroprosthetic Technology, Laboratory for Soft Bioelectronic Interfaces, Institute of Microengineering, Institute of Bioengineering, Center for Neuroprosthetics, Ecole Polytechnique Fédérale de Lausanne, 1202, Geneva, Switzerland
| |
Collapse
|
50
|
Obidin N, Tasnim F, Dagdeviren C. The Future of Neuroimplantable Devices: A Materials Science and Regulatory Perspective. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901482. [PMID: 31206827 DOI: 10.1002/adma.201901482] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/12/2019] [Indexed: 06/09/2023]
Abstract
The past two decades have seen unprecedented progress in the development of novel materials, form factors, and functionalities in neuroimplantable technologies, including electrocorticography (ECoG) systems, multielectrode arrays (MEAs), Stentrode, and deep brain probes. The key considerations for the development of such devices intended for acute implantation and chronic use, from the perspective of biocompatible hybrid materials incorporation, conformable device design, implantation procedures, and mechanical and biological risk factors, are highlighted. These topics are connected with the role that the U.S. Food and Drug Administration (FDA) plays in its regulation of neuroimplantable technologies based on the above parameters. Existing neuroimplantable devices and efforts to improve their materials and implantation protocols are first discussed in detail. The effects of device implantation with regards to biocompatibility and brain heterogeneity are then explored. Topics examined include brain-specific risk factors, such as bacterial infection, tissue scarring, inflammation, and vasculature damage, as well as efforts to manage these dangers through emerging hybrid, bioelectronic device architectures. The current challenges of gaining clinical approval by the FDA-in particular, with regards to biological, mechanical, and materials risk factors-are summarized. The available regulatory pathways to accelerate next-generation neuroimplantable devices to market are then discussed.
Collapse
Affiliation(s)
- Nikita Obidin
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Farita Tasnim
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Canan Dagdeviren
- MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|