1
|
Gedela NSS, Salim S, Radawiec RD, Richie J, Chestek C, Draelos A, Pelled G. Single unit electrophysiology recordings and computational modeling can predict octopus arm movement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612676. [PMID: 39345497 PMCID: PMC11430158 DOI: 10.1101/2024.09.13.612676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The octopus simplified nervous system holds the potential to reveal principles of motor circuits and improve brain-machine interface devices through computational modeling with machine learning and statistical analysis. Here, an array of carbon electrodes providing single-unit electrophysiology recordings were implanted into the octopus anterior nerve cord. The number of spikes and arm movements in response to stimulation at different locations along the arm were recorded. We observed that the number of spikes occurring within the first 100ms after stimulation were predictive of the resultant movement response. Computational models showed that temporal electrophysiological features could be used to predict whether an arm movement occurred with 88.64% confidence, and if it was a lateral arm movement or a grasping motion with 75.45% confidence. Both supervised and unsupervised methods were applied to gain streaming measurements of octopus arm movements and how their motor circuitry produces rich movement types in real time. Deep learning models and unsupervised dimension reduction identified a consistent set of features that could be used to distinguish different types of arm movements. These models generated predictions for how to evoke a particular, complex movement in an orchestrated sequence for an individual motor circuit.
Collapse
Affiliation(s)
- Nitish Satya Sai Gedela
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| | - Sachin Salim
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Ryan D Radawiec
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| | - Julianna Richie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Cynthia Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Anne Draelos
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Galit Pelled
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
- Department of Radiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
2
|
Vara H, Hernández-Labrado GR, Alves-Sampaio A, Collazos-Castro JE. Stability of Conducting Polymer-Coated Carbon Microfibers for Long-Term Electrical Stimulation of Injured Neural Tissue. Polymers (Basel) 2024; 16:2093. [PMID: 39065410 PMCID: PMC11280860 DOI: 10.3390/polym16142093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Electroactive microfiber-based scaffolds aid neural tissue repair. Carbon microfibers (CMFs) coated with the conducting polymer poly(3,4-ethylenedioxythiophene) doped with poly[(4-styrenesulfonic acid)-co-(maleic acid)] (PEDOT:PSS-co-MA) provide efficient support and guidance to regrowing axons across spinal cord lesions in rodents and pigs. We investigated the electrical and structural performance of PEDOT:PSS-co-MA-coated carbon MFs (PCMFs) for long-term, biphasic electrical stimulation (ES). Chronopotentiometry and electrochemical impedance spectroscopy (EIS) allowed the characterization of charge transfer in PCMFs during ES in vitro, and morphological changes were assessed by scanning electron microscopy (SEM). PCMFs that were 4 mm long withstood two-million-biphasic pulses without reaching cytotoxic voltages, with a 6 mm length producing optimal results. Although EIS and SEM unveiled some polymer deterioration in the 6 mm PCMFs, no significant changes in voltage excursions appeared. For the preliminary testing of the electrical performance of PCMFs in vivo, we used 12 mm long, 20-microfiber assemblies interconnected by metallic microwires. PCMFs-assemblies were implanted in two spinal cord-injured pigs and submitted to ES for 10 days. A cobalt-alloy interconnected assembly showed safe voltages for about 1.5 million-pulses and was electrically functional at 1-month post-implantation, suggesting its suitability for sub-chronic ES, as likely required for spinal cord repair. However, improving polymer adhesion to the carbon substrate is still needed to use PCMFs for prolonged ES.
Collapse
Affiliation(s)
- Hugo Vara
- Neural Repair and Biomaterials Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda S-N, 45071 Toledo, Spain; (H.V.); (A.A.-S.)
| | - Gabriel Raúl Hernández-Labrado
- Escuela de Ingeniería Industrial y Aeroespacial, Universidad de Castilla-La Mancha, Avda. Carlos III, 45071 Toledo, Spain;
| | - Alexandra Alves-Sampaio
- Neural Repair and Biomaterials Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda S-N, 45071 Toledo, Spain; (H.V.); (A.A.-S.)
| | - Jorge E. Collazos-Castro
- Neural Repair and Biomaterials Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Finca la Peraleda S-N, 45071 Toledo, Spain; (H.V.); (A.A.-S.)
| |
Collapse
|
3
|
Amjad U, Choi J, Gibson DJ, Murray R, Graybiel AM, Schwerdt HN. Synchronous Measurements of Extracellular Action Potentials and Neurochemical Activity with Carbon Fiber Electrodes in Nonhuman Primates. eNeuro 2024; 11:ENEURO.0001-24.2024. [PMID: 38918051 PMCID: PMC11232371 DOI: 10.1523/eneuro.0001-24.2024] [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: 12/27/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Measuring the dynamic relationship between neuromodulators, such as dopamine, and neuronal action potentials is imperative to understand how these fundamental modes of neural signaling interact to mediate behavior. We developed methods to measure concurrently dopamine and extracellular action potentials (i.e., spikes) in monkeys. Standard fast-scan cyclic voltammetric (FSCV) electrochemical (EChem) and electrophysiological (EPhys) recording systems are combined and used to collect spike and dopamine signals, respectively, from an array of carbon fiber (CF) sensors implanted in the monkey striatum. FSCV requires the application of small voltages at the implanted sensors to measure redox currents generated from target molecules, such as dopamine. These applied voltages create artifacts at neighboring EPhys measurement sensors which may lead to misclassification of these signals as physiological spikes. Therefore, simple automated temporal interpolation algorithms were designed to remove these artifacts and enable accurate spike extraction. We validated these methods using simulated artifacts and demonstrated an average spike recovery rate of 84.5%. We identified and discriminated cell type-specific units in the monkey striatum that were shown to correlate to specific behavioral task parameters related to reward size and eye movement direction. Synchronously recorded spike and dopamine signals displayed contrasting relations to the task variables, suggesting a complex relationship between these two modes of neural signaling. Future application of our methods will help advance our understanding of the interactions between neuromodulator signaling and neuronal activity, to elucidate more detailed mechanisms of neural circuitry and plasticity mediating behaviors in health and in disease.
Collapse
Affiliation(s)
- Usamma Amjad
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Jiwon Choi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815
| | - Daniel J Gibson
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Raymond Murray
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Ann M Graybiel
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Helen N Schwerdt
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815
| |
Collapse
|
4
|
Roy J, Sarah UT, Lissorgues G, Français O, Rezgui A, Poulichet P, Takhedmit H, Scorsone E, Rousseau L. Stability Study of Synthetic Diamond Using a Thermally Controlled Biological Environment: Application towards Long-Lasting Neural Prostheses. SENSORS (BASEL, SWITZERLAND) 2024; 24:3619. [PMID: 38894410 PMCID: PMC11175334 DOI: 10.3390/s24113619] [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: 03/15/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024]
Abstract
This paper demonstrates, for the first time, the stability of synthetic diamond as a passive layer within neural implants. Leveraging the exceptional biocompatibility of intrinsic nanocrystalline diamond, a comprehensive review of material aging analysis in the context of in-vivo implants is provided. This work is based on electric impedance monitoring through the formulation of an analytical model that scrutinizes essential parameters such as the deposited metal resistivity, insulation between conductors, changes in electrode geometry, and leakage currents. The evolution of these parameters takes place over an equivalent period of approximately 10 years. The analytical model, focusing on a fractional capacitor, provides nuanced insights into the surface conductivity variation. A comparative study is performed between a classical polymer material (SU8) and synthetic diamond. Samples subjected to dynamic impedance analysis reveal distinctive patterns over time, characterized by their physical degradation. The results highlight the very high stability of diamond, suggesting promise for the electrode's enduring viability. To support this analysis, microscopic and optical measurements conclude the paper and confirm the high stability of diamond and its strong potential as a material for neural implants with long-life use.
Collapse
Affiliation(s)
- Jordan Roy
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Umme Tabassum Sarah
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Gaëlle Lissorgues
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Olivier Français
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Abir Rezgui
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Patrick Poulichet
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Hakim Takhedmit
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| | - Emmanuel Scorsone
- Diamond Sensors Laboratory, CEA-LIST, F-91190 Gif-sur-Yvette, France;
| | - Lionel Rousseau
- ESYCOM Laboratory for Electronics, Communication and Microsystems, CNRS UMR 9007, F-77454 Marne-la-Vallée, France; (J.R.); (U.T.S.); (G.L.); (O.F.); (A.R.); (P.P.); (H.T.)
| |
Collapse
|
5
|
McNamara IN, Wellman SM, Li L, Eles JR, Savya S, Sohal HS, Angle MR, Kozai TDY. Electrode sharpness and insertion speed reduce tissue damage near high-density penetrating arrays. J Neural Eng 2024; 21:026030. [PMID: 38518365 DOI: 10.1088/1741-2552/ad36e1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
Objective. Over the past decade, neural electrodes have played a crucial role in bridging biological tissues with electronic and robotic devices. This study focuses on evaluating the optimal tip profile and insertion speed for effectively implanting Paradromics' high-density fine microwire arrays (FμA) prototypes into the primary visual cortex (V1) of mice and rats, addressing the challenges associated with the 'bed-of-nails' effect and tissue dimpling.Approach. Tissue response was assessed by investigating the impact of electrodes on the blood-brain barrier (BBB) and cellular damage, with a specific emphasis on tailored insertion strategies to minimize tissue disruption during electrode implantation.Main results.Electro-sharpened arrays demonstrated a marked reduction in cellular damage within 50μm of the electrode tip compared to blunt and angled arrays. Histological analysis revealed that slow insertion speeds led to greater BBB compromise than fast and pneumatic methods. Successful single-unit recordings validated the efficacy of the optimized electro-sharpened arrays in capturing neural activity.Significance.These findings underscore the critical role of tailored insertion strategies in minimizing tissue damage during electrode implantation, highlighting the suitability of electro-sharpened arrays for long-term implant applications. This research contributes to a deeper understanding of the complexities associated with high-channel-count microelectrode array implantation, emphasizing the importance of meticulous assessment and optimization of key parameters for effective integration and minimal tissue disruption. By elucidating the interplay between insertion parameters and tissue response, our study lays a strong foundation for the development of advanced implantable devices with a reduction in reactive gliosis and improved performance in neural recording applications.
Collapse
Affiliation(s)
- Ingrid N McNamara
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Lehong Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sajishnu Savya
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | | | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center of the Basis of Neural Cognition, Pittsburgh, PA, United States of America
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
6
|
Huan Y, Tibbetts BN, Richie JM, Chestek CA, Chiel HJ. Intracellular neural control of an active feeding structure in Aplysia using a carbon fiber electrode array. J Neurosci Methods 2024; 404:110077. [PMID: 38336092 PMCID: PMC11136531 DOI: 10.1016/j.jneumeth.2024.110077] [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: 11/10/2023] [Revised: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND To study neural control of behavior, intracellular recording and stimulation of many neurons in freely moving animals would be ideal. However, current technologies limit the number of neurons that can be monitored and manipulated. A new technology has become available for intracellular recording and stimulation which we demonstrate in the tractable nervous system of Aplysia. NEW METHOD Carbon fiber electrode arrays (whose tips are coated with platinum-iridium) were used with an in vitro feeding preparation to intracellularly record from and to control the activity of multiple neurons during feeding movements. RESULTS In an in vitro feeding preparation, the carbon fiber electrode arrays recorded action potentials and subthreshold synaptic potentials during feeding movements. Depolarizing or hyperpolarizing currents activated or inhibited identified neurons (respectively), manipulating the movements of the feeding apparatus. COMPARISON WITH EXISTING METHOD(S) Standard glass microelectrodes that are commonly used for intracellular recording are stiff, liable to break in response to movement, and require many micromanipulators to be precisely positioned. In contrast, carbon fiber arrays are less sensitive to movement, but are capable of multiple channels of intracellular recording and stimulation. CONCLUSIONS Carbon fiber arrays are a novel technology for intracellular recording that can be used in moving preparations. They can record both action potentials and synaptic activity in multiple neurons and can be used to stimulate multiple neurons in complex patterns.
Collapse
Affiliation(s)
- Yu Huan
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106-7080, USA
| | - Benjamin N Tibbetts
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106-7080, USA
| | - Julianna M Richie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hillel J Chiel
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106-7080, USA; Department of Neuroscience, Case Western Reserve University, Cleveland, OH 44106-7080, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106-7080, USA.
| |
Collapse
|
7
|
Kim J, Huang H, Gilbert ET, Arndt KC, English DF, Jia X. T-DOpE probes reveal sensitivity of hippocampal oscillations to cannabinoids in behaving mice. Nat Commun 2024; 15:1686. [PMID: 38402238 PMCID: PMC10894268 DOI: 10.1038/s41467-024-46021-4] [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: 07/19/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
Understanding the neural basis of behavior requires monitoring and manipulating combinations of physiological elements and their interactions in behaving animals. We developed a thermal tapering process enabling fabrication of low-cost, flexible probes combining ultrafine features: dense electrodes, optical waveguides, and microfluidic channels. Furthermore, we developed a semi-automated backend connection allowing scalable assembly. We demonstrate T-DOpE (Tapered Drug delivery, Optical stimulation, and Electrophysiology) probes achieve in single neuron-scale devices (1) high-fidelity electrophysiological recording (2) focal drug delivery and (3) optical stimulation. The device tip can be miniaturized (as small as 50 µm) to minimize tissue damage while the ~20 times larger backend allows for industrial-scale connectorization. T-DOpE probes implanted in mouse hippocampus revealed canonical neuronal activity at the level of local field potentials (LFP) and neural spiking. Taking advantage of the triple-functionality of these probes, we monitored LFP while manipulating cannabinoid receptors (CB1R; microfluidic agonist delivery) and CA1 neuronal activity (optogenetics). Focal infusion of CB1R agonist downregulated theta and sharp wave-ripple oscillations (SPW-Rs). Furthermore, we found that CB1R activation reduces sharp wave-ripples by impairing the innate SPW-R-generating ability of the CA1 circuit.
Collapse
Affiliation(s)
- Jongwoon Kim
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Hengji Huang
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Earl T Gilbert
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - Kaiser C Arndt
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | | | - Xiaoting Jia
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA, USA.
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA.
- Department of Materials Science and Engineering, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
8
|
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
|
9
|
Faul EBA, Broussard AM, Rivera DR, Pwint MY, Wu B, Cao Q, Bailey D, Cui XT, Castagnola E. Batch Fabrication of Microelectrode Arrays with Glassy Carbon Microelectrodes and Interconnections for Neurochemical Sensing: Promises and Challenges. MICROMACHINES 2024; 15:277. [PMID: 38399004 PMCID: PMC10892456 DOI: 10.3390/mi15020277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Flexible multielectrode arrays with glassy carbon (GC) electrodes and metal interconnection (hybrid MEAs) have shown promising performance in multi-channel neurochemical sensing. A primary challenge faced by hybrid MEAs fabrication is the adhesion of the metal traces with the GC electrodes, as prolonged electrical and mechanical stimulation can lead to adhesion failure. Previous devices with GC electrodes and interconnects made of a homogeneous material (all GC) demonstrated exceptional electrochemical stability but required miniaturization for enhanced tissue integration and chronic electrochemical sensing. In this study, we used two different methods for the fabrication of all GC-MEAs on thin flexible substrates with miniaturized features. The first method, like that previously reported, involves a double pattern-transfer photolithographic process, including transfer-bonding on temporary polymeric support. The second method requires a double-etching process, which uses a 2 µm-thick low stress silicon nitride coating of the Si wafer as the bottom insulator layer for the MEAs, bypassing the pattern-transfer and demonstrating a novel technique with potential advantages. We confirmed the feasibility of the two fabrication processes by verifying the practical conductivity of 3 µm-wide 2 µm-thick GC traces, the GC microelectrode functionality, and their sensing capability for the detection of serotonin using fast scan cyclic voltammetry. Through the exchange and discussion of insights regarding the strengths and limitations of these microfabrication methods, our goal is to propel the advancement of GC-based MEAs for the next generation of neural interface devices.
Collapse
Affiliation(s)
- Emma-Bernadette A. Faul
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (E.-B.A.F.); (A.M.B.); (D.R.R.)
| | - Austin M. Broussard
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (E.-B.A.F.); (A.M.B.); (D.R.R.)
| | - Daniel R. Rivera
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (E.-B.A.F.); (A.M.B.); (D.R.R.)
| | - May Yoon Pwint
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.Y.P.); (B.W.); (Q.C.); (X.T.C.)
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Bingchen Wu
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.Y.P.); (B.W.); (Q.C.); (X.T.C.)
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Qun Cao
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.Y.P.); (B.W.); (Q.C.); (X.T.C.)
| | - Davis Bailey
- Institute for Micromanufacturing, Louisiana Tech University, Ruston, LA 15213, USA;
| | - X. Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.Y.P.); (B.W.); (Q.C.); (X.T.C.)
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3110, USA
| | - Elisa Castagnola
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (E.-B.A.F.); (A.M.B.); (D.R.R.)
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (M.Y.P.); (B.W.); (Q.C.); (X.T.C.)
| |
Collapse
|
10
|
Richie J, Letner JG, Mclane-Svoboda A, Huan Y, Ghaffari DH, Valle ED, Patel PR, Chiel HJ, Pelled G, Weiland JD, Chestek CA. Fabrication and Validation of Sub-Cellular Carbon Fiber Electrodes. IEEE Trans Neural Syst Rehabil Eng 2024; 32:739-749. [PMID: 38294928 PMCID: PMC10919889 DOI: 10.1109/tnsre.2024.3360866] [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] [Indexed: 02/02/2024]
Abstract
Multielectrode arrays for interfacing with neurons are of great interest for a wide range of medical applications. However, current electrodes cause damage over time. Ultra small carbon fibers help to address issues but controlling the electrode site geometry is difficult. Here we propose a methodology to create small, pointed fiber electrodes (SPFe). We compare the SPFe to previously made blowtorched fibers in characterization. The SPFe result in small site sizes [Formula: see text] with consistently sharp points (20.8 ± 7.64°). Additionally, these electrodes were able to record and/or stimulate neurons multiple animal models including rat cortex, mouse retina, Aplysia ganglia and octopus axial cord. In rat cortex, these electrodes recorded significantly higher peak amplitudes than the traditional blowtorched fibers. These SPFe may be applicable to a wide range of applications requiring a highly specific interface with individual neurons.
Collapse
|
11
|
Jiao Y, Lei M, Zhu J, Chang R, Qu X. Advances in electrode interface materials and modification technologies for brain-computer interfaces. BIOMATERIALS TRANSLATIONAL 2023; 4:213-233. [PMID: 38282708 PMCID: PMC10817795 DOI: 10.12336/biomatertransl.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/13/2023] [Accepted: 11/24/2023] [Indexed: 01/30/2024]
Abstract
Recent advances in neuroelectrode interface materials and modification technologies are reviewed. Brain-computer interface is the new method of human-computer interaction, which not only can realise the exchange of information between the human brain and external devices, but also provides a brand-new means for the diagnosis and treatment of brain-related diseases. The neural electrode interface part of brain-computer interface is an important area for electrical, optical and chemical signal transmission between brain tissue system and external electronic devices, which determines the performance of brain-computer interface. In order to solve the problems of insufficient flexibility, insufficient signal recognition ability and insufficient biocompatibility of traditional rigid electrodes, researchers have carried out extensive studies on the neuroelectrode interface in terms of materials and modification techniques. This paper introduces the biological reactions that occur in neuroelectrodes after implantation into brain tissue and the decisive role of the electrode interface for electrode function. Following this, the latest research progress on neuroelectrode materials and interface materials is reviewed from the aspects of neuroelectrode materials and modification technologies, firstly taking materials as a clue, and then focusing on the preparation process of neuroelectrode coatings and the design scheme of functionalised structures.
Collapse
Affiliation(s)
- Yunke Jiao
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Miao Lei
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Jianwei Zhu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Ronghang Chang
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Xue Qu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Material Science and Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
- Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang Province, China
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai, China
| |
Collapse
|
12
|
Khatib M, Zhao ET, Wei S, Abramson A, Bishop ES, Chen CH, Thomas AL, Xu C, Park J, Lee Y, Hamnett R, Yu W, Root SE, Yuan L, Chakhtoura D, Kim KK, Zhong D, Nishio Y, Zhao C, Wu C, Jiang Y, Zhang A, Li J, Wang W, Salimi-Jazi F, Rafeeqi TA, Hemed NM, Tok JBH, Chen X, Kaltschmidt JA, Dunn JC, Bao Z. Spiral NeuroString: High-Density Soft Bioelectronic Fibers for Multimodal Sensing and Stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560482. [PMID: 37873341 PMCID: PMC10592902 DOI: 10.1101/2023.10.02.560482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Bioelectronic fibers hold promise for both research and clinical applications due to their compactness, ease of implantation, and ability to incorporate various functionalities such as sensing and stimulation. However, existing devices suffer from bulkiness, rigidity, limited functionality, and low density of active components. These limitations stem from the difficulty to incorporate many components on one-dimensional (1D) fiber devices due to the incompatibility of conventional microfabrication methods (e.g., photolithography) with curved, thin and long fiber structures. Herein, we introduce a fabrication approach, ‶spiral transformation″, to convert two-dimensional (2D) films containing microfabricated devices into 1D soft fibers. This approach allows for the creation of high density multimodal soft bioelectronic fibers, termed Spiral NeuroString (S-NeuroString), while enabling precise control over the longitudinal, angular, and radial positioning and distribution of the functional components. We show the utility of S-NeuroString for motility mapping, serotonin sensing, and tissue stimulation within the dynamic and soft gastrointestinal (GI) system, as well as for single-unit recordings in the brain. The described bioelectronic fibers hold great promises for next-generation multifunctional implantable electronics.
Collapse
Affiliation(s)
- Muhammad Khatib
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Eric Tianjiao Zhao
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Shiyuan Wei
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Alex Abramson
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Estelle Spear Bishop
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California, USA
| | - Chih-Hsin Chen
- Department of Surgery/Pediatric Surgery, Stanford University, Stanford, CA, USA
| | - Anne-Laure Thomas
- Department of Surgery/Pediatric Surgery, Stanford University, Stanford, CA, USA
| | - Chengyi Xu
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jaeho Park
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Yeongjun Lee
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Ryan Hamnett
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA
| | - Weilai Yu
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Samuel E. Root
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Lei Yuan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Dorine Chakhtoura
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Kyun Kyu Kim
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Donglai Zhong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Yuya Nishio
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Chuanzhen Zhao
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Can Wu
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Yuanwen Jiang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Anqi Zhang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Jinxing Li
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48823, USA
| | - Weichen Wang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | | | - Talha A. Rafeeqi
- Department of Surgery/Pediatric Surgery, Stanford University, Stanford, CA, USA
| | - Nofar Mintz Hemed
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Jeffrey B.-H. Tok
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Xiaoke Chen
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Julia A. Kaltschmidt
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, 94305, USA
| | - James C.Y. Dunn
- Department of Surgery/Pediatric Surgery, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Zhenan Bao
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
13
|
Seo KJ, Hill M, Ryu J, Chiang CH, Rachinskiy I, Qiang Y, Jang D, Trumpis M, Wang C, Viventi J, Fang H. A Soft, High-Density Neuroelectronic Array. NPJ FLEXIBLE ELECTRONICS 2023; 7:40. [PMID: 37692908 PMCID: PMC10487278 DOI: 10.1038/s41528-023-00271-2] [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: 03/27/2023] [Accepted: 07/23/2023] [Indexed: 09/12/2023]
Abstract
Techniques to study brain activities have evolved dramatically, yet tremendous challenges remain in acquiring high-throughput electrophysiological recordings minimally invasively. Here, we develop an integrated neuroelectronic array that is filamentary, high-density and flexible. Specifically, with a design of single-transistor multiplexing and current sensing, the total 256 neuroelectrodes achieve only a 2.3 × 0.3 mm2 area, unprecedentedly on a flexible substrate. A novel single-transistor multiplexing acquisition circuit further reduces noise from the electrodes, decreased the footprint of each pixel, and potentially increased the device lifetime. The filamentary neuroelectronic array also integrates with a rollable contact pad design, allowing the device to be injected through a syringe, enabling potential minimally invasive array delivery. Successful acute auditory experiments in rats validate the ability of the array to record neural signals with high tone decoding accuracy. Together, these results establish soft, high-density neuroelectronic arrays as promising devices for neuroscience research and clinical applications.
Collapse
Affiliation(s)
- Kyung Jin Seo
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Mackenna Hill
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Jaehyeon Ryu
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755 USA
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA 02115 USA
| | - Chia-Han Chiang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Iakov Rachinskiy
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Yi Qiang
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Dongyeol Jang
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755 USA
| | - Michael Trumpis
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Charles Wang
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Jonathan Viventi
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Hui Fang
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755 USA
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA 02115 USA
| |
Collapse
|
14
|
Jiracek-Sapieha L, Fluker K, Judy J. Reactive-Accelerated-Aging Testing of Thinned Tissue-Engineered Electronic Nerve Interfaces. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38082987 DOI: 10.1109/embc40787.2023.10340337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Tissue responses can cause a significant reduction in the performance of microelectrode-based devices implanted into neural tissue. Since the reduction of the thickness of implants has been shown to reduce tissue response, in this work we report on our effects to reduce the thickness of our tissue-engineered-electronic-nerve-interface (TEENI) devices and characterize their long-term reliability in a harsh environment. We were able to reduce the thickness of the TEENI threads that are to be located in nerve tissue from ~10 μm to ~2.5 μm in total thickness. To maintain the handleability needed during the assembly of the TEENI device into the hydrogel-based scaffold, we maintained full thickness elsewhere in the TEENI device and added support rails. During longitudinal reactive-accelerated-aging (RAA) experiments performed over 6 days and at 67°C, which corresponds to ~48 days in tissue, we observed that some channels maintain a stable impedance and others do not. Although analyses performed using a scanning electron microscope could clearly reveal delamination in some channels that exhibited large changes in impedance, it did not always correlate. Some channels with significant changes in impedance did not exhibit any observable delamination. Additional work is needed to study the relationship between changes in impedance and structural changes in the device, with the goal of improving device design to achieve longer-lasting devices.
Collapse
|
15
|
Kim J, Huang H, Gilbert E, Arndt K, English DF, Jia X. Tapered Drug delivery, Optical stimulation, and Electrophysiology (T-DOpE) probes reveal the importance of cannabinoid signaling in hippocampal CA1 oscillations in behaving mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544251. [PMID: 37333172 PMCID: PMC10274863 DOI: 10.1101/2023.06.08.544251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Understanding the neural basis of behavior requires monitoring and manipulating combinations of physiological elements and their interactions in behaving animals. Here we developed a thermal tapering process (TTP) which enables the fabrication of novel, low-cost, flexible probes that combine ultrafine features of dense electrodes, optical waveguides, and microfluidic channels. Furthermore, we developed a semi-automated backend connection allowing scalable assembly of the probes. We demonstrate that our T-DOpE ( T apered D rug delivery, Op tical stimulation, and E lectrophysiology) probe achieves in a single neuron-scale device (1) high-fidelity electrophysiological recording (2) focal drug delivery and (3) optical stimulation. With a tapered geometry, the device tip can be minimized (as small as 50 μm) to ensure minimal tissue damage while the backend is ~20 times larger allowing for direct integration with industrial-scale connectorization. Acute and chronic implantation of the probes in mouse hippocampus CA1 revealed canonical neuronal activity at the level of local field potentials and spiking. Taking advantage of the triple-functionality of the T-DOpE probe, we monitored local field potentials with simultaneous manipulation of endogenous type 1 cannabinoid receptors (CB1R; via microfluidic agonist delivery) and CA1 pyramidal cell membrane potential (optogenetic activation). Electro-pharmacological experiments revealed that focal infusion of CB1R agonist CP-55,940 in dorsal CA1 downregulated theta and sharp wave-ripple oscillations. Furthermore, using the full electro-pharmacological-optical feature set of the T-DOpE probe we found that CB1R activation reduces sharp wave-ripples (SPW-Rs) by impairing the innate SPW-R-generating ability of the CA1 circuit.
Collapse
|
16
|
Luan L, Yin R, Zhu H, Xie C. Emerging Penetrating Neural Electrodes: In Pursuit of Large Scale and Longevity. Annu Rev Biomed Eng 2023; 25:185-205. [PMID: 37289556 PMCID: PMC11078330 DOI: 10.1146/annurev-bioeng-090622-050507] [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] [Indexed: 06/10/2023]
Abstract
Penetrating neural electrodes provide a powerful approach to decipher brain circuitry by allowing for time-resolved electrical detections of individual action potentials. This unique capability has contributed tremendously to basic and translational neuroscience, enabling both fundamental understandings of brain functions and applications of human prosthetic devices that restore crucial sensations and movements. However, conventional approaches are limited by the scarce number of available sensing channels and compromised efficacy over long-term implantations. Recording longevity and scalability have become the most sought-after improvements in emerging technologies. In this review, we discuss the technological advances in the past 5-10 years that have enabled larger-scale, more detailed, and longer-lasting recordings of neural circuits at work than ever before. We present snapshots of the latest advances in penetration electrode technology, showcase their applications in animal models and humans, and outline the underlying design principles and considerations to fuel future technological development.
Collapse
Affiliation(s)
- Lan Luan
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, USA;
- Rice Neuroengineering Initiative, Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Rongkang Yin
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, USA;
- Rice Neuroengineering Initiative, Rice University, Houston, Texas, USA
| | - Hanlin Zhu
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, USA;
- Rice Neuroengineering Initiative, Rice University, Houston, Texas, USA
| | - Chong Xie
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, USA;
- Rice Neuroengineering Initiative, Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| |
Collapse
|
17
|
Dong T, Chen L, Patel PR, Richie JM, Chestek CA, Shih AJ. Automated assembly of high-density carbon fiber electrode arrays for single unit electrophysiological recordings. J Neural Eng 2023; 20:036012. [PMID: 37141883 DOI: 10.1088/1741-2552/acd279] [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: 03/23/2023] [Accepted: 05/04/2023] [Indexed: 05/06/2023]
Abstract
Objective.Carbon fiber (CF) is good for chronic neural recording due to the small diameter (7µm), high Young's modulus, and low electrical resistance, but most high-density carbon fiber (HDCF) arrays are manually assembled with labor-intensive procedures and limited by the accuracy and repeatability of the operator handling. A machine to automate the assembly is desired.Approach.The HDCF array assembly machine contains: (1) a roller-based CF extruder, (2) a motion system with three linear and one rotary stages, (3) an imaging system with two digital microscope cameras, and (4) a laser cutter. The roller-based extruder automatically feeds single CF as raw material. The motion system aligns the CF with the array backend then places it. The imaging system observes the relative position between the CF and the backend. The laser cutter cuts off the CF. Two image processing algorithms are implemented to align the CF with the support shanks and circuit connection pads.Main results.The machine was capable of precisely handling 6.8μm carbon fiber electrodes (CFEs). Each electrode was placed into a 12μm wide trenches in a silicon support shank. Two HDCF arrays with 16 CFEs populated on 3 mm shanks (with 80μm pitch) were fully assembled. Impedance measurements were found to be in good agreement with manual assembled arrays. One HDCF array was implanted in the motor cortex in an anesthetized rat and was able to detect single unit activity.Significance.This machine can eliminate the manual labor-intensive handling, alignment and placement of single CF during assembly, providing a proof-of-concepts towards fully automated HDCF array assembly and batch production.
Collapse
Affiliation(s)
- Tianshu Dong
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Lei Chen
- Department of Mechanical Engineering, University of Massachusetts Lowell, Lowell, MA, United States of America
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Julianna M Richie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, United States of America
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States of America
| | - Albert J Shih
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
18
|
Shen K, Chen O, Edmunds JL, Piech DK, Maharbiz MM. Translational opportunities and challenges of invasive electrodes for neural interfaces. Nat Biomed Eng 2023; 7:424-442. [PMID: 37081142 DOI: 10.1038/s41551-023-01021-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 02/15/2023] [Indexed: 04/22/2023]
Abstract
Invasive brain-machine interfaces can restore motor, sensory and cognitive functions. However, their clinical adoption has been hindered by the surgical risk of implantation and by suboptimal long-term reliability. In this Review, we highlight the opportunities and challenges of invasive technology for clinically relevant electrophysiology. Specifically, we discuss the characteristics of neural probes that are most likely to facilitate the clinical translation of invasive neural interfaces, describe the neural signals that can be acquired or produced by intracranial electrodes, the abiotic and biotic factors that contribute to their failure, and emerging neural-interface architectures.
Collapse
Affiliation(s)
- Konlin Shen
- University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.
| | - Oliver Chen
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, USA
| | - Jordan L Edmunds
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, USA
| | - David K Piech
- University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Berkeley, CA, USA
| | - Michel M Maharbiz
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
19
|
Jeakle EN, Abbott JR, Usoro JO, Wu Y, Haghighi P, Radhakrishna R, Sturgill BS, Nakajima S, Thai TTD, Pancrazio JJ, Cogan SF, Hernandez-Reynoso AG. Chronic Stability of Local Field Potentials Using Amorphous Silicon Carbide Microelectrode Arrays Implanted in the Rat Motor Cortex. MICROMACHINES 2023; 14:680. [PMID: 36985087 PMCID: PMC10054633 DOI: 10.3390/mi14030680] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 06/18/2023]
Abstract
Implantable microelectrode arrays (MEAs) enable the recording of electrical activity of cortical neurons, allowing the development of brain-machine interfaces. However, MEAs show reduced recording capabilities under chronic conditions, prompting the development of novel MEAs that can improve long-term performance. Conventional planar, silicon-based devices and ultra-thin amorphous silicon carbide (a-SiC) MEAs were implanted in the motor cortex of female Sprague-Dawley rats, and weekly anesthetized recordings were made for 16 weeks after implantation. The spectral density and bandpower between 1 and 500 Hz of recordings were compared over the implantation period for both device types. Initially, the bandpower of the a-SiC devices and standard MEAs was comparable. However, the standard MEAs showed a consistent decline in both bandpower and power spectral density throughout the 16 weeks post-implantation, whereas the a-SiC MEAs showed substantially more stable performance. These differences in bandpower and spectral density between standard and a-SiC MEAs were statistically significant from week 6 post-implantation until the end of the study at 16 weeks. These results support the use of ultra-thin a-SiC MEAs to develop chronic, reliable brain-machine interfaces.
Collapse
Affiliation(s)
- Eleanor N. Jeakle
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Justin R. Abbott
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Joshua O. Usoro
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Yupeng Wu
- Department of Materials Science and Engineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Pegah Haghighi
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Rahul Radhakrishna
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Brandon S. Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Shido Nakajima
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Teresa T. D. Thai
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Joseph J. Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Stuart F. Cogan
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| | - Ana G. Hernandez-Reynoso
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080-3021, USA
| |
Collapse
|
20
|
Letner JG, Patel PR, Hsieh JC, Smith Flores IM, della Valle E, Walker LA, Weiland JD, Chestek CA, Cai D. Post-explant profiling of subcellular-scale carbon fiber intracortical electrodes and surrounding neurons enables modeling of recorded electrophysiology. J Neural Eng 2023; 20:026019. [PMID: 36848679 PMCID: PMC10022369 DOI: 10.1088/1741-2552/acbf78] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/12/2023] [Accepted: 02/27/2023] [Indexed: 03/01/2023]
Abstract
Objective.Characterizing the relationship between neuron spiking and the signals that electrodes record is vital to defining the neural circuits driving brain function and informing clinical brain-machine interface design. However, high electrode biocompatibility and precisely localizing neurons around the electrodes are critical to defining this relationship.Approach.Here, we demonstrate consistent localization of the recording site tips of subcellular-scale (6.8µm diameter) carbon fiber electrodes and the positions of surrounding neurons. We implanted male rats with carbon fiber electrode arrays for 6 or 12+ weeks targeting layer V motor cortex. After explanting the arrays, we immunostained the implant site and localized putative recording site tips with subcellular-cellular resolution. We then 3D segmented neuron somata within a 50µm radius from implanted tips to measure neuron positions and health and compare to healthy cortex with symmetric stereotaxic coordinates.Main results.Immunostaining of astrocyte, microglia, and neuron markers confirmed that overall tissue health was indicative of high biocompatibility near the tips. While neurons near implanted carbon fibers were stretched, their number and distribution were similar to hypothetical fibers placed in healthy contralateral brain. Such similar neuron distributions suggest that these minimally invasive electrodes demonstrate the potential to sample naturalistic neural populations. This motivated the prediction of spikes produced by nearby neurons using a simple point source model fit using recorded electrophysiology and the mean positions of the nearest neurons observed in histology. Comparing spike amplitudes suggests that the radius at which single units can be distinguished from others is near the fourth closest neuron (30.7 ± 4.6µm,X-± S) in layer V motor cortex.Significance.Collectively, these data and simulations provide the first direct evidence that neuron placement in the immediate vicinity of the recording site influences how many spike clusters can be reliably identified by spike sorting.
Collapse
Affiliation(s)
- Joseph G Letner
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Jung-Chien Hsieh
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Israel M Smith Flores
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Elena della Valle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Logan A Walker
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Computational Medicine and Bioinformatics, Michigan Medicine, Ann Arbor, MI 48109, United States of America
| | - James D Weiland
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, United States of America
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States of America
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, United States of America
- Robotics Department, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Dawen Cai
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, United States of America
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, United States of America
| |
Collapse
|
21
|
Composite Fibrin and Carbon Microfibre Implant to Modulate Postraumatic Inflammation after Spinal Cord Injury. Cells 2023; 12:cells12060839. [PMID: 36980180 PMCID: PMC10047285 DOI: 10.3390/cells12060839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Poor functional recovery after spinal cord injury (SCI) drives the development of novel strategies to manage this devastating condition. We recently showed promising immunomodulatory and pro-regenerative actions of bio-functionalized carbon microfibres (MFs) implanted in a rodent model of SCI. In order to maximize tissue repair while easing MF implantation, we produced a composite implant based on the embedding of several MFs within a fibrin hydrogel. We used intravital imaging of fluorescent reporter mice at the early stages and spinal sections of the same animals 3 months later to characterize the neuroinflammatory response to the implant and its impact on axonal regeneration. Whereas fibrin alone was inert in the first week, its enzymatic degradation drove the chronic activation of microglial cells and axonal degeneration within 3 months. However, the presence of MFs inside the fibrin hydrogel slowed down fibrin degradation and boosted the early recruitment of immune cells. Noteworthy, there was an enhanced contribution of monocyte-derived dendritic cells (moDCs), preceding a faster transition toward an anti-inflammatory environment with increased axonal regeneration over 3 months. The inclusion of MF here ensured the long-term biocompatibility of fibrin hydrogels, which would otherwise preclude successful spinal cord regeneration.
Collapse
|
22
|
Mintz Hemed N, Melosh NA. An integrated perspective for the diagnosis and therapy of neurodevelopmental disorders - From an engineering point of view. Adv Drug Deliv Rev 2023; 194:114723. [PMID: 36746077 DOI: 10.1016/j.addr.2023.114723] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/14/2022] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
Neurodevelopmental disorders (NDDs) are complex conditions with largely unknown pathophysiology. While many NDD symptoms are familiar, the cause of these disorders remains unclear and may involve a combination of genetic, biological, psychosocial, and environmental risk factors. Current diagnosis relies heavily on behaviorally defined criteria, which may be biased by the clinical team's professional and cultural expectations, thus a push for new biological-based biomarkers for NDDs diagnosis is underway. Emerging new research technologies offer an unprecedented view into the electrical, chemical, and physiological activity in the brain and with further development in humans may provide clinically relevant diagnoses. These could also be extended to new treatment options, which can start to address the underlying physiological issues. When combined with current speech, language, occupational therapy, and pharmacological treatment these could greatly improve patient outcomes. The current review will discuss the latest technologies that are being used or may be used for NDDs diagnosis and treatment. The aim is to provide an inspiring and forward-looking view for future research in the field.
Collapse
Affiliation(s)
- Nofar Mintz Hemed
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA.
| | - Nicholas A Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
23
|
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
|
24
|
Shao Z, Chang Y, Venton BJ. Carbon microelectrodes with customized shapes for neurotransmitter detection: A review. Anal Chim Acta 2022; 1223:340165. [PMID: 35998998 PMCID: PMC9867599 DOI: 10.1016/j.aca.2022.340165] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 01/26/2023]
Abstract
Carbon is a popular electrode material for neurotransmitter detection due to its good electrochemical properties, high biocompatibility, and inert chemistry. Traditional carbon electrodes, such as carbon fibers, have smooth surfaces and fixed shapes. However, newer studies customize the shape and nanostructure the surface to enhance electrochemistry for different applications. In this review, we show how changing the structure of carbon electrodes with methods such as chemical vapor deposition (CVD), wet-etching, direct laser writing (DLW), and 3D printing leads to different electrochemical properties. The customized shapes include nanotips, complex 3D structures, porous structures, arrays, and flexible sensors with patterns. Nanostructuring enhances sensitivity and selectivity, depending on the carbon nanomaterial used. Carbon nanoparticle modifications enhance electron transfer kinetics and prevent fouling for neurochemicals that are easily polymerized. Porous electrodes trap analyte momentarily on the scale of an electrochemistry experiment, leading to thin layer electrochemical behavior that enhances secondary peaks from chemical reactions. Similar thin layer cell behavior is observed at cavity carbon nanopipette electrodes. Nanotip electrodes facilitate implantation closer to the synapse with reduced tissue damage. Carbon electrode arrays are used to measure from multiple neurotransmitter release sites simultaneously. Custom-shaped carbon electrodes are enabling new applications in neuroscience, such as distinguishing different catecholamines by secondary peaks, detection of vesicular release in single cells, and multi-region measurements in vivo.
Collapse
Affiliation(s)
- Zijun Shao
- Dept. of Chemistry, University of Virginia, Charlottesville, VA, 22904-4319, USA
| | - Yuanyu Chang
- Dept. of Chemistry, University of Virginia, Charlottesville, VA, 22904-4319, USA
| | - B Jill Venton
- Dept. of Chemistry, University of Virginia, Charlottesville, VA, 22904-4319, USA.
| |
Collapse
|
25
|
Robbins EM, Castagnola E, Cui XT. Accurate and stable chronic in vivo voltammetry enabled by a replaceable subcutaneous reference electrode. iScience 2022; 25:104845. [PMID: 35996579 PMCID: PMC9391596 DOI: 10.1016/j.isci.2022.104845] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 06/16/2022] [Accepted: 07/22/2022] [Indexed: 01/12/2023] Open
Abstract
In vivo sensing of neurotransmitters has provided valuable insight into both healthy and diseased brain. However, chronically implanted Ag/AgCl reference electrodes suffer from degradationgradation, resulting in errors in the potential at the working electrode. Here, we report a simple, effective way to protect in vivo sensing measurements from reference polarization with a replaceable subcutaneously implanted reference. We compared a brain-implanted reference and a subcutaneous reference and observed no difference in impedance or dopamine redox peak separation in an acute preparation. Chronically, peak background potential and dopamine oxidation potential shifts were eliminated for three weeks. Scanning electron microscopy shows changes in surface morphology and composition of chronically implanted Ag/AgCl electrodes, and postmortem histology reveals extensive cell death and gliosis in the surrounding tissue. As accurate reference potentials are critical to in vivo electrochemistry applications, this simple technique can improve a wide and diverse assortment of in vivo preparations.
Collapse
Affiliation(s)
- Elaine Marie Robbins
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Elisa Castagnola
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
- Corresponding author
| |
Collapse
|
26
|
Whitsitt QA, Koo B, Celik ME, Evans BM, Weiland JD, Purcell EK. Spatial Transcriptomics as a Novel Approach to Redefine Electrical Stimulation Safety. Front Neurosci 2022; 16:937923. [PMID: 35928007 PMCID: PMC9344921 DOI: 10.3389/fnins.2022.937923] [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: 05/06/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Current standards for safe delivery of electrical stimulation to the central nervous system are based on foundational studies which examined post-mortem tissue for histological signs of damage. This set of observations and the subsequently proposed limits to safe stimulation, termed the "Shannon limits," allow for a simple calculation (using charge per phase and charge density) to determine the intensity of electrical stimulation that can be delivered safely to brain tissue. In the three decades since the Shannon limits were reported, advances in molecular biology have allowed for more nuanced and detailed approaches to be used to expand current understanding of the physiological effects of stimulation. Here, we demonstrate the use of spatial transcriptomics (ST) in an exploratory investigation to assess the biological response to electrical stimulation in the brain. Electrical stimulation was delivered to the rat visual cortex with either acute or chronic electrode implantation procedures. To explore the influence of device type and stimulation parameters, we used carbon fiber ultramicroelectrode arrays (7 μm diameter) and microwire electrode arrays (50 μm diameter) delivering charge and charge density levels selected above and below reported tissue damage thresholds (range: 2-20 nC, 0.1-1 mC/cm2). Spatial transcriptomics was performed using Visium Spatial Gene Expression Slides (10x Genomics, Pleasanton, CA, United States), which enabled simultaneous immunohistochemistry and ST to directly compare traditional histological metrics to transcriptional profiles within each tissue sample. Our data give a first look at unique spatial patterns of gene expression that are related to cellular processes including inflammation, cell cycle progression, and neuronal plasticity. At the acute timepoint, an increase in inflammatory and plasticity related genes was observed surrounding a stimulating electrode compared to a craniotomy control. At the chronic timepoint, an increase in inflammatory and cell cycle progression related genes was observed both in the stimulating vs. non-stimulating microwire electrode comparison and in the stimulating microwire vs. carbon fiber comparison. Using the spatial aspect of this method as well as the within-sample link to traditional metrics of tissue damage, we demonstrate how these data may be analyzed and used to generate new hypotheses and inform safety standards for stimulation in cortex.
Collapse
Affiliation(s)
- Quentin A. Whitsitt
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Beomseo Koo
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Mahmut Emin Celik
- Department of Electrical and Electronics Engineering, Gazi University, Ankara, Turkey
| | - Blake M. Evans
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - James D. Weiland
- Department of Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Erin K. Purcell
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
27
|
Bhaskara S, Sakorikar T, Chatterjee S, Shabari Girishan K, Pandya HJ. Recent advancements in Micro-engineered devices for surface and deep brain animal studies: A review. SENSING AND BIO-SENSING RESEARCH 2022. [DOI: 10.1016/j.sbsr.2022.100483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
28
|
Yan D, Jiman AA, Bottorff EC, Patel PR, Meli D, Welle EJ, Ratze DC, Havton LA, Chestek CA, Kemp SWP, Bruns TM, Yoon E, Seymour JP. Ultraflexible and Stretchable Intrafascicular Peripheral Nerve Recording Device with Axon-Dimension, Cuff-Less Microneedle Electrode Array. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200311. [PMID: 35491522 PMCID: PMC9167574 DOI: 10.1002/smll.202200311] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/08/2022] [Indexed: 05/03/2023]
Abstract
Peripheral nerve mapping tools with higher spatial resolution are needed to advance systems neuroscience, and potentially provide a closed-loop biomarker in neuromodulation applications. Two critical challenges of microscale neural interfaces are 1) how to apply them to small peripheral nerves, and 2) how to minimize chronic reactivity. A flexible microneedle nerve array (MINA) is developed, which is the first high-density penetrating electrode array made with axon-sized silicon microneedles embedded in low-modulus thin silicone. The design, fabrication, acute recording, and chronic reactivity to an implanted MINA, are presented. Distinctive units are identified in the rat peroneal nerve. The authors also demonstrate a long-term, cuff-free, and suture-free fixation manner using rose bengal as a light-activated adhesive for two time-points. The tissue response is investigated at 1-week and 6-week time-points, including two sham groups and two MINA-implanted groups. These conditions are quantified in the left vagus nerve of rats using histomorphometry. Micro computed tomography (micro-CT) is added to visualize and quantify tissue encapsulation around the implant. MINA demonstrates a reduction in encapsulation thickness over previously quantified interfascicular methods. Future challenges include techniques for precise insertion of the microneedle electrodes and demonstrating long-term recording.
Collapse
Affiliation(s)
- Dongxiao Yan
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ahmad A Jiman
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Elizabeth C Bottorff
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dilara Meli
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Elissa J Welle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David C Ratze
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Leif A Havton
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- James J Peters Veterans Affairs Medical Center, Bronx, NY, 10468, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen W P Kemp
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Section of Plastic Surgery, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Tim M Bruns
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Center for Nanomedicine, Institute for Basic Science (IBS) and Graduate Program of Nano Biomedical Engineering (Nano BME), Advanced Science Institute, Yonsei University, Seoul, South Korea
| | - John P Seymour
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Neurosurgery, UTHealth, Houston, TX, 77030, USA
- Department of Electrical and Computer Engineering, Rice University, Houston, TX, 77030, USA
| |
Collapse
|
29
|
Koo B, Weiland JD. Progressive Retinal Degeneration Increases Cortical Response Latency of Light Stimulation but Not of Electric Stimulation. Transl Vis Sci Technol 2022; 11:19. [PMID: 35446408 PMCID: PMC9034728 DOI: 10.1167/tvst.11.4.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/31/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose The brain is known to change functionally and structurally in response to blindness, but less is known about the effects of restoration of cortical input on brain function. Here, we present a preliminary study to observe alterations in visual and electrical evoked cortical potentials as a function of age in a clinically relevant animal model of retinitis pigmentosa. Methods We recorded brain potentials elicited by light (visual evoked potentials [VEPs]) or corneal electrical stimulation (electrical evoked response [EER]) in retinal degenerate animal model LE-P23H-1. We used a linear mixed model to examine the effects of age on latency and amplitude of VEP and EER age groups P120, P180, and P360. Results VEP N1, P1, and N2 latency and amplitude were analyzed across animal age. For 1 Hz VEP, N1 latency increased significantly with animal age (slope = 0.053 ± 0.020 ms/day, P < 0.01). For 10 Hz VEP, N1, P1, and N2 latency increased significantly with animal age (slope = 0.104 ± 0.011, 0.135 ± 0.011, 0.087 ± 0.023 ms/day, and P < 0.001 for all VEP peaks). Conversely, EER latency did not change with age. Signal amplitude of VEP or EER did not change with age. Conclusions Cortical potentials evoked by electrical stimulation of the retina do not diminish in spite of continued retinal degeneration in P23H rats. Translational Relevance These findings suggest that retinal bioelectronic treatments of retinitis pigmentosa will activate cortex consistently despite variations in outer retinal degeneration. Clinical studies of retinal stimulation should consider varying retinitis pigmentosa genotypes as part of the experimental design.
Collapse
Affiliation(s)
- Beomseo Koo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - James D. Weiland
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Lim J, Lee J, Moon E, Barrow M, Atzeni G, Letner JG, Costello JT, Nason SR, Patel PR, Sun Y, Patil PG, Kim HS, Chestek CA, Phillips J, Blaauw D, Sylvester D, Jang T. A Light-Tolerant Wireless Neural Recording IC for Motor Prediction With Near-Infrared-Based Power and Data Telemetry. IEEE JOURNAL OF SOLID-STATE CIRCUITS 2022; 57:1061-1074. [PMID: 36186085 PMCID: PMC9518712 DOI: 10.1109/jssc.2022.3141688] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Miniaturized and wireless near-infrared (NIR) based neural recorders with optical powering and data telemetry have been introduced as a promising approach for safe long-term monitoring with the smallest physical dimension among state-of-the-art standalone recorders. However, a main challenge for the NIR based neural recording ICs is to maintain robust operation in the presence of light-induced parasitic short circuit current from junction diodes. This is especially true when the signal currents are kept small to reduce power consumption. In this work, we present a light-tolerant and low-power neural recording IC for motor prediction that can fully function in up to 300 μW/mm2 of light exposure. It achieves best-in-class power consumption of 0.57 μW at 38° C with a 4.1 NEF pseudo-resistorless amplifier, an on-chip neural feature extractor, and individual mote level gain control. Applying the 20-channel pre-recorded neural signals of a monkey, the IC predicts finger position and velocity with correlation coefficient up to 0.870 and 0.569, respectively, with individual mote level gain control enabled. In addition, wireless measurement is demonstrated through optical power and data telemetry using a custom PV/LED GaAs chip wire bonded to the proposed IC.
Collapse
Affiliation(s)
- Jongyup Lim
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Jungho Lee
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Eunseong Moon
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Michael Barrow
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Gabriele Atzeni
- Department of Information Technology and Electrical Engineering, ETH Zürich, 8092 Zürich, Switzerland
| | - Joseph G Letner
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Joseph T Costello
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Samuel R Nason
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Yi Sun
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Parag G Patil
- Department of Neurological Surgery, Neurology, Anesthesiology, and Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Hun-Seok Kim
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering and Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Jamie Phillips
- Department of Electrical and Computer Engineering, University of Delaware, Newark, DE 19716 USA
| | - David Blaauw
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Dennis Sylvester
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109 USA
| | - Taekwang Jang
- Department of Information Technology and Electrical Engineering, ETH Zürich, 8092 Zürich, Switzerland
| |
Collapse
|
31
|
Geramifard N, Dousti B, Nguyen CK, Abbott JR, Cogan S, Varner V. Insertion mechanics of amorphous SiC ultra-micro scale neural probes. J Neural Eng 2022; 19. [PMID: 35263724 PMCID: PMC9339220 DOI: 10.1088/1741-2552/ac5bf4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/09/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Trauma induced by the insertion of microelectrodes into cortical neural tissue is a significant problem. Further, micromotion and mechanical mismatch between microelectrode probes and neural tissue is implicated in an adverse foreign body response (FBR). Hence, intracortical ultra-microelectrode probes have been proposed as alternatives that minimize this FBR. However, significant challenges in implanting these flexible probes remain. We investigated the insertion mechanics of amorphous silicon carbide (a-SiC) probes with a view to defining probe geometries that can be inserted into cortex without buckling. APPROACH We determined the critical buckling force of a-SiC probes as a function of probe geometry and then characterized the buckling behavior of these probes by measuring force-displacement responses during insertion into agarose gel and rat cortex. MAIN RESULTS Insertion forces for a range of probe geometries were determined and compared with critical buckling forces to establish geometries that should avoid buckling during implantation into brain. The studies show that slower insertion speeds reduce the maximum insertion force for single-shank probes but increase cortical dimpling during insertion of multi-shank probes. SIGNIFICANCE Our results provide a guide for selecting probe geometries and insertion speeds that allow unaided implantation of probes into rat cortex. The design approach is applicable to other animal models where insertion of intracortical probes to a depth of 2 mm is required.
Collapse
Affiliation(s)
- Negar Geramifard
- Department of Bioeengineering, The University of Texas at Dallas Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Rd., BSB 13.601, Richardson, Texas, 75080-3021, UNITED STATES
| | - Behnoush Dousti
- The University of Texas at Dallas, Department of Bioengineering, Richardson, Texas, 75080-3021, UNITED STATES
| | - Christopher Khanhtuan Nguyen
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, Texas, 75080-3021, UNITED STATES
| | - Justin Robert Abbott
- Department of Bioengineering, The University of Texas at Dallas, 800 West Campbell Rd, Richardson, Texas, 75080, UNITED STATES
| | - Stuart Cogan
- Department of Bioengineering, The University of Texas at Dallas Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Road, Richardson, Texas, 75080-3021, UNITED STATES
| | - Victor Varner
- Department of Bioengineering, The University of Texas at Dallas Erik Jonsson School of Engineering and Computer Science, 800 West Campbell Rd, Richardson, Texas, 75080, UNITED STATES
| |
Collapse
|
32
|
Astro V, Alowaysi M, Fiacco E, Saera-Vila A, Cardona-Londoño KJ, Aiese Cigliano R, Adamo A. Pseudoautosomal Region 1 Overdosage Affects the Global Transcriptome in iPSCs From Patients With Klinefelter Syndrome and High-Grade X Chromosome Aneuploidies. Front Cell Dev Biol 2022; 9:801597. [PMID: 35186953 PMCID: PMC8850648 DOI: 10.3389/fcell.2021.801597] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/28/2021] [Indexed: 01/19/2023] Open
Abstract
Klinefelter syndrome (KS) is the most prevalent aneuploidy in males and is characterized by a 47,XXY karyotype. Less frequently, higher grade sex chromosome aneuploidies (HGAs) can also occur. Here, using a paradigmatic cohort of KS and HGA induced pluripotent stem cells (iPSCs) carrying 49,XXXXY, 48,XXXY, and 47,XXY karyotypes, we identified the genes within the pseudoautosomal region 1 (PAR1) as the most susceptible to dosage-dependent transcriptional dysregulation and therefore potentially responsible for the progressively worsening phenotype in higher grade X aneuploidies. By contrast, the biallelically expressed non-PAR escape genes displayed high interclonal and interpatient variability in iPSCs and differentiated derivatives, suggesting that these genes could be associated with variable KS traits. By interrogating KS and HGA iPSCs at the single-cell resolution we showed that PAR1 and non-PAR escape genes are not only resilient to the X-inactive specific transcript (XIST)-mediated inactivation but also that their transcriptional regulation is disjointed from the absolute XIST expression level. Finally, we explored the transcriptional effects of X chromosome overdosage on autosomes and identified the nuclear respiratory factor 1 (NRF1) as a key regulator of the zinc finger protein X-linked (ZFX). Our study provides the first evidence of an X-dosage-sensitive autosomal transcription factor regulating an X-linked gene in low- and high-grade X aneuploidies.
Collapse
Affiliation(s)
- Veronica Astro
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Maryam Alowaysi
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Elisabetta Fiacco
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | | | - Kelly J. Cardona-Londoño
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | | | - Antonio Adamo
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
- *Correspondence: Antonio Adamo,
| |
Collapse
|
33
|
Otte E, Vlachos A, Asplund M. Engineering strategies towards overcoming bleeding and glial scar formation around neural probes. Cell Tissue Res 2022; 387:461-477. [PMID: 35029757 PMCID: PMC8975777 DOI: 10.1007/s00441-021-03567-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022]
Abstract
Neural probes are sophisticated electrophysiological tools used for intra-cortical recording and stimulation. These microelectrode arrays, designed to penetrate and interface the brain from within, contribute at the forefront of basic and clinical neuroscience. However, one of the challenges and currently most significant limitations is their ‘seamless’ long-term integration into the surrounding brain tissue. Following implantation, which is typically accompanied by bleeding, the tissue responds with a scarring process, resulting in a gliotic region closest to the probe. This glial scarring is often associated with neuroinflammation, neurodegeneration, and a leaky blood–brain interface (BBI). The engineering progress on minimizing this reaction in the form of improved materials, microfabrication, and surgical techniques is summarized in this review. As research over the past decade has progressed towards a more detailed understanding of the nature of this biological response, it is time to pose the question: Are penetrating probes completely free from glial scarring at all possible?
Collapse
|
34
|
Fu X, Li G, Niu Y, Xu J, Wang P, Zhou Z, Ye Z, Liu X, Xu Z, Yang Z, Zhang Y, Lei T, Zhang B, Li Q, Cao A, Jiang T, Duan X. Carbon-Based Fiber Materials as Implantable Depth Neural Electrodes. Front Neurosci 2022; 15:771980. [PMID: 35002602 PMCID: PMC8730365 DOI: 10.3389/fnins.2021.771980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/29/2021] [Indexed: 01/02/2023] Open
Abstract
Implantable brain electrophysiology electrodes are valuable tools in both fundamental and applied neuroscience due to their ability to record neural activity with high spatiotemporal resolution from shallow and deep brain regions. Their use has been hindered, however, by the challenges in achieving chronically stable operations. Furthermore, implantable depth neural electrodes can only carry out limited data sampling within predefined anatomical regions, making it challenging to perform large-area brain mapping. Minimizing inflammatory responses and associated gliosis formation, and improving the durability and stability of the electrode insulation layers are critical to achieve long-term stable neural recording and stimulation. Combining electrophysiological measurements with simultaneous whole-brain imaging techniques, such as magnetic resonance imaging (MRI), provides a useful solution to alleviate the challenge in scalability of implantable depth electrodes. In recent years, various carbon-based materials have been used to fabricate flexible neural depth electrodes with reduced inflammatory responses and MRI-compatible electrodes, which allows structural and functional MRI mapping of the whole brain without obstructing any brain regions around the electrodes. Here, we conducted a systematic comparative evaluation on the electrochemical properties, mechanical properties, and MRI compatibility of different kinds of carbon-based fiber materials, including carbon nanotube fibers, graphene fibers, and carbon fibers. We also developed a strategy to improve the stability of the electrode insulation without sacrificing the flexibility of the implantable depth electrodes by sandwiching an inorganic barrier layer inside the polymer insulation film. These studies provide us with important insights into choosing the most suitable materials for next-generation implantable depth electrodes with unique capabilities for applications in both fundamental and translational neuroscience research.
Collapse
Affiliation(s)
- Xuefeng Fu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Gen Li
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Yutao Niu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Advanced Materials Division, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences (CAS), Suzhou, China
| | - Jingcao Xu
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Puxin Wang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zhaoxiao Zhou
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziming Ye
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Xiaojun Liu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Zheng Xu
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China
| | - Ziqian Yang
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yongyi Zhang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Advanced Materials Division, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences (CAS), Suzhou, China
| | - Ting Lei
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Baogui Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences (CAS), Beijing, China
| | - Qingwen Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, China.,Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Advanced Materials Division, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences (CAS), Suzhou, China
| | - Anyuan Cao
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Tianzai Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences (CAS), Beijing, China
| | - Xiaojie Duan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, China.,Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.,National Biomedical Imaging Center, Peking University, Beijing, China
| |
Collapse
|
35
|
Sharafkhani N, Kouzani AZ, Adams SD, Long JM, Lissorgues G, Rousseau L, Orwa JO. Neural tissue-microelectrode interaction: Brain micromotion, electrical impedance, and flexible microelectrode insertion. J Neurosci Methods 2022; 365:109388. [PMID: 34678387 DOI: 10.1016/j.jneumeth.2021.109388] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 10/20/2022]
Abstract
Insertion of a microelectrode into the brain to record/stimulate neurons damages neural tissue and blood vessels and initiates the brain's wound healing response. Due to the large difference between the stiffness of neural tissue and microelectrode, brain micromotion also leads to neural tissue damage and associated local immune response. Over time, following implantation, the brain's response to the tissue damage can result in microelectrode failure. Reducing the microelectrode's cross-sectional dimensions to single-digit microns or using soft materials with elastic modulus close to that of the neural tissue are effective methods to alleviate the neural tissue damage and enhance microelectrode longevity. However, the increase in electrical impedance of the microelectrode caused by reducing the microelectrode contact site's dimensions can decrease the signal-to-noise ratio. Most importantly, the reduced dimensions also lead to a reduction in the critical buckling force, which increases the microelectrode's propensity to buckling during insertion. After discussing brain micromotion, the main source of neural tissue damage, surface modification of the microelectrode contact site is reviewed as a key method for addressing the increase in electrical impedance issue. The review then focuses on recent approaches to aiding insertion of flexible microelectrodes into the brain, including bending stiffness modification, effective length reduction, and application of a magnetic field to pull the electrode. An understanding of the advantages and drawbacks of the developed strategies offers a guide for dealing with the buckling phenomenon during implantation.
Collapse
Affiliation(s)
- Naser Sharafkhani
- School of Engineering, Deakin University, Geelong, VIC 3216, Australia.
| | - Abbas Z Kouzani
- School of Engineering, Deakin University, Geelong, VIC 3216, Australia
| | - Scott D Adams
- School of Engineering, Deakin University, Geelong, VIC 3216, Australia
| | - John M Long
- School of Engineering, Deakin University, Geelong, VIC 3216, Australia
| | | | | | - Julius O Orwa
- School of Engineering, Deakin University, Geelong, VIC 3216, Australia.
| |
Collapse
|
36
|
Huan Y, Gill JP, Fritzinger JB, Patel PR, Richie JM, Valle ED, Weiland JD, Chestek CA, Chiel HJ. Carbon fiber electrodes for intracellular recording and stimulation. J Neural Eng 2021; 18:10.1088/1741-2552/ac3dd7. [PMID: 34826825 PMCID: PMC10729305 DOI: 10.1088/1741-2552/ac3dd7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/26/2021] [Indexed: 01/18/2023]
Abstract
Objective.To understand neural circuit dynamics, it is critical to manipulate and record many individual neurons. Traditional recording methods, such as glass microelectrodes, can only control a small number of neurons. More recently, devices with high electrode density have been developed, but few of them can be used for intracellular recording or stimulation in intact nervous systems. Carbon fiber electrodes (CFEs) are 8µm-diameter electrodes that can be assembled into dense arrays (pitches ⩾ 80µm). They have good signal-to-noise ratios (SNRs) and provide stable extracellular recordings both acutely and chronically in neural tissuein vivo(e.g. rat motor cortex). The small fiber size suggests that arrays could be used for intracellular stimulation.Approach.We tested CFEs for intracellular stimulation using the large identified and electrically compact neurons of the marine molluskAplysia californica. Neuron cell bodies inAplysiarange from 30µm to over 250µm. We compared the efficacy of CFEs to glass microelectrodes by impaling the same neuron's cell body with both electrodes and connecting them to a DC coupled amplifier.Main results.We observed that intracellular waveforms were essentially identical, but the amplitude and SNR in the CFE were lower than in the glass microelectrode. CFE arrays could record from 3 to 8 neurons simultaneously for many hours, and many of these recordings were intracellular, as shown by simultaneous glass microelectrode recordings. CFEs coated with platinum-iridium could stimulate and had stable impedances over many hours. CFEs not within neurons could record local extracellular activity. Despite the lower SNR, the CFEs could record synaptic potentials. CFEs were less sensitive to mechanical perturbations than glass microelectrodes.Significance.The ability to do stable multi-channel recording while stimulating and recording intracellularly make CFEs a powerful new technology for studying neural circuit dynamics.
Collapse
Affiliation(s)
- Yu Huan
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Jeffrey P Gill
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States of America
| | - Johanna B Fritzinger
- Department of Neurosciences, University of Rochester, Rochester, NY, United States of America
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Julianna M Richie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Elena Della Valle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - James D Weiland
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States of America
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, United States of America
- Neurosciences Program, University of Michigan, Ann Arbor, MI, United States of America
- Robotics Program, University of Michigan, Ann Arbor, MI, United States of America
| | - Hillel J Chiel
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|
37
|
Lucio Boschen S, Trevathan J, Hara SA, Asp A, Lujan JL. Defining a Path Toward the Use of Fast-Scan Cyclic Voltammetry in Human Studies. Front Neurosci 2021; 15:728092. [PMID: 34867151 PMCID: PMC8633532 DOI: 10.3389/fnins.2021.728092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Fast Scan Cyclic Voltammetry (FSCV) has been used for decades as a neurochemical tool for in vivo detection of phasic changes in electroactive neurotransmitters in animal models. Recently, multiple research groups have initiated human neurochemical studies using FSCV or demonstrated interest in bringing FSCV into clinical use. However, there remain technical challenges that limit clinical implementation of FSCV by creating barriers to appropriate scientific rigor and patient safety. In order to progress with clinical FSCV, these limitations must be first addressed through (1) appropriate pre-clinical studies to ensure accurate measurement of neurotransmitters and (2) the application of a risk management framework to assess patient safety. The intent of this work is to bring awareness of the current issues associated with FSCV to the scientific, engineering, and clinical communities and encourage them to seek solutions or alternatives that ensure data accuracy, rigor and reproducibility, and patient safety.
Collapse
Affiliation(s)
- Suelen Lucio Boschen
- Applied Computational Neurophysiology and Neuromodulation Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - James Trevathan
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Seth A Hara
- Division of Engineering, Mayo Clinic, Rochester, MN, United States
| | - Anders Asp
- Applied Computational Neurophysiology and Neuromodulation Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - J Luis Lujan
- Applied Computational Neurophysiology and Neuromodulation Laboratory, Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
38
|
della Valle E, Koo B, Patel PR, Whitsitt Q, Purcell EK, Chestek CA, Weiland JD. Electrodeposited Platinum Iridium Enables Microstimulation With Carbon Fiber Electrodes. FRONTIERS IN NANOTECHNOLOGY 2021. [DOI: 10.3389/fnano.2021.782883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ultrasmall microelectrode arrays have the potential to improve the spatial resolution of microstimulation. Carbon fiber (CF) microelectrodes with cross-sections of less than 8 μm have been demonstrated to penetrate cortical tissue and evoke minimal scarring in chronic implant tests. In this study, we investigate the stability and performance of neural stimulation electrodes comprised of electrodeposited platinum-iridium (PtIr) on carbon fibers. We conducted pulse testing and characterized charge injection in vitro and recorded voltage transients in vitro and in vivo. Standard electrochemical measurements (impedance spectroscopy and cyclic voltammetry) and visual inspection (scanning electron microscopy) were used to assess changes due to pulsing. Similar to other studies, the application of pulses caused a decrease in impedance and a reduction in voltage transients, but analysis of the impedance data suggests that these changes are due to surface modification and not permanent changes to the electrode. Comparison of scanning electron microscope images before and after pulse testing confirmed electrode stability.
Collapse
|
39
|
Hyakumura T, Aregueta-Robles U, Duan W, Villalobos J, Adams WK, Poole-Warren L, Fallon JB. Improving Deep Brain Stimulation Electrode Performance in vivo Through Use of Conductive Hydrogel Coatings. Front Neurosci 2021; 15:761525. [PMID: 34803592 PMCID: PMC8602793 DOI: 10.3389/fnins.2021.761525] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Active implantable neurological devices like deep brain stimulators have been used over the past few decades to treat movement disorders such as those in people with Parkinson’s disease and more recently, in psychiatric conditions like obsessive compulsive disorder. Electrode-tissue interfaces that support safe and effective targeting of specific brain regions are critical to success of these devices. Development of directional electrodes that activate smaller volumes of brain tissue requires electrodes to operate safely with higher charge densities. Coatings such as conductive hydrogels (CHs) provide lower impedances and higher charge injection limits (CILs) than standard platinum electrodes and support safer application of smaller electrode sizes. The aim of this study was to examine the chronic in vivo performance of a new low swelling CH coating that supports higher safe charge densities than traditional platinum electrodes. A range of hydrogel blends were engineered and their swelling and electrical performance compared. Electrochemical performance and stability of high and low swelling formulations were compared during insertion into a model brain in vitro and the formulation with lower swelling characteristics was chosen for the in vivo study. CH-coated or uncoated Pt electrode arrays were implanted into the brains of 14 rats, and their electrochemical performance was tested weekly for 8 weeks. Tissue response and neural survival was assessed histologically following electrode array removal. CH coating resulted in significantly lower voltage transient impedance, higher CIL, lower electrochemical impedance spectroscopy, and higher charge storage capacity compared to uncoated Pt electrodes in vivo, and this advantage was maintained over the 8-week implantation. There was no significant difference in evoked potential thresholds, signal-to-noise ratio, tissue response or neural survival between CH-coated and uncoated Pt groups. The significant electrochemical advantage and stability of CH coating in the brain supports the suitability of this coating technology for future development of smaller, higher fidelity electrode arrays with higher charge density requirement.
Collapse
Affiliation(s)
- Tomoko Hyakumura
- The Bionics Institute of Australia, East Melbourne, VIC, Australia.,Department of Medical Bionics, The University of Melbourne, Parkville, VIC, Australia
| | - Ulises Aregueta-Robles
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - Wenlu Duan
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - Joel Villalobos
- The Bionics Institute of Australia, East Melbourne, VIC, Australia.,Department of Medical Bionics, The University of Melbourne, Parkville, VIC, Australia
| | - Wendy K Adams
- The Bionics Institute of Australia, East Melbourne, VIC, Australia
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW, Australia.,Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, NSW, Australia
| | - James B Fallon
- The Bionics Institute of Australia, East Melbourne, VIC, Australia.,Department of Medical Bionics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
40
|
Richie JM, Patel PR, Welle EJ, Dong T, Chen L, Shih AJ, Chestek CA. Open-source Toolkit: Benchtop Carbon Fiber Microelectrode Array for Nerve Recording. J Vis Exp 2021:10.3791/63099. [PMID: 34779441 PMCID: PMC10771276 DOI: 10.3791/63099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Conventional peripheral nerve probes are primarily fabricated in a cleanroom, requiring the use of multiple expensive and highly specialized tools. This paper presents a cleanroom "light" fabrication process of carbon fiber neural electrode arrays that can be learned quickly by an inexperienced cleanroom user. This carbon fiber electrode array fabrication process requires just one cleanroom tool, a Parylene C deposition machine, that can be learned quickly or outsourced to a commercial processing facility at marginal cost. This fabrication process also includes hand-populating printed circuit boards, insulation, and tip optimization. The three different tip optimizations explored here (Nd:YAG laser, blowtorch, and UV laser) result in a range of tip geometries and 1 kHz impedances, with blowtorched fibers resulting in the lowest impedance. While previous experiments have proven laser and blowtorch electrode efficacy, this paper also shows that UV laser-cut fibers can record neural signals in vivo. Existing carbon fiber arrays either do not have individuated electrodes in favor of bundles or require cleanroom fabricated guides for population and insulation. The proposed arrays use only tools that can be used at a benchtop for fiber population. This carbon fiber electrode array fabrication process allows for quick customization of bulk array fabrication at a reduced price compared to commercially available probes.
Collapse
Affiliation(s)
- Julianna M Richie
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Paras R Patel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Elissa J Welle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor
| | - Tianshu Dong
- Department of Mechanical Engineering, University of Michigan, Ann Arbor
| | - Lei Chen
- Department of Mechanical Engineering, University of Massachusetts Lowell
| | - Albert J Shih
- Department of Mechanical Engineering, University of Michigan, Ann Arbor
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor; Neuroscience Graduate Program, University of Michigan, Ann Arbor; Robotics Graduate Program, University of Michigan, Ann Arbor;
| |
Collapse
|
41
|
Zou L, Tian H, Guan S, Ding J, Gao L, Wang J, Fang Y. Self-assembled multifunctional neural probes for precise integration of optogenetics and electrophysiology. Nat Commun 2021; 12:5871. [PMID: 34620851 PMCID: PMC8497603 DOI: 10.1038/s41467-021-26168-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/17/2021] [Indexed: 11/12/2022] Open
Abstract
Optogenetics combined with electrical recording has emerged as a powerful tool for investigating causal relationships between neural circuit activity and function. However, the size of optogenetically manipulated tissue is typically 1-2 orders of magnitude larger than that can be electrically recorded, rendering difficulty for assigning functional roles of recorded neurons. Here we report a viral vector-delivery optrode (VVD-optrode) system for precise integration of optogenetics and electrophysiology in the brain. Our system consists of flexible microelectrode filaments and fiber optics that are simultaneously self-assembled in a nanoliter-scale, viral vector-delivery polymer carrier. The highly localized delivery and neuronal expression of opsin genes at microelectrode-tissue interfaces ensure high spatial congruence between optogenetically manipulated and electrically recorded neuronal populations. We demonstrate that this multifunctional system is capable of optogenetic manipulation and electrical recording of spatially defined neuronal populations for three months, allowing precise and long-term studies of neural circuit functions. The authors present a viral vector-delivery optrode system to integrate optogenetics and electrophysiology. The flexible microelectrode filaments and fiber optics self-assemble in a nanoliter-scale, viral vector-delivery polymer carrier for localized delivery and expression of opsin genes at microelectrode-tissue interfaces.
Collapse
Affiliation(s)
- Liang Zou
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huihui Tian
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Shouliang Guan
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianfei Ding
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Lei Gao
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinfen Wang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Ying Fang
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
42
|
Wunderlich H, Kozielski KL. Next generation material interfaces for neural engineering. Curr Opin Biotechnol 2021; 72:29-38. [PMID: 34601203 DOI: 10.1016/j.copbio.2021.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/06/2021] [Accepted: 09/07/2021] [Indexed: 11/28/2022]
Abstract
Neural implant technology is rapidly progressing, and gaining broad interest in research fields such as electrical engineering, materials science, neurobiology, and data science. As the potential applications of neural devices have increased, new technologies to make neural intervention longer-lasting and less invasive have brought attention to neural interface engineering. This review will focus on recent developments in materials for neural implants, highlighting new technologies in the fields of soft electrodes, mechanical and chemical engineering of interface coatings, and remotely powered devices. In this context, novel implantation strategies, manufacturing methods, and combinatorial device functions will also be discussed.
Collapse
Affiliation(s)
- Hannah Wunderlich
- Department of Bioengineering and Biosystems, Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kristen L Kozielski
- Department of Electrical and Computer Engineering, Technical University of Munich, Munich, Germany.
| |
Collapse
|
43
|
Valle ED, Welle EJ, Chestek CA, Weiland JD. Compositional and morphological properties of platinum-iridium electrodeposited on carbon fiber microelectrodes. J Neural Eng 2021; 18:10.1088/1741-2552/ac20bb. [PMID: 34428753 PMCID: PMC10756281 DOI: 10.1088/1741-2552/ac20bb] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 08/24/2021] [Indexed: 11/12/2022]
Abstract
Objective. Neural interfaces based on carbon fiber (CF) electrodes have demonstrated key positive attributes such as minimal foreign body response and mechanical strength to self-insert in brain tissue. However, carbon does not form a low impedance electrode interface with neural tissue. Electrodeposited platinum iridium (PtIr) has been used to improve electrode interface properties for metallic bioelectrodes.Approach. In this study, a PtIr electrodeposition process has been performed on CF microelectrode arrays to improve the interfacial properties of these arrays. We study the film morphology and composition as well as electrode durability and impedance.Results. A PtIr coating with a composition of 70% Pt, 30% Ir and a thickness of ∼400 nm was observed. Pt and Ir were evenly distributed within the film. Impedance was decreased by 89% @ 1 kHz. Accelerated soak testing in a heated (T= 50∘C) saline solution showed impedance increase (@ 1 kHz) of ∼12% after 36 days (89 equivalent) of soaking.
Collapse
Affiliation(s)
- Elena della Valle
- Biomedical Engineering Department, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Elissa J Welle
- Biomedical Engineering Department, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Cynthia A Chestek
- Biomedical Engineering Department, University of Michigan, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - James D Weiland
- Biomedical Engineering Department, University of Michigan, Ann Arbor, MI, United States of America
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
44
|
Devi M, Vomero M, Fuhrer E, Castagnola E, Gueli C, Nimbalkar S, Hirabayashi M, Kassegne S, Stieglitz T, Sharma S. Carbon-based neural electrodes: promises and challenges. J Neural Eng 2021; 18. [PMID: 34404037 DOI: 10.1088/1741-2552/ac1e45] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/17/2021] [Indexed: 01/01/2023]
Abstract
Neural electrodes are primary functional elements of neuroelectronic devices designed to record neural activity based on electrochemical signals. These electrodes may also be utilized for electrically stimulating the neural cells, such that their response can be simultaneously recorded. In addition to being medically safe, the electrode material should be electrically conductive and electrochemically stable under harsh biological environments. Mechanical flexibility and conformability, resistance to crack formation and compatibility with common microfabrication techniques are equally desirable properties. Traditionally, (noble) metals have been the preferred for neural electrode applications due to their proven biosafety and a relatively high electrical conductivity. Carbon is a recent addition to this list, which is far superior in terms of its electrochemical stability and corrosion resistance. Carbon has also enabled 3D electrode fabrication as opposed to the thin-film based 2D structures. One of carbon's peculiar aspects is its availability in a wide range of allotropes with specialized properties that render it highly versatile. These variations, however, also make it difficult to understand carbon itself as a unique material, and thus, each allotrope is often regarded independently. Some carbon types have already shown promising results in bioelectronic medicine, while many others remain potential candidates. In this topical review, we first provide a broad overview of the neuroelectronic devices and the basic requirements of an electrode material. We subsequently discuss the carbon family of materials and their properties that are useful in neural applications. Examples of devices fabricated using bulk and nano carbon materials are reviewed and critically compared. We then summarize the challenges, future prospects and next-generation carbon technology that can be helpful in the field of neural sciences. The article aims at providing a common platform to neuroscientists, electrochemists, biologists, microsystems engineers and carbon scientists to enable active and comprehensive efforts directed towards carbon-based neuroelectronic device fabrication.
Collapse
Affiliation(s)
- Mamta Devi
- School of Engineering, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh 175075, India
| | - Maria Vomero
- Bioelectronic Systems Laboratory, Columbia University, 500 West 120th Street, New York, NY 10027, United States of America
| | - Erwin Fuhrer
- School of Computing and Electrical Engineering, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh 175075 India
| | - Elisa Castagnola
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Calogero Gueli
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering-IMTEK, University of Freiburg, Georges-Koehler-Allee 080, 79110 Freiburg, Germany
| | - Surabhi Nimbalkar
- NanoFAB.SDSU Research Lab, Department of Mechanical Engineering, San Diego State University and NSF-ERC Center for Neurotechnology (CNT), 5500 Campanile Drive, San Diego, CA 92182, United States of America
| | - Mieko Hirabayashi
- NanoFAB.SDSU Research Lab, Department of Mechanical Engineering, San Diego State University and NSF-ERC Center for Neurotechnology (CNT), 5500 Campanile Drive, San Diego, CA 92182, United States of America
| | - Sam Kassegne
- NanoFAB.SDSU Research Lab, Department of Mechanical Engineering, San Diego State University and NSF-ERC Center for Neurotechnology (CNT), 5500 Campanile Drive, San Diego, CA 92182, United States of America
| | - Thomas Stieglitz
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering-IMTEK, University of Freiburg, Georges-Koehler-Allee 080, 79110 Freiburg, Germany.,BrainLinks-BrainTools Center, University of Freiburg, Georges-Koehler-Allee 080, 79110 Freiburg, Germany.,Bernstein Center Freiburg, University of Freiburg, Hansastr. 9a, 79104 Freiburg, Germany
| | - Swati Sharma
- School of Engineering, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh 175075, India
| |
Collapse
|
45
|
Intracortical Microelectrode Array Unit Yield under Chronic Conditions: A Comparative Evaluation. MICROMACHINES 2021; 12:mi12080972. [PMID: 34442594 PMCID: PMC8400387 DOI: 10.3390/mi12080972] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 01/01/2023]
Abstract
While microelectrode arrays (MEAs) offer the promise of elucidating functional neural circuitry and serve as the basis for a cortical neuroprosthesis, the challenge of designing and demonstrating chronically reliable technology remains. Numerous studies report “chronic” data but the actual time spans and performance measures corresponding to the experimental work vary. In this study, we reviewed the experimental durations that constitute chronic studies across a range of MEA types and animal species to gain an understanding of the widespread variability in reported study duration. For rodents, which are the most commonly used animal model in chronic studies, we examined active electrode yield (AEY) for different array types as a means to contextualize the study duration variance, as well as investigate and interpret the performance of custom devices in comparison to conventional MEAs. We observed wide-spread variance within species for the chronic implantation period and an AEY that decayed linearly in rodent models that implanted commercially-available devices. These observations provide a benchmark for comparing the performance of new technologies and highlight the need for consistency in chronic MEA studies. Additionally, to fully derive performance under chronic conditions, the duration of abiotic failure modes, biological processes induced by indwelling probes, and intended application of the device are key determinants.
Collapse
|
46
|
Abstract
Brain-machine interfaces (BMI) are being developed to restore upper limb function for persons with spinal cord injury or other motor degenerative conditions. BMI and implantable sensors for myoelectric prostheses directly extract information from the central or peripheral nervous system to provide users with high fidelity control of their prosthetic device. Control algorithms have been highly transferable between the 2 technologies but also face common issues. In this review of the current state of the art in each field, the authors point out similarities and differences between the 2 technologies that may guide the implementation of common solutions to these challenges.
Collapse
Affiliation(s)
- Alex K Vaskov
- Robotics Institute, University of Michigan, 2505 Hayward St, Ann Arbor, MI 48109, USA
| | - Cynthia A Chestek
- Robotics Institute, University of Michigan, 2505 Hayward St, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Ave, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan, 204 Washtenaw Ave, Ann Arbor, MI 48109, USA.
| |
Collapse
|
47
|
Chen L, Hartner J, Dong T, Li A, Watson B, Shih A. Flexible High-Resolution Force and Dimpling Measurement System for Pia and Dura Penetration During In Vivo Microelectrode Insertion Into Rat Brain. IEEE Trans Biomed Eng 2021; 68:2602-2612. [PMID: 33798065 PMCID: PMC8323825 DOI: 10.1109/tbme.2021.3070781] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Understanding the in vivo force and tissue dimpling during micro-electrode implantation into the brain are important for neuro-electrophysiology to minimize damage while enabling accurate placement and stable chronic extracellular electrophysiological recordings. Prior studies were unable to measure the sub-mN forces exerted during in vivo insertion of small electrodes. Here, we have investigated the in vivo force and dimpling depth profiles during brain surface membrane rupture (including dura) in anesthetized rats. METHODS A μN-resolution cantilever beam-based measurement system was designed, built, and calibrated and adapted for in vivo use. A total of 244 in vivo insertion tests were conducted on 8 anesthetized rats with 121 through pia mater and 123 through dura and pia combined. RESULTS Both microwire tip sharpening and diameter reduction reduced membrane rupture force (insertion force) and eased brain surface penetration. But dimpling depth and rupture force are not always strongly correlated. Multi-shank silicon probes showed smaller dimpling and rupture force per shank than single shank devices. CONCLUSION A force measurement system with flexible range and μN-level resolution (up to 0.032 μN) was achieved and proved feasible. For both pia-only and dura-pia penetrations in anesthetized rats, the rupture force and membrane dimpling depth at rupture are linearly related to the microwire diameter. SIGNIFICANCE We have developed a new system with both μN-level resolution and capacity to be used in vivo for measurement of force profiles of various neural interfaces into the brain. This allows quantification of brain tissue cutting and provides design guidelines for optimal neural interfaces.
Collapse
|
48
|
Restoring upper extremity function with brain-machine interfaces. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 159:153-186. [PMID: 34446245 DOI: 10.1016/bs.irn.2021.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
One of the most exciting advances to emerge in neural interface technologies has been the development of real-time brain-machine interface (BMI) neuroprosthetic devices to restore upper extremity function. BMI neuroprostheses, made possible by synergistic advances in neural recording technologies, high-speed computation and signal processing, and neuroscience, have permitted the restoration of volitional movement to patients suffering the loss of upper-extremity function. In this chapter, we review the scientific and technological advances underlying these remarkable devices. After presenting an introduction to the current state of the field, we provide an accessible technical discussion of the two fundamental requirements of a successful neuroprosthesis: signal extraction from the brain and signal decoding that results in robust prosthetic control. We close with a presentation of emerging technologies that are likely to substantially advance the field.
Collapse
|
49
|
Ravagli E, Mastitskaya S, Thompson N, Welle EJ, Chestek CA, Aristovich K, Holder D. Fascicle localisation within peripheral nerves through evoked activity recordings: A comparison between electrical impedance tomography and multi-electrode arrays. J Neurosci Methods 2021; 358:109140. [PMID: 33774053 PMCID: PMC8249910 DOI: 10.1016/j.jneumeth.2021.109140] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/07/2021] [Accepted: 03/12/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND The lack of understanding of fascicular organisation in peripheral nerves limits the potential of vagus nerve stimulation therapy. Two promising methods may be employed to identify the functional anatomy of fascicles within the nerve: fast neural electrical impedance tomography (EIT), and penetrating multi-electrode arrays (MEA). These could provide a means to image the compound action potential within fascicles in the nerve. NEW METHOD We compared the ability to localise fascicle activity between silicon shanks (SS) and carbon fibre (CF) multi-electrode arrays and fast neural EIT, with micro-computed tomography (MicroCT) as an independent reference. Fast neural EIT in peripheral nerves was only recently developed and MEA technology has been used only sparingly in nerves and not for source localisation. Assessment was performed in rat sciatic nerves while evoking neural activity in the tibial and peroneal fascicles. RESULTS Recorded compound action potentials were larger with CF compared to SS (∼700 μV vs ∼300 μV); however, background noise was greater (6.3 μV vs 1.7 μV) leading to lower SNR. Maximum spatial discrimination between Centres-of-Mass of fascicular activity was achieved by fast neural EIT (402 ± 30 μm) and CF MEA (414 ± 123 μm), with no statistical difference between MicroCT (625 ± 17 μm) and CF (p > 0.05) and between CF and EIT (p > 0.05). Compared to CF MEAs, SS MEAs had a lower discrimination power (103 ± 51 μm, p < 0.05). COMPARISON WITH EXISTING METHODS EIT and CF MEAs showed localisation power closest to MicroCT. Silicon MEAs adopted in this study failed to discriminate fascicle location. Re-design of probe geometry may improve results. CONCLUSIONS Nerve EIT is an accurate tool for assessment of fascicular position within nerves. Accuracy of EIT and CF MEA is similar to the reference method. We give technical recommendations for performing multi-electrode recordings in nerves.
Collapse
Affiliation(s)
- Enrico Ravagli
- Medical Physics and Biomedical Engineering, University College London, UK.
| | | | - Nicole Thompson
- Medical Physics and Biomedical Engineering, University College London, UK
| | - Elissa J Welle
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Cynthia A Chestek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kirill Aristovich
- Medical Physics and Biomedical Engineering, University College London, UK
| | - David Holder
- Medical Physics and Biomedical Engineering, University College London, UK
| |
Collapse
|
50
|
Welle EJ, Woods JE, Jiman AA, Richie JM, Bottorff EC, Ouyang Z, Seymour JP, Patel PR, Bruns TM, Chestek CA. Sharpened and Mechanically Durable Carbon Fiber Electrode Arrays for Neural Recording. IEEE Trans Neural Syst Rehabil Eng 2021; 29:993-1003. [PMID: 34014825 PMCID: PMC8459724 DOI: 10.1109/tnsre.2021.3082056] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bioelectric medicine treatments target disorders of the nervous system unresponsive to pharmacological methods. While current stimulation paradigms effectively treat many disorders, the underlying mechanisms are relatively unknown, and current neuroscience recording electrodes are often limited in their specificity to gross averages across many neurons or axons. Here, we develop a novel, durable carbon fiber electrode array adaptable to many neural structures for precise neural recording. Carbon fibers ( [Formula: see text] diameter) were sharpened using a reproducible blowtorchmethod that uses the reflection of fibers against the surface of a water bath. The arrays were developed by partially embedding carbon fibers in medical-grade silicone to improve durability. We recorded acute spontaneous electrophysiology from the rat cervical vagus nerve (CVN), feline dorsal root ganglia (DRG), and rat brain. Blowtorching resulted in fibers of 72.3 ± 33.5-degree tip angle with [Formula: see text] exposed carbon. Observable neural clusters were recorded using sharpened carbon fiber electrodes fromrat CVN ( [Formula: see text]), feline DRG ( [Formula: see text]), and rat brain ( [Formula: see text]). Recordings from the feline DRG included physiologically relevant signals from increased bladder pressure and cutaneous brushing. These results suggest that this carbon fiber array is a uniquely durable and adaptable neural recordingdevice. In the future, this device may be useful as a bioelectric medicine tool for diagnosis and closed-loop neural control of therapeutic treatments and monitoring systems.
Collapse
|