1
|
Jung M, Abu Shihada J, Decke S, Koschinski L, Graff PS, Maruri Pazmino S, Höllig A, Koch H, Musall S, Offenhäusser A, Rincón Montes V. Flexible 3D Kirigami Probes for In Vitro and In Vivo Neural Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2418524. [PMID: 40223534 DOI: 10.1002/adma.202418524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/24/2025] [Indexed: 04/15/2025]
Abstract
3D microelectrode arrays (MEAs) are gaining popularity as brain-machine interfaces and platforms for studying electrophysiological activity. Interactions with neural tissue depend on the electrochemical, mechanical, and spatial features of the recording platform. While planar or protruding 2D MEAs are limited in their ability to capture neural activity across layers, existing 3D platforms still require advancements in manufacturing scalability, spatial resolution, and tissue integration. In this work, a customizable, scalable, and straightforward approach to fabricate flexible 3D kirigami MEAs containing both surface and penetrating electrodes, designed to interact with the 3D space of neural tissue, is presented. These novel probes feature up to 512 electrodes distributed across 128 shanks in a single flexible device, with shank heights reaching up to 1 mm. The 3D kirigami MEAs are successfully deployed in several neural applications, both in vitro and in vivo, and identified spatially dependent electrophysiological activity patterns. Flexible 3D kirigami MEAs are therefore a powerful tool for large-scale electrical sampling of complex neural tissues while improving tissue integration and offering enhanced capabilities for analyzing neural disorders and disease models where high spatial resolution is required.
Collapse
Affiliation(s)
- Marie Jung
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - Jamal Abu Shihada
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - Simon Decke
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - Lina Koschinski
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
- Helmholtz Nano Facility (HNF), Forschungszentrum Jülich, Jülich, Germany
| | - Peter Severin Graff
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | | | - Anke Höllig
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital, Aachen, Germany
| | - Simon Musall
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
- Institute for Zoology, RWTH Aachen University, Aachen, Germany
- Faculty of Medicine, Institute of Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Andreas Offenhäusser
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
| | - Viviana Rincón Montes
- Bioelectronics, Institute of Biological Information Processing-3, Forschungszentrum Jülich, Jülich, Germany
| |
Collapse
|
2
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
3
|
Hoeferlin GF, Grabinski SE, Druschel LN, Duncan JL, Burkhart G, Weagraff GR, Lee AH, Hong C, Bambroo M, Olivares H, Bajwa T, Coleman J, Li L, Memberg W, Sweet J, Hamedani HA, Acharya AP, Hernandez-Reynoso AG, Donskey C, Jaskiw G, Ricky Chan E, Shoffstall AJ, Bolu Ajiboye A, von Recum HA, Zhang L, Capadona JR. Bacteria invade the brain following intracortical microelectrode implantation, inducing gut-brain axis disruption and contributing to reduced microelectrode performance. Nat Commun 2025; 16:1829. [PMID: 39979293 PMCID: PMC11842729 DOI: 10.1038/s41467-025-56979-4] [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: 03/04/2024] [Accepted: 01/29/2025] [Indexed: 02/22/2025] Open
Abstract
Brain-machine interface performance can be affected by neuroinflammatory responses due to blood-brain barrier (BBB) damage following intracortical microelectrode implantation. Recent findings suggest that certain gut bacterial constituents might enter the brain through damaged BBB. Therefore, we hypothesized that damage to the BBB caused by microelectrode implantation could facilitate microbiome entry into the brain. In our study, we found bacterial sequences, including gut-related ones, in the brains of mice with implanted microelectrodes. These sequences changed over time. Mice treated with antibiotics showed a reduced presence of these bacteria and had a different inflammatory response, which temporarily improved microelectrode recording performance. However, long-term antibiotic use worsened performance and disrupted neurodegenerative pathways. Many bacterial sequences found were not present in the gut or in unimplanted brains. Together, the current study established a paradigm-shifting mechanism that may contribute to chronic intracortical microelectrode recording performance and affect overall brain health following intracortical microelectrode implantation.
Collapse
Grants
- R01 NS131502 NINDS NIH HHS
- R25 CA221718 NCI NIH HHS
- T32 EB004314 NIBIB NIH HHS
- This study was supported in part by Merit Review Award GRANT12418820 (Capadona), Biomedical Science and Engineering Summer Program for Rehabilitation Interventions GRANT14089804 (Capadona/Hess-Dunning), and Senior Research Career Scientist Award # GRANT12635707 (Capadona) from the United States (US) Department of Veterans Affairs Rehabilitation Research and Development Service. Additionally, this work was also supported in part by the National Institute of Health, National Institute of Neurological Disorders and Stroke GRANT12635723 (Capadona/Pancrazio and diversity supplement Hernandez-Reynoso) and NS131502 (Ware/Pancrazio/Capadona), the National Cancer Institute NCI R25 CA221718 (Berger) provided support for Weagraff, the Congressionally Directed Medical Research Program (CDMRP) – Spinal Cord Injury Research Program (SCIRP), administered through the Department of Defense Award # SC180308 (Ajiboye) and the National Institute for Biomedical Imaging and Bioengineering, T32EB004314, provided support for both Hoeferlin and Burkhart (Capadona/Kirsch). Microbiome analyses were partially supported by the junior faculty’s startup funding from the CWRU School of Medicine, BGT630267 (Zhang). Finally, partial funding was provided from discretionary funding from the Donnell Institute Professorship endowment (Capadona) and the Case School of Engineering Research Incentive Program (Capadona).
Collapse
Affiliation(s)
- George F Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Sarah E Grabinski
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Jonathan L Duncan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Grace Burkhart
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Gwendolyn R Weagraff
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Biology, University of Florida, Gainesville, FL, USA
| | - Alice H Lee
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Christopher Hong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Meera Bambroo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Hannah Olivares
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Tejas Bajwa
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Jennifer Coleman
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - William Memberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Jennifer Sweet
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Neurological Surgery, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Hoda Amani Hamedani
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Materials Science and Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Abhinav P Acharya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Ana G Hernandez-Reynoso
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Curtis Donskey
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Division of Infectious Diseases & HIV Medicine in the Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - George Jaskiw
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - A Bolu Ajiboye
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Horst A von Recum
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Liangliang Zhang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA.
| |
Collapse
|
4
|
Yi D, Yao Y, Wang Y, Chen L. Design, Fabrication, and Implantation of Invasive Microelectrode Arrays as in vivo Brain Machine Interfaces: A Comprehensive Review. JOURNAL OF MANUFACTURING PROCESSES 2024; 126:185-207. [PMID: 39185373 PMCID: PMC11340637 DOI: 10.1016/j.jmapro.2024.07.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Invasive Microelectrode Arrays (MEAs) have been a significant and useful tool for us to gain a fundamental understanding of how the brain works through high spatiotemporal resolution neuron-level recordings and/or stimulations. Through decades of research, various types of microwire, silicon, and flexible substrate-based MEAs have been developed using the evolving new materials, novel design concepts, and cutting-edge advanced manufacturing capabilities. Surgical implantation of the latest minimal damaging flexible MEAs through the hard-to-penetrate brain membranes introduces new challenges and thus the development of implantation strategies and instruments for the latest MEAs. In this paper, studies on the design considerations and enabling manufacturing processes of various invasive MEAs as in vivo brain-machine interfaces have been reviewed to facilitate the development as well as the state-of-art of such brain-machine interfaces from an engineering perspective. The challenges and solution strategies developed for surgically implanting such interfaces into the brain have also been evaluated and summarized. Finally, the research gaps have been identified in the design, manufacturing, and implantation perspectives, and future research prospects in invasive MEA development have been proposed.
Collapse
Affiliation(s)
- Dongyang Yi
- Department of Mechanical and Industrial Engineering, University of Massachusetts Lowell, Lowell, MA 01854
| | - Yao Yao
- Department of Industrial and Systems Engineering, University of Missouri, Columbia, MO 65211
| | - Yi Wang
- Department of Industrial and Systems Engineering, University of Missouri, Columbia, MO 65211
| | - Lei Chen
- Department of Mechanical and Industrial Engineering, University of Massachusetts Lowell, Lowell, MA 01854
| |
Collapse
|
5
|
Song SS, Druschel LN, Kasthuri NM, Wang JJ, Conard JH, Chan ER, Acharya AP, Capadona JR. Comprehensive proteomic analysis of the differential expression of 62 proteins following intracortical microelectrode implantation. Sci Rep 2024; 14:17596. [PMID: 39080300 PMCID: PMC11289480 DOI: 10.1038/s41598-024-68017-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Intracortical microelectrodes (IMEs) are devices designed to be implanted into the cerebral cortex for various neuroscience and neuro-engineering applications. A critical feature of IMEs is their ability to detect neural activity from individual neurons. Currently, IMEs are limited by chronic failure, largely considered to be caused by the prolonged neuroinflammatory response to the implanted devices. Over the past few years, the characterization of the neuroinflammatory response has grown in sophistication, with the most recent advances focusing on mRNA expression following IME implantation. While gene expression studies increase our broad understanding of the relationship between IMEs and cortical tissue, advanced proteomic techniques have not been reported. Proteomic evaluation is necessary to describe the diverse changes in protein expression specific to neuroinflammation, neurodegeneration, or tissue and cellular viability, which could lead to the further development of targeted intervention strategies designed to improve IME functionality. In this study, we have characterized the expression of 62 proteins within 180 μm of the IME implant site at 4-, 8-, and 16-weeks post-implantation. We identified potential targets for immunotherapies, as well as key pathways that contribute to neuronal dieback around the IME implant.
Collapse
Affiliation(s)
- Sydney S Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Niveda M Kasthuri
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Jaime J Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA
| | - Jacob H Conard
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Abhinav P Acharya
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA.
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
6
|
Coles L, Ventrella D, Carnicer-Lombarte A, Elmi A, Troughton JG, Mariello M, El Hadwe S, Woodington BJ, Bacci ML, Malliaras GG, Barone DG, Proctor CM. Origami-inspired soft fluidic actuation for minimally invasive large-area electrocorticography. Nat Commun 2024; 15:6290. [PMID: 39060241 PMCID: PMC11282215 DOI: 10.1038/s41467-024-50597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Electrocorticography is an established neural interfacing technique wherein an array of electrodes enables large-area recording from the cortical surface. Electrocorticography is commonly used for seizure mapping however the implantation of large-area electrocorticography arrays is a highly invasive procedure, requiring a craniotomy larger than the implant area to place the device. In this work, flexible thin-film electrode arrays are combined with concepts from soft robotics, to realize a large-area electrocorticography device that can change shape via integrated fluidic actuators. We show that the 32-electrode device can be packaged using origami-inspired folding into a compressed state and implanted through a small burr-hole craniotomy, then expanded on the surface of the brain for large-area cortical coverage. The implantation, expansion, and recording functionality of the device is confirmed in-vitro and in porcine in-vivo models. The integration of shape actuation into neural implants provides a clinically viable pathway to realize large-area neural interfaces via minimally invasive surgical techniques.
Collapse
Affiliation(s)
- Lawrence Coles
- Department of Engineering, University of Cambridge, Cambridge, UK
- Institute of Biomedical Engineering, Engineering Science Department, University of Oxford, Oxford, UK
| | - Domenico Ventrella
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | | | - Alberto Elmi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | - Joe G Troughton
- Department of Engineering, University of Cambridge, Cambridge, UK
- Institute of Biomedical Engineering, Engineering Science Department, University of Oxford, Oxford, UK
| | - Massimo Mariello
- Institute of Biomedical Engineering, Engineering Science Department, University of Oxford, Oxford, UK
| | - Salim El Hadwe
- Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ben J Woodington
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Maria L Bacci
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | | | - Damiano G Barone
- Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Christopher M Proctor
- Department of Engineering, University of Cambridge, Cambridge, UK.
- Institute of Biomedical Engineering, Engineering Science Department, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Hofmann UG, Stieglitz T. Why some BCI should still be called BMI. Nat Commun 2024; 15:6207. [PMID: 39043690 PMCID: PMC11266610 DOI: 10.1038/s41467-024-50603-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024] Open
Affiliation(s)
- Ulrich G Hofmann
- Section for Neuroelectronic Systems, Department of Neurosurgery, Medical Center University of Freiburg and Medical Faculty, University of Freiburg, Freiburg, Germany.
- BrainLinks-BrainTools, Freiburg, Germany.
| | - Thomas Stieglitz
- BrainLinks-BrainTools, Freiburg, Germany.
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering-IMTEK, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
8
|
Meijs S, Andreis FR, Janjua TAM, Jensen W. Reliability of a cranial window for chronic epidural recordings from the pig primary somatosensory cortex. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039725 DOI: 10.1109/embc53108.2024.10782109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Chronically implanted microelectrodes face adverse biological responses and various kinds of device failure. To overcome these challenges, a cranial window was developed allowing repeated access to the primary somatosensory cortex (S1) of the pig. This study evaluated the reliability of the signals recorded using repeated temporary placement of a micro-electrocorticography (µECoG) array via a cranial window. Seven pigs were implanted with a cranial window, which was accessed twice after implantation at 2-3 week intervals. Brain responses were evoked by electrical stimulation to the ulnar nerve. At each session, three trials were recorded, consisting of a hundred stimulations. Averaged responses of P1 amplitude, latency, and variance between channels were obtained. Signal characteristics were stable within and between sessions. No systematic errors were found for the P1 amplitude and channel variability. For the P1 latency, a systematic decrease in latency was found between session 1 (25±2 ms) and sessions 2 and 3 (24±2 ms). These results show that the temporary placement of microelectrodes to record brain signals is a good alternative to permanent implantation.
Collapse
|
9
|
Galindo AN, Frey Rubio DA, Hettiaratchi MH. Biomaterial strategies for regulating the neuroinflammatory response. MATERIALS ADVANCES 2024; 5:4025-4054. [PMID: 38774837 PMCID: PMC11103561 DOI: 10.1039/d3ma00736g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/07/2024] [Indexed: 05/24/2024]
Abstract
Injury and disease in the central nervous system (CNS) can result in a dysregulated inflammatory environment that inhibits the repair of functional tissue. Biomaterials present a promising approach to tackle this complex inhibitory environment and modulate the mechanisms involved in neuroinflammation to halt the progression of secondary injury and promote the repair of functional tissue. In this review, we will cover recent advances in biomaterial strategies, including nanoparticles, hydrogels, implantable scaffolds, and neural probe coatings, that have been used to modulate the innate immune response to injury and disease within the CNS. The stages of inflammation following CNS injury and the main inflammatory contributors involved in common neurodegenerative diseases will be discussed, as understanding the inflammatory response to injury and disease is critical for identifying therapeutic targets and designing effective biomaterial-based treatment strategies. Biomaterials and novel composites will then be discussed with an emphasis on strategies that deliver immunomodulatory agents or utilize cell-material interactions to modulate inflammation and promote functional tissue repair. We will explore the application of these biomaterial-based strategies in the context of nanoparticle- and hydrogel-mediated delivery of small molecule drugs and therapeutic proteins to inflamed nervous tissue, implantation of hydrogels and scaffolds to modulate immune cell behavior and guide axon elongation, and neural probe coatings to mitigate glial scarring and enhance signaling at the tissue-device interface. Finally, we will present a future outlook on the growing role of biomaterial-based strategies for immunomodulation in regenerative medicine and neuroengineering applications in the CNS.
Collapse
Affiliation(s)
- Alycia N Galindo
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
| | - David A Frey Rubio
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
| | - Marian H Hettiaratchi
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon Eugene OR USA
- Department of Chemistry and Biochemistry, University of Oregon Eugene OR USA
| |
Collapse
|
10
|
Song S, Druschel L, Kasthuri N, Wang J, Conard J, Chan E, Acharya A, Capadona J. Comprehensive Proteomic Analysis of the Differential Expression of 83 Proteins Following Intracortical Microelectrode Implantation. RESEARCH SQUARE 2024:rs.3.rs-4039586. [PMID: 38559066 PMCID: PMC10980140 DOI: 10.21203/rs.3.rs-4039586/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Intracortical microelectrodes (IMEs) are devices designed to be implanted into the cerebral cortex for various neuroscience and neuro-engineering applications. A critical feature of these devices is their ability to detect neural activity from individual neurons. Currently, IMEs are limited by chronic failure, largely considered to be caused by the prolonged neuroinflammatory response to the implanted devices. Over the decades, characterization of the neuroinflammatory response has grown in sophistication, with the most recent advances including advanced genomics and spatially resolved transcriptomics. While gene expression studies increase our broad understanding of the relationship between IMEs and cortical tissue, advanced proteomic techniques have not been reported. Proteomic evaluation is necessary to describe the diverse changes in protein expression specific to neuroinflammation, neurodegeneration, or tissue and cellular viability, which could lead to the development of more targeted intervention strategies designed to improve IME function. In this study, we have characterized the expression of 83 proteins within 180 μm of the IME implant site at 4-, 8-, and 16-weeks post-implantation. We identified potential targets for immunotherapies, as well as key pathways and functions that contribute to neuronal dieback around the IME implant.
Collapse
|
11
|
Capadona J, Hoeferlin G, Grabinski S, Druschel L, Duncan J, Burkhart G, Weagraff G, Lee A, Hong C, Bambroo M, Olivares H, Bajwa T, Memberg W, Sweet J, Hamedani HA, Acharya A, Hernandez-Reynoso A, Donskey C, Jaskiw G, Chan R, Ajiboye A, von Recum H, Zhang L. Bacteria Invade the Brain Following Sterile Intracortical Microelectrode Implantation. RESEARCH SQUARE 2024:rs.3.rs-3980065. [PMID: 38496527 PMCID: PMC10942555 DOI: 10.21203/rs.3.rs-3980065/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Brain-machine interface performance is largely affected by the neuroinflammatory responses resulting in large part from blood-brain barrier (BBB) damage following intracortical microelectrode implantation. Recent findings strongly suggest that certain gut bacterial constituents penetrate the BBB and are resident in various brain regions of rodents and humans, both in health and disease. Therefore, we hypothesized that damage to the BBB caused by microelectrode implantation could amplify dysregulation of the microbiome-gut-brain axis. Here, we report that bacteria, including those commonly found in the gut, enter the brain following intracortical microelectrode implantation in mice implanted with single-shank silicon microelectrodes. Systemic antibiotic treatment of mice implanted with microelectrodes to suppress bacteria resulted in differential expression of bacteria in the brain tissue and a reduced acute inflammatory response compared to untreated controls, correlating with temporary improvements in microelectrode recording performance. Long-term antibiotic treatment resulted in worsening microelectrode recording performance and dysregulation of neurodegenerative pathways. Fecal microbiome composition was similar between implanted mice and an implanted human, suggesting translational findings. However, a significant portion of invading bacteria was not resident in the brain or gut. Together, the current study established a paradigm-shifting mechanism that may contribute to chronic intracortical microelectrode recording performance and affect overall brain health following intracortical microelectrode implantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ricky Chan
- Institute for Computational Biology, Case Western Reserve University
| | | | | | | |
Collapse
|
12
|
Hernandez-Reynoso AG, Sturgill BS, Hoeferlin GF, Druschel LN, Krebs OK, Menendez DM, Thai TTD, Smith TJ, Duncan J, Zhang J, Mittal G, Radhakrishna R, Desai MS, Cogan SF, Pancrazio JJ, Capadona JR. The effect of a Mn(III)tetrakis(4-benzoic acid)porphyrin (MnTBAP) coating on the chronic recording performance of planar silicon intracortical microelectrode arrays. Biomaterials 2023; 303:122351. [PMID: 37931456 PMCID: PMC10842897 DOI: 10.1016/j.biomaterials.2023.122351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 11/08/2023]
Abstract
Intracortical microelectrode arrays (MEAs) are used to record neural activity. However, their implantation initiates a neuroinflammatory cascade, involving the accumulation of reactive oxygen species, leading to interface failure. Here, we coated commercially-available MEAs with Mn(III)tetrakis(4-benzoic acid)porphyrin (MnTBAP), to mitigate oxidative stress. First, we assessed the in vitro cytotoxicity of modified sample substrates. Then, we implanted 36 rats with uncoated, MnTBAP-coated ("Coated"), or (3-Aminopropyl)triethoxysilane (APTES)-coated devices - an intermediate step in the coating process. We assessed electrode performance during the acute (1-5 weeks), sub-chronic (6-11 weeks), and chronic (12-16 weeks) phases after implantation. Three subsets of animals were euthanized at different time points to assess the acute, sub-chronic and chronic immunohistological responses. Results showed that MnTBAP coatings were not cytotoxic in vitro, and their implantation in vivo improved the proportion of electrodes during the sub-chronic and chronic phases; APTES coatings resulted in failure of the neural interface during the chronic phase. In addition, MnTBAP coatings improved the quality of the signal throughout the study and reduced the neuroinflammatory response around the implant as early as two weeks, an effect that remained consistent for months post-implantation. Together, these results suggest that MnTBAP coatings are a potentially useful modification to improve MEA reliability.
Collapse
Affiliation(s)
- Ana G Hernandez-Reynoso
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Brandon S Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - George F Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Olivia K Krebs
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Dhariyat M Menendez
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Teresa T D Thai
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Thomas J Smith
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Jonathan Duncan
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Jichu Zhang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Gaurav Mittal
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| | - Rahul Radhakrishna
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Mrudang Spandan Desai
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Stuart F Cogan
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Joseph J Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, United States.
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH, 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, 10701 East Blvd, Cleveland, OH, 44106, United States.
| |
Collapse
|
13
|
Perna A, Angotzi GN, Berdondini L, Ribeiro JF. Advancing the interfacing performances of chronically implantable neural probes in the era of CMOS neuroelectronics. Front Neurosci 2023; 17:1275908. [PMID: 38027514 PMCID: PMC10644322 DOI: 10.3389/fnins.2023.1275908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Tissue penetrating microelectrode neural probes can record electrophysiological brain signals at resolutions down to single neurons, making them invaluable tools for neuroscience research and Brain-Computer-Interfaces (BCIs). The known gradual decrease of their electrical interfacing performances in chronic settings, however, remains a major challenge. A key factor leading to such decay is Foreign Body Reaction (FBR), which is the cascade of biological responses that occurs in the brain in the presence of a tissue damaging artificial device. Interestingly, the recent adoption of Complementary Metal Oxide Semiconductor (CMOS) technology to realize implantable neural probes capable of monitoring hundreds to thousands of neurons simultaneously, may open new opportunities to face the FBR challenge. Indeed, this shift from passive Micro Electro-Mechanical Systems (MEMS) to active CMOS neural probe technologies creates important, yet unexplored, opportunities to tune probe features such as the mechanical properties of the probe, its layout, size, and surface physicochemical properties, to minimize tissue damage and consequently FBR. Here, we will first review relevant literature on FBR to provide a better understanding of the processes and sources underlying this tissue response. Methods to assess FBR will be described, including conventional approaches based on the imaging of biomarkers, and more recent transcriptomics technologies. Then, we will consider emerging opportunities offered by the features of CMOS probes. Finally, we will describe a prototypical neural probe that may meet the needs for advancing clinical BCIs, and we propose axial insertion force as a potential metric to assess the influence of probe features on acute tissue damage and to control the implantation procedure to minimize iatrogenic injury and subsequent FBR.
Collapse
Affiliation(s)
- Alberto Perna
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Istituto Italiano di Tecnologia, Genova, Italy
| | - Gian Nicola Angotzi
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| | - Luca Berdondini
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| | - João Filipe Ribeiro
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| |
Collapse
|
14
|
Song S, Druschel LN, Chan ER, Capadona JR. Differential expression of genes involved in the chronic response to intracortical microelectrodes. Acta Biomater 2023; 169:348-362. [PMID: 37507031 PMCID: PMC10528922 DOI: 10.1016/j.actbio.2023.07.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Brain-Machine Interface systems (BMIs) are clinically valuable devices that can provide functional restoration for patients with spinal cord injury or improved integration for patients requiring prostheses. Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for precisely controlling BMIs. However, intracortical microelectrodes have a demonstrated history of progressive decline in the recording performance with time, inhibiting their usefulness. One major contributor to decreased performance is the neuroinflammatory response to the implanted microelectrodes. The neuroinflammatory response can lead to neurodegeneration and the formation of a glial scar at the implant site. Historically, histological imaging of relatively few known cellular and protein markers has characterized the neuroinflammatory response to implanted microelectrode arrays. However, neuroinflammation requires many molecular players to coordinate the response - meaning traditional methods could result in an incomplete understanding. Taking advantage of recent advancements in tools to characterize the relative or absolute DNA/RNA expression levels, a few groups have begun to explore gene expression at the microelectrode-tissue interface. We have utilized a custom panel of ∼813 neuroinflammatory-specific genes developed with NanoString for bulk tissue analysis at the microelectrode-tissue interface. Our previous studies characterized the acute innate immune response to intracortical microelectrodes. Here we investigated the gene expression at the microelectrode-tissue interface in wild-type (WT) mice chronically implanted with nonfunctioning probes. We found 28 differentially expressed genes at chronic time points (4WK, 8WK, and 16WK), many in the complement and extracellular matrix system. Further, the expression levels were relatively stable over time. Genes identified here represent chronic molecular players at the microelectrode implant sites and potential therapeutic targets for the long-term integration of microelectrodes. STATEMENT OF SIGNIFICANCE: Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for the precise control of Brain-Machine Interface systems (BMIs). However, intracortical microelectrodes have a demonstrated history of progressive declines in the recording performance with time, inhibiting their usefulness. One major contributor to the decline in these devices is the neuroinflammatory response against the implanted microelectrodes. Historically, neuroinflammation to implanted microelectrode arrays has been characterized by histological imaging of relatively few known cellular and protein markers. Few studies have begun to develop a more in-depth understanding of the molecular pathways facilitating device-mediated neuroinflammation. Here, we are among the first to identify genetic pathways that could represent targets to improve the host response to intracortical microelectrodes, and ultimately device performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
15
|
Kim Y, Mueller NN, Schwartzman WE, Sarno D, Wynder R, Hoeferlin GF, Gisser K, Capadona JR, Hess-Dunning A. Fabrication Methods and Chronic In Vivo Validation of Mechanically Adaptive Microfluidic Intracortical Devices. MICROMACHINES 2023; 14:1015. [PMID: 37241639 PMCID: PMC10223487 DOI: 10.3390/mi14051015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023]
Abstract
Intracortical neural probes are both a powerful tool in basic neuroscience studies of brain function and a critical component of brain computer interfaces (BCIs) designed to restore function to paralyzed patients. Intracortical neural probes can be used both to detect neural activity at single unit resolution and to stimulate small populations of neurons with high resolution. Unfortunately, intracortical neural probes tend to fail at chronic timepoints in large part due to the neuroinflammatory response that follows implantation and persistent dwelling in the cortex. Many promising approaches are under development to circumvent the inflammatory response, including the development of less inflammatory materials/device designs and the delivery of antioxidant or anti-inflammatory therapies. Here, we report on our recent efforts to integrate the neuroprotective effects of both a dynamically softening polymer substrate designed to minimize tissue strain and localized drug delivery at the intracortical neural probe/tissue interface through the incorporation of microfluidic channels within the probe. The fabrication process and device design were both optimized with respect to the resulting device mechanical properties, stability, and microfluidic functionality. The optimized devices were successfully able to deliver an antioxidant solution throughout a six-week in vivo rat study. Histological data indicated that a multi-outlet design was most effective at reducing markers of inflammation. The ability to reduce inflammation through a combined approach of drug delivery and soft materials as a platform technology allows future studies to explore additional therapeutics to further enhance intracortical neural probes performance and longevity for clinical applications.
Collapse
Affiliation(s)
- Youjoung Kim
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Natalie N Mueller
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - William E Schwartzman
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Danielle Sarno
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Reagan Wynder
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - George F Hoeferlin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Kaela Gisser
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Allison Hess-Dunning
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
16
|
Thompson C, Evans B, Zhao D, Purcell E. Spatiotemporal Expression of RNA-Seq Identified Proteins at the Electrode Interface. Acta Biomater 2023; 164:209-222. [PMID: 37116634 DOI: 10.1016/j.actbio.2023.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/30/2023]
Abstract
Implantation of electrodes in the brain can be used to record from or stimulate neural tissues to treat neurological disease and injury. However, the tissue response to implanted devices can limit their functional longevity. Recent RNA-seq datasets identify hundreds of genes associated with gliosis, neuronal function, myelination, and cellular metabolism that are spatiotemporally expressed in neural tissues following the insertion of microelectrodes. To validate mRNA as a predictor of protein expression, this study evaluates a sub-set of RNA-seq identified proteins (RSIP) at 24-hours, 1-week, and 6-weeks post-implantation using quantitative immunofluorescence methods. This study found that expression of RSIPs associated with glial activation (Glial fibrillary acidic protein (GFAP), Polypyrmidine tract binding protein-1 (Ptbp1)), neuronal structure (Neurofilament heavy chain (Nefh), Proteolipid protein-1 (Plp1), Myelin Basic Protein (MBP)), and iron metabolism (Transferrin (TF), Ferritin heavy chain-1 (Fth1)) reinforce transcriptional data. This study also provides additional context to the cellular distribution of RSIPs using a MATLAB-based approach to quantify immunofluorescence intensity within specific cell types. Ptbp1, TF, and Fth1 were found to be spatiotemporally distributed within neurons, astrocytes, microglia, and oligodendrocytes at the device interface relative to distal and contralateral tissues. The altered distribution of RSIPs relative to distal tissue is largely localized within 100µm of the device injury, which approaches the functional recording range of implanted electrodes. This study provides evidence that RNA-sequencing can be used to predict protein-level changes in cortical tissues and that RSIPs can be further investigated to identify new biomarkers of the tissue response that influence signal quality. STATEMENT OF SIGNIFICANCE: : Microelectrode arrays implanted into the brain are useful tools that can be used to study neuroscience and to treat pathological conditions in a clinical setting. The tissue response to these devices, however, can severely limit their functional longevity. Transcriptomics has deepened the understandings of the tissue response by revealing numerous genes which are differentially expressed following device insertion. This manuscript provides validation for the use of transcriptomics to characterize the tissue response by evaluating a subset of known differentially expressed genes at the protein level around implanted electrodes over time. In additional to validating mRNA-to-protein relationships at the device interface, this study has identified emerging trends in the spatiotemporal distribution of proteins involved with glial activation, neuronal remodeling, and essential iron binding proteins around implanted silicon devices. This study additionally provides a new MATLAB based methodology to quantify protein distribution within discrete cell types at the device interface which may be used as biomarkers for further study or therapeutic intervention in the future.
Collapse
Affiliation(s)
- Cort Thompson
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Blake Evans
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Dorothy Zhao
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America
| | - Erin Purcell
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, United States of America; Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, United States of America.
| |
Collapse
|
17
|
Riggins TE, Whitsitt QA, Saxena A, Hunter E, Hunt B, Thompson CH, Moore MG, Purcell EK. Gene Expression Changes in Cultured Reactive Rat Astrocyte Models and Comparison to Device-Associated Effects in the Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.522870. [PMID: 36712012 PMCID: PMC9881929 DOI: 10.1101/2023.01.06.522870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Implanted microelectrode arrays hold immense therapeutic potential for many neurodegenerative diseases. However, a foreign body response limits long-term device performance. Recent literature supports the role of astrocytes in the response to damage to the central nervous system (CNS) and suggests that reactive astrocytes exist on a spectrum of phenotypes, from beneficial to neurotoxic. The goal of our study was to gain insight into the subtypes of reactive astrocytes responding to electrodes implanted in the brain. In this study, we tested the transcriptomic profile of two reactive astrocyte culture models (cytokine cocktail or lipopolysaccharide, LPS) utilizing RNA sequencing, which we then compared to differential gene expression surrounding devices inserted into rat motor cortex via spatial transcriptomics. We interpreted changes in the genetic expression of the culture models to that of 24 hour, 1 week and 6 week rat tissue samples at multiple distances radiating from the injury site. We found overlapping expression of up to ∼250 genes between in vitro models and in vivo effects, depending on duration of implantation. Cytokine-induced cells shared more genes in common with chronically implanted tissue (≥1 week) in comparison to LPS-exposed cells. We revealed localized expression of a subset of these intersecting genes (e.g., Serping1, Chi3l1, and Cyp7b1) in regions of device-encapsulating, glial fibrillary acidic protein (GFAP)-expressing astrocytes identified with immunohistochemistry. We applied a factorization approach to assess the strength of the relationship between reactivity markers and the spatial distribution of GFAP-expressing astrocytes in vivo . We also provide lists of hundreds of differentially expressed genes between reactive culture models and untreated controls, and we observed 311 shared genes between the cytokine induced model and the LPS-reaction induced control model. Our results show that comparisons of reactive astrocyte culture models with spatial transcriptomics data can reveal new biomarkers of the foreign body response to implantable neurotechnology. These comparisons also provide a strategy to assess the development of in vitro models of the tissue response to implanted electrodes.
Collapse
|
18
|
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
|
19
|
Moore MG, Thompson CH, Reimers MA, Purcell EK. Differential Co-Expression Analysis of RNA-Seq Data Reveals Novel Potential Biomarkers of Device-Tissue Interaction. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:3072-3076. [PMID: 36085767 PMCID: PMC9724584 DOI: 10.1109/embc48229.2022.9871437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The biological response to electrodes implanted in the brain has been a long-standing barrier to achieving a stable tissue device-interface. Understanding the mechanisms underlying this response could explain phenomena including recording instability and loss, shifting stimulation thresholds, off-target effects of neuromodulation, and stimulation-induced depression of neural excitability. Our prior work detected differential expression in hundreds of genes following device implantation. Here, we extend upon that work by providing new analyses using differential co-expression analysis, which identifies changes in the correlation structure between groups of genes detected at the interface in comparison to control tissues. We used an "eigengene" approach to identify hub genes associated with each module. Our work adds to a growing body of literature which applies new techniques in molecular biology and computational analysis to long-standing issues surrounding electrode integration with the brain.
Collapse
|