1
|
Deak F. Alzheimer's disease and other memory disorders in the age of AI: reflection and perspectives on the 120th anniversary of the birth of Dr. John von Neumann. GeroScience 2024:10.1007/s11357-024-01378-8. [PMID: 39419932 DOI: 10.1007/s11357-024-01378-8] [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: 07/19/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
Two themes are coming to the forefront in this decade: Cognitive impairment of an aging population and the quantum leap in developing artificial intelligence (AI). Both can be described as growing exponentially and presenting serious challenges. Although many questions have been addressed about the dangers of AI, we want to go beyond the fearful aspects of this topic and focus on the possible contribution of AI to solve the problem of chronic disorders of the elderly leading to cognitive impairment, like Alzheimer's disease, Parkinson's disease, and Lewy body dementia. Our second goal is to look at the ways in which modern neuroscience can influence the future design of computers and the development of AI. We wish to honor the memory of Dr. John von Neumann, who came up with many breakthrough details of the first electronic computer. Remarkably, Dr. von Neumann dedicated his last book to the comparison of the human brain and the computer as it stood in those years of the mid-1950s. We will point out how his ideas are more relevant than ever in the age of supercomputers, AI and brain implants.
Collapse
Affiliation(s)
- Ferenc Deak
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
2
|
Alfihed S, Majrashi M, Ansary M, Alshamrani N, Albrahim SH, Alsolami A, Alamari HA, Zaman A, Almutairi D, Kurdi A, Alzaydi MM, Tabbakh T, Al-Otaibi F. Non-Invasive Brain Sensing Technologies for Modulation of Neurological Disorders. BIOSENSORS 2024; 14:335. [PMID: 39056611 PMCID: PMC11274405 DOI: 10.3390/bios14070335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/01/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024]
Abstract
The non-invasive brain sensing modulation technology field is experiencing rapid development, with new techniques constantly emerging. This study delves into the field of non-invasive brain neuromodulation, a safer and potentially effective approach for treating a spectrum of neurological and psychiatric disorders. Unlike traditional deep brain stimulation (DBS) surgery, non-invasive techniques employ ultrasound, electrical currents, and electromagnetic field stimulation to stimulate the brain from outside the skull, thereby eliminating surgery risks and enhancing patient comfort. This study explores the mechanisms of various modalities, including transcranial direct current stimulation (tDCS) and transcranial magnetic stimulation (TMS), highlighting their potential to address chronic pain, anxiety, Parkinson's disease, and depression. We also probe into the concept of closed-loop neuromodulation, which personalizes stimulation based on real-time brain activity. While we acknowledge the limitations of current technologies, our study concludes by proposing future research avenues to advance this rapidly evolving field with its immense potential to revolutionize neurological and psychiatric care and lay the foundation for the continuing advancement of innovative non-invasive brain sensing technologies.
Collapse
Affiliation(s)
- Salman Alfihed
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Majed Majrashi
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Muhammad Ansary
- Neuroscience Center Research Unit, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Naif Alshamrani
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Shahad H. Albrahim
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Abdulrahman Alsolami
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Hala A. Alamari
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Adnan Zaman
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Dhaifallah Almutairi
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Abdulaziz Kurdi
- Advanced Materials Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Mai M. Alzaydi
- Bioengineering Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia
| | - Thamer Tabbakh
- Microelectronics and Semiconductor Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (S.A.)
| | - Faisal Al-Otaibi
- Neuroscience Center Research Unit, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
3
|
Alahi MEE, Rizu MI, Tina FW, Huang Z, Nag A, Afsarimanesh N. Recent Advancements in Graphene-Based Implantable Electrodes for Neural Recording/Stimulation. SENSORS (BASEL, SWITZERLAND) 2023; 23:9911. [PMID: 38139756 PMCID: PMC10747868 DOI: 10.3390/s23249911] [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: 10/02/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
Implantable electrodes represent a groundbreaking advancement in nervous system research, providing a pivotal tool for recording and stimulating human neural activity. This capability is integral for unraveling the intricacies of the nervous system's functionality and for devising innovative treatments for various neurological disorders. Implantable electrodes offer distinct advantages compared to conventional recording and stimulating neural activity methods. They deliver heightened precision, fewer associated side effects, and the ability to gather data from diverse neural sources. Crucially, the development of implantable electrodes necessitates key attributes: flexibility, stability, and high resolution. Graphene emerges as a highly promising material for fabricating such electrodes due to its exceptional properties. It boasts remarkable flexibility, ensuring seamless integration with the complex and contoured surfaces of neural tissues. Additionally, graphene exhibits low electrical resistance, enabling efficient transmission of neural signals. Its transparency further extends its utility, facilitating compatibility with various imaging techniques and optogenetics. This paper showcases noteworthy endeavors in utilizing graphene in its pure form and as composites to create and deploy implantable devices tailored for neural recordings and stimulations. It underscores the potential for significant advancements in this field. Furthermore, this paper delves into prospective avenues for refining existing graphene-based electrodes, enhancing their suitability for neural recording applications in in vitro and in vivo settings. These future steps promise to revolutionize further our capacity to understand and interact with the neural research landscape.
Collapse
Affiliation(s)
- Md Eshrat E. Alahi
- School of Engineering and Technology, Walailak University, 222 Thaiburi, Thasala District, Nakhon Si Thammarat 80160, Thailand
| | - Mubdiul Islam Rizu
- Microsystems Nanotechnologies for Chemical Analysis (MINOS), Universitat Rovira I Virgili, Avinguda Països Catalans, 26—Campus Sescelades, 43007 Tarragona, Spain;
| | - Fahmida Wazed Tina
- Creative Innovation in Science and Technology Program, Faculty of Science and Technology, Nakhon Si Thammarat Rajabhat University, Nakhon Si Thammarat 80280, Thailand;
| | - Zhaoling Huang
- School of Mechanical and Electrical Engineering, Guilin University of Electronic Technology, Guilin 541004, China;
| | - Anindya Nag
- Faculty of Electrical and Computer Engineering, Technische Universität Dresden, 01062 Dresden, Germany;
- Centre for Tactile Internet with Human-in-the-Loop (CeTI), Technische Universität Dresden, 01069 Dresden, Germany
| | - Nasrin Afsarimanesh
- School of Civil and Mechanical Engineering, Curtin University, Perth, WA 6102, Australia;
| |
Collapse
|
4
|
Boulingre M, Portillo-Lara R, Green RA. Biohybrid neural interfaces: improving the biological integration of neural implants. Chem Commun (Camb) 2023; 59:14745-14758. [PMID: 37991846 PMCID: PMC10720954 DOI: 10.1039/d3cc05006h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Implantable neural interfaces (NIs) have emerged in the clinic as outstanding tools for the management of a variety of neurological conditions caused by trauma or disease. However, the foreign body reaction triggered upon implantation remains one of the major challenges hindering the safety and longevity of NIs. The integration of tools and principles from biomaterial design and tissue engineering has been investigated as a promising strategy to develop NIs with enhanced functionality and performance. In this Feature Article, we highlight the main bioengineering approaches for the development of biohybrid NIs with an emphasis on relevant device design criteria. Technical and scientific challenges associated with the fabrication and functional assessment of technologies composed of both artificial and biological components are discussed. Lastly, we provide future perspectives related to engineering, regulatory, and neuroethical challenges to be addressed towards the realisation of the promise of biohybrid neurotechnology.
Collapse
Affiliation(s)
- Marjolaine Boulingre
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Roberto Portillo-Lara
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Rylie A Green
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| |
Collapse
|
5
|
Abstract
Injectable bioprobes record single-neuron activity from within blood vessels.
Collapse
Affiliation(s)
- Brian P Timko
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
6
|
Zhang Q, Jing W, Wu S, Zhu M, Jiang J, Liu X, Yu D, Cheng L, Feng B, Wen J, Xiong F, Lu Y, Du H. Development of a synchronous recording and photo-stimulating electrode in multiple brain neurons. Front Neurosci 2023; 17:1195095. [PMID: 37383109 PMCID: PMC10293621 DOI: 10.3389/fnins.2023.1195095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/25/2023] [Indexed: 06/30/2023] Open
Abstract
The investigation of brain networks and neural circuits involves the crucial aspects of observing and modulating neurophysiological activity. Recently, opto-electrodes have emerged as an efficient tool for electrophysiological recording and optogenetic stimulation, which has greatly facilitated the analysis of neural coding. However, implantation and electrode weight control have posed significant challenges in achieving long-term and multi-regional brain recording and stimulation. To address this issue, we have developed a mold and custom-printed circuit board-based opto-electrode. We report successful opto-electrode placement and high-quality electrophysiological recordings from the default mode network (DMN) of the mouse brain. This novel opto-electrode facilitates synchronous recording and stimulation in multiple brain regions and holds promise for advancing future research on neural circuits and networks.
Collapse
Affiliation(s)
- Qingping Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Jing
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Shiping Wu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzheng Zhu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Jingrui Jiang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Liu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Dian Yu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Long Cheng
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Feng
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianbin Wen
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Xiong
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, National Center for Magnetic Resonance in Wuhan, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Huiyun Du
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Wuhan Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Wuhan, China
| |
Collapse
|
7
|
Mitchell P, Lee SCM, Yoo PE, Morokoff A, Sharma RP, Williams DL, MacIsaac C, Howard ME, Irving L, Vrljic I, Williams C, Bush S, Balabanski AH, Drummond KJ, Desmond P, Weber D, Denison T, Mathers S, O’Brien TJ, Mocco J, Grayden DB, Liebeskind DS, Opie NL, Oxley TJ, Campbell BCV. Assessment of Safety of a Fully Implanted Endovascular Brain-Computer Interface for Severe Paralysis in 4 Patients: The Stentrode With Thought-Controlled Digital Switch (SWITCH) Study. JAMA Neurol 2023; 80:270-278. [PMID: 36622685 PMCID: PMC9857731 DOI: 10.1001/jamaneurol.2022.4847] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/18/2022] [Indexed: 01/10/2023]
Abstract
Importance Brain-computer interface (BCI) implants have previously required craniotomy to deliver penetrating or surface electrodes to the brain. Whether a minimally invasive endovascular technique to deliver recording electrodes through the jugular vein to superior sagittal sinus is safe and feasible is unknown. Objective To assess the safety of an endovascular BCI and feasibility of using the system to control a computer by thought. Design, Setting, and Participants The Stentrode With Thought-Controlled Digital Switch (SWITCH) study, a single-center, prospective, first in-human study, evaluated 5 patients with severe bilateral upper-limb paralysis, with a follow-up of 12 months. From a referred sample, 4 patients with amyotrophic lateral sclerosis and 1 with primary lateral sclerosis met inclusion criteria and were enrolled in the study. Surgical procedures and follow-up visits were performed at the Royal Melbourne Hospital, Parkville, Australia. Training sessions were performed at patients' homes and at a university clinic. The study start date was May 27, 2019, and final follow-up was completed January 9, 2022. Interventions Recording devices were delivered via catheter and connected to subcutaneous electronic units. Devices communicated wirelessly to an external device for personal computer control. Main Outcomes and Measures The primary safety end point was device-related serious adverse events resulting in death or permanent increased disability. Secondary end points were blood vessel occlusion and device migration. Exploratory end points were signal fidelity and stability over 12 months, number of distinct commands created by neuronal activity, and use of system for digital device control. Results Of 4 patients included in analyses, all were male, and the mean (SD) age was 61 (17) years. Patients with preserved motor cortex activity and suitable venous anatomy were implanted. Each completed 12-month follow-up with no serious adverse events and no vessel occlusion or device migration. Mean (SD) signal bandwidth was 233 (16) Hz and was stable throughout study in all 4 patients (SD range across all sessions, 7-32 Hz). At least 5 attempted movement types were decoded offline, and each patient successfully controlled a computer with the BCI. Conclusions and Relevance Endovascular access to the sensorimotor cortex is an alternative to placing BCI electrodes in or on the dura by open-brain surgery. These final safety and feasibility data from the first in-human SWITCH study indicate that it is possible to record neural signals from a blood vessel. The favorable safety profile could promote wider and more rapid translation of BCI to people with paralysis. Trial Registration ClinicalTrials.gov Identifier: NCT03834857.
Collapse
Affiliation(s)
- Peter Mitchell
- Department of Radiology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Sarah C. M. Lee
- Neurology, Calvary Healthcare Bethlehem, Parkdale, Australia
| | | | - Andrew Morokoff
- Parkville Neurosurgery, The University of Melbourne, Royal Melbourne Hospital, Parkville, Australia
| | - Rahul P. Sharma
- Stanford Healthcare Cardiovascular Medicine, Stanford University, Stanford, California
| | - Daryl L. Williams
- Department of Anaesthesia and Pain Management, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Christopher MacIsaac
- Intensive Care Department, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Mark E. Howard
- Victorian Respiratory Support Service, Austin Health, Heidelberg, Australia
| | - Lou Irving
- Peter MacCallum Cancer Centre, The University of Melbourne, The Royal Melbourne Hospital, Melbourne, Australia
| | - Ivan Vrljic
- Department of Radiology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Cameron Williams
- Department of Neurology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Steven Bush
- Department of Neurology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Anna H. Balabanski
- Department of Neurology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
- Melbourne Brain Centre, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
- Department of Neuroscience, Alfred Brain, Alfred Health, Melbourne, Australia
| | - Katharine J. Drummond
- Department of Neurosurgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Patricia Desmond
- Department of Radiology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| | - Douglas Weber
- Department of Biomedical Engineering, College of Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Timothy Denison
- Institute of Biomedical Engineering, The University of Oxford, Oxford, United Kingdom
| | - Susan Mathers
- Neurology, Calvary Healthcare Bethlehem, Parkdale, Australia
| | - Terence J. O’Brien
- Department of Neurology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
- Department of Medicine, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
- Department of Neuroscience, The Central Clinical School, Monash University and Alfred Health, Melbourne, Australia
| | - J. Mocco
- Department of Neurosurgery, Klingenstein Clinical Center, The Mount Sinai Hospital, New York, New York
| | - David B. Grayden
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Parkville, Australia
| | - David S. Liebeskind
- UCLA Comprehensive Stroke Center, Department of Neurology, University of California, Los Angeles
| | - Nicholas L. Opie
- Vascular Bionics Laboratory, Department of Medicine, The University of Melbourne, Melbourne, Australia
- Synchron, Carlton, Australia
| | - Thomas J. Oxley
- Synchron Inc, New York, New York
- Vascular Bionics Laboratory, Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Bruce C. V. Campbell
- Department of Neurology, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
- Melbourne Brain Centre, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Australia
| |
Collapse
|
8
|
Zhao C, Man T, Cao Y, Weiss PS, Monbouquette HG, Andrews AM. Flexible and Implantable Polyimide Aptamer-Field-Effect Transistor Biosensors. ACS Sens 2022; 7:3644-3653. [PMID: 36399772 PMCID: PMC9982941 DOI: 10.1021/acssensors.2c01909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Monitoring neurochemical signaling across time scales is critical to understanding how brains encode and store information. Flexible (vs stiff) devices have been shown to improve in vivo monitoring, particularly over longer times, by reducing tissue damage and immunological responses. Here, we report our initial steps toward developing flexible and implantable neuroprobes with aptamer-field-effect transistor (FET) biosensors for neurotransmitter monitoring. A high-throughput process was developed to fabricate thin, flexible polyimide probes using microelectromechanical-system (MEMS) technologies, where 150 flexible probes were fabricated on each 4 in. Si wafer. Probes were 150 μm wide and 7 μm thick with two FETs per tip. The bending stiffness was 1.2 × 10-11 N·m2. Semiconductor thin films (3 nm In2O3) were functionalized with DNA aptamers for target recognition, which produces aptamer conformational rearrangements detected via changes in FET conductance. Flexible aptamer-FET neuroprobes detected serotonin at femtomolar concentrations in high-ionic strength artificial cerebrospinal fluid. A straightforward implantation process was developed, where microfabricated Si carrier devices assisted with implantation such that flexible neuroprobes detected physiological relevant serotonin in a tissue-hydrogel brain mimic.
Collapse
Affiliation(s)
- Chuanzhen Zhao
- 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
| | - Tianxing Man
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Yan Cao
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States,Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - 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,Departments of Bioengineering and Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Harold G. Monbouquette
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States,Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Anne M. Andrews
- 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 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,To whom correspondence should be addressed to:
| |
Collapse
|
9
|
Savya SP, Li F, Lam S, Wellman SM, Stieger KC, Chen K, Eles JR, Kozai TDY. In vivo spatiotemporal dynamics of astrocyte reactivity following neural electrode implantation. Biomaterials 2022; 289:121784. [PMID: 36103781 PMCID: PMC10231871 DOI: 10.1016/j.biomaterials.2022.121784] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/02/2022]
Abstract
Brain computer interfaces (BCIs), including penetrating microelectrode arrays, enable both recording and stimulation of neural cells. However, device implantation inevitably causes injury to brain tissue and induces a foreign body response, leading to reduced recording performance and stimulation efficacy. Astrocytes in the healthy brain play multiple roles including regulating energy metabolism, homeostatic balance, transmission of neural signals, and neurovascular coupling. Following an insult to the brain, they are activated and gather around the site of injury. These reactive astrocytes have been regarded as one of the main contributors to the formation of a glial scar which affects the performance of microelectrode arrays. This study investigates the dynamics of astrocytes within the first 2 weeks after implantation of an intracortical microelectrode into the mouse brain using two-photon microscopy. From our observation astrocytes are highly dynamic during this period, exhibiting patterns of process extension, soma migration, morphological activation, and device encapsulation that are spatiotemporally distinct from other glial cells, such as microglia or oligodendrocyte precursor cells. This detailed characterization of astrocyte reactivity will help to better understand the tissue response to intracortical devices and lead to the development of more effective intervention strategies to improve the functional performance of neural interfacing technology.
Collapse
Affiliation(s)
- Sajishnu P Savya
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Northwestern University, USA
| | - Fan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Computational Modeling & Simulation PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephanie Lam
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
10
|
Microelectrode implants, inflammatory response and long-lasting effects on NADPH diaphorase neurons in the rat frontal cortex. Exp Brain Res 2022; 240:2569-2580. [PMID: 35947168 DOI: 10.1007/s00221-022-06434-3] [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: 05/17/2022] [Accepted: 08/03/2022] [Indexed: 11/04/2022]
Abstract
At present, one of the main therapeutic challenges comprises the development of technologies to improve the life quality of people suffering from different types of body paralysis, through the reestablishment of sensory and motor functions. In this regard, brain-machine interfaces (BMI) offer hope to effectively mitigate body paralysis through the control of paralyzed body parts by brain activity. Invasive BMI use chronic multielectrode implants to record neural activity directly from the brain tissue. While such invasive devices provide the highest amount of usable neural activity for BMI control, they also involve direct damage to the nervous tissue. In the cerebral cortex, high levels of the enzyme NADPH diaphorase (NADPH-d) characterize a particular class of interneurons that regulates neuronal excitability and blood supply. To gain insight into the biocompatibility of invasive BMI, we assessed the impact of chronic implanted tungsten multielectrode bundles on the distribution and morphology of NADPH-d-reactive neurons in the rat frontal cortex. NADPH-d neuronal labeling was correlated with glial response markers and with indices of healthy neuronal activity measured by electrophysiological recordings performed up to 3 months after multielectrode implantation. Chronic electrode arrays caused a small and quite localized structural disturbance on the implanted site, with neuronal loss and glial activation circumscribed to the site of implant. Electrodes remained viable during the entire period of implantation. Moreover, neither the distribution nor the morphology of NADPH-d neurons was altered. Overall, our findings provide additional evidence that tungsten multielectrodes can be employed as a viable element for long-lasting therapeutic BMI applications.
Collapse
|
11
|
Darbin O, Hatanaka N, Takara S, Kaneko N, Chiken S, Naritoku D, Martino A, Nambu A. Subthalamic nucleus deep brain stimulation driven by primary motor cortex γ2 activity in parkinsonian monkeys. Sci Rep 2022; 12:6493. [PMID: 35444245 PMCID: PMC9021287 DOI: 10.1038/s41598-022-10130-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 03/31/2022] [Indexed: 11/17/2022] Open
Abstract
In parkinsonism, subthalamic nucleus (STN) electrical deep brain stimulation (DBS) improves symptoms, but may be associated with side effects. Adaptive DBS (aDBS), which enables modulation of stimulation, may limit side effects, but limited information is available about clinical effectiveness and efficaciousness. We developed a brain-machine interface for aDBS, which enables modulation of stimulation parameters of STN-DBS in response to γ2 band activity (80-200 Hz) of local field potentials (LFPs) recorded from the primary motor cortex (M1), and tested its effectiveness in parkinsonian monkeys. We trained two monkeys to perform an upper limb reaching task and rendered them parkinsonian with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Bipolar intracortical recording electrodes were implanted in the M1, and a recording chamber was attached to access the STN. In aDBS, the M1 LFPs were recorded, filtered into the γ2 band, and discretized into logic pulses by a window discriminator, and the pulses were used to modulate the interval and amplitude of DBS pulses. In constant DBS (cDBS), constant stimulus intervals and amplitudes were used. Reaction and movement times during the task were measured and compared between aDBS and cDBS. The M1-γ2 activities were increased before and during movements in parkinsonian monkeys and these activities modulated the aDBS pulse interval, amplitude, and dispersion. With aDBS and cDBS, reaction and movement times were significantly decreased in comparison to DBS-OFF. The electric charge delivered was lower with aDBS than cDBS. M1-γ2 aDBS in parkinsonian monkeys resulted in clinical benefits that did not exceed those from cDBS. However, M1-γ2 aDBS achieved this magnitude of benefit for only two thirds of the charge delivered by cDBS. In conclusion, M1-γ2 aDBS is an effective therapeutic approach which requires a lower electrical charge delivery than cDBS for comparable clinical benefits.
Collapse
Affiliation(s)
- Olivier Darbin
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan. .,Department of Neurology, University South Alabama College of Medicine, 307 University Blvd, Mobile, AL, 36688, USA.
| | - Nobuhiko Hatanaka
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Sayuki Takara
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan.,Department of Physiology, Faculty of Medecine, Kindai University, Osaka-Sayama, Osaka, Japan
| | - Nobuya Kaneko
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Satomi Chiken
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan.,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan
| | - Dean Naritoku
- Department of Neurology, University South Alabama College of Medicine, 307 University Blvd, Mobile, AL, 36688, USA
| | - Anthony Martino
- Department of Neurosurgery, University South Alabama College of Medicine, Mobile, AL, USA
| | - Atsushi Nambu
- Division of System Neurophysiology, National Institute for Physiological Sciences, Okazaki, Aichi, Japan. .,Department of Physiological Sciences, SOKENDAI (Graduate University for Advanced Studies), Okazaki, Aichi, Japan.
| |
Collapse
|
12
|
Sturgill B, Radhakrishna R, Thai TTD, Patnaik SS, Capadona JR, Pancrazio JJ. Characterization of Active Electrode Yield for Intracortical Arrays: Awake versus Anesthesia. MICROMACHINES 2022; 13:mi13030480. [PMID: 35334770 PMCID: PMC8955818 DOI: 10.3390/mi13030480] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/04/2022]
Abstract
Intracortical microelectrode arrays are used for recording neural signals at single-unit resolution and are promising tools for studying brain function and developing neuroprosthetics. Research is being done to increase the chronic performance and reliability of these probes, which tend to decrease or fail within several months of implantation. Although recording paradigms vary, studies focused on assessing the reliability and performance of these devices often perform recordings under anesthesia. However, anesthetics—such as isoflurane—are known to alter neural activity and electrophysiologic function. Therefore, we compared the neural recording performance under anesthesia (2% isoflurane) followed by awake conditions for probes implanted in the motor cortex of both male and female Sprague-Dawley rats. While the single-unit spike rate was significantly higher by almost 600% under awake compared to anesthetized conditions, we found no difference in the active electrode yield between the two conditions two weeks after surgery. Additionally, the signal-to-noise ratio was greater under anesthesia due to the noise levels being nearly 50% greater in awake recordings, even though there was a 14% increase in the peak-to-peak voltage of distinguished single units when awake. We observe that these findings are similar for chronic time points as well. Our observations indicate that either anesthetized or awake recordings are acceptable for studies assessing the chronic reliability and performance of intracortical microelectrode arrays.
Collapse
Affiliation(s)
- Brandon Sturgill
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Rahul Radhakrishna
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Teresa Thuc Doan Thai
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Sourav S. Patnaik
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA;
| | - Joseph J. Pancrazio
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA; (B.S.); (R.R.); (T.T.D.T.); (S.S.P.)
- Correspondence:
| |
Collapse
|
13
|
Kunigk NG, Urdaneta ME, Malone IG, Delgado F, Otto KJ. Reducing Behavioral Detection Thresholds per Electrode via Synchronous, Spatially-Dependent Intracortical Microstimulation. Front Neurosci 2022; 16:876142. [PMID: 35784835 PMCID: PMC9247280 DOI: 10.3389/fnins.2022.876142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/31/2022] [Indexed: 12/04/2022] Open
Abstract
Intracortical microstimulation (ICMS) has shown promise in restoring quality of life to patients suffering from paralysis, specifically when used in the primary somatosensory cortex (S1). However, these benefits can be hampered by long-term degradation of electrode performance due to the brain's foreign body response. Advances in microfabrication techniques have allowed for the development of neuroprostheses with subcellular electrodes, which are characterized by greater versatility and a less detrimental immune response during chronic use. These probes are hypothesized to enable more selective, higher-resolution stimulation of cortical tissue with long-term implants. However, microstimulation using physiologically relevant charges with these smaller-scale devices can damage electrode sites and reduce the efficacy of the overall device. Studies have shown promise in bypassing this limitation by spreading the stimulation charge between multiple channels in an implanted electrode array, but to our knowledge the usefulness of this strategy in laminar arrays with electrode sites spanning each layer of the cortex remains unexplored. To investigate the efficacy of simultaneous multi-channel ICMS in electrode arrays with stimulation sites spanning cortical depth, we implanted laminar electrode arrays in the primary somatosensory cortex of rats trained in a behavioral avoidance paradigm. By measuring detection thresholds, we were able to quantify improvements in ICMS performance using a simultaneous multi-channel stimulation paradigm. The charge required per site to elicit detection thresholds was halved when stimulating from two adjacent electrode sites, although the overall charge used by the implant was increased. This reduction in threshold charge was more pronounced when stimulating with more than two channels and lessened with greater distance between stimulating channels. Our findings suggest that these improvements are based on the synchronicity and polarity of each stimulus, leading us to conclude that these improvements in stimulation efficiency per electrode are due to charge summation as opposed to a summation of neural responses to stimulation. Additionally, the per-site charge reductions are seen regardless of the cortical depth of each utilized channel. This evocation of physiological detection thresholds with lower stimulation currents per electrode site has implications for the feasibility of stimulation regimes in future advanced neuroprosthetic devices, which could benefit from reducing the charge output per site.
Collapse
Affiliation(s)
- Nicolas G. Kunigk
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Morgan E. Urdaneta
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Ian G. Malone
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, United States
| | - Francisco Delgado
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Kevin J. Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, United States
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, United States
- Department of Neurology, University of Florida, Gainesville, FL, United States
- McKnight Brain Institute, University of Florida, Gainesville, FL, United States
- *Correspondence: Kevin J. Otto,
| |
Collapse
|
14
|
Jang JW, Kang YN, Seo HW, Kim B, Choe HK, Park SH, Lee MG, Kim S. Long-term in-vivorecording performance of flexible penetrating microelectrode arrays. J Neural Eng 2021; 18. [PMID: 34795067 DOI: 10.1088/1741-2552/ac3656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/02/2021] [Indexed: 11/12/2022]
Abstract
Objective. Neural interfaces are an essential tool to enable the human body to directly communicate with machines such as computers or prosthetic robotic arms. Since invasive electrodes can be located closer to target neurons, they have advantages such as precision in stimulation and high signal-to-noise ratio (SNR) in recording, while they often exhibit unstable performance in long-termin-vivoimplantation because of the tissue damage caused by the electrodes insertion. In the present study, we investigated the electrical functionality of flexible penetrating microelectrode arrays (FPMAs) up to 3 months inin-vivoconditions.Approach. Thein-vivoexperiment was performed by implanting FPMAs in five rats. Thein-vivoimpedance as well as the action potential (AP) amplitude and SNR were analyzed over weeks. Additionally, APs were tracked over time to investigate the possibility of single neuron recording.Main results. It was observed that the FPMAs exhibited dramatic increases in impedance for the first 4 weeks after implantation, accompanied by decreases in AP amplitude. However, the increase/decrease in AP amplitude was always accompanied by the increase/decrease in background noise, resulting in quite consistently maintained SNRs. After 4 weeks of implantation, we observed two distinctive issues regarding long-term implantation, each caused by chronic tissue responses or by the delamination of insulation layer. The results demonstrate that the FPMAs successfully recorded neuronal signals up to 12 weeks, with very stably maintained SNRs, reduced by only 16.1% on average compared to the first recordings, although biological tissue reactions or physical degradation of the FPMA were present.Significance. The fabricated FPMAs successfully recorded intracortical signals for 3 months. The SNR was maintained up to 3 months and the chronic function of FPMA was comparable with other silicon based implantable electrodes.
Collapse
Affiliation(s)
- Jae-Won Jang
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Yoo Na Kang
- Department of Medical Assistant Robot, Korea Institute of Machinery and Materials (KIMM), Daegu, Republic of Korea
| | - Hee Won Seo
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Boil Kim
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Han Kyoung Choe
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Sang Hyun Park
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Maan-Gee Lee
- Department of Pharmacology, School of MedicineKyungpook National University, Daegu, Republic of Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Sohee Kim
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| |
Collapse
|
15
|
Cajigas I, Davis KC, Meschede-Krasa B, Prins NW, Gallo S, Naeem JA, Palermo A, Wilson A, Guerra S, Parks BA, Zimmerman L, Gant K, Levi AD, Dietrich WD, Fisher L, Vanni S, Tauber JM, Garwood IC, Abel JH, Brown EN, Ivan ME, Prasad A, Jagid J. Implantable brain-computer interface for neuroprosthetic-enabled volitional hand grasp restoration in spinal cord injury. Brain Commun 2021; 3:fcab248. [PMID: 34870202 PMCID: PMC8637800 DOI: 10.1093/braincomms/fcab248] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/27/2021] [Accepted: 08/19/2021] [Indexed: 11/12/2022] Open
Abstract
Loss of hand function after cervical spinal cord injury severely impairs functional independence. We describe a method for restoring volitional control of hand grasp in one 21-year-old male subject with complete cervical quadriplegia (C5 American Spinal Injury Association Impairment Scale A) using a portable fully implanted brain-computer interface within the home environment. The brain-computer interface consists of subdural surface electrodes placed over the dominant-hand motor cortex and connects to a transmitter implanted subcutaneously below the clavicle, which allows continuous reading of the electrocorticographic activity. Movement-intent was used to trigger functional electrical stimulation of the dominant hand during an initial 29-weeks laboratory study and subsequently via a mechanical hand orthosis during in-home use. Movement-intent information could be decoded consistently throughout the 29-weeks in-laboratory study with a mean accuracy of 89.0% (range 78-93.3%). Improvements were observed in both the speed and accuracy of various upper extremity tasks, including lifting small objects and transferring objects to specific targets. At-home decoding accuracy during open-loop trials reached an accuracy of 91.3% (range 80-98.95%) and an accuracy of 88.3% (range 77.6-95.5%) during closed-loop trials. Importantly, the temporal stability of both the functional outcomes and decoder metrics were not explored in this study. A fully implanted brain-computer interface can be safely used to reliably decode movement-intent from motor cortex, allowing for accurate volitional control of hand grasp.
Collapse
Affiliation(s)
- Iahn Cajigas
- Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA
| | - Kevin C Davis
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Benyamin Meschede-Krasa
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Noeline W Prins
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
- Department of Electrical and Information Engineering, Faculty of Engineering, University of Ruhuna, Hapugala, Galle 80000, Sri Lanka
| | - Sebastian Gallo
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Jasim Ahmad Naeem
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Anne Palermo
- Department of Physical Therapy, University of Miami, Miami, FL 33146, USA
| | - Audrey Wilson
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Santiago Guerra
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Brandon A Parks
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Lauren Zimmerman
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Katie Gant
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Allan D Levi
- Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Letitia Fisher
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Steven Vanni
- Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - John Michael Tauber
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Indie C Garwood
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John H Abel
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Emery N Brown
- Department of Brain and Cognitive Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael E Ivan
- Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Jonathan Jagid
- Department of Neurological Surgery, University of Miami, Miami, FL 33136, USA
- Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
16
|
Hao Z, Wang S, Zhang K, Zhou J, Li D, He J, Gao L, Wang L. Biofabrication of a Low Modulus Bioelectroprobe for Neurons to Grow Into. MATERIALS 2021; 14:ma14164718. [PMID: 34443240 PMCID: PMC8400188 DOI: 10.3390/ma14164718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/29/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
Implantable nerve electrodes, as a bridge between the brain and external devices, have been widely used in areas such as brain function exploration, neurological disease treatment and human–computer interaction. However, the mechanical properties mismatch between the electrode material and the brain tissue seriously affects the stability of electrode signal acquisition and the effectiveness of long-term service in vivo. In this study, a modified neuroelectrode was developed with conductive biomaterials. The electrode has good biocompatibility and a gradient microstructure suitable for cell growth. Compared with metal electrodes, bioelectrodes not only greatly reduced the elastic modulus (<10 kpa) but also increased the conductivity of the electrode by 200 times. Through acute electrophysiological analysis and a 12-week chronic in vivo experiment, the bioelectrode clearly recorded the rat’s brain electrical signals, effectively avoided the generation of glial scars and induced neurons to move closer to the electrode. The new conductive biomaterial electrodes developed in this research make long-term implantation of cortical nerve electrodes possible.
Collapse
Affiliation(s)
- Zhiyan Hao
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
| | - Sen Wang
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi’an 710032, China;
| | - Jiajia Zhou
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
| | - Dichen Li
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jiankang He
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
| | - Lin Gao
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ling Wang
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an 710054, China; (Z.H.); (S.W.); (J.Z.); (D.L.); (J.H.); (L.G.)
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: ; Tel.: +86-29-8339-5382
| |
Collapse
|
17
|
Sridharan A, Muthuswamy J. Soft, Conductive, Brain-Like, Coatings at Tips of Microelectrodes Improve Electrical Stability under Chronic, In Vivo Conditions. MICROMACHINES 2021; 12:761. [PMID: 34203234 PMCID: PMC8306035 DOI: 10.3390/mi12070761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 01/27/2023]
Abstract
Several recent studies have reported improved histological and electrophysiological outcomes with soft neural interfaces that have elastic moduli ranging from 10 s of kPa to hundreds of MPa. However, many of these soft interfaces use custom fabrication processes. We test the hypothesis that a readily adoptable fabrication process for only coating the tips of microelectrodes with soft brain-like (elastic modulus of ~5 kPa) material improves the long-term electrical performance of neural interfaces. Conventional tungsten microelectrodes (n = 9 with soft coatings and n = 6 uncoated controls) and Pt/Ir microelectrodes (n = 16 with soft coatings) were implanted in six animals for durations ranging from 5 weeks to over 1 year in a subset of rats. Electrochemical impedance spectroscopy was used to assess the quality of the brain tissue-electrode interface under chronic conditions. Neural recordings were assessed for unit activity and signal quality. Electrodes with soft, silicone coatings showed relatively stable electrical impedance characteristics over 6 weeks to >1 year compared to the uncoated control electrodes. Single unit activity recorded by coated electrodes showed larger peak-to-peak amplitudes and increased number of detectable neurons compared to uncoated controls over 6-7 weeks. We demonstrate the feasibility of using a readily translatable process to create brain-like soft interfaces that can potentially overcome variable performance associated with chronic rigid neural interfaces.
Collapse
Affiliation(s)
| | - Jit Muthuswamy
- School of Biological and Health Systems Engineering, Ira A. Fulton School of Engineering, Arizona State University, Tempe, AZ 85287-9709, USA;
| |
Collapse
|
18
|
Cajigas I, Vedantam A. Brain-Computer Interface, Neuromodulation, and Neurorehabilitation Strategies for Spinal Cord Injury. Neurosurg Clin N Am 2021; 32:407-417. [PMID: 34053728 DOI: 10.1016/j.nec.2021.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
As neural bypass interfacing, neuromodulation, and neurorehabilitation continue to evolve, there is growing recognition that combination therapies may achieve superior results. This article briefly introduces these broad areas of active research and lays out some of the current evidence for their use for patients with spinal cord injury.
Collapse
Affiliation(s)
- Iahn Cajigas
- Department of Neurosurgery, University of Miami, 1095 Northwest 14th Terrace (D4-6), Miami, FL 33136, USA.
| | - Aditya Vedantam
- Department of Neurosurgery, University of Miami, 1095 Northwest 14th Terrace (D4-6), Miami, FL 33136, USA
| |
Collapse
|
19
|
Redolfi Riva E, Micera S. Progress and challenges of implantable neural interfaces based on nature-derived materials. Bioelectron Med 2021; 7:6. [PMID: 33902750 PMCID: PMC8077843 DOI: 10.1186/s42234-021-00067-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/31/2021] [Indexed: 12/31/2022] Open
Abstract
Neural interfaces are bioelectronic devices capable of stimulating a population of neurons or nerve fascicles and recording electrical signals in a specific area. Despite their success in restoring sensory-motor functions in people with disabilities, their long-term exploitation is still limited by poor biocompatibility, mechanical mismatch between the device and neural tissue and the risk of a chronic inflammatory response upon implantation.In this context, the use of nature-derived materials can help address these issues. Examples of these materials, such as extracellular matrix proteins, peptides, lipids and polysaccharides, have been employed for decades in biomedical science. Their excellent biocompatibility, biodegradability in the absence of toxic compound release, physiochemical properties that are similar to those of human tissues and reduced immunogenicity make them outstanding candidates to improve neural interface biocompatibility and long-term implantation safety. The objective of this review is to highlight progress and challenges concerning the impact of nature-derived materials on neural interface design. The use of these materials as biocompatible coatings and as building blocks of insulation materials for use in implantable neural interfaces is discussed. Moreover, future perspectives are presented to show the increasingly important uses of these materials for neural interface fabrication and their possible use for other applications in the framework of neural engineering.
Collapse
Affiliation(s)
- Eugenio Redolfi Riva
- The BioRobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy.
| | - Silvestro Micera
- The BioRobotics Institute and Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Pisa, Italy
- Bertarelli Foundation Chair in Translational Neuroengineering, Centre for Neuroprosthetics and Institute of Bioengineering, School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
20
|
Alvarez NT, Buschbeck E, Miller S, Le AD, Gupta VK, Ruhunage C, Vilinsky I, Ma Y. Carbon Nanotube Fibers for Neural Recording and Stimulation. ACS APPLIED BIO MATERIALS 2020; 3:6478-6487. [PMID: 35021779 DOI: 10.1021/acsabm.0c00861] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recordings and stimulations of neuronal electrical activity are topics of great interest in neuroscience. Many recording techniques, and even treatment of neurological disorders, can benefit from a microelectrode that is flexible, chemically inert, and electrically conducting and preferentially transfers electrons via capacitive charge injection. Commercial electrodes that currently exist and other electrodes that are being tested with the purpose of facilitating and improving the electron transport between solid materials and biological tissues still have some limitations. This paper discusses carbon nanotube (CNT)-based microelectrodes to record and stimulate neurons and compares their electron transport capabilities to noble metals such as Au and Ag. The recording ability of electrodes is tested through electroretinography on Sarcophaga bullata fly eyes by using Au and Ag wires and CNT fibers as electrodes. Stimulation is demonstrated through the implantation of Au wire and CNT fibers into the antennas of the Madagascar hissing cockroach (Gromphadorhina portentosa) to control their locomotion. Our results demonstrate that a particular property of the CNT fiber is its high rate of electron transfer, leading to an order of magnitude lower impedance compared to Au and Ag and an impressive 15.09 charge injection capacity. We also established that this carbon nanomaterial assembly performs well for in vivo electrophysiology, rendering it a promising prospect for neurophysiological applications.
Collapse
Affiliation(s)
- Noe T Alvarez
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Elke Buschbeck
- Department of Biology, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Sydney Miller
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Anh Duc Le
- Department of Biology, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Vandna K Gupta
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Chethani Ruhunage
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Ilya Vilinsky
- Department of Biology, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Yishan Ma
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221, United States
| |
Collapse
|
21
|
Apollo NV, Murphy B, Prezelski K, Driscoll N, Richardson AG, Lucas TH, Vitale F. Gels, jets, mosquitoes, and magnets: a review of implantation strategies for soft neural probes. J Neural Eng 2020; 17:041002. [PMID: 32759476 PMCID: PMC8152109 DOI: 10.1088/1741-2552/abacd7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Implantable neuroelectronic interfaces have enabled breakthrough advances in the clinical diagnosis and treatment of neurological disorders, as well as in fundamental studies of brain function, behavior, and disease. Intracranial electroencephalography (EEG) mapping with stereo-EEG (sEEG) depth electrodes is routinely adopted for precise epilepsy diagnostics and surgical treatment, while deep brain stimulation has become the standard of care for managing movement disorders. Intracortical microelectrode arrays for high-fidelity recordings of neural spiking activity have led to impressive demonstrations of the power of brain-machine interfaces for motor and sensory functional recovery. Yet, despite the rapid pace of technology development, the issue of establishing a safe, long-term, stable, and functional interface between neuroelectronic devices and the host brain tissue still remains largely unresolved. A body of work spanning at least the last 15 years suggests that safe, chronic integration between invasive electrodes and the brain requires a close match between the mechanical properties of man-made components and the neural tissue. In other words, the next generation of invasive electrodes should be soft and compliant, without sacrificing biological and chemical stability. Soft neuroelectronic interfaces, however, pose a new and significant surgical challenge: bending and buckling during implantation that can preclude accurate and safe device placement. In this topical review, we describe the next generation of soft electrodes and the surgical implantation methods for safe and precise insertion into brain structures. We provide an overview of the most recent innovations in the field of insertion strategies for flexible neural electrodes such as dissolvable or biodegradable carriers, microactuators, biologically-inspired support structures, and electromagnetic drives. In our analysis, we also highlight approaches developed in different fields, such as robotic surgery, which could be potentially adapted and translated to the insertion of flexible neural probes.
Collapse
Affiliation(s)
- Nicholas V Apollo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, 19104, United States of America
| | - Brendan Murphy
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, 19104, United States of America
- These authors contributed equally
| | - Kayla Prezelski
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, 19104, United States of America
- These authors contributed equally
| | - Nicolette Driscoll
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, 19104, United States of America
| | - Andrew G Richardson
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
| | - Timothy H Lucas
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
| | - Flavia Vitale
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Center for Neurotrauma, Neurodegeneration, and Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, 19104, United States of America
- These authors contributed equally
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States of America
- Department of Physical Medicine & Rehabilitation, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, United States of America
| |
Collapse
|
22
|
Harris AR. Current perspectives on the safe electrical stimulation of peripheral nerves with platinum electrodes. ACTA ACUST UNITED AC 2020. [DOI: 10.2217/bem-2020-0007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review details some peripheral nervous system (PNS) targets and electrode designs used for electrical stimulation. It investigates limitations in current knowledge of safe electrical stimulation and possible future electrode developments. Current PNS targets are large, leading to poor resolution and off-target side-effects. Most clinical devices are platinum or platinum/iridium embedded in an insulation material. Their safety is usually guided by the Shannon plot, which is not valid for the PNS. New electrode designs are needed to target smaller nerve fibers, enabling higher resolution electrical therapies with fewer off-target side-effects. Damage can occur through biological and electrochemical mechanisms. Greater mechanistic understanding is required to ensure safe and efficacious, long-term electrical stimulation with new electrode materials, geometries and stimulation waveforms.
Collapse
Affiliation(s)
- Alexander R Harris
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, University of Wollongong, NSW 2522, Australia
| |
Collapse
|
23
|
Liu Q, Zhao C, Chen M, Liu Y, Zhao Z, Wu F, Li Z, Weiss PS, Andrews AM, Zhou C. Flexible Multiplexed In 2O 3 Nanoribbon Aptamer-Field-Effect Transistors for Biosensing. iScience 2020; 23:101469. [PMID: 33083757 PMCID: PMC7509003 DOI: 10.1016/j.isci.2020.101469] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/13/2020] [Indexed: 11/05/2022] Open
Abstract
Flexible sensors are essential for advancing implantable and wearable bioelectronics toward monitoring chemical signals within and on the body. Developing biosensors for monitoring multiple neurotransmitters in real time represents a key in vivo application that will increase understanding of information encoded in brain neurochemical fluxes. Here, arrays of devices having multiple In2O3 nanoribbon field-effect transistors (FETs) were fabricated on 1.4-μm-thick polyethylene terephthalate (PET) substrates using shadow mask patterning techniques. Thin PET-FET devices withstood crumpling and bending such that stable transistor performance with high mobility was maintained over >100 bending cycles. Real-time detection of the small-molecule neurotransmitters serotonin and dopamine was achieved by immobilizing recently identified high-affinity nucleic-acid aptamers on individual In2O3 nanoribbon devices. Limits of detection were 10 fM for serotonin and dopamine with detection ranges spanning eight orders of magnitude. Simultaneous sensing of temperature, pH, serotonin, and dopamine enabled integration of physiological and neurochemical data from individual bioelectronic devices. We fabricated flexible In2O3 nanoribbon transistors using cleanroom-free processes Flexible In2O3 transistors withstood crumpling and bending with stable performance Flexible aptamer biosensors detect neurotransmitters in real time Multiplexed sensors monitor temperature, pH, serotonin, and dopamine simultaneously
Collapse
Affiliation(s)
- Qingzhou Liu
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Chuanzhen Zhao
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mingrui Chen
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Yihang Liu
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhiyuan Zhao
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Fanqi Wu
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhen Li
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul S Weiss
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Departments of Bioengineering and Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Anne M Andrews
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chongwu Zhou
- Ming Hsieh Department of Electrical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
24
|
Liu C, Nguyen MA, Alvarez-Ciara A, Franklin M, Bennett C, Domena JB, Kleinhenz NC, Blanco Colmenares GA, Duque S, Chebbi AF, Bernard B, Olivier JH, Prasad A. Surface Modifications of an Organic Polymer-Based Microwire Platform for Sustained Release of an Anti-Inflammatory Drug. ACS APPLIED BIO MATERIALS 2020; 3:4613-4625. [DOI: 10.1021/acsabm.0c00506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Chuan Liu
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Michelle A. Nguyen
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Anabel Alvarez-Ciara
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Melissa Franklin
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Justin B. Domena
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Noah C. Kleinhenz
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Gabriel A. Blanco Colmenares
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Sebastian Duque
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Aisha F. Chebbi
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Brianna Bernard
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Jean-Hubert Olivier
- Department of Chemistry, University of Miami, Cox Science Center, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
- The Miami Project to Cure Paralysis, University of Miami, Miami, Florida 33136, United States
| |
Collapse
|
25
|
Khan MS, Kumar R, Manno SH, Ahmed I, Lun Law AW, Cruces RR, Ma V, Cho WC, Cheng SH, Lau C. Glymphatic clearance of simulated silicon dispersion in mouse brain analyzed by laser induced breakdown spectroscopy. Heliyon 2020; 6:e03702. [PMID: 32322711 PMCID: PMC7168738 DOI: 10.1016/j.heliyon.2020.e03702] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/29/2019] [Accepted: 03/26/2020] [Indexed: 11/20/2022] Open
Abstract
Silicon-based devices, such as neural probes, are increasingly used as electrodes for receiving electrical signals from neural tissue. Neural probes used chronically have been known to induce inflammation and elicit an immune response. The current study detects and evaluates silicon dispersion from a concentrated source in the mouse brain using laser induced breakdown spectroscopy. Element lines for Si (I) were found at the injection site at approximately 288 nm at 3hr post-implantation, even with tissue perfusion, indicating possible infusion into neural tissue. At 24hr and 1-week post-implantation, no silicon lines were found, indicating clearance. An isolated immune response was found by CD68 macrophage response at 24hr post injection. Future studies should measure chronic silicon exposure to determine if the inflammatory response is proportional to silicon administration. The present type of protocol, coupling laser induced breakdown spectroscopy, neuroimaging, histology, immunohistochemistry, and determination of clearance could be used to investigate the glymphatic system and different tissue states such as in disease (e.g. Alzheimer's).
Collapse
Affiliation(s)
| | - Rachit Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sinai H.C. Manno
- Department of Physics, City University of Hong Kong, Kowloon, HKSAR, China
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, HKSAR, China
| | - Irfan Ahmed
- Electrical Engineering Department, Sukkur IBA University, Sukkur 65200, Sindh, Pakistan
| | - Alan Wing Lun Law
- Department of Physics, City University of Hong Kong, Kowloon, HKSAR, China
| | - Raul R. Cruces
- McConnell Brain Imaging Center, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Victor Ma
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, HKSAR, China
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, HKSAR, China
| | - Shuk Han Cheng
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, HKSAR, China
- State Key Laboratory of Marine Pollution (SKLMP), City University of Hong Kong, Kowloon, HKSAR, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, HKSAR, China
| | - Condon Lau
- Department of Physics, City University of Hong Kong, Kowloon, HKSAR, China
- Corresponding author.
| |
Collapse
|
26
|
Zanos S. Closed-Loop Neuromodulation in Physiological and Translational Research. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034314. [PMID: 30559253 DOI: 10.1101/cshperspect.a034314] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Neuromodulation, the focused delivery of energy to neural tissue to affect neural or physiological processes, is a common method to study the physiology of the nervous system. It is also successfully used as treatment for disorders in which the nervous system is affected or implicated. Typically, neurostimulation is delivered in open-loop mode (i.e., according to a predetermined schedule and independently of the state of the organ or physiological system whose function is sought to be modulated). However, the physiology of the nervous system or the modulated organ can be dynamic, and the same stimulus may have different effects depending on the underlying state. As a result, open-loop stimulation may fail to restore the desired function or cause side effects. In such cases, a neuromodulation intervention may be preferable to be administered in closed-loop mode. In a closed-loop neuromodulation (CLN) system, stimulation is delivered when certain physiological states or conditions are met (responsive neurostimulation); the stimulation parameters can also be adjusted dynamically to optimize the effect of stimulation in real time (adaptive neurostimulation). In this review, the reasons and the conditions for using CLN are discussed, the basic components of a CLN system are described, and examples of CLN systems used in physiological and translational research are presented.
Collapse
Affiliation(s)
- Stavros Zanos
- Translational Neurophysiology Laboratory, Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York 11030
| |
Collapse
|
27
|
Xu K, Li S, Dong S, Zhang S, Pan G, Wang G, Shi L, Guo W, Yu C, Luo J. Bioresorbable Electrode Array for Electrophysiological and Pressure Signal Recording in the Brain. Adv Healthc Mater 2019; 8:e1801649. [PMID: 31168937 DOI: 10.1002/adhm.201801649] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/29/2019] [Indexed: 01/29/2023]
Abstract
Medical implantation of an electrocorticography (ECoG) recording system for brain monitoring is an effective clinical tool for seizure focus location and brain disease diagnosis. Planar and flexible ECoG electrodes can minimize the risks of infection and serious inflammatory response, and their good shape adaptability allows the device to fit complex cortex shape and structure to record brain signals with high spatial and temporal resolution. However, these ECoG electrodes require an additional surgery to remove the implant, which imposes potential medical risks. Here, a novel flexible and bioresorbable ECoG device integrated with an intracortical pressure sensor for monitoring swelling of the cortex during operation is reported. The ECoG device is fabricated with poly(l-lactide) and polycaprolactone composite and transient metal molybdenum. In vivo tests on rats show that the ECoG system can record the dynamic changes in brain signals for the different epilepsy stages with high resolution, while the malleable pressure sensor shows a linear relationship between the pressure and resistance in in vitro tests. In vitro degradation experiments show that the ECoG system can work stably for about five days before loss of efficacy, and the whole ECoG system degrades completely in a phosphate buffer solution in about 100 days.
Collapse
Affiliation(s)
- Kedi Xu
- Key Laboratory of Biomedical Engineering of Education MinistryQiushi Academy for Advanced StudiesDepartment of Biomedical EngineeringZhejiang University Hangzhou 310027 China
| | - Shijian Li
- College of Computer ScienceZhejiang University Hangzhou 310027 China
| | - Shurong Dong
- College of Information Science and Electronic EngineeringZhejiang University Hangzhou 310027 China
| | - Shaomin Zhang
- Key Laboratory of Biomedical Engineering of Education MinistryQiushi Academy for Advanced StudiesDepartment of Biomedical EngineeringZhejiang University Hangzhou 310027 China
| | - Gang Pan
- College of Computer ScienceZhejiang University Hangzhou 310027 China
| | - Guangming Wang
- College of Information Science and Electronic EngineeringZhejiang University Hangzhou 310027 China
| | - Lin Shi
- College of Information Science and Electronic EngineeringZhejiang University Hangzhou 310027 China
| | - Wei Guo
- College of Information Science and Electronic EngineeringZhejiang University Hangzhou 310027 China
| | - Chaonan Yu
- Key Laboratory of Biomedical Engineering of Education MinistryQiushi Academy for Advanced StudiesDepartment of Biomedical EngineeringZhejiang University Hangzhou 310027 China
| | - Jikui Luo
- Institute of Renewable Energy and Environmental TechnologyBolton University Deane Road Bolton BL3 5AB UK
| |
Collapse
|
28
|
Beygi M, Bentley JT, Frewin CL, Kuliasha CA, Takshi A, Bernardin EK, La Via F, Saddow SE. Fabrication of a Monolithic Implantable Neural Interface from Cubic Silicon Carbide. MICROMACHINES 2019; 10:E430. [PMID: 31261887 PMCID: PMC6680876 DOI: 10.3390/mi10070430] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/18/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023]
Abstract
One of the main issues with micron-sized intracortical neural interfaces (INIs) is their long-term reliability, with one major factor stemming from the material failure caused by the heterogeneous integration of multiple materials used to realize the implant. Single crystalline cubic silicon carbide (3C-SiC) is a semiconductor material that has been long recognized for its mechanical robustness and chemical inertness. It has the benefit of demonstrated biocompatibility, which makes it a promising candidate for chronically-stable, implantable INIs. Here, we report on the fabrication and initial electrochemical characterization of a nearly monolithic, Michigan-style 3C-SiC microelectrode array (MEA) probe. The probe consists of a single 5 mm-long shank with 16 electrode sites. An ~8 µm-thick p-type 3C-SiC epilayer was grown on a silicon-on-insulator (SOI) wafer, which was followed by a ~2 µm-thick epilayer of heavily n-type (n+) 3C-SiC in order to form conductive traces and the electrode sites. Diodes formed between the p and n+ layers provided substrate isolation between the channels. A thin layer of amorphous silicon carbide (a-SiC) was deposited via plasma-enhanced chemical vapor deposition (PECVD) to insulate the surface of the probe from the external environment. Forming the probes on a SOI wafer supported the ease of probe removal from the handle wafer by simple immersion in HF, thus aiding in the manufacturability of the probes. Free-standing probes and planar single-ended test microelectrodes were fabricated from the same 3C-SiC epiwafers. Cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS) were performed on test microelectrodes with an area of 491 µm2 in phosphate buffered saline (PBS) solution. The measurements showed an impedance magnitude of 165 kΩ ± 14.7 kΩ (mean ± standard deviation) at 1 kHz, anodic charge storage capacity (CSC) of 15.4 ± 1.46 mC/cm2, and a cathodic CSC of 15.2 ± 1.03 mC/cm2. Current-voltage tests were conducted to characterize the p-n diode, n-p-n junction isolation, and leakage currents. The turn-on voltage was determined to be on the order of ~1.4 V and the leakage current was less than 8 μArms. This all-SiC neural probe realizes nearly monolithic integration of device components to provide a likely neurocompatible INI that should mitigate long-term reliability issues associated with chronic implantation.
Collapse
Affiliation(s)
- Mohammad Beygi
- Department of Electrical Engineering, University of South Florida, Tampa, FL 33620, USA
| | - John T Bentley
- Department of Medical Engineering, University of South Florida, Tampa, FL 33620, USA
| | | | - Cary A Kuliasha
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Arash Takshi
- Department of Electrical Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Evans K Bernardin
- Department of Medical Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Francesco La Via
- CNR Institute for Microelectronics and Microsystems, Catania, Sicily 95121, Italy
| | - Stephen E Saddow
- Department of Electrical Engineering, University of South Florida, Tampa, FL 33620, USA.
- Department of Medical Engineering, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
29
|
Wang K, Frewin CL, Esrafilzadeh D, Yu C, Wang C, Pancrazio JJ, Romero-Ortega M, Jalili R, Wallace G. High-Performance Graphene-Fiber-Based Neural Recording Microelectrodes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805867. [PMID: 30803072 DOI: 10.1002/adma.201805867] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/10/2019] [Indexed: 05/24/2023]
Abstract
Fabrication of flexible and free-standing graphene-fiber- (GF-) based microelectrode arrays with a thin platinum coating, acting as a current collector, results in a structure with low impedance, high surface area, and excellent electrochemical properties. This modification results in a strong synergistic effect between these two constituents leading to a robust and superior hybrid material with better performance than either graphene electrodes or Pt electrodes. The low impedance and porous structure of the GF results in an unrivalled charge injection capacity of 10.34 mC cm-2 with the ability to record and detect neuronal activity. Furthermore, the thin Pt layer transfers the collected signals along the microelectrode efficiently. In vivo studies show that microelectrodes implanted in the rat cerebral cortex can detect neuronal activity with remarkably high signal-to-noise ratio (SNR) of 9.2 dB in an area as small as an individual neuron.
Collapse
Affiliation(s)
- Kezhong Wang
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Christopher L Frewin
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2031, Australia
| | - Changchun Yu
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Caiyun Wang
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Mario Romero-Ortega
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX, 75080, USA
| | - Rouhollah Jalili
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2031, Australia
| | - Gordon Wallace
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| |
Collapse
|
30
|
Martins NRB, Angelica A, Chakravarthy K, Svidinenko Y, Boehm FJ, Opris I, Lebedev MA, Swan M, Garan SA, Rosenfeld JV, Hogg T, Freitas RA. Human Brain/Cloud Interface. Front Neurosci 2019; 13:112. [PMID: 30983948 PMCID: PMC6450227 DOI: 10.3389/fnins.2019.00112] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/30/2019] [Indexed: 12/25/2022] Open
Abstract
The Internet comprises a decentralized global system that serves humanity's collective effort to generate, process, and store data, most of which is handled by the rapidly expanding cloud. A stable, secure, real-time system may allow for interfacing the cloud with the human brain. One promising strategy for enabling such a system, denoted here as a "human brain/cloud interface" ("B/CI"), would be based on technologies referred to here as "neuralnanorobotics." Future neuralnanorobotics technologies are anticipated to facilitate accurate diagnoses and eventual cures for the ∼400 conditions that affect the human brain. Neuralnanorobotics may also enable a B/CI with controlled connectivity between neural activity and external data storage and processing, via the direct monitoring of the brain's ∼86 × 109 neurons and ∼2 × 1014 synapses. Subsequent to navigating the human vasculature, three species of neuralnanorobots (endoneurobots, gliabots, and synaptobots) could traverse the blood-brain barrier (BBB), enter the brain parenchyma, ingress into individual human brain cells, and autoposition themselves at the axon initial segments of neurons (endoneurobots), within glial cells (gliabots), and in intimate proximity to synapses (synaptobots). They would then wirelessly transmit up to ∼6 × 1016 bits per second of synaptically processed and encoded human-brain electrical information via auxiliary nanorobotic fiber optics (30 cm3) with the capacity to handle up to 1018 bits/sec and provide rapid data transfer to a cloud based supercomputer for real-time brain-state monitoring and data extraction. A neuralnanorobotically enabled human B/CI might serve as a personalized conduit, allowing persons to obtain direct, instantaneous access to virtually any facet of cumulative human knowledge. Other anticipated applications include myriad opportunities to improve education, intelligence, entertainment, traveling, and other interactive experiences. A specialized application might be the capacity to engage in fully immersive experiential/sensory experiences, including what is referred to here as "transparent shadowing" (TS). Through TS, individuals might experience episodic segments of the lives of other willing participants (locally or remote) to, hopefully, encourage and inspire improved understanding and tolerance among all members of the human family.
Collapse
Affiliation(s)
- Nuno R. B. Martins
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Center for Research and Education on Aging (CREA), University of California, Berkeley and LBNL, Berkeley, CA, United States
| | | | - Krishnan Chakravarthy
- UC San Diego Health Science, San Diego, CA, United States
- VA San Diego Healthcare System, San Diego, CA, United States
| | | | | | - Ioan Opris
- Miami Project to Cure Paralysis, University of Miami, Miami, FL, United States
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - Mikhail A. Lebedev
- Center for Neuroengineering, Duke University, Durham, NC, United States
- Center for Bioelectric Interfaces of the Institute for Cognitive Neuroscience of the National Research University Higher School of Economics, Moscow, Russia
- Department of Information and Internet Technologies of Digital Health Institute, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Melanie Swan
- Department of Philosophy, Purdue University, West Lafayette, IN, United States
| | - Steven A. Garan
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Center for Research and Education on Aging (CREA), University of California, Berkeley and LBNL, Berkeley, CA, United States
| | - Jeffrey V. Rosenfeld
- Monash Institute of Medical Engineering, Monash University, Clayton, VIC, Australia
- Department of Neurosurgery, Alfred Hospital, Melbourne, VIC, Australia
- Department of Surgery, Monash University, Clayton, VIC, Australia
- Department of Surgery, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Tad Hogg
- Institute for Molecular Manufacturing, Palo Alto, CA, United States
| | | |
Collapse
|
31
|
Swain S, Gupta RK, Ratnayake K, Priyanka PD, Singh R, Jana S, Mitra K, Karunarathne A, Giri L. Confocal Imaging and k-Means Clustering of GABA B and mGluR Mediated Modulation of Ca 2+ Spiking in Hippocampal Neurons. ACS Chem Neurosci 2018; 9:3094-3107. [PMID: 30044088 DOI: 10.1021/acschemneuro.8b00297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Imaging cytosolic calcium in neurons is emerging as a new tool in neurological disease diagnosis, drug screening, and toxicity testing. Ca2+ oscillation signatures show a significant variation depending on GPCR targeting agonists. Quantification of Ca2+ spike trains in ligand induced Ca2+ oscillations remains challenging due to their inherent heterogeneity in primary culture. Moreover, there is no framework available for identification of optimal number of clusters and distance metric to cluster Ca2+ spike trains. Using quantitative confocal imaging and clustering analysis, we show the characterization of Ca2+ spiking in GPCR targeting drug-treated primary culture of hippocampal neurons. A systematic framework for selection of the clustering method instead of an intuition-based method was used to optimize the cluster number and distance metric. The results discern neurons with diverse Ca2+ response patterns, including higher amplitude fast spiking and lower spiking responses, and their relative percentage in a neuron population in absence and presence of GPCR-targeted drugs. The proposed framework was employed to show that the clustering pattern of Ca2+ spiking can be controlled using GABAB and mGluR targeting drugs. This approach can be used for unbiased measurement of neural activity and identification of spiking population with varying amplitude and frequencies, providing a platform for high-content drug screening.
Collapse
Affiliation(s)
- Sarpras Swain
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Rishikesh Kumar Gupta
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Kasun Ratnayake
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Pantula Devi Priyanka
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Ranjana Singh
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Soumya Jana
- Department of Electrical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Kishalay Mitra
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| | - Ajith Karunarathne
- Department of Chemistry and Biochemistry, University of Toledo, Toledo, Ohio 43606, United States
| | - Lopamudra Giri
- Department of Chemical Engineering, Indian Institute of Technology, Hyderabad 502285, India
| |
Collapse
|
32
|
Vitale F, Shen W, Driscoll N, Burrell JC, Richardson AG, Adewole O, Murphy B, Ananthakrishnan A, Oh H, Wang T, Lucas TH, Cullen DK, Allen MG, Litt B. Biomimetic extracellular matrix coatings improve the chronic biocompatibility of microfabricated subdural microelectrode arrays. PLoS One 2018; 13:e0206137. [PMID: 30383805 PMCID: PMC6211660 DOI: 10.1371/journal.pone.0206137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 10/08/2018] [Indexed: 01/15/2023] Open
Abstract
Intracranial electrodes are a vital component of implantable neurodevices, both for acute diagnostics and chronic treatment with open and closed-loop neuromodulation. Their performance is hampered by acute implantation trauma and chronic inflammation in response to implanted materials and mechanical mismatch between stiff synthetic electrodes and pulsating, natural soft host neural tissue. Flexible electronics based on thin polymer films patterned with microscale conductive features can help alleviate the mechanically induced trauma; however, this strategy alone does not mitigate inflammation at the device-tissue interface. In this study, we propose a biomimetic approach that integrates microscale extracellular matrix (ECM) coatings on microfabricated flexible subdural microelectrodes. Taking advantage of a high-throughput process employing micro-transfer molding and excimer laser micromachining, we fabricate multi-channel subdural microelectrodes primarily composed of ECM protein material and demonstrate that the electrochemical and mechanical properties match those of standard, uncoated controls. In vivo ECoG recordings in rodent brain confirm that the ECM microelectrode coatings and the protein interface do not alter signal fidelity. Astrogliotic, foreign body reaction to ECM coated devices is reduced, compared to uncoated controls, at 7 and 30 days, after subdural implantation in rat somatosensory cortex. We propose microfabricated, flexible, biomimetic electrodes as a new strategy to reduce inflammation at the device-tissue interface and improve the long-term stability of implantable subdural electrodes.
Collapse
Affiliation(s)
- Flavia Vitale
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurology, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Physical Medicine & Rehabilitation, University of Pennsylvania, Philadelphia PA, United States of America
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
| | - Wendy Shen
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia PA, United States of America
| | - Nicolette Driscoll
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Justin C. Burrell
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - Andrew G. Richardson
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - Oladayo Adewole
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Brendan Murphy
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Akshay Ananthakrishnan
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia PA, United States of America
| | - Hanju Oh
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia PA, United States of America
| | - Theodore Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Timothy H. Lucas
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - D. Kacy Cullen
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - Mark G. Allen
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Brian Litt
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurology, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| |
Collapse
|
33
|
Shen W, Das S, Vitale F, Richardson A, Ananthakrishnan A, Struzyna LA, Brown DP, Song N, Ramkumar M, Lucas T, Cullen DK, Litt B, Allen MG. Microfabricated intracortical extracellular matrix-microelectrodes for improving neural interfaces. MICROSYSTEMS & NANOENGINEERING 2018; 4:30. [PMID: 31057918 PMCID: PMC6220172 DOI: 10.1038/s41378-018-0030-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/30/2018] [Accepted: 08/05/2018] [Indexed: 05/30/2023]
Abstract
Intracortical neural microelectrodes, which can directly interface with local neural microcircuits with high spatial and temporal resolution, are critical for neuroscience research, emerging clinical applications, and brain computer interfaces (BCI). However, clinical applications of these devices remain limited mostly by their inability to mitigate inflammatory reactions and support dense neuronal survival at their interfaces. Herein we report the development of microelectrodes primarily composed of extracellular matrix (ECM) proteins, which act as a bio-compatible and an electrochemical interface between the microelectrodes and physiological solution. These ECM-microelectrodes are batch fabricated using a novel combination of micro-transfer-molding and excimer laser micromachining to exhibit final dimensions comparable to those of commercial silicon-based microelectrodes. These are further integrated with a removable insertion stent which aids in intracortical implantation. Results from electrochemical models and in vivo recordings from the rat's cortex indicate that ECM encapsulations have no significant effect on the electrochemical impedance characteristics of ECM-microelectrodes at neurologically relevant frequencies. ECM-microelectrodes are found to support a dense layer of neuronal somata and neurites on the electrode surface with high neuronal viability and exhibited markedly diminished neuroinflammation and glial scarring in early chronic experiments in rats.
Collapse
Affiliation(s)
- Wen Shen
- Krishna P. Singh Center for Nanotechnology, University of Pennsylvania, Philadelphia, PA 19104 USA
- Present Address: Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, TX 76019 USA
| | - Suradip Das
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Flavia Vitale
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Andrew Richardson
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Akshay Ananthakrishnan
- Department of Mechanical Engineering and Applied Mechanics, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Laura A. Struzyna
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Daniel P. Brown
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Naixin Song
- Department of Electrical and Systems Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Murari Ramkumar
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Timothy Lucas
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - D. Kacy Cullen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Brian Litt
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Mark G. Allen
- Krishna P. Singh Center for Nanotechnology, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
34
|
Sharma M, Gardner AT, Strathman HJ, Warren DJ, Silver J, Walker RM. Acquisition of Neural Action Potentials Using Rapid Multiplexing Directly at the Electrodes. MICROMACHINES 2018; 9:E477. [PMID: 30424410 PMCID: PMC6215140 DOI: 10.3390/mi9100477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/15/2018] [Accepted: 09/17/2018] [Indexed: 02/02/2023]
Abstract
Neural recording systems that interface with implanted microelectrodes are used extensively in experimental neuroscience and neural engineering research. Interface electronics that are needed to amplify, filter, and digitize signals from multichannel electrode arrays are a critical bottleneck to scaling such systems. This paper presents the design and testing of an electronic architecture for intracortical neural recording that drastically reduces the size per channel by rapidly multiplexing many electrodes to a single circuit. The architecture utilizes mixed-signal feedback to cancel electrode offsets, windowed integration sampling to reduce aliased high-frequency noise, and a successive approximation analog-to-digital converter with small capacitance and asynchronous control. Results are presented from a 180 nm CMOS integrated circuit prototype verified using in vivo experiments with a tungsten microwire array implanted in rodent cortex. The integrated circuit prototype achieves <0.004 mm² area per channel, 7 µW power dissipation per channel, 5.6 µVrms input referred noise, 50 dB common mode rejection ratio, and generates 9-bit samples at 30 kHz per channel by multiplexing at 600 kHz. General considerations are discussed for rapid time domain multiplexing of high-impedance microelectrodes. Overall, this work describes a promising path forward for scaling neural recording systems to numbers of electrodes that are orders of magnitude larger.
Collapse
Affiliation(s)
- Mohit Sharma
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Avery Tye Gardner
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Hunter J Strathman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - David J Warren
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jason Silver
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Ross M Walker
- Department of Electrical and Computer Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
35
|
Goss-Varley M, Shoffstall AJ, Dona KR, McMahon JA, Lindner SC, Ereifej ES, Capadona JR. Rodent Behavioral Testing to Assess Functional Deficits Caused by Microelectrode Implantation in the Rat Motor Cortex. J Vis Exp 2018:57829. [PMID: 30176008 PMCID: PMC6128113 DOI: 10.3791/57829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Medical devices implanted in the brain hold tremendous potential. As part of a Brain Machine Interface (BMI) system, intracortical microelectrodes demonstrate the ability to record action potentials from individual or small groups of neurons. Such recorded signals have successfully been used to allow patients to interface with or control computers, robotic limbs, and their own limbs. However, previous animal studies have shown that a microelectrode implantation in the brain not only damages the surrounding tissue but can also result in functional deficits. Here, we discuss a series of behavioral tests to quantify potential motor impairments following the implantation of intracortical microelectrodes into the motor cortex of a rat. The methods for open field grid, ladder crossing, and grip strength testing provide valuable information regarding the potential complications resulting from a microelectrode implantation. The results of the behavioral testing are correlated with endpoint histology, providing additional information on the pathological outcomes and impacts of this procedure on the adjacent tissue.
Collapse
Affiliation(s)
- Monika Goss-Varley
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University
| | - Andrew J Shoffstall
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University
| | - Keith R Dona
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University
| | - Justin A McMahon
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University
| | - Sydney C Lindner
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University
| | - Evon S Ereifej
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University
| | - Jeffrey R Capadona
- Advanced Platform Technology Center, Rehabilitation Research and Development, Louis Stokes Cleveland Department of Veterans Affairs Medical Center; Department of Biomedical Engineering, Case Western Reserve University;
| |
Collapse
|
36
|
Hermann JK, Lin S, Soffer A, Wong C, Srivastava V, Chang J, Sunil S, Sudhakar S, Tomaszewski WH, Protasiewicz G, Selkirk SM, Miller RH, Capadona JR. The Role of Toll-Like Receptor 2 and 4 Innate Immunity Pathways in Intracortical Microelectrode-Induced Neuroinflammation. Front Bioeng Biotechnol 2018; 6:113. [PMID: 30159311 PMCID: PMC6104445 DOI: 10.3389/fbioe.2018.00113] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022] Open
Abstract
We have recently demonstrated that partial inhibition of the cluster of differentiation 14 (CD14) innate immunity co-receptor pathway improves the long-term performance of intracortical microelectrodes better than complete inhibition. We hypothesized that partial activation of the CD14 pathway was critical to a neuroprotective response to the injury associated with initial and sustained device implantation. Therefore, here we investigated the role of two innate immunity receptors that closely interact with CD14 in inflammatory activation. We implanted silicon planar non-recording neural probes into knockout mice lacking Toll-like receptor 2 (Tlr2-/-), knockout mice lacking Toll-like receptor 4 (Tlr4-/-), and wildtype (WT) control mice, and evaluated endpoint histology at 2 and 16 weeks after implantation. Tlr4-/- mice exhibited significantly lower BBB permeability at acute and chronic time points, but also demonstrated significantly lower neuronal survival at the chronic time point. Inhibition of the Toll-like receptor 2 (TLR2) pathway had no significant effect compared to control animals. Additionally, when investigating the maturation of the neuroinflammatory response from 2 to 16 weeks, transgenic knockout mice exhibited similar histological trends to WT controls, except that knockout mice did not exhibit changes in microglia and macrophage activation over time. Together, our results indicate that complete genetic removal of Toll-like receptor 4 (TLR4) was detrimental to the integration of intracortical neural probes, while inhibition of TLR2 had no impact within the tests performed in this study. Therefore, approaches focusing on incomplete or acute inhibition of TLR4 may still improve intracortical microelectrode integration and long term recording performance.
Collapse
Affiliation(s)
- John K. Hermann
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Shushen Lin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Arielle Soffer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Chun Wong
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Vishnupriya Srivastava
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Jeremy Chang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Smrithi Sunil
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Shruti Sudhakar
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - William H. Tomaszewski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Grace Protasiewicz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Stephen M. Selkirk
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
- Department of Neurology, Case Western Reserve University, Cleveland, OH, United States
- Spinal Cord Injury Division, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Robert H. Miller
- Neurosciences, George Washington University, Washington, DC, United States
| | - Jeffrey R. Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
- Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, United States
| |
Collapse
|
37
|
Abedini M, Tekieh T, Sasanpour P. Recording Neural Activity Based on Surface Plasmon Resonance by Optical Fibers-A Computational Analysis. Front Comput Neurosci 2018; 12:61. [PMID: 30123119 PMCID: PMC6085840 DOI: 10.3389/fncom.2018.00061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/11/2018] [Indexed: 02/05/2023] Open
Abstract
An all optical, non-destructive method for monitoring neural activity has been proposed and its performance in detection has been analyzed computationally. The proposed method is based on excitation of Surface Plasmon Resonance (SPR) through the structure of optical fibers. The sensor structure consists of a multimode optical fiber where, the cladding of fiber has been removed and thin film of gold structure has been deposited on the surface. Impinging the laser light with appropriate wavelength inside the fiber and based on the total internal reflection, the evanescent wave will excite surface plasmons in the gold thin film. The absorption of light by surface plasmons in the gold structure is severely dependent on the dielectric properties at its vicinity. The electrical activity of neural cells (action potential) can modulate the dielectric properties at its vicinity and hence can modify the absorption of light inside the optical fiber. We have computationally analyzed the performance of the proposed sensor with different available geometries using Finite Element Method (FEM). In this regard, we have shown that the optical response of proposed sensor will track the action potential of the neuron at its vicinity. Based on different geometrical structure, the sensor has absorption in different regions of visible spectrum.
Collapse
Affiliation(s)
- Mitra Abedini
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Shahid Beheshti Medical University, Tehran, Iran
| | - Tahereh Tekieh
- Complex System Group, Department of Physics, Sydney University, Sydney, NSW, Australia
| | - Pezhman Sasanpour
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Shahid Beheshti Medical University, Tehran, Iran.,School of Nanoscience, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| |
Collapse
|
38
|
Caldwell R, Mandal H, Sharma R, Solzbacher F, Tathireddy P, Rieth L. Analysis of Al 2O 3-parylene C bilayer coatings and impact of microelectrode topography on long term stability of implantable neural arrays. J Neural Eng 2018; 14:046011. [PMID: 28351998 DOI: 10.1088/1741-2552/aa69d3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Performance of many dielectric coatings for neural electrodes degrades over time, contributing to loss of neural signals and evoked percepts. Studies using planar test substrates have found that a novel bilayer coating of atomic-layer deposited (ALD) Al2O3 and parylene C is a promising candidate for neural electrode applications, exhibiting superior stability to parylene C alone. However, initial results from bilayer encapsulation testing on non-planar devices have been less positive. Our aim was to evaluate ALD Al2O3-parylene C coatings using novel test paradigms, to rigorously evaluate dielectric coatings for neural electrode applications by incorporating neural electrode topography into test structure design. APPROACH Five test devices incorporated three distinct topographical features common to neural electrodes, derived from the utah electrode array (UEA). Devices with bilayer (52 nm Al2O3 + 6 µm parylene C) were evaluated against parylene C controls (N ⩾ 6 per device type). Devices were aged in phosphate buffered saline at 67 °C for up to 311 d, and monitored through: (1) leakage current to evaluate encapsulation lifetimes (>1 nA during 5VDC bias indicated failure), and (2) wideband (1-105 Hz) impedance. MAIN RESULTS Mean-times-to-failure (MTTFs) ranged from 12 to 506 d for bilayer-coated devices, versus 10 to >2310 d for controls. Statistical testing (log-rank test, α = 0.05) of failure rates gave mixed results but favored the control condition. After failure, impedance loss for bilayer devices continued for months and manifested across the entire spectrum, whereas the effect was self-limiting after several days, and restricted to frequencies <100 Hz for controls. These results correlated well with observations of UEAs encapsulated with bilayer and control films. SIGNIFICANCE We observed encapsulation failure modes and behaviors comparable to neural electrode performance which were undetected in studies with planar test devices. We found the impact of parylene C defects to be exacerbated by ALD Al2O3, and conclude that inferior bilayer performance arises from degradation of ALD Al2O3 when directly exposed to saline. This is an important consideration, given that neural electrodes with bilayer coatings are expected to have ALD Al2O3 exposed at dielectric boundaries that delineate electrode sites. Process improvements and use of different inorganic coatings to decrease dissolution in physiological fluids may improve performance. Testing frameworks which take neural electrode complexities into account will be well suited to reliably evaluate such encapsulation schemes.
Collapse
Affiliation(s)
- Ryan Caldwell
- Department of Bioengineering, University of Utah, Salt Lake City, UT, United States of America
| | | | | | | | | | | |
Collapse
|
39
|
Luo Z, Weiss DE, Liu Q, Tian B. Biomimetic Approaches Toward Smart Bio-hybrid Systems. NANO RESEARCH 2018; 11:3009-3030. [PMID: 30906509 PMCID: PMC6430233 DOI: 10.1007/s12274-018-2004-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 05/30/2023]
Abstract
Bio-integrated materials and devices can blur the interfaces between living and artificial systems. Microfluidics, bioelectronics and engineered nanostructures, with close interactions with biology at the cellular or tissue levels, have already yielded a spectrum of new applications. Many new designs emerge, including those of organ-on-a-chip systems, biodegradable implants, electroceutical devices, minimally invasive neuro-prosthetic tools, and soft robotics. In this review, we highlight a few recent advances on the fabrication and application of the smart bio-hybrid systems, with a particular emphasis on the three-dimensional (3D) bio-integrated devices that mimick the 3D feature of tissue scaffolds. Moreover, neurons integrated with engineered nanostructures for wireless neuromodulation and dynamic neural output will be briefly discussed. We will also go over the progress in the construction of cell-enabled soft robotics, where a tight coupling of the synthetic and biological parts is crucial for efficient functions. Finally, we summarize the approaches for enhancing bio-integration with biomimetic micro- and nanostructures.
Collapse
Affiliation(s)
- Zhiqiang Luo
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Dara E. Weiss
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Qingyun Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, PR China
| | - Bozhi Tian
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- The James Franck Institute, The University of Chicago, Chicago, IL 60637, USA
- The Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
40
|
Cody PA, Eles JR, Lagenaur CF, Kozai TDY, Cui XT. Unique electrophysiological and impedance signatures between encapsulation types: An analysis of biological Utah array failure and benefit of a biomimetic coating in a rat model. Biomaterials 2018; 161:117-128. [PMID: 29421549 PMCID: PMC5817007 DOI: 10.1016/j.biomaterials.2018.01.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/16/2022]
Abstract
Intracortical microelectrode arrays, especially the Utah array, remain the most common choice for obtaining high dimensional recordings of spiking neural activity for brain computer interface and basic neuroscience research. Despite the widespread use and established design, mechanical, material and biological challenges persist that contribute to a steady decline in recording performance (as evidenced by both diminished signal amplitude and recorded cell population over time) or outright array failure. Device implantation injury causes acute cell death and activation of inflammatory microglia and astrocytes that leads to a chronic neurodegeneration and inflammatory glial aggregation around the electrode shanks and often times fibrous tissue growth above the pia along the bed of the array within the meninges. This multifaceted deleterious cascade can result in substantial variability in performance even under the same experimental conditions. We track both impedance signatures and electrophysiological performance of 4 × 4 floating microelectrode Utah arrays implanted in the primary monocular visual cortex (V1m) of Long-Evans rats over a 12-week period. We employ a repeatable visual stimulation method to compare signal-to-noise ratio as well as single- and multi-unit yield from weekly recordings. To explain signal variability with biological response, we compare arrays categorized as either Type 1, partial fibrous encapsulation, or Type 2, complete fibrous encapsulation and demonstrate performance and impedance signatures unique to encapsulation type. We additionally assess benefits of a biomolecule coating intended to minimize distance to recordable units and observe a temporary improvement on multi-unit recording yield and single-unit amplitude.
Collapse
Affiliation(s)
- Patrick A Cody
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carl F Lagenaur
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - X Tracy Cui
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
41
|
Wellman SM, Eles JR, Ludwig KA, Seymour JP, Michelson NJ, McFadden WE, Vazquez AL, Kozai TDY. A Materials Roadmap to Functional Neural Interface Design. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1701269. [PMID: 29805350 PMCID: PMC5963731 DOI: 10.1002/adfm.201701269] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Advancement in neurotechnologies for electrophysiology, neurochemical sensing, neuromodulation, and optogenetics are revolutionizing scientific understanding of the brain while enabling treatments, cures, and preventative measures for a variety of neurological disorders. The grand challenge in neural interface engineering is to seamlessly integrate the interface between neurobiology and engineered technology, to record from and modulate neurons over chronic timescales. However, the biological inflammatory response to implants, neural degeneration, and long-term material stability diminish the quality of interface overtime. Recent advances in functional materials have been aimed at engineering solutions for chronic neural interfaces. Yet, the development and deployment of neural interfaces designed from novel materials have introduced new challenges that have largely avoided being addressed. Many engineering efforts that solely focus on optimizing individual probe design parameters, such as softness or flexibility, downplay critical multi-dimensional interactions between different physical properties of the device that contribute to overall performance and biocompatibility. Moreover, the use of these new materials present substantial new difficulties that must be addressed before regulatory approval for use in human patients will be achievable. In this review, the interdependence of different electrode components are highlighted to demonstrate the current materials-based challenges facing the field of neural interface engineering.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - James R Eles
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - Kip A Ludwig
- Department of Neurologic Surgery, 200 First St. SW, Rochester, MN 55905
| | - John P Seymour
- Electrical & Computer Engineering, 1301 Beal Ave., 2227 EECS, Ann Arbor, MI 48109
| | - Nicholas J Michelson
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - William E McFadden
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - Alberto L Vazquez
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - Takashi D Y Kozai
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| |
Collapse
|
42
|
Lo MC, Wang S, Singh S, Damodaran VB, Ahmed I, Coffey K, Barker D, Saste K, Kals K, Kaplan HM, Kohn J, Shreiber DI, Zahn JD. Evaluating the in vivo glial response to miniaturized parylene cortical probes coated with an ultra-fast degrading polymer to aid insertion. J Neural Eng 2018; 15:036002. [PMID: 29485103 DOI: 10.1088/1741-2552/aa9fad] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Despite the feasibility of short-term neural recordings using implantable microelectrodes, attaining reliable, chronic recordings remains a challenge. Most neural recording devices suffer from a long-term tissue response, including gliosis, at the device-tissue interface. It was hypothesized that smaller, more flexible intracortical probes would limit gliosis by providing a better mechanical match with surrounding tissue. APPROACH This paper describes the in vivo evaluation of flexible parylene microprobes designed to improve the interface with the adjacent neural tissue to limit gliosis and thereby allow for improved recording longevity. The probes were coated with an ultrafast degrading tyrosine-derived polycarbonate (E5005(2K)) polymer that provides temporary mechanical support for device implantation, yet degrades within 2 h post-implantation. A parametric study of probes of varying dimensions and polymer coating thicknesses were implanted in rat brains. The glial tissue response and neuronal loss were assessed from 72 h to 24 weeks post-implantation via immunohistochemistry. MAIN RESULTS Experimental results suggest that both probe and polymer coating sizes affect the extent of gliosis. When an appropriate sized coating dimension (100 µm × 100 µm) and small probe (30 µm × 5 µm) was implanted, a minimal post-implantation glial response was observed. No discernible gliosis was detected when compared to tissue where a sham control consisting of a solid degradable polymer shuttle of the same dimensions was inserted. A larger polymer coating (200 µm × 200 µm) device induced a more severe glial response at later time points, suggesting that the initial insertion trauma can affect gliosis even when the polymer shuttle degrades rapidly. A larger degree of gliosis was also observed when comparing a larger sized probe (80 µm × 5 µm) to a smaller probe (30 µm × 5 µm) using the same polymer coating size (100 µm × 100 µm). There was no significant neuronal loss around the implantation sites for most device candidates except the group with largest polymer coating and probe sizes. SIGNIFICANCE These results suggest that: (1) the degree of mechanical trauma at device implantation and mechanical mismatches at the probe-tissue interface affect long term gliosis; (2) smaller, more flexible probes may minimize the glial response to provide improved tissue biocompatibility when used for chronic neural signal recording; and (3) some degree of glial scarring did not significantly affect neuronal distribution around the probe.
Collapse
Affiliation(s)
- Meng-Chen Lo
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
A Mosquito Inspired Strategy to Implant Microprobes into the Brain. Sci Rep 2018; 8:122. [PMID: 29317748 PMCID: PMC5760625 DOI: 10.1038/s41598-017-18522-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/13/2017] [Indexed: 02/05/2023] Open
Abstract
Mosquitos are among the deadliest insects on the planet due to their ability to transmit diseases like malaria through their bite. In order to bite, a mosquito must insert a set of micro-sized needles through the skin to reach vascular structures. The mosquito uses a combination of mechanisms including an insertion guide to enable it to bite and feed off of larger animals. Here, we report on a biomimetic strategy inspired by the mosquito insertion guide to enable the implantation of intracortical microelectrodes into the brain. Next generation microelectrode designs leveraging ultra-small dimensions and/or flexible materials offer the promise of increased performance, but present difficulties in reliable implantation. With the biomimetic guide in place, the rate of successful microprobe insertion increased from 37.5% to 100% due to the rise in the critical buckling force of the microprobes by 3.8-fold. The prototype guides presented here provide a reproducible method to augment the insertion of small, flexible devices into the brain. In the future, similar approaches may be considered and applied to the insertion of other difficult to implant medical devices.
Collapse
|
44
|
Prospects for a Robust Cortical Recording Interface. Neuromodulation 2018. [DOI: 10.1016/b978-0-12-805353-9.00028-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
45
|
Cork SC, Eftekhar A, Mirza KB, Zuliani C, Nikolic K, Gardiner JV, Bloom SR, Toumazou C. Extracellular pH monitoring for use in closed-loop vagus nerve stimulation. J Neural Eng 2017; 15:016001. [DOI: 10.1088/1741-2552/aa8239] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
46
|
Mols K, Musa S, Nuttin B, Lagae L, Bonin V. In vivo characterization of the electrophysiological and astrocytic responses to a silicon neuroprobe implanted in the mouse neocortex. Sci Rep 2017; 7:15642. [PMID: 29142267 PMCID: PMC5688150 DOI: 10.1038/s41598-017-15121-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 10/23/2017] [Indexed: 12/04/2022] Open
Abstract
Silicon neuroprobes hold great potential for studies of large-scale neural activity and brain computer interfaces, but data on brain response in chronic implants is limited. Here we explored with in vivo cellular imaging the response to multisite silicon probes for neural recordings. We tested a chronic implant for mice consisting of a CMOS-compatible silicon probe rigidly implanted in the cortex under a cranial imaging window. Multiunit recordings of cortical neurons with the implant showed no degradation of electrophysiological signals weeks after implantation (mean spike and noise amplitudes of 186 ± 42 µVpp and 16 ± 3.2 µVrms, respectively, n = 5 mice). Two-photon imaging through the cranial window allowed longitudinal monitoring of fluorescently-labeled astrocytes from the second week post implantation for 8 weeks (n = 3 mice). The imaging showed a local increase in astrocyte-related fluorescence that remained stable from the second to the tenth week post implantation. These results demonstrate that, in a standard electrophysiology protocol in mice, rigidly implanted silicon probes can provide good short to medium term chronic recording performance with a limited astrocyte inflammatory response. The precise factors influencing the response to silicon probe implants remain to be elucidated.
Collapse
Affiliation(s)
- Katrien Mols
- Neuro-Electronics Research Flanders, Kapeldreef 75, 3001, Leuven, Belgium.,imec, Department of Life Science Technologies, Kapeldreef 75, 3001, Leuven, Belgium.,KU Leuven, Department of Neurosciences, 3000, Leuven, Belgium
| | - Silke Musa
- imec, Department of Life Science Technologies, Kapeldreef 75, 3001, Leuven, Belgium
| | - Bart Nuttin
- KU Leuven, Department of Neurosciences, 3000, Leuven, Belgium
| | - Liesbet Lagae
- imec, Department of Life Science Technologies, Kapeldreef 75, 3001, Leuven, Belgium.,KU Leuven, Department of Physics and Astronomy, 3001, Leuven, Belgium
| | - Vincent Bonin
- Neuro-Electronics Research Flanders, Kapeldreef 75, 3001, Leuven, Belgium. .,Vlaams Instituut voor Biotechnologie (VIB), 3001, Leuven, Belgium. .,KU Leuven, Department of Biology, 3000, Leuven, Belgium.
| |
Collapse
|
47
|
Microelectrode implantation in motor cortex causes fine motor deficit: Implications on potential considerations to Brain Computer Interfacing and Human Augmentation. Sci Rep 2017; 7:15254. [PMID: 29127346 PMCID: PMC5681545 DOI: 10.1038/s41598-017-15623-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 10/25/2017] [Indexed: 12/22/2022] Open
Abstract
Intracortical microelectrodes have shown great success in enabling locked-in patients to interact with computers, robotic limbs, and their own electrically driven limbs. The recent advances have inspired world-wide enthusiasm resulting in billions of dollars invested in federal and industrial sponsorships to understanding the brain for rehabilitative applications. Additionally, private philanthropists have also demonstrated excitement in the field by investing in the use of brain interfacing technologies as a means to human augmentation. While the promise of incredible technologies is real, caution must be taken as implications regarding optimal performance and unforeseen side effects following device implantation into the brain are not fully characterized. The current study is aimed to quantify any motor deficit caused by microelectrode implantation in the motor cortex of healthy rats compared to non-implanted controls. Following electrode insertion, rats were tested on an open-field grid test to study gross motor function and a ladder test to study fine motor function. It was discovered that rats with chronically indwelling intracortical microelectrodes exhibited up to an incredible 527% increase in time to complete the fine motor task. This initial study defines the need for further and more robust behavioral testing of potential unintentional harm caused by microelectrode implantation.
Collapse
|
48
|
Affiliation(s)
- Eduardo Fernández
- Bioengineering Institute; Miguel Hernández University of Elche and CIBER BBN; Elche 03202 Spain
| | - Pablo Botella
- Instituto de Tecnología Química; Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas; Valencia 46022 Spain
| |
Collapse
|
49
|
Loris ZB, Danzi M, Sick J, Dietrich WD, Bramlett HM, Sick T. Automated approach to detecting behavioral states using EEG-DABS. Heliyon 2017; 3:e00344. [PMID: 28725869 PMCID: PMC5507012 DOI: 10.1016/j.heliyon.2017.e00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 06/05/2017] [Accepted: 06/28/2017] [Indexed: 11/30/2022] Open
Abstract
Electrocorticographic (ECoG) signals represent cortical electrical dipoles generated by synchronous local field potentials that result from simultaneous firing of neurons at distinct frequencies (brain waves). Since different brain waves correlate to different behavioral states, ECoG signals presents a novel strategy to detect complex behaviors. We developed a program, EEG Detection Analysis for Behavioral States (EEG-DABS) that advances Fast Fourier Transforms through ECoG signals time series, separating it into (user defined) frequency bands and normalizes them to reduce variability. EEG-DABS determines events if segments of an experimental ECoG record have significantly different power bands than a selected control pattern of EEG. Events are identified at every epoch and frequency band and then are displayed as output graphs by the program. Certain patterns of events correspond to specific behaviors. Once a predetermined pattern was selected for a behavioral state, EEG-DABS correctly identified the desired behavioral event. The selection of frequency band combinations for detection of the behavior affects accuracy of the method. All instances of certain behaviors, such as freezing, were correctly identified from the event patterns generated with EEG-DABS. Detecting behaviors is typically achieved by visually discerning unique animal phenotypes, a process that is time consuming, unreliable, and subjective. EEG-DABS removes variability by using defined parameters of EEG/ECoG for a desired behavior over chronic recordings. EEG-DABS presents a simple and automated approach to quantify different behavioral states from ECoG signals.
Collapse
Affiliation(s)
- Zachary B Loris
- Department of Neurological Surgery, 1095 NW 14th Terrace, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.,The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, Florida, 33136, USA.,Neuroscience Program, 1120 NW 14th Street, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Mathew Danzi
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, Florida, 33136, USA.,Neuroscience Program, 1120 NW 14th Street, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.,Center for Computational Science, University of Miami, Miami, Florida, 33146, USA
| | - Justin Sick
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, Florida, 33136, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, 1095 NW 14th Terrace, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.,The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, Florida, 33136, USA.,Neuroscience Program, 1120 NW 14th Street, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, 1095 NW 14th Terrace, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.,The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, Florida, 33136, USA.,Department of Neurology, 1150 NW 14th Street, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.,Bruce W. Carter Department of Veterans Affairs Medical Center, 1201 NW 16th Street, Miami, Florida, 33125, USA
| | - Thomas Sick
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, Florida, 33136, USA.,Department of Neurology, 1150 NW 14th Street, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.,Neuroscience Program, 1120 NW 14th Street, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA
| |
Collapse
|
50
|
Thompson CH, Khan SA, Khan WA, Li W, Purcell EK. Neuronal excitability and network formation on optically transparent electrode materials. INTERNATIONAL IEEE/EMBS CONFERENCE ON NEURAL ENGINEERING : [PROCEEDINGS]. INTERNATIONAL IEEE EMBS CONFERENCE ON NEURAL ENGINEERING 2017; 2017:154-157. [PMID: 30338028 PMCID: PMC6190689 DOI: 10.1109/ner.2017.8008315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
With the advent of genetically-encoded optical tools to trigger or report neuronal activity, new designs for multielectrode arrays (MEAs) used in neural interfacing incorporate both optical and electrical modes of stimulating or recording neural activity. Likewise, the need to improve upon the biocompatibility of implanted MEAs has moved the field towards the use of softer, more compliant materials in device fabrication. However, there is limited available information on the impact of the materials used in MEAs on the function of interfaced individual neurons and neuronal networks. We assessed the responses of rat cortical neurons on optically transparent materials commonly used in the construction of "next-generation" devices: indium tin oxide (ITO), parylene-C, and polydimethylsiloxane (PDMS). We found that neuronal network formation and spiking responses to electrical stimulation were enhanced in neurons cultured on ITO. We observed reduced excitability and synaptic connectivity between neurons cultured on PDMS. We hypothesize that the superior conductivity of ITO and suboptimal neuronal attachment to PDMS contributed to our results.
Collapse
Affiliation(s)
| | - Sahar A Khan
- Michigan State University, East Lansing, MI 48824 USA
| | - Wasif A Khan
- Michigan State University, East Lansing, MI 48824 USA
| | - Wen Li
- Electrical and Computer Engineering Department, Michigan State University, East Lansing, MI 48824 USA
| | - Erin K Purcell
- Department of Biomedical Engineering and the Department of Electrical and Computer Engineering at Michigan State University, East Lansing, MI 48824 USA (phone: 586-202-4232; fax: 517-353-1980; )
| |
Collapse
|