1
|
Villa J, Cury J, Kessler L, Tan X, Richter CP. Enhancing biocompatibility of the brain-machine interface: A review. Bioact Mater 2024; 42:531-549. [PMID: 39308547 PMCID: PMC11416625 DOI: 10.1016/j.bioactmat.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
In vivo implantation of microelectrodes opens the door to studying neural circuits and restoring damaged neural pathways through direct electrical stimulation and recording. Although some neuroprostheses have achieved clinical success, electrode material properties, inflammatory response, and glial scar formation at the electrode-tissue interfaces affect performance and sustainability. Those challenges can be addressed by improving some of the materials' mechanical, physical, chemical, and electrical properties. This paper reviews materials and designs of current microelectrodes and discusses perspectives to advance neuroprosthetics performance.
Collapse
Affiliation(s)
- Jordan Villa
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Joaquin Cury
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Lexie Kessler
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Xiaodong Tan
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
| | - Claus-Peter Richter
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Biomedical Engineering, Northwestern University, USA
| |
Collapse
|
2
|
Li F, Gallego J, Tirko NN, Greaser J, Bashe D, Patel R, Shaker E, Van Valkenburg GE, Alsubhi AS, Wellman S, Singh V, Padilla CG, Gheres KW, Broussard JI, Bagwell R, Mulvihill M, Kozai TDY. Low-intensity pulsed ultrasound stimulation (LIPUS) modulates microglial activation following intracortical microelectrode implantation. Nat Commun 2024; 15:5512. [PMID: 38951525 PMCID: PMC11217463 DOI: 10.1038/s41467-024-49709-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
Microglia are important players in surveillance and repair of the brain. Implanting an electrode into the cortex activates microglia, produces an inflammatory cascade, triggers the foreign body response, and opens the blood-brain barrier. These changes can impede intracortical brain-computer interfaces performance. Using two-photon imaging of implanted microelectrodes, we test the hypothesis that low-intensity pulsed ultrasound stimulation can reduce microglia-mediated neuroinflammation following the implantation of microelectrodes. In the first week of treatment, we found that low-intensity pulsed ultrasound stimulation increased microglia migration speed by 128%, enhanced microglia expansion area by 109%, and a reduction in microglial activation by 17%, indicating improved tissue healing and surveillance. Microglial coverage of the microelectrode was reduced by 50% and astrocytic scarring by 36% resulting in an increase in recording performance at chronic time. The data indicate that low-intensity pulsed ultrasound stimulation helps reduce the foreign body response around chronic intracortical microelectrodes.
Collapse
Affiliation(s)
- Fan Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- Computational Modeling and Simulation PhD Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jazlyn Gallego
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Natasha N Tirko
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | | | - Derek Bashe
- Washington University in St. Louis, St. Louis, MO, USA
| | - Rudra Patel
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eric Shaker
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | - Vanshika Singh
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Camila Garcia Padilla
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | | | | | | | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA.
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Shi D, Narayanan S, Woeppel K, Cui XT. Improving the Biocompatibility and Functionality of Neural Interface Devices with Silica Nanoparticles. Acc Chem Res 2024; 57:1684-1695. [PMID: 38814586 PMCID: PMC11191400 DOI: 10.1021/acs.accounts.4c00160] [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: 03/10/2024] [Revised: 05/03/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024]
Abstract
ConspectusNeural interface technologies enable bidirectional communication between the nervous system and external instrumentation. Advancements in neural interface devices not only open new frontiers for neuroscience research, but also hold great promise for clinical diagnosis, therapy, and rehabilitation for various neurological disorders. However, the performance of current neural electrode devices, often termed neural probes, is far from satisfactory. Glial scarring, neuronal degeneration, and electrode degradation eventually cause the devices to lose their connection with the brain. To improve the chronic performance of neural probes, efforts need to be made on two fronts: enhancing the physiochemical properties of the electrode materials and mitigating the undesired host tissue response.In this Account, we discuss our efforts in developing silica-nanoparticle-based (SiNP) coatings aimed at enhancing neural probe electrochemical properties and promoting device-tissue integration. Our work focuses on three approaches:(1) SiNPs' surface texturization to enhance biomimetic protein coatings for promoting neural integration. Through covalent immobilization, SiNP introduces biologically relevant nanotopography to neural probe surfaces, enhancing neuronal cell attachments and inhibiting microglia. The SiNP base coating further increases the binding density and stability of bioactive molecules such as L1CAM and facilitates the widespread dissemination of biomimetic coatings. (2) Doping SiNPs into conductive polymer electrode coatings improves the electrochemical properties and stability. As neural interface devices are moving to subcellular sizes to escape the immune response and high electrode site density to increase spatial resolution, the electrode sites need to be very small. The smaller electrode size comes at the cost of a high electrode impedance, elevated thermal noise, and insufficient charge injection capacity. Electrochemically deposited conductive polymer films reduce electrode impedance but do not endure prolonged electrical cycling. When incorporated into conductive polymer coatings as a dopant, the SiNP provides structural support for the polymer thin films, significantly increasing their stability and durability. Low interfacial impedance maintained by the conducting polymer/SiNP composite is critical for extended electrode longevity and effective charge injection in chronic neural stimulation applications. (3) Porous nanoparticles are used as drug carriers in conductive polymer coatings for local drug/neurochemical delivery. When triggered by external electrical stimuli, drug molecules and neurochemicals can be released in a controlled manner. Such precise focal manipulation of cellular and vascular behavior enables us to probe brain circuitry and develop therapeutic applications.We foresee tremendous opportunities for further advancing the functionality of SiNP coatings by incorporating new nanoscale components and integrating the coating with other design strategies. With an enriched nanoscale toolbox and optimized design strategies, we can create customizable multifunctional and multimodal neural interfaces that can operate at multiple spatial levels and seamlessly integrate with the host tissue for extended applications.
Collapse
Affiliation(s)
- Delin Shi
- University
of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, Pennsylvania 15260, United States
- Center
for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, Pennsylvania 15213, United States
| | - Sharada Narayanan
- University
of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, Pennsylvania 15260, United States
- Center
for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, Pennsylvania 15213, United States
| | - Kevin Woeppel
- University
of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, Pennsylvania 15260, United States
- Center
for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, Pennsylvania 15213, United States
| | - Xinyan Tracy Cui
- University
of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, Pennsylvania 15260, United States
- Center
for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
4
|
Dhawan V, Martin PN, Hu X, Cui XT. Investigation of a chondroitin sulfate-based bioactive coating for neural interface applications. J Mater Chem B 2024; 12:5535-5550. [PMID: 38747002 PMCID: PMC11152038 DOI: 10.1039/d4tb00501e] [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: 03/08/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024]
Abstract
Invasive neural implants allow for high-resolution bidirectional communication with the nervous tissue and have demonstrated the ability to record neural activity, stimulate neurons, and sense neurochemical species with high spatial selectivity and resolution. However, upon implantation, they are exposed to a foreign body response which can disrupt the seamless integration of the device with the native tissue and lead to deterioration in device functionality for chronic implantation. Modifying the device surface by incorporating bioactive coatings has been a promising approach to camouflage the device and improve integration while maintaining device performance. In this work, we explored the novel application of a chondroitin sulfate (CS) based hydrophilic coating, with anti-fouling and neurite-growth promoting properties for neural recording electrodes. CS-coated samples exhibited significantly reduced protein-fouling in vitro which was maintained for up to 4-weeks. Cell culture studies revealed a significant increase in neurite attachment and outgrowth and a significant decrease in microglia attachment and activation for the CS group as compared to the control. After 1-week of in vivo implantation in the mouse cortex, the coated probes demonstrated significantly lower biofouling as compared to uncoated controls. Like the in vitro results, increased neuronal population (neuronal nuclei and neurofilament) and decreased microglial activation were observed. To assess the coating's effect on the recording performance of silicon microelectrodes, we implanted coated and uncoated electrodes in the mouse striatum for 1 week and performed impedance and recording measurements. We observed significantly lower impedance in the coated group, likely due to the increased wettability of the coated surface. The peak-to-peak amplitude and the noise floor levels were both lower in the CS group compared to the controls, which led to a comparable signal-to-noise ratio between the two groups. The overall single unit yield (% channels recording a single unit) was 74% for the CS and 67% for the control group on day 1. Taken together, this study demonstrates the effectiveness of the polysaccharide-based coating in reducing biofouling and improving biocompatibility for neural electrode devices.
Collapse
Affiliation(s)
- Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Paige Nicole Martin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Xiaoming Hu
- Department of Neurology, University of Pittsburgh, PA, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Du Z, Chen G, Li Y, Zheng N, Cheng JX, Yang C. Photoacoustic: A Versatile Nongenetic Method for High-Precision Neuromodulation. Acc Chem Res 2024; 57:1595-1607. [PMID: 38759211 PMCID: PMC11154953 DOI: 10.1021/acs.accounts.4c00119] [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: 02/21/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
High-precision neuromodulation plays a pivotal role in elucidating fundamental principles of neuroscience and treating specific neurological disorders. Optical neuromodulation, enabled by spatial resolution defined by the diffraction limit at the submicrometer scale, is a general strategy to achieve such precision. Optogenetics offers single-neuron spatial resolution with cellular specificity, whereas the requirement of genetic transfection hinders its clinical application. Direct photothermal modulation, an alternative nongenetic optical approach, often associates a large temperature increase with the risk of thermal damage to surrounding tissues.Photoacoustic (also called optoacoustic) neural stimulation is an emerging technology for neural stimulation with the following key features demonstrated. First, the photoacoustic approach demonstrated high efficacy without the need for genetic modification. The generated pulsed ultrasound upon ns laser pulses with energy ranging from a few μJ to tens of μJ is sufficient to activate wild-type neurons. Second, the photoacoustic approach provides sub-100-μm spatial precision. It overcomes the fundamental wave diffraction limit of ultrasound by harnessing the localized ultrasound field generated through light absorption. A spatial precision of 400 μm has been achieved in rodent brains using a fiber-based photoacoustic emitter. Single-cell stimulation in neuronal cultures in vitro and in brain slices ex vivo is achieved using tapered fiber-based photoacoustic emitters. This precision is 10 to 100 times better than that for piezo-based low-frequency ultrasound and is essential to pinpoint a specific region or cell population in a living brain. Third, compared to direct photothermal stimulation via temperature increase, photoacoustic stimulation requires 40 times less laser energy dose to evoke neuron activities and is associated with a minimal temperature increase of less than 1 °C, preventing potential thermal damage to neurons. Fourth, photoacoustics is a versatile approach and can be designed in various platforms aiming at specific applications. Our team has shown the design of fiber-based photoacoustic emitters, photoacoustic nanotransducers, soft biocompatible photoacoustic films, and soft photoacoustic lenses. Since they interact with neurons through ultrasound without the need for direct contact, photoacoustic enables noninvasive transcranial and dura-penetrating brain stimulation without compromising high precision.In this Account, we will first review the basic principles of photoacoustic and discuss the key design elements of PA transducers for neural modulation guided by the principle. We will also highlight how these design goals were achieved from a materials chemistry perspective. The design of different PA interfaces, their unique capability, and their applications in neural systems will be reviewed. In the end, we will discuss the remaining challenges and future perspectives for this technology.
Collapse
Affiliation(s)
- Zhiyi Du
- Department
of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Guo Chen
- Department
of Electrical and Computer Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Yueming Li
- Department
of Electrical and Computer Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Nan Zheng
- Division
of Materials Science and Engineering, Boston
University, Boston, Massachusetts 02215, United States
| | - Ji-Xin Cheng
- Department
of Electrical and Computer Engineering, Boston University, Boston, Massachusetts 02215, United States
- Department
of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Chen Yang
- Department
of Chemistry, Boston University, Boston, Massachusetts 02215, United States
- Department
of Electrical and Computer Engineering, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
6
|
McNamara IN, Wellman SM, Li L, Eles JR, Savya S, Sohal HS, Angle MR, Kozai TDY. Electrode sharpness and insertion speed reduce tissue damage near high-density penetrating arrays. J Neural Eng 2024; 21:026030. [PMID: 38518365 DOI: 10.1088/1741-2552/ad36e1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
Objective. Over the past decade, neural electrodes have played a crucial role in bridging biological tissues with electronic and robotic devices. This study focuses on evaluating the optimal tip profile and insertion speed for effectively implanting Paradromics' high-density fine microwire arrays (FμA) prototypes into the primary visual cortex (V1) of mice and rats, addressing the challenges associated with the 'bed-of-nails' effect and tissue dimpling.Approach. Tissue response was assessed by investigating the impact of electrodes on the blood-brain barrier (BBB) and cellular damage, with a specific emphasis on tailored insertion strategies to minimize tissue disruption during electrode implantation.Main results.Electro-sharpened arrays demonstrated a marked reduction in cellular damage within 50μm of the electrode tip compared to blunt and angled arrays. Histological analysis revealed that slow insertion speeds led to greater BBB compromise than fast and pneumatic methods. Successful single-unit recordings validated the efficacy of the optimized electro-sharpened arrays in capturing neural activity.Significance.These findings underscore the critical role of tailored insertion strategies in minimizing tissue damage during electrode implantation, highlighting the suitability of electro-sharpened arrays for long-term implant applications. This research contributes to a deeper understanding of the complexities associated with high-channel-count microelectrode array implantation, emphasizing the importance of meticulous assessment and optimization of key parameters for effective integration and minimal tissue disruption. By elucidating the interplay between insertion parameters and tissue response, our study lays a strong foundation for the development of advanced implantable devices with a reduction in reactive gliosis and improved performance in neural recording applications.
Collapse
Affiliation(s)
- Ingrid N McNamara
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Lehong Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sajishnu Savya
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | | | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center of the Basis of Neural Cognition, Pittsburgh, PA, United States of America
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
7
|
Sturgill B, Hernandez-Reynoso AG, Druschel LN, Smith TJ, Boucher PE, Hoeferlin GF, Thai TTD, Jiang MS, Hess JL, Alam NN, Menendez DM, Duncan JL, Cogan SF, Pancrazio JJ, Capadona JR. Reactive Amine Functionalized Microelectrode Arrays Provide Short-Term Benefit but Long-Term Detriment to In Vivo Recording Performance. ACS APPLIED BIO MATERIALS 2024; 7:1052-1063. [PMID: 38290529 PMCID: PMC10880090 DOI: 10.1021/acsabm.3c01014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Intracortical microelectrode arrays (MEAs) are used for recording neural signals. However, indwelling devices result in chronic neuroinflammation, which leads to decreased recording performance through degradation of the device and surrounding tissue. Coating the MEAs with bioactive molecules is being explored to mitigate neuroinflammation. Such approaches often require an intermediate functionalization step such as (3-aminopropyl)triethoxysilane (APTES), which serves as a linker. However, the standalone effect of this intermediate step has not been previously characterized. Here, we investigated the effect of coating MEAs with APTES by comparing APTES-coated to uncoated controls in vivo and ex vivo. First, we measured water contact angles between silicon uncoated and APTES-coated substrates to verify the hydrophilic characteristics of the APTES coating. Next, we implanted MEAs in the motor cortex (M1) of Sprague-Dawley rats with uncoated or APTES-coated devices. We assessed changes in the electrochemical impedance and neural recording performance over a chronic implantation period of 16 weeks. Additionally, histology and bulk gene expression were analyzed to understand further the reactive tissue changes arising from the coating. Results showed that APTES increased the hydrophilicity of the devices and decreased electrochemical impedance at 1 kHz. APTES coatings proved detrimental to the recording performance, as shown by a constant decay up to 16 weeks postimplantation. Bulk gene analysis showed differential changes in gene expression between groups that were inconclusive with regard to the long-term effect on neuronal tissue. Together, these results suggest that APTES coatings are ultimately detrimental to chronic neural recordings. Furthermore, interpretations of studies using APTES as a functionalization step should consider the potential consequences if the final functionalization step is incomplete.
Collapse
Affiliation(s)
- Brandon
S. Sturgill
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Ana G. Hernandez-Reynoso
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Lindsey N. Druschel
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Thomas J. Smith
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Pierce E. Boucher
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - George F. Hoeferlin
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Teresa Thuc Doan Thai
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Madison S. Jiang
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Jordan L. Hess
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Neeha N. Alam
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Dhariyat M. Menendez
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Jonathan L. Duncan
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Stuart F. Cogan
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Joseph J. Pancrazio
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Jeffrey R. Capadona
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| |
Collapse
|
8
|
Tsui CT, Mirkiani S, Roszko DA, Churchward MA, Mushahwar VK, Todd KG. In vitro biocompatibility evaluation of functional electrically stimulating microelectrodes on primary glia. Front Bioeng Biotechnol 2024; 12:1351087. [PMID: 38314352 PMCID: PMC10834782 DOI: 10.3389/fbioe.2024.1351087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Neural interfacing devices interact with the central nervous system to alleviate functional deficits arising from disease or injury. This often entails the use of invasive microelectrode implants that elicit inflammatory responses from glial cells and leads to loss of device function. Previous work focused on improving implant biocompatibility by modifying electrode composition; here, we investigated the direct effects of electrical stimulation on glial cells at the electrode interface. A high-throughput in vitro system that assesses primary glial cell response to biphasic stimulation waveforms at 0 mA, 0.15 mA, and 1.5 mA was developed and optimized. Primary mixed glial cell cultures were generated from heterozygous CX3CR-1+/EGFP mice, electrically stimulated for 4 h/day over 3 days using 75 μm platinum-iridium microelectrodes, and biomarker immunofluorescence was measured. Electrodes were then imaged on a scanning electron microscope to assess sustained electrode damage. Fluorescence and electron microscopy analyses suggest varying degrees of localized responses for each biomarker assayed (Hoescht, EGFP, GFAP, and IL-1β), a result that expands on comparable in vivo models. This system allows for the comparison of a breadth of electrical stimulation parameters, and opens another avenue through which neural interfacing device developers can improve biocompatibility and longevity of electrodes in tissue.
Collapse
Affiliation(s)
- Christopher T. Tsui
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| | - Soroush Mirkiani
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| | - David A. Roszko
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| | - Matthew A. Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
- Department of Biological and Environmental Sciences, Concordia University of Edmonton, Edmonton, AB, Canada
| | - Vivian K. Mushahwar
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Kathryn G. Todd
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
- Sensory Motor Adaptive Rehabilitation Technology (SMART) Network, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
9
|
Aktas B, Ozgun A, Kilickap BD, Garipcan B. Cell adhesion molecule immobilized gold surfaces for enhanced neuron-electrode interfaces. J Biomed Mater Res B Appl Biomater 2024; 112:e35310. [PMID: 37950592 DOI: 10.1002/jbm.b.35310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 11/12/2023]
Abstract
To provide a long-term solution for increasing the biocompatibility of neuroprosthetics, approaches to reduce the side effects of invasive neuro-implantable devices are still in need of improvement. Physical, chemical, and bioactive design aspects of the biomaterials are proven to be important for providing proper cell-to-cell, cell-to-material interactions. Particularly, modification of implant surfaces with bioactive cues, especially cell adhesion molecules (CAMs) that capitalize on native neural adhesion mechanisms, are promising candidates in favor of providing efficient interfaces. Within this concept, this study utilized specific CAMs, namely N-Cadherin (Neural cadherin, N-Cad) and neural cell adhesion molecule (NCAM), to enhance neuron-electrode contact by mimicking the cell-to-ECM interactions for improving the survival of cells and promoting neurite outgrowth. For this purpose, representative gold electrode surfaces were modified with N-Cadherin, NCAM, and the mixture (1:1) of these molecules. Modifications were characterized, and the effect of surface modification on both differentiated and undifferentiated neuroblastoma SH-SY5Y cell lines were compared. The findings demonstrated the successful modification of these molecules which subsequently exhibited biocompatible properties as evidenced by the cell viability results. In cell culture experiments, the CAMs displayed promising results in promoting neurite outgrowth compared to conventional poly-l-lysine coated surfaces, especially NCAM and N-Cad/NCAM modified surfaces clearly showed significant improvement. Overall, this optimized approach is expected to provide an insight into the action mechanisms of cells against the local environment and advance processes for the fabrication of alternative neural interfaces.
Collapse
Affiliation(s)
- Bengu Aktas
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Alp Ozgun
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Bora Garipcan
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| |
Collapse
|
10
|
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
|
11
|
Li F, Gallego J, Tirko NN, Greaser J, Bashe D, Patel R, Shaker E, Van Valkenburg GE, Alsubhi AS, Wellman S, Singh V, Padill CG, Gheres KW, Bagwell R, Mulvihill M, Kozai TDY. Low-intensity pulsed ultrasound stimulation (LIPUS) modulates microglial activation following intracortical microelectrode implantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.05.570162. [PMID: 38105969 PMCID: PMC10723293 DOI: 10.1101/2023.12.05.570162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Microglia are important players in surveillance and repair of the brain. Their activation mediates neuroinflammation caused by intracortical microelectrode implantation, which impedes the application of intracortical brain-computer interfaces (BCIs). While low-intensity pulsed ultrasound stimulation (LIPUS) can attenuate microglial activation, its potential to modulate the microglia-mediated neuroinflammation and enhance the bio-integration of microelectrodes remains insufficiently explored. We found that LIPUS increased microglia migration speed from 0.59±0.04 to 1.35±0.07 µm/hr on day 1 and enhanced microglia expansion area from 44.50±6.86 to 93.15±8.77 µm 2 /min on day 7, indicating improved tissue healing and surveillance. Furthermore, LIPUS reduced microglial activation by 17% on day 6, vessel-associated microglia ratio from 70.67±6.15 to 40.43±3.87% on day 7, and vessel diameter by 20% on day 28. Additionally, microglial coverage of the microelectrode was reduced by 50% in week 1, indicating better tissue-microelectrode integration. These data reveal that LIPUS helps resolve neuroinflammation around chronic intracortical microelectrodes.
Collapse
|
12
|
Lee CH, Park YK, Lee K. Recent strategies for neural dynamics observation at a larger scale and wider scope. Biosens Bioelectron 2023; 240:115638. [PMID: 37647685 DOI: 10.1016/j.bios.2023.115638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/15/2023] [Accepted: 08/24/2023] [Indexed: 09/01/2023]
Abstract
The tremendous technical progress in neuroscience offers opportunities to observe a more minor or/and broader dynamic picture of the brain. Moreover, the large-scale neural activity of individual neurons enables the dissection of detailed mechanistic links between neural populations and behaviors. To measure neural activity in-vivo, multi-neuron recording, and neuroimaging techniques are employed and developed to acquire more neurons. The tools introduced concurrently recorded dozens to hundreds of neurons in the coordinated brain regions and elucidated the neuronal ensembles from a massive population perspective of diverse neurons at cellular resolution. In particular, the increasing spatiotemporal resolution of neuronal monitoring across the whole brain dramatically facilitates our understanding of additional nervous system functions in health and disease. Here, we will introduce state-of-the-art neuroscience tools involving large-scale neural population recording and the long-range connections spanning multiple brain regions. Their synergic effects provide to clarify the controversial circuitry underlying neuroscience. These challenging neural tools present a promising outlook for the fundamental dynamic interplay across levels of synaptic cellular, circuit organization, and brain-wide. Hence, more observations of neural dynamics will provide more clues to elucidate brain functions and push forward innovative technology at the intersection of neural engineering disciplines. We hope this review will provide insight into the use or development of recent neural techniques considering spatiotemporal scales of brain observation.
Collapse
Affiliation(s)
- Chang Hak Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Young Kwon Park
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea
| | - Kwang Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu, South Korea.
| |
Collapse
|
13
|
Woeppel K, Dhawan V, Shi D, Cui XT. Nanotopography-enhanced biomimetic coating maintains bioactivity after weeks of dry storage and improves chronic neural recording. Biomaterials 2023; 302:122326. [PMID: 37716282 PMCID: PMC10993103 DOI: 10.1016/j.biomaterials.2023.122326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
We developed a nanoparticle base layer technology capable of maintaining the bioactivity of protein-based neural probe coating intended to improve neural recording quality. When covalently bound on thiolated nanoparticle (TNP) modified surfaces, neural adhesion molecule L1 maintained bioactivity throughout 8 weeks of dry storage at room temperature, while those bound to unmodified surfaces lost 66% bioactivity within 3 days. We tested the TNP + L1 coating in mouse brains on two different neural electrode arrays after two different dry storage durations (3 and 28 days). The results show that dry-stored coating is as good as the freshly prepared, and even after 28 days of storage, the number of single units per channel and signal-to-noise ratio of the TNP + L1 coated arrays were significantly higher by 32% and 40% respectively than uncoated controls over 16 weeks. This nanoparticle base layer approach enables the dissemination of biomolecule-functionalized neural probes to users worldwide and may also benefit a broad range of applications that rely on surface-bound biomolecules.
Collapse
Affiliation(s)
- Kevin Woeppel
- University of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, PA, 15213, USA
| | - Vaishnavi Dhawan
- University of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, PA, 15213, USA
| | - Delin Shi
- University of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, PA, 15213, USA
| | - Xinyan Tracy Cui
- University of Pittsburgh, Department of Bioengineering, 4200 Fifth Avenue, Pittsburgh, PA, 15260, USA; Center for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, PA, 15213, USA; McGowan Institute for Regenerative Medicine, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
14
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. Biomaterials 2023; 301:122210. [PMID: 37413842 PMCID: PMC10528716 DOI: 10.1016/j.biomaterials.2023.122210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over the chronic implantation period. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Zheng N, Jiang Y, Jiang S, Kim J, Chen G, Li Y, Cheng J, Jia X, Yang C. Multifunctional Fiber-Based Optoacoustic Emitter as a Bidirectional Brain Interface. Adv Healthc Mater 2023; 12:e2300430. [PMID: 37451259 PMCID: PMC10592200 DOI: 10.1002/adhm.202300430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
A bidirectional brain interface with both "write" and "read" functions can be an important tool for fundamental studies and potential clinical treatments for neurological diseases. Herein, a miniaturized multifunctional fiber-based optoacoustic emitter (mFOE) is reported thatintegrates simultaneous optoacoustic stimulation for "write" and electrophysiology recording of neural circuits for "read". Because of the intrinsic ability of neurons to respond to acoustic wave, there is no requirement of the viral transfection. The orthogonality between optoacoustic waves and electrical field provides a solution to avoid the interference between electrical stimulation and recording. The stimulation function of the mFOE is first validated in cultured ratcortical neurons using calcium imaging. In vivo application of mFOE for successful simultaneous optoacoustic stimulation and electrical recording of brain activities is confirmed in mouse hippocampus in both acute and chronical applications up to 1 month. Minor brain tissue damage is confirmed after these applications. The capability of simultaneous neural stimulation and recording enabled by mFOE opens up new possibilities for the investigation of neural circuits and brings new insights into the study of ultrasound neurostimulation.
Collapse
Affiliation(s)
- Nan Zheng
- Division of Materials Science and EngineeringBoston UniversityBostonMA02215USA
| | - Ying Jiang
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
| | - Shan Jiang
- Bradley Department of Electrical and Computer EngineeringVirginia TechBlacksburgVA24061USA
| | - Jongwoon Kim
- Bradley Department of Electrical and Computer EngineeringVirginia TechBlacksburgVA24061USA
| | - Guo Chen
- Department of Electrical and Computer EngineeringBoston UniversityBostonMAUSA
| | - Yueming Li
- Department of Mechanical EngineeringBoston UniversityBostonMA02215USA
| | - Ji‐Xin Cheng
- Department of Biomedical EngineeringBoston UniversityBostonMA02215USA
- Department of Electrical and Computer EngineeringBoston UniversityBostonMAUSA
| | - Xiaoting Jia
- Bradley Department of Electrical and Computer EngineeringVirginia TechBlacksburgVA24061USA
- Department of Materials Science and EngineeringVirginia TechBlacksburgVA24061USA
| | - Chen Yang
- Department of Electrical and Computer EngineeringBoston UniversityBostonMAUSA
- Department of ChemistryBoston UniversityBostonMA02215USA
| |
Collapse
|
16
|
Ghazal M, Susloparova A, Lefebvre C, Daher Mansour M, Ghodhbane N, Melot A, Scholaert C, Guérin D, Janel S, Barois N, Colin M, Buée L, Yger P, Halliez S, Coffinier Y, Pecqueur S, Alibart F. Electropolymerization processing of side-chain engineered EDOT for high performance microelectrode arrays. Biosens Bioelectron 2023; 237:115538. [PMID: 37506488 DOI: 10.1016/j.bios.2023.115538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/04/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Microelectrode Arrays (MEAs) are popular tools for in vitro extracellular recording. They are often optimized by surface engineering to improve affinity with neurons and guarantee higher recording quality and stability. Recently, PEDOT:PSS has been used to coat microelectrodes due to its good biocompatibility and low impedance, which enhances neural coupling. Herein, we investigate on electro-co-polymerization of EDOT with its triglymated derivative to control valence between monomer units and hydrophilic functions on a conducting polymer. Molecular packing, cation complexation, dopant stoichiometry are governed by the glycolation degree of the electro-active coating of the microelectrodes. Optimal monomer ratio allows fine-tuning the material hydrophilicity and biocompatibility without compromising the electrochemical impedance of microelectrodes nor their stability while interfaced with a neural cell culture. After incubation, sensing readout on the modified electrodes shows higher performances with respect to unmodified electropolymerized PEDOT, with higher signal-to-noise ratio (SNR) and higher spike counts on the same neural culture. Reported SNR values are superior to that of state-of-the-art PEDOT microelectrodes and close to that of state-of-the-art 3D microelectrodes, with a reduced fabrication complexity. Thanks to this versatile technique and its impact on the surface chemistry of the microelectrode, we show that electro-co-polymerization trades with many-compound properties to easily gather them into single macromolecular structures. Applied on sensor arrays, it holds great potential for the customization of neurosensors to adapt to environmental boundaries and to optimize extracted sensing features.
Collapse
Affiliation(s)
- Mahdi Ghazal
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France
| | - Anna Susloparova
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France
| | - Camille Lefebvre
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Michel Daher Mansour
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France
| | - Najami Ghodhbane
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France
| | - Alexis Melot
- Laboratoire Nanotechnologies & Nanosystèmes (LN2, UMI 3463) | CNRS, Université de Sherbrooke, J1X0A5, Sherbrooke, Canada
| | - Corentin Scholaert
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France
| | - David Guérin
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France
| | - Sébastien Janel
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Nicolas Barois
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - Morvane Colin
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Pierre Yger
- Plasticity & SubjectivitY Team, Lille Neuroscience & Cognition Research Centre, University of Lille, INSERM U1172, Lille, France; Institut de La Vision, Sorbonne Université, INSERM, Centre National de La Recherche Scientifique, Paris, France
| | - Sophie Halliez
- University of Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Yannick Coffinier
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France.
| | - Sébastien Pecqueur
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France.
| | - Fabien Alibart
- Institute of Electronics, Microelectronics and Nanotechnology (IEMN, UMR 8520) | Univ. Lille, CNRS, Univ. Polytechnique Hauts-de-France, 59000, Lille, France; Laboratoire Nanotechnologies & Nanosystèmes (LN2, UMI 3463) | CNRS, Université de Sherbrooke, J1X0A5, Sherbrooke, Canada
| |
Collapse
|
17
|
Trotier A, Bagnoli E, Walski T, Evers J, Pugliese E, Lowery M, Kilcoyne M, Fitzgerald U, Biggs M. Micromotion Derived Fluid Shear Stress Mediates Peri-Electrode Gliosis through Mechanosensitive Ion Channels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301352. [PMID: 37518828 PMCID: PMC10520674 DOI: 10.1002/advs.202301352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/11/2023] [Indexed: 08/01/2023]
Abstract
The development of bioelectronic neural implant technologies has advanced significantly over the past 5 years, particularly in brain-machine interfaces and electronic medicine. However, neuroelectrode-based therapies require invasive neurosurgery and can subject neural tissues to micromotion-induced mechanical shear, leading to chronic inflammation, the formation of a peri-electrode void and the deposition of reactive glial scar tissue. These structures act as physical barriers, hindering electrical signal propagation and reducing neural implant functionality. Although well documented, the mechanisms behind the initiation and progression of these processes are poorly understood. Herein, in silico analysis of micromotion-induced peri-electrode void progression and gliosis is described. Subsequently, ventral mesencephalic cells exposed to milliscale fluid shear stress in vitro exhibited increased expression of gliosis-associated proteins and overexpression of mechanosensitive ion channels PIEZO1 (piezo-type mechanosensitive ion channel component 1) and TRPA1 (transient receptor potential ankyrin 1), effects further confirmed in vivo in a rat model of peri-electrode gliosis. Furthermore, in vitro analysis indicates that chemical inhibition/activation of PIEZO1 affects fluid shear stress mediated astrocyte reactivity in a mitochondrial-dependent manner. Together, the results suggest that mechanosensitive ion channels play a major role in the development of a peri-electrode void and micromotion-induced glial scarring at the peri-electrode region.
Collapse
Affiliation(s)
- Alexandre Trotier
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
- Galway Neuroscience CentreUniversity of GalwayGalwayH91 W2TYIreland
| | - Enrico Bagnoli
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
- Galway Neuroscience CentreUniversity of GalwayGalwayH91 W2TYIreland
| | - Tomasz Walski
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
- Department of Biomedical EngineeringFaculty of Fundamental Problems of TechnologyWrocław University of Science and TechnologyWroclaw50‐370Poland
| | - Judith Evers
- School of Electrical and Electronic EngineeringUniversity College DublinDublin 4Ireland
| | - Eugenia Pugliese
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
| | - Madeleine Lowery
- School of Electrical and Electronic EngineeringUniversity College DublinDublin 4Ireland
| | - Michelle Kilcoyne
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
- Galway Neuroscience CentreUniversity of GalwayGalwayH91 W2TYIreland
- Carbohydrate Signalling GroupDiscipline of MicrobiologyUniversity of GalwayGalwayH91 W2TYIreland
| | - Una Fitzgerald
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
- Galway Neuroscience CentreUniversity of GalwayGalwayH91 W2TYIreland
| | - Manus Biggs
- SFI Research Centre for Medical Devices (CÚRAM)University of GalwayGalwayH91 W2TYIreland
- Galway Neuroscience CentreUniversity of GalwayGalwayH91 W2TYIreland
| |
Collapse
|
18
|
Composite Fibrin and Carbon Microfibre Implant to Modulate Postraumatic Inflammation after Spinal Cord Injury. Cells 2023; 12:cells12060839. [PMID: 36980180 PMCID: PMC10047285 DOI: 10.3390/cells12060839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Poor functional recovery after spinal cord injury (SCI) drives the development of novel strategies to manage this devastating condition. We recently showed promising immunomodulatory and pro-regenerative actions of bio-functionalized carbon microfibres (MFs) implanted in a rodent model of SCI. In order to maximize tissue repair while easing MF implantation, we produced a composite implant based on the embedding of several MFs within a fibrin hydrogel. We used intravital imaging of fluorescent reporter mice at the early stages and spinal sections of the same animals 3 months later to characterize the neuroinflammatory response to the implant and its impact on axonal regeneration. Whereas fibrin alone was inert in the first week, its enzymatic degradation drove the chronic activation of microglial cells and axonal degeneration within 3 months. However, the presence of MFs inside the fibrin hydrogel slowed down fibrin degradation and boosted the early recruitment of immune cells. Noteworthy, there was an enhanced contribution of monocyte-derived dendritic cells (moDCs), preceding a faster transition toward an anti-inflammatory environment with increased axonal regeneration over 3 months. The inclusion of MF here ensured the long-term biocompatibility of fibrin hydrogels, which would otherwise preclude successful spinal cord regeneration.
Collapse
|
19
|
Sharon A, Jankowski MM, Shmoel N, Erez H, Spira ME. Significantly reduced inflammatory foreign-body-response to neuroimplants and improved recording performance in young compared to adult rats. Acta Biomater 2023; 158:292-307. [PMID: 36632879 DOI: 10.1016/j.actbio.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
The multicellular inflammatory encapsulation of implanted intracortical multielectrode arrays (MEA) is associated with severe deterioration of their field potentials' (FP) recording performance, which thus limits the use of brain implants in basic research and clinical applications. Therefore, extensive efforts have been made to identify the conditions in which the inflammatory foreign body response (FBR) is alleviated, or to develop methods to mitigate the formation of the inflammatory barrier. Here, for the first time, we show that (1) in young rats (74±8 gr, 4 weeks old at the onset of the experiments), cortical tissue recovery following MEA implantation proceeds with ameliorated inflammatory scar as compared to adult rats (242 ± 18 gr, 9 weeks old at the experimental onset); (2) in contrast to adult rats in which the Colony Stimulating factor 1 Receptor (CSF1R) antagonist chow eliminated ∼95% of the cortical microglia but not microglia adhering to the implant surfaces, in young rats the microglia adhering to the implant were eliminated along with the parenchymal microglia population. The removal of microglia adhering to the implant surfaces was correlated with improved recording performance by in-house fabricated Perforated Polyimide MEA Platforms (PPMP). These results support the hypothesis that microglia adhering to the surface of the electrodes, rather than the multicellular inflammatory scar, is the major underlying mechanism that deteriorates implant recording performance, and that young rats provide an advantageous model to study months-long, multisite electrophysiology in freely behaving rats. STATEMENT OF SIGNIFICANCE: Multisite electrophysiological recordings and stimulation devices play central roles in basic brain research and medical applications. The insertion of multielectrode-array platforms into the brain's parenchyma unavoidably injures the tissue, and initiates a multicellular inflammatory cascade culminating in the formation of an encapsulating scar tissue (the foreign body response-FBR). The dominant view, which directs most current research efforts to mitigate the FBR, holds that the FBR is the major hurdle to effective electrophysiological use of neuroprobes. By contrast, this report demonstrates that microglia adhering to the surface of a neuroimplants, rather than the multicellular FBR, underlie the performance deterioration of neuroimplants. These findings pave the way to the development of novel and focused strategies to overcome the functional deterioration of neuroimplants.
Collapse
Affiliation(s)
- Aviv Sharon
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maciej M Jankowski
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel; Edmond and Lily Safra Center for Brain Sciences, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nava Shmoel
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Erez
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Micha E Spira
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
20
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating Clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526463. [PMID: 36778360 PMCID: PMC9915570 DOI: 10.1101/2023.01.31.526463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over chronic implantation. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period. Abstract Figure
Collapse
|
21
|
The Interactions of the 70 kDa Fragment of Cell Adhesion Molecule L1 with Topoisomerase 1, Peroxisome Proliferator-Activated Receptor γ and NADH Dehydrogenase (Ubiquinone) Flavoprotein 2 Are Involved in Gene Expression and Neuronal L1-Dependent Functions. Int J Mol Sci 2023; 24:ijms24032097. [PMID: 36768419 PMCID: PMC9916828 DOI: 10.3390/ijms24032097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
The cell adhesion molecule L1 is essential not only for neural development, but also for synaptic functions and regeneration after trauma in adulthood. Abnormalities in L1 functions cause developmental and degenerative disorders. L1's functions critically depend on proteolysis which underlies dynamic cell interactions and signal transduction. We showed that a 70 kDa fragment (L1-70) supports mitochondrial functions and gene transcription. To gain further insights into L1-70's functions, we investigated several binding partners. Here we show that L1-70 interacts with topoisomerase 1 (TOP1), peroxisome proliferator-activated receptor γ (PPARγ) and NADH dehydrogenase (ubiquinone) flavoprotein 2 (NDUFV2). TOP1, PPARγ and NDUFV2 siRNAs reduced L1-dependent neurite outgrowth, and the topoisomerase inhibitors topotecan and irinotecan inhibited L1-dependent neurite outgrowth, neuronal survival and migration. In cultured neurons, L1 siRNA reduces the expression levels of the long autism genes neurexin-1 (Nrxn1) and neuroligin-1 (Nlgn1) and of the mitochondrially encoded gene NADH:ubiquinone oxidoreductase core subunit 2 (ND2). In mutant mice lacking L1-70, Nrxn1 and Nlgn1, but not ND2, mRNA levels are reduced. Since L1-70's interactions with TOP1, PPARγ and NDUFV2 contribute to the expression of two essential long autism genes and regulate important neuronal functions, we propose that L1 may not only ameliorate neurological problems, but also psychiatric dysfunctions.
Collapse
|
22
|
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
|
23
|
Kushwah N, Woeppel K, Dhawan V, Shi D, Cui XT. Effects of neuronal cell adhesion molecule L1 and nanoparticle surface modification on microglia. Acta Biomater 2022; 149:273-286. [PMID: 35764240 PMCID: PMC10018678 DOI: 10.1016/j.actbio.2022.06.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022]
Abstract
Microelectrode arrays for neural recording suffer from low yield and stability partly due to the inflammatory host responses. A neuronal cell adhesion molecule L1 coating has been shown to promote electrode-neuron integration, reduce microglia activation and improve recording. Coupling L1 to surface via a nanoparticle (NP) base layer further increased the protein surface density and stability. However, the exact L1-microglia interaction in these coatings has not been studied. Here we cultured primary microglia on L1 modified surfaces (with and without NP) and characterized microglia activation upon phorbol myristate acetate (PMA) and lipopolysaccharide (LPS) stimulation. Results showed L1 coatings reduced microglia's superoxide production in response to PMA and presented intrinsic antioxidant properties. Meanwhile, L1 decreased iNOS, NO, and pro-inflammatory cytokines (TNF alpha, IL-6, IL-1 beta), while increased anti-inflammatory cytokines (TGF beta 1, IL-10) in LPS stimulated microglia. Furthermore, L1 increased Arg-1 expression and phagocytosis upon LPS stimulation. Rougher NP surface showed lower number of microglia attached per area than their smooth counterpart, lower IL-6 release and superoxide production, and higher intrinsic reducing potential. Finally, we examined the effect of L1 and nanoparticle modifications on microglia response in vivo over 8 weeks with 2-photon imaging. Microglial coverage on the implant surface was found to be lower on the L1 modified substrates relative to unmodified, consistent with the in vitro observation. Our results indicate L1 significantly reduces superoxide production and inflammatory response of microglia and promotes wound healing, while L1 immobilization via a nanoparticle base layer brings added benefit without adverse effects. STATEMENT OF SIGNIFICANCE: Surface modification of microelectrode arrays with L1 has been shown to reduce microglia coverage on neural probe surface in vivo and improves neural recording, but the specific mechanism of action is not fully understood. The results in this study show that surface bound L1 reduces superoxide production from cultured microglia via direct reduction reaction and signaling pathways, increases anti-inflammatory cytokine release and phagocytosis in response to PMA or LPS stimulation. Additionally, roughening the surface with nanoparticles prior to L1 immobilization further increased the benefit of L1 in reducing microglia activation and oxidative stress. Together, our findings shed light on the mechanisms of action of nanotextured and neuroadhesive neural implant coatings and guide future development of seamless tissue interface.
Collapse
Affiliation(s)
- Neetu Kushwah
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Kevin Woeppel
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States
| | - Vaishnavi Dhawan
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States
| | - Delin Shi
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States
| | - Xinyan Tracy Cui
- Neural Tissue/Electrode Interface and Neural Tissue Engineering lab, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, United States; McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, United States; Center for Neural Basis of Cognition, Pittsburgh, PA 15213, United States.
| |
Collapse
|
24
|
Krahe DD, Woeppel KM, Yang Q, Kushwah N, Cui XT. Melatonin Decreases Acute Inflammatory Response to Neural Probe Insertion. Antioxidants (Basel) 2022; 11:antiox11081628. [PMID: 36009346 PMCID: PMC9405074 DOI: 10.3390/antiox11081628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022] Open
Abstract
Neural electrode insertion trauma impedes the recording and stimulation capabilities of numerous diagnostic and treatment avenues. Implantation leads to the activation of inflammatory markers and cell types, which is detrimental to neural tissue health and recording capabilities. Oxidative stress and inflammation at the implant site have been shown to decrease with chronic administration of antioxidant melatonin at week 16, but its effects on the acute landscape have not been studied. To assess the effect of melatonin administration in the acute phase, specifically the first week post-implantation, we utilized histological and q-PCR methods to quantify cellular and molecular indicators of inflammation and oxidative stress in the tissue surrounding implanted probes in C57BL/6 mice as well as two-photon microscopy to track the microglial responses to the probes in real-time in transgenic mice expressing GFP with CX3CR1 promotor. Histological results indicate that melatonin effectively maintained neuron density surrounding the electrode, inhibited accumulation and activation of microglia and astrocytes, and reduced oxidative tissue damage. The expression of the pro-inflammatory cytokines, TNF-α and IL-6, were significantly reduced in melatonin-treated animals. Additionally, microglial encapsulation of the implant surface was inhibited by melatonin as compared to control animals following implantation. Our results combined with previous research suggest that melatonin is a particularly suitable drug for modulating inflammatory activity around neural electrode implants both acutely and chronically, translating to more stable and reliable interfaces.
Collapse
Affiliation(s)
- Daniela D. Krahe
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Kevin M. Woeppel
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Center for the Neural Basis of Cognition, Pittsburgh, PA 15213, USA
| | - Qianru Yang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Center for the Neural Basis of Cognition, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA
| | - Neetu Kushwah
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Center for the Neural Basis of Cognition, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA 15219, USA
- Correspondence:
| |
Collapse
|
25
|
Long-term microglial phase-specific dynamics during single vessel occlusion and recanalization. Commun Biol 2022; 5:841. [PMID: 35986097 PMCID: PMC9391347 DOI: 10.1038/s42003-022-03784-0] [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: 11/25/2021] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
Vascular occlusion leading to brain dysfunctions is usually considered evoking microglia-induced inflammation response. However, it remains unclear how microglia interact with blood vessels in the development of vascular occlusion-related brain disorders. Here, we illuminate long-term spatiotemporal dynamics of microglia during single vessel occlusion and recanalization. Microglia display remarkable response characteristics in different phases, including acute reaction, rapid diffusion, transition and chronic effect. Fibrinogen-induced microglial cluster promotes major histocompatibility complex II (MHCII) expression. Microglial soma represents a unique filament-shape migration and has slower motility compared to the immediate reaction of processes to occlusion. We capture proliferative microglia redistribute territory. Microglial cluster resolves gradually and microglia recover to resting state both in the morphology and function in the chronic effect phase. Therefore, our study offers a comprehensive analysis of spatiotemporal dynamics of microglia and potential mechanisms to both vessel occlusion and recanalization. Microglial phase-specific response suggests the morphological feature-oriented phased intervention would be an attractive option for vascular occlusion-related diseases treatments. The spatiotemporal dynamics of the microglial inflammatory response to single vessel occlusion and recanalization are analysed, revealing four different response phases.
Collapse
|
26
|
Parylene C as an Insulating Polymer for Implantable Neural Interfaces: Acute Electrochemical Impedance Behaviors in Saline and Pig Brain In Vitro. Polymers (Basel) 2022; 14:polym14153033. [PMID: 35893997 PMCID: PMC9332801 DOI: 10.3390/polym14153033] [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: 06/10/2022] [Revised: 07/09/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Parylene is used as encapsulating material for medical devices due to its excellent biocompatibility and insulativity. Its performance as the insulating polymer of implantable neural interfaces has been studied in electrolyte solutions and in vivo. Biological tissue in vitro, as a potential environment for characterization and application, is convenient to access in the fabrication lab of polymer and neural electrodes, but there has been little study investigating the behaviors of Parylene in the tissue in vitro. Here, we investigated the electrochemical impedance behaviors of Parylene C polymer coating both in normal saline and in a chilled pig brain in vitro by performing electrochemical impedance spectroscopy (EIS) measurements of platinum (Pt) wire neural electrodes. The electrochemical impedance at the representative frequencies is discussed, which helps to construct the equivalent circuit model. Statistical analysis of fitted parameters of the equivalent circuit model showed good reliability of Parylene C as an insulating polymer in both electrolyte models. The electrochemical impedance measured in pig brain in vitro shows marked differences from that of saline.
Collapse
|
27
|
Zheng XS, Yang Q, Vazquez A, Cui XT. Imaging the stability of chronic electrical microstimulation using electrodes coated with PEDOT/CNT and iridium oxide. iScience 2022; 25:104539. [PMID: 35769881 PMCID: PMC9234710 DOI: 10.1016/j.isci.2022.104539] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/22/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic microstimulation is faced with challenges that require an additional understanding of stability and safety. We implanted silicon arrays coated with poly(3,4-ethylenedioxythiophene) (PEDOT)/Carbon Nanotubes (CNT), or PCand IrOx into the cortex of GCaMP6s mice and electrically stimulated them for up to 12 weeks. We quantified neuronal responses to stimulation using two-photon imaging and mesoscale fluorescence microscopy and characterized electrode performance over time. We observed dynamic changes in stimulation stability over time and a significant advantage in energy efficiency using PC coated electrodes over IrOx coated electrodes. In a subset of mice, we observed abnormal ictal cortical responses or cortical spreading depression using stimulation parameters commonly used in intracortical stimulation applications, suggesting the need to investigate the potential neuronal damage and redefine the stimulation safety limit. This study not only revealed the dynamic changes in stimulation efficiency after implantation but also reiterates the potential for PC as a high-efficiency material in chronic neuromodulation.
Collapse
Affiliation(s)
- Xin Sally Zheng
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Qianru Yang
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
- Center for Neural Basis of Cognition, 115 Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Alberto Vazquez
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
- Center for Neural Basis of Cognition, 115 Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
- Department of Radiology, University of Pittsburgh, 200 Lothrop St, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, 3025 East Carson Street, Pittsburgh, PA 15219, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, 5057 Biomedical Science Tower 3, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
- Center for Neural Basis of Cognition, 115 Mellon Institute, 4400 Fifth Avenue, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, 3025 East Carson Street, Pittsburgh, PA 15219, USA
| |
Collapse
|
28
|
Abstract
Neuroprosthetic devices that record and modulate neural activities have demonstrated immense potential for bypassing or restoring lost neurological functions due to neural injuries and disorders. However, implantable electrical devices interfacing with brain tissue are susceptible to a series of inflammatory tissue responses along with mechanical or electrical failures which can affect the device performance over time. Several biomaterial strategies have been implemented to improve device-tissue integration for high quality and stable performance. Ranging from developing smaller, softer, and more flexible electrode designs to introducing bioactive coatings and drug-eluting layers on the electrode surface, such strategies have shown different degrees of success but with limitations. With their hydrophilic properties and specific bioactivities, carbohydrates offer a potential solution for addressing some of the limitations of the existing biomolecular approaches. In this review, we summarize the role of polysaccharides in the central nervous system, with a primary focus on glycoproteins and proteoglycans, to shed light on their untapped potential as biomaterials for neural implants. Utilization of glycosaminoglycans for neural interface and tissue regeneration applications is comprehensively reviewed to provide the current state of carbohydrate-based biomaterials for neural implants. Finally, we will discuss the challenges and opportunities of applying carbohydrate-based biomaterials for neural tissue interfaces.
Collapse
Affiliation(s)
- Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Urdaneta ME, Kunigk NG, Currlin S, Delgado F, Fried SI, Otto KJ. The Long-Term Stability of Intracortical Microstimulation and the Foreign Body Response Are Layer Dependent. Front Neurosci 2022; 16:908858. [PMID: 35769707 PMCID: PMC9234554 DOI: 10.3389/fnins.2022.908858] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Intracortical microstimulation (ICMS) of the somatosensory cortex (S1) can restore sensory function in patients with paralysis. Studies assessing the stability of ICMS have reported heterogeneous responses across electrodes and over time, potentially hindering the implementation and translatability of these technologies. The foreign body response (FBR) and the encapsulating glial scar have been associated with a decay in chronic performance of implanted electrodes. Moreover, the morphology, intrinsic properties, and function of cells vary across cortical layers, each potentially affecting the sensitivity to ICMS as well as the degree of the FBR across cortical depth. However, layer-by-layer comparisons of the long-term stability of ICMS as well as the extent of the astrocytic glial scar change across cortical layers have not been well explored. Here, we implanted silicon microelectrodes with electrode sites spanning all the layers of S1 in rats. Using a behavioral paradigm, we obtained ICMS detection thresholds from all cortical layers for up to 40 weeks. Our results showed that the sensitivity and long-term performance of ICMS is indeed layer dependent. Overall, detection thresholds decreased during the first 7 weeks post-implantation (WPI). This was followed by a period in which thresholds remained stable or increased depending on the interfacing layer: thresholds in L1 and L6 exhibited the most consistent increases over time, while those in L4 and L5 remained the most stable. Furthermore, histological investigation of the tissue surrounding the electrode showed a biological response of microglia and macrophages which peaked at L1, while the area of the astrocytic glial scar peaked at L2/3. Interestingly, the biological response of these FBR markers is less exacerbated at L4 and L5, suggesting a potential link between the FBR and the long-term stability of ICMS. These findings suggest that interfacing depth can play an important role in the design of chronically stable implantable microelectrodes.
Collapse
Affiliation(s)
- Morgan E. Urdaneta
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- *Correspondence: Morgan E. Urdaneta,
| | - Nicolas G. Kunigk
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Seth Currlin
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
| | - Francisco Delgado
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Shelley I. Fried
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Boston Veterans Affairs Healthcare System, Boston, MA, United States
| | - Kevin J. Otto
- Department of Neuroscience, University of Florida, Gainesville, FL, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, United States
- Department of Neurology, University of Florida, Gainesville, FL, United States
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, United States
- Kevin J. Otto,
| |
Collapse
|
30
|
Rommelfanger NJ, Keck CH, Chen Y, Hong G. Learning from the brain's architecture: bioinspired strategies towards implantable neural interfaces. Curr Opin Biotechnol 2021; 72:8-12. [PMID: 34365114 PMCID: PMC8671194 DOI: 10.1016/j.copbio.2021.07.020] [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: 02/28/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 11/16/2022]
Abstract
While early neural interfaces consisted of rigid, monolithic probes, recent implantable technologies include meshes, gels, and threads that imitate various properties of the neural tissue itself. Such mimicry brings new capabilities to the traditional electrophysiology toolbox, with benefits for both neuroscience studies and clinical treatments. Specifically, by matching the multi-dimensional mechanical properties of the brain, neural implants can preserve the endogenous environment while functioning over chronic timescales. Further, topological mimicry of neural structures enables seamless integration into the tissue and provides proximal access to neurons for high-quality recordings. Ultimately, we envision that neuromorphic devices incorporating functional, mechanical, and topological mimicry of the brain may facilitate stable operation of advanced brain machine interfaces with minimal disruption of the native tissue.
Collapse
Affiliation(s)
- Nicholas J Rommelfanger
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Carl Hc Keck
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Yihang Chen
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
Shi D, Dhawan V, Cui XT. Bio-integrative design of the neural tissue-device interface. Curr Opin Biotechnol 2021; 72:54-61. [PMID: 34710753 PMCID: PMC8671324 DOI: 10.1016/j.copbio.2021.10.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/19/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
Neural implants enable bidirectional communications with nervous tissue and have demonstrated tremendous potential in research and clinical applications. To obtain high fidelity and stable information exchange, we need to minimize the undesired host responses and achieve intimate neuron-device interaction. This paper highlights the key bio-integrative strategies aimed at seamless integration through intelligent device designs to minimize the immune responses, as well as incorporate bioactive elements to actively modulate cellular reactions. These approaches span from surface modification and bioactive agent delivery, to biomorphic and biohybrid designs. Many of these strategies have shown effectiveness in functional outcome measures, others are exploratory but with fascinating potentials. The combination of bio-integrative strategies may synergistically promote the next generation of neural interfaces.
Collapse
Affiliation(s)
- Delin Shi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; Center for Neural Basis of Cognition, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States.
| |
Collapse
|
32
|
Clay M, Monbouquette HG. Simulated Performance of Electroenzymatic Glutamate Biosensors In Vivo Illuminates the Complex Connection to Calibration In Vitro. ACS Chem Neurosci 2021; 12:4275-4285. [PMID: 34734695 DOI: 10.1021/acschemneuro.1c00365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Detailed simulations show that the relationship between electroenzymatic glutamate (Glut) sensor performance in vitro and that modeled in vivo is complicated by the influence of both resistances to mass transfer and clearance rates of Glut and H2O2 in the brain extracellular space (ECS). Mathematical modeling provides a powerful means to illustrate how these devices are expected to respond to a variety of conditions in vivo in ways that cannot be accomplished readily using existing experimental techniques. Through the use of transient model simulations in one spatial dimension, it is shown that the sensor response in vivo may exhibit much greater dependence on H2O2 mass transfer and clearance in the surrounding tissue than previously thought. This dependence may lead to sensor signals more than double the expected values (based on prior sensor calibration in vitro) for Glut release events within a few microns of the sensor surface. The sensor response in general is greatly affected by the distance between the device and location of Glut release, and apparent concentrations reported by simulated sensors consistently are well below the actual Glut levels for events occurring at distances greater than a few microns. Simulations of transient Glut concentrations, including a physiologically relevant bolus release, indicate that detection of Glut signaling likely is limited to events within 30 μm of the sensor surface based on representative sensor detection limits. It follows that important limitations also exist with respect to interpretation of decays in sensor signals, including relation of such data to actual Glut concentration declines in vivo. Thus, the use of sensor signal data to determine quantitatively the rates of Glut uptake from the brain ECS likely is problematic. The model is designed to represent a broad range of relevant physiological conditions, and although limited to one dimension, provides much needed guidance regarding the interpretation in general of electroenzymatic sensor data gathered in vivo.
Collapse
Affiliation(s)
- Mackenzie Clay
- Chemical and Biomolecular Engineering Department, University of California, Los Angeles, California 90095−1592, United States
| | - Harold G. Monbouquette
- Chemical and Biomolecular Engineering Department, University of California, Los Angeles, California 90095−1592, United States
| |
Collapse
|
33
|
Long-term in vivo two-photon imaging of the neuroinflammatory response to intracortical implants and micro-vessel disruptions in awake mice. Biomaterials 2021; 276:121060. [PMID: 34419839 DOI: 10.1016/j.biomaterials.2021.121060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/16/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022]
Abstract
Our understanding of biomaterials in the brain have been greatly enhanced by advancements in in vivo imaging technologies such as two-photon microscopy. However, when applied to chronic studies, two-photon microscopy enables high-resolution imaging only in superficial regions due to inflammatory responses introduced by the craniotomy and insertion of foreign biomaterials. Microprisms provide a unique vertical view from brain surface to ~1 mm deep or more (depending on the size of the microprisms) which may break through this limitation on imaging depth. Although microprism has been used in the field of neuroscience, the in vivo foreign body responses to the microprism implant have yet to be fully elucidated. This is of important concern in broader applications of this approach, especially for neuroinflammation-sensitive studies. In this work, we first assessed the activation of microglia/macrophages for 16 weeks after microprism implantation using two-photon microscopy in awake CX3CR1-GFP mice. The imaging window became clear from bleedings after ~2 weeks and the maximum imaging distance (in the horizontal direction) stabilized at around 500 μm after ~5 weeks. We also quantified the microglial morphology from week 3 to week 16 post-implantation. Compared to non-implant controls, microglia near the microprism showed higher cell density, smaller soma, and shorter and less branched processes in the early-chronic phase. After week 5, microglial morphology further than 100 μm from the microprism was generally similar to microglia in the control group. In addition, time-lapse imaging confirmed that microglial processes were surveying normally from week 3, even for microglia as close as 50 μm away. These morphological analyses and dynamic imaging results suggest that microglia around chronically implanted microprism eventually exhibit inactive phenotypes. Next, we examined microglial/macrophage responses following laser induced micro-vessel disruptions as an example application of microprism implantation for neuroinflammation related studies. Through the microprism, we captured microglial/macrophage polarization and migration, as well as blood flow changes after the insult for additional 16 weeks. To our surprise, microglia/macrophage aggregation around the insult site was sustained over the 16-week observation period. This work demonstrates the feasibility of using microprisms for long-term characterizations of inflammatory responses to other injuries including implantable devices at deeper depths than that achievable by conventional two-photon microscopy.
Collapse
|
34
|
Woeppel KM, Cui XT. Nanoparticle and Biomolecule Surface Modification Synergistically Increases Neural Electrode Recording Yield and Minimizes Inflammatory Host Response. Adv Healthc Mater 2021; 10:e2002150. [PMID: 34190425 DOI: 10.1002/adhm.202002150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/08/2021] [Indexed: 11/08/2022]
Abstract
Due to their ability to interface with neural tissues, neural electrodes are the key tool used for neurophysiological studies, electrochemical detection, brain computer interfacing, and countless neuromodulation therapies and diagnostic procedures. However, the long-term applications of neural electrodes are limited by the inflammatory host tissue response, decreasing detectable electrical signals, and insulating the device from the native environment. Surface modification methods are proposed to limit these detrimental responses but each has their own limitations. Here, a combinatorial approach is presented toward creating a stable interface between the electrode and host tissues. First, a thiolated nanoparticle (TNP) coating is utilized to increase the surface area and roughness. Next, the neural adhesion molecule L1 is immobilized to the nanoparticle modified substrate. In vitro, the combined nanotopographical and bioactive modifications (TNP+L1) elevate the bioactivity of L1, which is maintained for 28 d. In vivo, TNP+L1 modification improves the recording performance of the neural electrode arrays compared to TNP or L1 modification alone. Postmortem histology reveals greater neural cell density around the TNP+L1 coating while eliminating any inflammatory microglial encapsulation after 4 weeks. These results demonstrate that nanotopographical and bioactive modifications synergistically produce a seamless neural tissue interface for chronic neural implants.
Collapse
Affiliation(s)
- Kevin M. Woeppel
- Department of Bioengineering University of Pittsburgh Pittsburgh PA 15260 USA
- Center for the Neural basis of Cognition Pittsburgh PA 15260 USA
| | - Xinyan Tracy Cui
- Department of Bioengineering University of Pittsburgh Pittsburgh PA 15260 USA
- Center for the Neural basis of Cognition Pittsburgh PA 15260 USA
- McGowan Institute for Regenerative Medicine Pittsburgh PA 15260 USA
| |
Collapse
|
35
|
Zheng XS, Tan C, Castagnola E, Cui XT. Electrode Materials for Chronic Electrical Microstimulation. Adv Healthc Mater 2021; 10:e2100119. [PMID: 34029008 PMCID: PMC8257249 DOI: 10.1002/adhm.202100119] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/20/2021] [Indexed: 02/06/2023]
Abstract
Electrical microstimulation has enabled partial restoration of vision, hearing, movement, somatosensation, as well as improving organ functions by electrically modulating neural activities. However, chronic microstimulation is faced with numerous challenges. The implantation of an electrode array into the neural tissue triggers an inflammatory response, which can be exacerbated by the delivery of electrical currents. Meanwhile, prolonged stimulation may lead to electrode material degradation., which can be accelerated by the hostile inflammatory environment. Both material degradation and adverse tissue reactions can compromise stimulation performance over time. For stable chronic electrical stimulation, an ideal microelectrode must present 1) high charge injection limit, to efficiently deliver charge without exceeding safety limits for both tissue and electrodes, 2) small size, to gain high spatial selectivity, 3) excellent biocompatibility that ensures tissue health immediately next to the device, and 4) stable in vivo electrochemical properties over the application period. In this review, the challenges in chronic microstimulation are described in detail. To aid material scientists interested in neural stimulation research, the in vitro and in vivo testing methods are introduced for assessing stimulation functionality and longevity and a detailed overview of recent advances in electrode material research and device fabrication for improving chronic microstimulation performance is provided.
Collapse
Affiliation(s)
- Xin Sally Zheng
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Ave. Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Chao Tan
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Ave. Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Elisa Castagnola
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Ave. Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Ave. Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
36
|
Costanza A, Radomska M, Bondolfi G, Zenga F, Amerio A, Aguglia A, Serafini G, Amore M, Berardelli I, Pompili M, Nguyen KD. Suicidality Associated With Deep Brain Stimulation in Extrapyramidal Diseases: A Critical Review and Hypotheses on Neuroanatomical and Neuroimmune Mechanisms. Front Integr Neurosci 2021; 15:632249. [PMID: 33897384 PMCID: PMC8060445 DOI: 10.3389/fnint.2021.632249] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Deep brain stimulation (DBS) is a very well-established and effective treatment for patients with extrapyramidal diseases. Despite its generally favorable clinical efficacy, some undesirable outcomes associated with DBS have been reported. Among such complications are incidences of suicidal ideation (SI) and behavior (SB) in patients undergoing this neurosurgical procedure. However, causal associations between DBS and increased suicide risk are not demonstrated and they constitute a debated issue. In light of these observations, the main objective of this work is to provide a comprehensive and unbiased overview of the literature on suicide risk in patients who received subthalamic nucleus (STN) and internal part of globus pallidum (GPi) DBS treatment. Additionally, putative mechanisms that might be involved in the development of SI and SB in these patients as well as caveats associated with these hypotheses are introduced. Finally, we briefly propose some clinical implications, including therapeutic strategies addressing these potential disease mechanisms. While a mechanistic connection between DBS and suicidality remains a controversial topic that requires further investigation, it is of critical importance to consider suicide risk as an integral component of candidate selection and post-operative care in DBS.
Collapse
Affiliation(s)
- Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland.,Department of Psychiatry, ASO Santi Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Italy
| | - Michalina Radomska
- Faculty of Psychology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Guido Bondolfi
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland.,Department of Psychiatry, Service of Liaison Psychiatry and Crisis Intervention (SPLIC), Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Francesco Zenga
- Department of Neurosurgery, University and City of Health and Science Hospital, Turin, Italy
| | - Andrea Amerio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Mood Disorders Program, Tufts Medical Center, Boston, MA, United States
| | - Andrea Aguglia
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Serafini
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Isabella Berardelli
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Khoa D Nguyen
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, United States.,Tranquis Therapeutics, Palo Alto, CA, United States.,Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
37
|
Sharon A, Jankowski MM, Shmoel N, Erez H, Spira ME. Inflammatory Foreign Body Response Induced by Neuro-Implants in Rat Cortices Depleted of Resident Microglia by a CSF1R Inhibitor and Its Implications. Front Neurosci 2021; 15:646914. [PMID: 33841088 PMCID: PMC8032961 DOI: 10.3389/fnins.2021.646914] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/25/2021] [Indexed: 12/30/2022] Open
Abstract
Inflammatory encapsulation of implanted cortical-neuro-probes [the foreign body response (FBR)] severely limits their use in basic brain research and in clinical applications. A better understanding of the inflammatory FBR is needed to effectively mitigate these critical limitations. Combining the use of the brain permeant colony stimulating factor 1 receptor inhibitor PLX5622 and a perforated polyimide-based multielectrode array platform (PPMP) that can be sectioned along with the surrounding tissue, we examined the contribution of microglia to the formation of inflammatory FBR. To that end, we imaged the inflammatory processes induced by PPMP implantations after eliminating 89-94% of the cortical microglia by PLX5622 treatment. The observations showed that: (I) inflammatory encapsulation of implanted PPMPs proceeds by astrocytes in microglia-free cortices. The activated astrocytes adhered to the PPMP's surfaces. This suggests that the roles of microglia in the FBR might be redundant. (II) PPMP implantation into control or continuously PLX5622-treated rats triggered a localized surge of microglia mitosis. The daughter cells that formed a "cloud" of short-lived (T 1 / 2 ≤ 14 days) microglia around and in contact with the implant surfaces were PLX5622 insensitive. (III) Neuron degeneration by PPMP implantation and the ensuing recovery in time, space, and density progressed in a similar manner in the cortices following 89-94% depletion of microglia. This implies that microglia do not serve a protective role with respect to the neurons. (IV) Although the overall cell composition and dimensions of the encapsulating scar in PLX5622-treated rats differed from the controls, the recorded field potential (FP) qualities and yield were undistinguishable. This is accounted for by assuming that the FP amplitudes in the control and PLX5622-treated rats were related to the seal resistance formed at the interface between the adhering microglia and/or astrocytes and the PPMP platform rather than across the scar tissue. These observations suggest that the prevention of both astrocytes and microglia adhesion to the electrodes is required to improve FP recording quality and yield.
Collapse
Affiliation(s)
- Aviv Sharon
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maciej M. Jankowski
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nava Shmoel
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Erez
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Micha E. Spira
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
38
|
Dryg I, Xie Y, Bergmann M, Urban G, Shain W, Hofmann UG. Long-term in vivomonitoring of gliotic sheathing of ultrathin entropic coated brain microprobes with fiber-based optical coherence tomography. J Neural Eng 2021; 18. [PMID: 33657543 DOI: 10.1088/1741-2552/abebc2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/03/2021] [Indexed: 11/11/2022]
Abstract
Objective.Microfabricated neuroprosthetic devices have made possible important observations on neuron activity; however, long-term high-fidelity recording performance of these devices has yet to be realized. Tissue-device interactions appear to be a primary source of lost recording performance. The current state of the art for visualizing the tissue response surrounding brain implants in animals is immunohistochemistry + confocal microscopy, which is mainly performed after sacrificing the animal. Monitoring the tissue response as it develops could reveal important features of the response which may inform improvements in electrode design.Approach.Optical coherence tomography (OCT), an imaging technique commonly used in ophthalmology, has already been adapted for imaging of brain tissue. Here, we use OCT to achieve real-time,in vivomonitoring of the tissue response surrounding chronically implanted neural devices. The employed tissue-response-provoking implants are coated with a plasma-deposited nanofilm, which has been demonstrated as a biocompatible and anti-inflammatory interface for indwelling devices. We evaluate the method by comparing the OCT results to traditional histology qualitatively and quantitatively.Main results.The differences in OCT signal across the implantation period between the plasma group and the control reveal that the plasma-type coating of otherwise rigid brain probes (glass) only slightly improve the glial encapsulation in the brain parenchyma indicating that geometrical or mechanical influences are dominating the encapsulation process.Significance.Our approach can long-term monitor and compare the tissue-response to chronically-implanted neural probes with and withour plasma coating in living animal models. Our findings provide valuable insigh to the well acknowledged yet not solved challenge.
Collapse
Affiliation(s)
- Ian Dryg
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Yijing Xie
- Section for Neuroelectronic Systems, Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany
| | - Michael Bergmann
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
| | - Gerald Urban
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
| | - William Shain
- Department of Bioengineering, University of Washington, Seattle, WA, United States of America.,Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States of America
| | - Ulrich G Hofmann
- Section for Neuroelectronic Systems, Department of Neurosurgery, Medical Center, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
39
|
Khodadadei F, Liu AP, Harris CA. A high-resolution real-time quantification of astrocyte cytokine secretion under shear stress for investigating hydrocephalus shunt failure. Commun Biol 2021; 4:387. [PMID: 33758339 PMCID: PMC7988003 DOI: 10.1038/s42003-021-01888-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/11/2021] [Indexed: 12/18/2022] Open
Abstract
It has been hypothesized that physiological shear forces acting on medical devices implanted in the brain significantly accelerate the rate to device failure in patients with chronically indwelling neuroprosthetics. In hydrocephalus shunt devices, shear forces arise from cerebrospinal fluid flow. The shunt's unacceptably high failure rate is mostly due to obstruction with adherent inflammatory cells. Astrocytes are the dominant cell type bound directly to obstructing shunts, rapidly manipulating their activation via shear stress-dependent cytokine secretion. Here we developed a total internal reflection fluorescence microscopy combined with a microfluidic shear device chip (MSDC) for quantitative analysis and direct spatial-temporal mapping of secreted cytokines at the single-cell level under physiological shear stress to identify the root cause for shunt failure. Real-time secretion imaging at 1-min time intervals enabled successful detection of a significant increase of astrocyte IL-6 cytokine secretion under shear stress greater than 0.5 dyne/cm2, validating our hypothesis and highlighting the importance of reducing shear stress activation of cells.
Collapse
Affiliation(s)
- Fatemeh Khodadadei
- Dept. of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Allen P Liu
- Dept. of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Dept. of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Dept. of Biophysics, University of Michigan, Ann Arbor, MI, USA
- Dept. of Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Carolyn A Harris
- Dept. of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
- Dept. of Biomedical Engineering, Wayne State University, Detroit, MI, USA.
- Dept. of Neurosurgery, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
40
|
Dubaniewicz M, Eles JR, Lam S, Song S, Cambi F, Sun D, Wellman SM, Kozai TDY. Inhibition of Na +/H +exchanger modulates microglial activation and scar formation following microelectrode implantation. J Neural Eng 2021; 18. [PMID: 33621208 DOI: 10.1088/1741-2552/abe8f1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/23/2021] [Indexed: 11/12/2022]
Abstract
Objective.Intracortical microelectrodes are an important tool for neuroscience research and have great potential for clinical use. However, the use of microelectrode arrays to treat neurological disorders and control prosthetics is limited by biological challenges such as glial scarring, which can impair chronic recording performance. Microglia activation is an early and prominent contributor to glial scarring. After insertion of an intracortical microelectrode, nearby microglia transition into a state of activation, migrate, and encapsulate the device. Na+/H+exchanger isoform-1 (NHE-1) is involved in various microglial functions, including their polarity and motility, and has been implicated in pro-inflammatory responses to tissue injury. HOE-642 (cariporide) is an inhibitor of NHE-1 and has been shown to depress microglial activation and inflammatory response in brain injury models.Approach.In this study, the effects of HOE-642 treatment on microglial interactions to intracortical microelectrodes was evaluated using two-photon microscopyin vivo.Main results.The rate at which microglia processes and soma migrate in response to electrode implantation was unaffected by HOE-642 administration. However, HOE-642 administration effectively reduced the radius of microglia activation at 72 h post-implantation from 222.2µm to 177.9µm. Furthermore, treatment with HOE-642 significantly reduced microglial encapsulation of implanted devices at 5 h post-insertion from 50.7 ± 6.0% to 8.9 ± 6.1%, which suggests an NHE-1-specific mechanism mediating microglia reactivity and gliosis during implantation injury.Significance.This study implicates NHE-1 as a potential target of interest in microglial reactivity and HOE-642 as a potential treatment to attenuate the glial response and scar formation around implanted intracortical microelectrodes.
Collapse
Affiliation(s)
- Mitchell Dubaniewicz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Stephanie Lam
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, United States of America.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Dandan Sun
- Veterans Administration Pittsburgh, Pittsburgh, PA, United States of America.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.,NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
41
|
Eles JR, Stieger KC, Kozai TDY. The temporal pattern of Intracortical Microstimulation pulses elicits distinct temporal and spatial recruitment of cortical neuropil and neurons. J Neural Eng 2020; 18. [PMID: 33075762 DOI: 10.1088/1741-2552/abc29c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The spacing or distribution of stimulation pulses of therapeutic neurostimulation waveforms-referred to here as the Temporal Pattern (TP)-has emerged as an important parameter for tuning the response to deep-brain stimulation and intracortical microstimulation (ICMS). While it has long been assumed that modulating the TP of ICMS may be effective by altering the rate coding of the neural response, it is unclear how it alters the neural response at the neural network level. The present study is designed to elucidate the neural response to TP at the network level. APPROACH We use in vivo two-photon imaging of ICMS in mice expressing the calcium sensor Thy1-GCaMP or the glutamate sensor hSyn-iGluSnFr to examine the layer II/III neural response to stimulations with different TPs. We study the neuronal calcium and glutamate response to TPs with the same average frequency (10Hz) and same total charge injection, but varying degrees of bursting. We also investigate one control pattern with an average frequency of 100Hz and 10X the charge injection. MAIN RESULTS Stimulation trains with the same average frequency (10 Hz) and same total charge injection but distinct temporal patterns recruits distinct sets of neurons. More-than-half (60% of 309 cells) prefer one temporal pattern over the other. Despite their distinct spatial recruitment patterns, both cells exhibit similar ability to follow 30s trains of both TPs without failing, and they exhibit similar levels of glutamate release during stimulation. Both neuronal calcium and glutamate release train to the bursting TP pattern (~21-fold increase in relative power at the frequency of bursting. Bursting also results in a statistically significant elevation in the correlation between somatic calcium activity and neuropil activity, which we explore as a metric for inhibitory-excitatory tone. Interestingly, soma-neuropil correlation during the bursting pattern is a statistically significant predictor of cell preference for TP, which exposes a key link between inhibitory-excitatory tone. Finally, using mesoscale imaging, we show that both TPs result in distal inhibition during stimulation, which reveals complex spatial and temporal interactions between temporal pattern and inhibitory-excitatory tone in ICMS. SIGNIFICANCE Our results may ultimately suggest that TP is a valuable parameter space to modulate inhibitory-excitatory tone as well as distinct network activity in ICMS. This presents a broader mechanism of action than rate coding, as previously thought. By implicating these additional mechanisms, TP may have broader utility in the clinic and should be pursued to expand the efficacy of ICMS therapies.
Collapse
Affiliation(s)
- James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Kevin C Stieger
- Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, UNITED STATES
| | - Takashi D Yoshida Kozai
- Department of Bioengineering, University of Pittsburgh, 3501 Fifth Ave, 5059-BST3, Pittsburgh, PA 15213, USA, Pittsburgh, Pennsylvania, UNITED STATES
| |
Collapse
|
42
|
Huang SH, Shmoel N, Jankowski MM, Erez H, Sharon A, Abu-Salah W, Nelken I, Weiss A, Spira ME. Immunohistological and Ultrastructural Study of the Inflammatory Response to Perforated Polyimide Cortical Implants: Mechanisms Underlying Deterioration of Electrophysiological Recording Quality. Front Neurosci 2020; 14:926. [PMID: 32982683 PMCID: PMC7489236 DOI: 10.3389/fnins.2020.00926] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The deterioration of field potential (FP) recording quality and yield by in vivo multielectrode arrays (MEA) within days to weeks of implantation severely limits progress in basic and applied brain research. The prevailing hypothesis is that implantation of MEA platforms initiate and perpetuate inflammatory processes which culminate in the formation of scar tissue (the foreign body response, FBR) around the implant. The FBR leads to progressive degradation of the recording qualities by displacing neurons away from the electrode surfaces, increasing the resistance between neurons (current source) and the sensing pads and by reducing the neurons’ excitable membrane properties and functional synaptic connectivity through the release of pro-inflammatory cytokines. Meticulous attempts to causally relate the cellular composition, cell density, and electrical properties of the FBR have failed to unequivocally correlate the deterioration of recording quality with the histological severity of the FBR. Based on confocal and electron microscope analysis of thin sections of polyimide based MEA implants along with the surrounding brain tissue at different points in time after implantation, we propose that abrupt FP amplitude attenuation occurs at the implant/brain-parenchyma junction as a result of high seal resistance insulation formed by adhering microglia to the implant surfaces. In contrast to the prevailing hypothesis, that FP decrease occurs across the encapsulating scar of the implanted MEA, this mechanism potentially explains why no correlations have been found between the dimensions and density of the FBR and the recording quality. Recognizing that the seal resistance formed by adhering-microglia to the implant constitutes a downstream element undermining extracellular FP recordings, suggests that approaches to mitigate the formation of the insulating glial could lead to improved recording quality and yield.
Collapse
Affiliation(s)
- Shun-Ho Huang
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nava Shmoel
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maciej M Jankowski
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel.,Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Erez
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aviv Sharon
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wesal Abu-Salah
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Israel Nelken
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aryeh Weiss
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel
| | - Micha E Spira
- Department of Neurobiology, The Alexander Silberman Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Harvey M. Kruger Family Center for Nanoscience, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
43
|
Bettinger CJ, Ecker M, Kozai TDY, Malliaras GG, Meng E, Voit W. Recent advances in neural interfaces-Materials chemistry to clinical translation. MRS BULLETIN 2020; 45:655-668. [PMID: 34690420 PMCID: PMC8536148 DOI: 10.1557/mrs.2020.195] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Implantable neural interfaces are important tools to accelerate neuroscience research and translate clinical neurotechnologies. The promise of a bidirectional communication link between the nervous system of humans and computers is compelling, yet important materials challenges must be first addressed to improve the reliability of implantable neural interfaces. This perspective highlights recent progress and challenges related to arguably two of the most common failure modes for implantable neural interfaces: (1) compromised barrier layers and packaging leading to failure of electronic components; (2) encapsulation and rejection of the implant due to injurious tissue-biomaterials interactions, which erode the quality and bandwidth of signals across the biology-technology interface. Innovative materials and device design concepts could address these failure modes to improve device performance and broaden the translational prospects of neural interfaces. A brief overview of contemporary neural interfaces is presented and followed by recent progress in chemistry, materials, and fabrication techniques to improve in vivo reliability, including novel barrier materials and harmonizing the various incongruences of the tissue-device interface. Challenges and opportunities related to the clinical translation of neural interfaces are also discussed.
Collapse
Affiliation(s)
- Christopher J Bettinger
- Department of Materials Science and Engineering, and Department of Biomedical Engineering, Carnegie Mellon University, USA
| | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, USA
| | | | | | - Ellis Meng
- Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, USA
| | - Walter Voit
- Department of Mechanical Engineering, The University of Texas at Dallas, USA
| |
Collapse
|
44
|
Stieger KC, Eles JR, Ludwig KA, Kozai TDY. In vivo microstimulation with cathodic and anodic asymmetric waveforms modulates spatiotemporal calcium dynamics in cortical neuropil and pyramidal neurons of male mice. J Neurosci Res 2020; 98:2072-2095. [PMID: 32592267 DOI: 10.1002/jnr.24676] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022]
Abstract
Electrical stimulation has been critical in the development of an understanding of brain function and disease. Despite its widespread use and obvious clinical potential, the mechanisms governing stimulation in the cortex remain largely unexplored in the context of pulse parameters. Modeling studies have suggested that modulation of stimulation pulse waveform may be able to control the probability of neuronal activation to selectively stimulate either cell bodies or passing fibers depending on the leading polarity. Thus, asymmetric waveforms with equal charge per phase (i.e., increasing the leading phase duration and proportionately decreasing the amplitude) may be able to activate a more spatially localized or distributed population of neurons if the leading phase is cathodic or anodic, respectively. Here, we use two-photon and mesoscale calcium imaging of GCaMP6s expressed in excitatory pyramidal neurons of male mice to investigate the role of pulse polarity and waveform asymmetry on the spatiotemporal properties of direct neuronal activation with 10-Hz electrical stimulation. We demonstrate that increasing cathodic asymmetry effectively reduces neuronal activation and results in a more spatially localized subpopulation of activated neurons without sacrificing the density of activated neurons around the electrode. Conversely, increasing anodic asymmetry increases the spatial spread of activation and highly resembles spatiotemporal calcium activity induced by conventional symmetric cathodic stimulation. These results suggest that stimulation polarity and asymmetry can be used to modulate the spatiotemporal dynamics of neuronal activity thus increasing the effective parameter space of electrical stimulation to restore sensation and study circuit dynamics.
Collapse
Affiliation(s)
- Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.,Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kip A Ludwig
- Department of Biomedical Engineering, University of Wisconsin Madison, Madison, WI, USA.,Department of Neurological Surgery, University of Wisconsin Madison, Madison, WI, USA.,Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.,Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA.,Center for Neuroscience, 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
|
45
|
Yang Q, Wu B, Eles JR, Vazquez AL, Kozai TDY, Cui XT. Zwitterionic Polymer Coating Suppresses Microglial Encapsulation to Neural Implants In Vitro and In Vivo. ADVANCED BIOSYSTEMS 2020; 4:e1900287. [PMID: 32363792 PMCID: PMC7686959 DOI: 10.1002/adbi.201900287] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 01/08/2023]
Abstract
For brain computer interfaces (BCI), the immune response to implanted electrodes is a major biological cause of device failure. Bioactive coatings such as neural adhesion molecule L1 have been shown to improve the biocompatibility, but are difficult to handle or produce in batches. Here, a synthetic zwitterionic polymer coating, poly(sulfobetaine methacrylate) (PSBMA) is developed for neural implants with the goal of reducing the inflammatory host response. In tests in vitro, the zwitterionic coating inhibits protein adsorption and the attachment of fibroblasts and microglia, and remains stable for at least 4 weeks. In vivo two-photon microscopy on CX3CR1-GFP mice shows that the zwitterionic coating significantly suppresses the microglial encapsulation of neural microelectrodes over a 6 h observation period. Furthermore, the lower microglial encapsulation on zwitterionic polymer-coated microelectrodes is revealed to originate from a reduction in the size but not the number of microglial end feet. This work provides a facile method for coating neural implants with zwitterionic polymers and illustrates the initial interaction between microglia and coated surface at high temporal and spatial resolution.
Collapse
Affiliation(s)
- Qianru Yang
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - Bingchen Wu
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - James R Eles
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - Alberto L Vazquez
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 East Carson Street, Pittsburgh, PA, 15219, USA
| | - Takashi D Y Kozai
- Center for Biotechnology and Bioengineering, University of Pittsburgh, 300 Technology Dr, Pittsburgh, PA, 15213, USA
| | - X Tracy Cui
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| |
Collapse
|
46
|
Wellman SM, Guzman K, Stieger KC, Brink LE, Sridhar S, Dubaniewicz MT, Li L, Cambi F, Kozai TDY. Cuprizone-induced oligodendrocyte loss and demyelination impairs recording performance of chronically implanted neural interfaces. Biomaterials 2020; 239:119842. [PMID: 32065972 PMCID: PMC7540937 DOI: 10.1016/j.biomaterials.2020.119842] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
Biological inflammation induced during penetrating cortical injury can disrupt functional neuronal and glial activity within the cortex, resulting in potential recording failure of chronically implanted neural interfaces. Oligodendrocytes provide critical support for neuronal health and function through direct contact with neuronal soma and axons within the cortex. Given their fundamental role to regulate neuronal activity via myelin, coupled with their heightened vulnerability to metabolic brain injury due to high energetic demands, oligodendrocytes are hypothesized as a possible source of biological failure in declining recording performances of intracortical microelectrode devices. To determine the extent of their contribution to neuronal activity and function, a cuprizone-inducible model of oligodendrocyte depletion and demyelination in mice was performed prior to microelectrode implantation. At 5 weeks of cuprizone exposure, mice demonstrated significantly reduced cortical oligodendrocyte density and myelin expression. Mice were then implanted with functional recording microelectrodes in the visual cortex and neuronal activity was evaluated up to 7 weeks alongside continued cuprizone administration. Cuprizone-induced oligodendrocyte loss and demyelination was associated with significantly reduced recording performances at the onset of implantation, which remained relatively stable over time. In contast, recording performances for mice on a normal diet were intially elevated before decreasing over time to the recording level of tcuprizone-treated mice. Further electrophysiological analysis revealed deficits in multi-unit firing rates, frequency-dependent disruptions in neuronal oscillations, and altered laminar communication within the cortex of cuprizone-treated mice. Post-mortem immunohistochemistry revealed robust depletion of oligodendrocytes around implanted microelectrode arrays alongside comparable neuronal densities to control mice, suggesting that oligodendrocyte loss was a possible contributor to chronically impaired device performances. This study highlights potentially significant contributions from the oligodendrocyte lineage population concerning the biological integration and long-term functional performance of neural interfacing technology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Kelly Guzman
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA
| | - Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | | | - Sadhana Sridhar
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Lehong Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
47
|
Thompson CH, Riggins TE, Patel PR, Chestek CA, Li W, Purcell E. Toward guiding principles for the design of biologically-integrated electrodes for the central nervous system. J Neural Eng 2020; 17:021001. [PMID: 31986501 PMCID: PMC7523527 DOI: 10.1088/1741-2552/ab7030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Innovation in electrode design has produced a myriad of new and creative strategies for interfacing the nervous system with softer, less invasive, more broadly distributed sites with high spatial resolution. However, despite rapid growth in the use of implanted electrode arrays in research and clinical applications, there are no broadly accepted guiding principles for the design of biocompatible chronic recording interfaces in the central nervous system (CNS). Studies suggest that the architecture and flexibility of devices play important roles in determining effective tissue integration: device feature dimensions (varying from 'sub'- to 'supra'-cellular scales, <10 µm to >100 µm), Young's modulus, and bending modulus have all been identified as key features of design. However, critical knowledge gaps remain in the field with respect to the underlying motivation for these designs: (1) a systematic study of the relationship between device design features (materials, architecture, flexibility), biointegration, and signal quality needs to be performed, including controls for interaction effects between design features, (2) benchmarks for success need to be determined (biological integration, recording performance, longevity, stability), and (3) user results, particularly those that champion a specific design or electrode modification, need to be replicated across laboratories. Finally, the ancillary effects of factors such as tethering, site impedance and insertion method need to be considered. Here, we briefly review observations to-date of device design effects on tissue integration and performance, and then highlight the need for comprehensive and systematic testing of these effects moving forward.
Collapse
Affiliation(s)
- Cort H Thompson
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States of America
| | | | | | | | | | | |
Collapse
|
48
|
Neuroadhesive protein coating improves the chronic performance of neuroelectronics in mouse brain. Biosens Bioelectron 2020; 155:112096. [PMID: 32090868 DOI: 10.1016/j.bios.2020.112096] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/15/2022]
Abstract
Intracortical microelectrodes are being developed to both record and stimulate neurons to understand brain circuitry or restore lost functions. However, the success of these probes is hampered partly due to the inflammatory host tissue responses to implants. To minimize the foreign body reactions, L1, a brain derived neuronal specific cell adhesion molecule, has been covalently bound to the neural electrode array surface. Here we evaluated the chronic recording performance of L1-coated silicon based laminar neural electrode arrays implanted into V1m cortex of mice. The L1 coating enhanced the overall visually evoked single-unit (SU) yield and SU amplitude, as well as signal-to-noise-ratio (SNR) in the mouse brain compared to the uncoated arrays across the 0-1500 μm depth. The improvement in recording is most dramatic in the hippocampus region, where the control group showed severe recording yield decrease after one week, while the L1 implants maintained a high SU yield throughout the 16 weeks. Immunohistological analysis revealed significant increases of axonal and neuronal density along with significantly lowered microglia activation around the L1 probe after 16 weeks. These results collectively confirm the effectiveness of L1 based biomimetic coating on minimizing inflammatory tissue response and improving neural recording quality and longevity. Improving chronic recording will benefit the brain-computer interface technologies and neuroscience studies involving chronic tracking of neural activities.
Collapse
|
49
|
Eles JR, Kozai TDY. In vivo imaging of calcium and glutamate responses to intracortical microstimulation reveals distinct temporal responses of the neuropil and somatic compartments in layer II/III neurons. Biomaterials 2020; 234:119767. [PMID: 31954232 DOI: 10.1016/j.biomaterials.2020.119767] [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] [Received: 09/03/2019] [Revised: 12/22/2019] [Accepted: 01/05/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Intracortical microelectrode implants can generate a tissue response hallmarked by glial scarring and neuron cell death within 100-150 μm of the biomaterial device. Many have proposed that any performance decline in intracortical microstimulation (ICMS) due to this foreign body tissue response could be offset by increasing the stimulation amplitude. The mechanisms of this approach are unclear, however, as there has not been consensus on how increasing amplitude affects the spatial and temporal recruitment patterns of ICMS. APPROACH We clarify these unknowns using in vivo two-photon imaging of mice transgenically expressing the calcium sensor GCaMP6s in Thy1 neurons or virally expressing the glutamate sensor iGluSnFr in neurons. Calcium and neurotransmitter activity are tracked in the neuronal somas and neuropil during long-train stimulation in Layer II/III of somatosensory cortex. MAIN RESULTS Neural calcium activity and glutamate release are dense and strongest within 20-40 μm around the electrode, falling off with distance from the electrode. Neuronal calcium increases with higher amplitude stimulations. During prolonged stimulation trains, a sub-population of somas fail to maintain calcium activity. Interestingly, neuropil calcium activity is 3-fold less correlated to somatic calcium activity for cells that drop-out during the long stimulation train compared to cells that sustain activity throughout the train. Glutamate release is apparent only within 20 μm of the electrode and is sustained for at least 10s after cessation of the 15 and 20 μA stimulation train, but not lower amplitudes. SIGNIFICANCE These results demonstrate that increasing amplitude can increase the radius and intensity of neural recruitment, but it also alters the temporal response of some neurons. Further, dense glutamate release is highest within the first 20 μm of the electrode site even at high amplitudes, suggesting that there may be spatial limitations to the amplitude parameter space. The glutamate elevation outlasts stimulation, suggesting that high-amplitude stimulation may affect neurotransmitter re-uptake. This ultimately suggests that increasing the amplitude of ICMS device stimulation may fundamentally alter the temporal neural response, which could have implications for using amplitude to improve the ICMS effect or "offset" the effects of glial scarring.
Collapse
Affiliation(s)
- James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, Carnegie Mellon University, Pittsburgh, PA, USA; Center for Neuroscience, 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
|
50
|
Golabchi A, Wu B, Cao B, Bettinger CJ, Cui XT. Zwitterionic polymer/polydopamine coating reduce acute inflammatory tissue responses to neural implants. Biomaterials 2019; 225:119519. [PMID: 31600673 PMCID: PMC6896321 DOI: 10.1016/j.biomaterials.2019.119519] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
The inflammatory brain tissue response to implanted neural electrode devices has hindered the longevity of these implants. Zwitterionic polymers have a potent anti-fouling effect that decreases the foreign body response to subcutaneous implants. In this study, we developed a nanoscale anti-fouling coating composed of zwitterionic poly (sulfobetaine methacrylate) (PSB) and polydopamine (PDA) for neural probes. The addition of PDA improved the stability of the coating compared to PSB alone, without compromising the anti-fouling properties of the film. PDA-PSB coating reduced protein adsorption by 89% compared to bare Si samples, while fibroblast adhesion was reduced by 86%. PDA-PSB coated silicon based neural probes were implanted into mouse brain, and the inflammatory tissue responses to the implants were assessed by immunohistochemistry one week after implantation. The PSB-PDA coated implants showed a significantly decreased expression of glial fibrillary acidic protein (GFAP), a marker for reactive astrocytes, within 70 μm from the electrode-tissue interface (p < 0.05). Additionally, the coating reduced the microglia activation as shown in decreased Iba-1 and lectin staining, and improved blood-brain barrier integrity indicated by reduced immunoglobulin (IgG) leakage into the tissue around the probes. These findings demonstrate that anti-fouling zwitterionic coating is effective in suppressing the acute inflammatory brain tissue response to implants, and should be further investigated for its potential to improve chronic performance of neural implants.
Collapse
Affiliation(s)
- Asiyeh Golabchi
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA
| | - Bingchen Wu
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA
| | - Bin Cao
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA
| | - Christopher J Bettinger
- Department of Biomedical Engineering, Department of Material Science and Engineering, Carnegie Mellon University, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, USA; Center for Neural Basis of Cognition, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, USA.
| |
Collapse
|