1
|
Chen JY, Chen YC, Wu SL. Deep Brain Stimulation in Advanced Parkinson's Disease: An Uncommon Case of Allergic Encephalitis. J Mov Disord 2024; 17:345-347. [PMID: 38616161 PMCID: PMC11300388 DOI: 10.14802/jmd.23237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/09/2024] [Accepted: 04/12/2024] [Indexed: 04/16/2024] Open
Affiliation(s)
- Jyun-Yi Chen
- Department of Neurology, Changhua Christian Hospital, Changhua City, Taiwan
| | - Yen-Chung Chen
- Department of Neurology, Changhua Christian Hospital, Changhua City, Taiwan
| | - Shey-Lin Wu
- Department of Neurology, Zhangbin Show Chwan Memorial Hospital, Changhua City, Taiwan
| |
Collapse
|
2
|
Song S, Druschel LN, Chan ER, Capadona JR. Differential expression of genes involved in the chronic response to intracortical microelectrodes. Acta Biomater 2023; 169:348-362. [PMID: 37507031 PMCID: PMC10528922 DOI: 10.1016/j.actbio.2023.07.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Brain-Machine Interface systems (BMIs) are clinically valuable devices that can provide functional restoration for patients with spinal cord injury or improved integration for patients requiring prostheses. Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for precisely controlling BMIs. However, intracortical microelectrodes have a demonstrated history of progressive decline in the recording performance with time, inhibiting their usefulness. One major contributor to decreased performance is the neuroinflammatory response to the implanted microelectrodes. The neuroinflammatory response can lead to neurodegeneration and the formation of a glial scar at the implant site. Historically, histological imaging of relatively few known cellular and protein markers has characterized the neuroinflammatory response to implanted microelectrode arrays. However, neuroinflammation requires many molecular players to coordinate the response - meaning traditional methods could result in an incomplete understanding. Taking advantage of recent advancements in tools to characterize the relative or absolute DNA/RNA expression levels, a few groups have begun to explore gene expression at the microelectrode-tissue interface. We have utilized a custom panel of ∼813 neuroinflammatory-specific genes developed with NanoString for bulk tissue analysis at the microelectrode-tissue interface. Our previous studies characterized the acute innate immune response to intracortical microelectrodes. Here we investigated the gene expression at the microelectrode-tissue interface in wild-type (WT) mice chronically implanted with nonfunctioning probes. We found 28 differentially expressed genes at chronic time points (4WK, 8WK, and 16WK), many in the complement and extracellular matrix system. Further, the expression levels were relatively stable over time. Genes identified here represent chronic molecular players at the microelectrode implant sites and potential therapeutic targets for the long-term integration of microelectrodes. STATEMENT OF SIGNIFICANCE: Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for the precise control of Brain-Machine Interface systems (BMIs). However, intracortical microelectrodes have a demonstrated history of progressive declines in the recording performance with time, inhibiting their usefulness. One major contributor to the decline in these devices is the neuroinflammatory response against the implanted microelectrodes. Historically, neuroinflammation to implanted microelectrode arrays has been characterized by histological imaging of relatively few known cellular and protein markers. Few studies have begun to develop a more in-depth understanding of the molecular pathways facilitating device-mediated neuroinflammation. Here, we are among the first to identify genetic pathways that could represent targets to improve the host response to intracortical microelectrodes, and ultimately device performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
3
|
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
|
4
|
Alavarse AC, Frachini ECG, Silva JB, Pereira RDS, Ulrich H, Petri DFS. Amino acid decorated xanthan gum coatings: Molecular arrangement and cell adhesion. CARBOHYDRATE POLYMER TECHNOLOGIES AND APPLICATIONS 2022. [DOI: 10.1016/j.carpta.2022.100227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
5
|
Khodadadei F, Arshad R, Morales DM, Gluski J, Marupudi NI, McAllister JP, Limbrick DD, Harris CA. The effect of A1 and A2 reactive astrocyte expression on hydrocephalus shunt failure. Fluids Barriers CNS 2022; 19:78. [PMID: 36171630 PMCID: PMC9516791 DOI: 10.1186/s12987-022-00367-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background The composition of tissue obstructing neuroprosthetic devices is largely composed of inflammatory cells with a significant astrocyte component. In a first-of-its-kind study, we profile the astrocyte phenotypes present on hydrocephalus shunts. Methods qPCR and RNA in-situ hybridization were used to quantify pro-inflammatory (A1) and anti-inflammatory (A2) reactive astrocyte phenotypes by analyzing C3 and EMP1 genes, respectively. Additionally, CSF cytokine levels were quantified using ELISA. In an in vitro model of astrocyte growth on shunts, different cytokines were used to prevent the activation of resting astrocytes into the A1 and A2 phenotypes. Obstructed and non-obstructed shunts were characterized based on the degree of actual tissue blockage on the shunt surface instead of clinical diagnosis. Results The results showed a heterogeneous population of A1 and A2 reactive astrocytes on the shunts with obstructed shunts having a significantly higher proportion of A2 astrocytes compared to non-obstructed shunts. In addition, the pro-A2 cytokine IL-6 inducing proliferation of astrocytes was found at higher concentrations among CSF from obstructed samples. Consequently, in the in vitro model of astrocyte growth on shunts, cytokine neutralizing antibodies were used to prevent activation of resting astrocytes into the A1 and A2 phenotypes which resulted in a significant reduction in both A1 and A2 growth. Conclusions Therefore, targeting cytokines involved with astrocyte A1 and A2 activation is a promising intervention aimed to prevent shunt obstruction. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00367-3.
Collapse
Affiliation(s)
- Fatemeh Khodadadei
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
| | - Rooshan Arshad
- School of Medicine, Wayne State University, Detroit, MI, USA
| | - Diego M Morales
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob Gluski
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Neena I Marupudi
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - James P McAllister
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Carolyn A Harris
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA. .,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Biomedical Engineering, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
6
|
Cornelison C, Fadel S. Clickable Biomaterials for Modulating Neuroinflammation. Int J Mol Sci 2022; 23:8496. [PMID: 35955631 PMCID: PMC9369181 DOI: 10.3390/ijms23158496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
Crosstalk between the nervous and immune systems in the context of trauma or disease can lead to a state of neuroinflammation or excessive recruitment and activation of peripheral and central immune cells. Neuroinflammation is an underlying and contributing factor to myriad neuropathologies including neurodegenerative diseases like Alzheimer's disease and Parkinson's disease; autoimmune diseases like multiple sclerosis; peripheral and central nervous system infections; and ischemic and traumatic neural injuries. Therapeutic modulation of immune cell function is an emerging strategy to quell neuroinflammation and promote tissue homeostasis and/or repair. One such branch of 'immunomodulation' leverages the versatility of biomaterials to regulate immune cell phenotypes through direct cell-material interactions or targeted release of therapeutic payloads. In this regard, a growing trend in biomaterial science is the functionalization of materials using chemistries that do not interfere with biological processes, so-called 'click' or bioorthogonal reactions. Bioorthogonal chemistries such as Michael-type additions, thiol-ene reactions, and Diels-Alder reactions are highly specific and can be used in the presence of live cells for material crosslinking, decoration, protein or cell targeting, and spatiotemporal modification. Hence, click-based biomaterials can be highly bioactive and instruct a variety of cellular functions, even within the context of neuroinflammation. This manuscript will review recent advances in the application of click-based biomaterials for treating neuroinflammation and promoting neural tissue repair.
Collapse
Affiliation(s)
- Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | | |
Collapse
|
7
|
Kiang L, Woodington B, Carnicer-Lombarte A, Malliaras G, Barone DG. Spinal cord bioelectronic interfaces: opportunities in neural recording and clinical challenges. J Neural Eng 2022; 19. [PMID: 35320780 DOI: 10.1088/1741-2552/ac605f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/23/2022] [Indexed: 11/11/2022]
Abstract
Bioelectronic stimulation of the spinal cord has demonstrated significant progress in restoration of motor function in spinal cord injury (SCI). The proximal, uninjured spinal cord presents a viable target for the recording and generation of control signals to drive targeted stimulation. Signals have been directly recorded from the spinal cord in behaving animals and correlated with limb kinematics. Advances in flexible materials, electrode impedance and signal analysis will allow SCR to be used in next-generation neuroprosthetics. In this review, we summarize the technological advances enabling progress in SCR and describe systematically the clinical challenges facing spinal cord bioelectronic interfaces and potential solutions, from device manufacture, surgical implantation to chronic effects of foreign body reaction and stress-strain mismatches between electrodes and neural tissue. Finally, we establish our vision of bi-directional closed-loop spinal cord bioelectronic bypass interfaces that enable the communication of disrupted sensory signals and restoration of motor function in SCI.
Collapse
Affiliation(s)
- Lei Kiang
- Orthopaedic Surgery, Singapore General Hospital, Outram Road, Singapore, Singapore, 169608, SINGAPORE
| | - Ben Woodington
- Department of Engineering, University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, Cambridge, CB2 1TN, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Alejandro Carnicer-Lombarte
- Clinical Neurosciences, University of Cambridge, Bioelectronics Laboratory, Cambridge, CB2 0PY, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - George Malliaras
- University of Cambridge, University of Cambridge, Cambridge, CB2 1TN, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| | - Damiano G Barone
- Department of Engineering, University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, Cambridge, Cambridgeshire, CB2 1TN, UNITED KINGDOM OF GREAT BRITAIN AND NORTHERN IRELAND
| |
Collapse
|
8
|
Flexible Neural Probes with Optical Artifact-Suppressing Modification and Biofriendly Polypeptide Coating. MICROMACHINES 2022; 13:mi13020199. [PMID: 35208323 PMCID: PMC8877708 DOI: 10.3390/mi13020199] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/23/2022] [Accepted: 01/23/2022] [Indexed: 11/24/2022]
Abstract
The advent of optogenetics provides a well-targeted tool to manipulate neurons because of its high time resolution and cell-type specificity. Recently, closed-loop neural manipulation techniques consisting of optical stimulation and electrical recording have been widely used. However, metal microelectrodes exposed to light radiation could generate photoelectric noise, thus causing loss or distortion of neural signal in recording channels. Meanwhile, the biocompatibility of neural probes remains to be improved. Here, five kinds of neural interface materials are deposited on flexible polyimide-based neural probes and illuminated with a series of blue laser pulses to study their electrochemical performance and photoelectric noises for single-unit recording. The results show that the modifications can not only improve the electrochemical performance, but can also reduce the photoelectric artifacts. In particular, the double-layer composite consisting of platinum-black and conductive polymer has the best comprehensive performance. Thus, a layer of polypeptide is deposited on the entire surface of the double-layer modified neural probes to further improve their biocompatibility. The results show that the biocompatible polypeptide coating has little effect on the electrochemical performance of the neural probe, and it may serve as a drug carrier due to its special micromorphology.
Collapse
|
9
|
Yu X, Cheng F, He W. Bisepoxide-Jeffamine microgel synthesis and application toward heterogeneous surface morphology for differential neuronal/non-neuronal cell responses in vitro. Colloids Surf B Biointerfaces 2021; 207:112009. [PMID: 34339968 DOI: 10.1016/j.colsurfb.2021.112009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/16/2021] [Accepted: 07/24/2021] [Indexed: 11/30/2022]
Abstract
Herein, a new non-vinylic type of cationic microgels (MG) was readily prepared from ethylene glycol diglycidyl ether and Jeffamine T-403 in water. The MG was responsive to both temperature and pH, and oxidatively stable as demonstrated by the hydrogen peroxide study. Using glass as a model substrate, its surface was easily imparted with a heterogeneous morphology by simply adsorbing MG dispersed in basic solution. Specifically, the morphology features patches made of a monolayer of connected yet individually recognizable MG. Through in vitro cell studies, we show that a mere change of the extent of surface coverage by such a patchy morphology can strike a balance in promoting adhesion and differentiation of neuron-like PC-12 cells and primary cortical neurons of chick embryo, without soliciting proliferative response from non-neuronal cells of NIH3T3 fibroblast and CTX astrocyte. This simple yet unconventional surface morphology created by MG could be leveraged in the future as an alternative strategy for neural interface engineering.
Collapse
Affiliation(s)
- Xueying Yu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, Liaoning, 116023, China; School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning, 116023, China
| | - Fang Cheng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, Liaoning, 116023, China; School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning, 116023, China
| | - Wei He
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, Liaoning, 116023, China; School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning, 116023, China.
| |
Collapse
|
10
|
Kolaya E, Firestein BL. Deep brain stimulation: Challenges at the tissue-electrode interface and current solutions. Biotechnol Prog 2021; 37:e3179. [PMID: 34056871 DOI: 10.1002/btpr.3179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 11/08/2022]
Abstract
Deep brain stimulation (DBS) is used to treat the motor symptoms of Parkinson's disease patients by stimulating the subthalamic nucleus. However, optimization of DBS is still needed since the performance of the neural electrodes is limited by the body's response to the implant. This review discusses the issues with DBS, such as placement of electrodes, foreign body response, and electrode degradation. The current solutions to these technical issues include modifications to electrode material, coatings, and geometry.
Collapse
Affiliation(s)
- Emily Kolaya
- Biomedical Engineering Graduate Program, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Bonnie L Firestein
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
11
|
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: 3.7] [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
|
12
|
Lee A, Septiadi D, Taladriz‐Blanco P, Almeida M, Haeni L, Spuch‐Calvar M, Abdussalam W, Rothen‐Rutishauser B, Petri‐Fink A. Particle Stiffness and Surface Topography Determine Macrophage-Mediated Removal of Surface Adsorbed Particles. Adv Healthc Mater 2021; 10:e2001667. [PMID: 33434386 PMCID: PMC11469114 DOI: 10.1002/adhm.202001667] [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: 09/18/2020] [Revised: 12/10/2020] [Indexed: 01/09/2023]
Abstract
Cellular surface recognition and behavior are driven by a host of physical and chemical features which have been exploited to influence particle-cell interactions. Mechanical and topographical cues define the physical milieu which plays an important role in defining a range of cellular activities such as material recognition, adhesion, and migration through cytoskeletal organization and signaling. In order to elucidate the effect of local mechanical and topographical features generated by the adsorption of particles to an underlying surface on primary human monocyte-derived macrophages (MDM), a series of poly(N-isopropylacrylamide) (pNIPAM) particles with differing rigidity are self-assembled to form a defined particle-decorated surface. Assembly of particle-decorated surfaces is facilitated by modification of the underlying glass to possess a positive charge through functionalization using 3-aminopropyltriethoxysilane (APTES) or coating with poly(L-lysine) (PLL). MDMs are noted to preferentially remove particles with higher degrees of crosslinking (stiffer) than those with lower degrees of crosslinking (softer). Alterations to the surface density of particles enabled a greater area of the particle-decorated surface to be cleared. Uniquely, the impact of particle adsorption is evinced to have a direct impact on topographical recognition of the surface, suggesting a novel approach for controllably affecting cell-surface recognition and response.
Collapse
Affiliation(s)
- Aaron Lee
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4Fribourg1700Switzerland
| | - Dedy Septiadi
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4Fribourg1700Switzerland
| | | | - Mauro Almeida
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4Fribourg1700Switzerland
| | - Laetitia Haeni
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4Fribourg1700Switzerland
| | - Miguel Spuch‐Calvar
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4Fribourg1700Switzerland
| | - Wildan Abdussalam
- Department of High Energy DensityHelmholtz‐Zentrum Dresden‐RossendorfBautzner Landstraße 400Dresden01328Germany
| | | | - Alke Petri‐Fink
- Adolphe Merkle InstituteUniversity of FribourgChemin des Verdiers 4Fribourg1700Switzerland
- Department of ChemistryUniversity of FribourgChemin du Musée 9Fribourg1700Switzerland
| |
Collapse
|
13
|
Krukiewicz K, Britton J, Więcławska D, Skorupa M, Fernandez J, Sarasua JR, Biggs MJP. Electrical percolation in extrinsically conducting, poly(ε-decalactone) composite neural interface materials. Sci Rep 2021; 11:1295. [PMID: 33446813 PMCID: PMC7809477 DOI: 10.1038/s41598-020-80361-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/18/2020] [Indexed: 11/08/2022] Open
Abstract
By providing a bidirectional communication channel between neural tissues and a biomedical device, it is envisaged that neural interfaces will be fundamental in the future diagnosis and treatment of neurological disorders. Due to the mechanical mismatch between neural tissue and metallic neural electrodes, soft electrically conducting materials are of great benefit in promoting chronic device functionality. In this study, carbon nanotubes (CNT), silver nanowires (AgNW) and poly(hydroxymethyl 3,4-ethylenedioxythiophene) microspheres (MSP) were employed as conducting fillers within a poly(ε-decalactone) (EDL) matrix, to form a soft and electrically conducting composite. The effect of a filler type on the electrical percolation threshold, and composite biocompatibility was investigated in vitro. EDL-based composites exhibited favourable electrochemical characteristics: EDL/CNT-the lowest film resistance (1.2 ± 0.3 kΩ), EDL/AgNW-the highest charge storage capacity (10.7 ± 0.3 mC cm- 2), and EDL/MSP-the highest interphase capacitance (1478.4 ± 92.4 µF cm-2). All investigated composite surfaces were found to be biocompatible, and to reduce the presence of reactive astrocytes relative to control electrodes. The results of this work clearly demonstrated the ability of high aspect ratio structures to form an extended percolation network within a polyester matrix, resulting in the formulation of composites with advantageous mechanical, electrochemical and biocompatibility properties.
Collapse
Affiliation(s)
- Katarzyna Krukiewicz
- Centre for Research in Medical Devices, National University of Ireland, Newcastle Road, Galway, H91 W2TY, Ireland.
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M.Strzody 9, 44-100, Gliwice, Poland.
| | - James Britton
- Centre for Research in Medical Devices, National University of Ireland, Newcastle Road, Galway, H91 W2TY, Ireland
| | - Daria Więcławska
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M.Strzody 9, 44-100, Gliwice, Poland
| | - Małgorzata Skorupa
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, M.Strzody 9, 44-100, Gliwice, Poland
| | - Jorge Fernandez
- Department of Mining-Metallurgy Engineering and Materials Science, School of Engineering, POLYMAT, University of the Basque Country (UPV/EHU), Alameda de Urquijo s/n, 48013, Bilbao, Spain
- Polimerbio, S.L, Paseo Mikeletegi 83, 20009, Donostia-San Sebastian, Spain
| | - Jose-Ramon Sarasua
- Department of Mining-Metallurgy Engineering and Materials Science, School of Engineering, POLYMAT, University of the Basque Country (UPV/EHU), Alameda de Urquijo s/n, 48013, Bilbao, Spain
| | - Manus J P Biggs
- Centre for Research in Medical Devices, National University of Ireland, Newcastle Road, Galway, H91 W2TY, Ireland
| |
Collapse
|
14
|
Schmitt C, Rasch F, Cossais F, Held-Feindt J, Lucius R, Vázquez AR, Nia AS, Lohe MR, Feng X, Mishra YK, Adelung R, Schütt F, Hattermann K. Glial cell responses on tetrapod-shaped graphene oxide and reduced graphene oxide 3D scaffolds in brain in vitro and ex vivo models of indirect contact. Biomed Mater 2020; 16:015008. [DOI: 10.1088/1748-605x/aba796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
15
|
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: 5.0] [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
|
16
|
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: 15.0] [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
|
17
|
Hermann JK, Ravikumar M, Shoffstall AJ, Ereifej ES, Kovach KM, Chang J, Soffer A, Wong C, Srivastava V, Smith P, Protasiewicz G, Jiang J, Selkirk SM, Miller RH, Sidik S, Ziats NP, Taylor DM, Capadona JR. Inhibition of the cluster of differentiation 14 innate immunity pathway with IAXO-101 improves chronic microelectrode performance. J Neural Eng 2019; 15:025002. [PMID: 29219114 DOI: 10.1088/1741-2552/aaa03e] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Neuroinflammatory mechanisms are hypothesized to contribute to intracortical microelectrode failures. The cluster of differentiation 14 (CD14) molecule is an innate immunity receptor involved in the recognition of pathogens and tissue damage to promote inflammation. The goal of the study was to investigate the effect of CD14 inhibition on intracortical microelectrode recording performance and tissue integration. APPROACH Mice implanted with intracortical microelectrodes in the motor cortex underwent electrophysiological characterization for 16 weeks, followed by endpoint histology. Three conditions were examined: (1) wildtype control mice, (2) knockout mice lacking CD14, and (3) wildtype control mice administered a small molecule inhibitor to CD14 called IAXO-101. MAIN RESULTS The CD14 knockout mice exhibited acute but not chronic improvements in intracortical microelectrode performance without significant differences in endpoint histology. Mice receiving IAXO-101 exhibited significant improvements in recording performance over the entire 16 week duration without significant differences in endpoint histology. SIGNIFICANCE Full removal of CD14 is beneficial at acute time ranges, but limited CD14 signaling is beneficial at chronic time ranges. Innate immunity receptor inhibition strategies have the potential to improve long-term intracortical microelectrode performance.
Collapse
Affiliation(s)
- John K Hermann
- Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Rehabilitation Research and Development, 10701 East Blvd. Mail Stop 151 AW/APT, Cleveland OH 44106, United States of America. Department of Biomedical Engineering, Case Western Reserve University, School of Engineering, 2071 Martin Luther King Jr Drive, Wickenden Bldg, Cleveland OH 44106, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kyrey T, Witte J, Pipich V, Feoktystov A, Koutsioubas A, Vezhlev E, Frielinghaus H, von Klitzing R, Wellert S, Holderer O. Influence of the cross-linker content on adsorbed functionalised microgel coatings. POLYMER 2019. [DOI: 10.1016/j.polymer.2019.02.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
19
|
Vitale F, Shen W, Driscoll N, Burrell JC, Richardson AG, Adewole O, Murphy B, Ananthakrishnan A, Oh H, Wang T, Lucas TH, Cullen DK, Allen MG, Litt B. Biomimetic extracellular matrix coatings improve the chronic biocompatibility of microfabricated subdural microelectrode arrays. PLoS One 2018; 13:e0206137. [PMID: 30383805 PMCID: PMC6211660 DOI: 10.1371/journal.pone.0206137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 10/08/2018] [Indexed: 01/15/2023] Open
Abstract
Intracranial electrodes are a vital component of implantable neurodevices, both for acute diagnostics and chronic treatment with open and closed-loop neuromodulation. Their performance is hampered by acute implantation trauma and chronic inflammation in response to implanted materials and mechanical mismatch between stiff synthetic electrodes and pulsating, natural soft host neural tissue. Flexible electronics based on thin polymer films patterned with microscale conductive features can help alleviate the mechanically induced trauma; however, this strategy alone does not mitigate inflammation at the device-tissue interface. In this study, we propose a biomimetic approach that integrates microscale extracellular matrix (ECM) coatings on microfabricated flexible subdural microelectrodes. Taking advantage of a high-throughput process employing micro-transfer molding and excimer laser micromachining, we fabricate multi-channel subdural microelectrodes primarily composed of ECM protein material and demonstrate that the electrochemical and mechanical properties match those of standard, uncoated controls. In vivo ECoG recordings in rodent brain confirm that the ECM microelectrode coatings and the protein interface do not alter signal fidelity. Astrogliotic, foreign body reaction to ECM coated devices is reduced, compared to uncoated controls, at 7 and 30 days, after subdural implantation in rat somatosensory cortex. We propose microfabricated, flexible, biomimetic electrodes as a new strategy to reduce inflammation at the device-tissue interface and improve the long-term stability of implantable subdural electrodes.
Collapse
Affiliation(s)
- Flavia Vitale
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurology, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Physical Medicine & Rehabilitation, University of Pennsylvania, Philadelphia PA, United States of America
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
| | - Wendy Shen
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia PA, United States of America
| | - Nicolette Driscoll
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Justin C. Burrell
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - Andrew G. Richardson
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - Oladayo Adewole
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Brendan Murphy
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Akshay Ananthakrishnan
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia PA, United States of America
| | - Hanju Oh
- Department of Mechanical Engineering and Applied Mechanics, University of Pennsylvania, Philadelphia PA, United States of America
| | - Theodore Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Timothy H. Lucas
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - D. Kacy Cullen
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurosurgery, University of Pennsylvania, Philadelphia PA, United States of America
| | - Mark G. Allen
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia PA, United States of America
| | - Brian Litt
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Neurology, University of Pennsylvania, Philadelphia PA, United States of America
- Department of Bioengineering, University of Pennsylvania, Philadelphia PA, United States of America
| |
Collapse
|
20
|
Park HJ, Kang H, Jo J, Chung E, Kim S. Planar coil-based contact-mode magnetic stimulation: synaptic responses in hippocampal slices and thermal considerations. Sci Rep 2018; 8:13423. [PMID: 30194395 PMCID: PMC6128857 DOI: 10.1038/s41598-018-31536-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 08/16/2018] [Indexed: 12/30/2022] Open
Abstract
Implantable magnetic stimulation is an emerging type of neuromodulation using coils that are small enough to be implanted in the brain. A major advantage of this method is that stimulation performance could be sustained even though the coil is encapsulated by gliosis due to foreign body reactions. Magnetic fields can induce indirect electric fields and currents in neurons. Compared to transcranial magnetic stimulation, the coil size used in implantable magnetic stimulation can be greatly reduced. However, the size reduction is accompanied by an increase in coil resistance. Hence, the coil could potentially damage neurons from the excess heat generated. Therefore, it is necessary to study the stimulation performance and possible thermal damage by implantable magnetic stimulation. Here, we devised contact-mode magnetic stimulation (CMS), wherein magnetic stimulation was applied to hippocampal slices through a customized planar-type coil underneath the slice in the contact mode. With acute hippocampal slices, we investigated the synaptic responses to examine the field excitatory postsynaptic responses of CMS and the temperature rise during CMS. A long-lasting synaptic depression was exhibited in the CA1 stratum radiatum after CMS, while the temperature remained in a safe range so as not to seriously affect the neural responses.
Collapse
Affiliation(s)
- Hee-Jin Park
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Heekyung Kang
- Department of Biomedical Science and Neurology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jihoon Jo
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Euiheon Chung
- School of Mechanical Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.
- Department of Biomedical Science and Engineering, Institute of Integrated Technology, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.
| | - Sohee Kim
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
21
|
A novel neural electrode with micro-motion-attenuation capability based on compliant mechanisms—physical design concepts and evaluations. Med Biol Eng Comput 2018; 56:1911-1923. [DOI: 10.1007/s11517-018-1826-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/28/2018] [Indexed: 12/23/2022]
|
22
|
Wellman SM, Eles JR, Ludwig KA, Seymour JP, Michelson NJ, McFadden WE, Vazquez AL, Kozai TDY. A Materials Roadmap to Functional Neural Interface Design. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1701269. [PMID: 29805350 PMCID: PMC5963731 DOI: 10.1002/adfm.201701269] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Advancement in neurotechnologies for electrophysiology, neurochemical sensing, neuromodulation, and optogenetics are revolutionizing scientific understanding of the brain while enabling treatments, cures, and preventative measures for a variety of neurological disorders. The grand challenge in neural interface engineering is to seamlessly integrate the interface between neurobiology and engineered technology, to record from and modulate neurons over chronic timescales. However, the biological inflammatory response to implants, neural degeneration, and long-term material stability diminish the quality of interface overtime. Recent advances in functional materials have been aimed at engineering solutions for chronic neural interfaces. Yet, the development and deployment of neural interfaces designed from novel materials have introduced new challenges that have largely avoided being addressed. Many engineering efforts that solely focus on optimizing individual probe design parameters, such as softness or flexibility, downplay critical multi-dimensional interactions between different physical properties of the device that contribute to overall performance and biocompatibility. Moreover, the use of these new materials present substantial new difficulties that must be addressed before regulatory approval for use in human patients will be achievable. In this review, the interdependence of different electrode components are highlighted to demonstrate the current materials-based challenges facing the field of neural interface engineering.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - James R Eles
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - Kip A Ludwig
- Department of Neurologic Surgery, 200 First St. SW, Rochester, MN 55905
| | - John P Seymour
- Electrical & Computer Engineering, 1301 Beal Ave., 2227 EECS, Ann Arbor, MI 48109
| | - Nicholas J Michelson
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - William E McFadden
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - Alberto L Vazquez
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| | - Takashi D Y Kozai
- Department of Bioengineering, Center for the Basis of Neural Cognition, McGowan Institute of Regenerative Medicine, NeuroTech Center, University of Pittsburgh Brain Institute, Center for Neuroscience at the University of Pittsburgh, University of Pittsburgh, 208 Center for Biotechnology, 300 Technology Dr., Pittsburgh, PA 15219, United States
| |
Collapse
|
23
|
Serruya MD, Harris JP, Adewole DO, Struzyna LA, Burrell JC, Nemes A, Petrov D, Kraft RH, Chen HI, Wolf JA, Cullen DK. Engineered Axonal Tracts as "Living Electrodes" for Synaptic-Based Modulation of Neural Circuitry. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1701183. [PMID: 34045935 PMCID: PMC8152180 DOI: 10.1002/adfm.201701183] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Brain-computer interface and neuromodulation strategies relying on penetrating non-organic electrodes/optrodes are limited by an inflammatory foreign body response that ultimately diminishes performance. A novel "biohybrid" strategy is advanced, whereby living neurons, biomaterials, and microelectrode/optical technology are used together to provide a biologically-based vehicle to probe and modulate nervous-system activity. Microtissue engineering techniques are employed to create axon-based "living electrodes", which are columnar microstructures comprised of neuronal population(s) projecting long axonal tracts within the lumen of a hydrogel designed to chaperone delivery into the brain. Upon microinjection, the axonal segment penetrates to prescribed depth for synaptic integration with local host neurons, with the perikaryal segment remaining externalized below conforming electrical-optical arrays. In this paradigm, only the biological component ultimately remains in the brain, potentially attenuating a chronic foreign-body response. Axon-based living electrodes are constructed using multiple neuronal subtypes, each with differential capacity to stimulate, inhibit, and/or modulate neural circuitry based on specificity uniquely afforded by synaptic integration, yet ultimately computer controlled by optical/electrical components on the brain surface. Current efforts are assessing the efficacy of this biohybrid interface for targeted, synaptic-based neuromodulation, and the specificity, spatial density and long-term fidelity versus conventional microelectronic or optical substrates alone.
Collapse
Affiliation(s)
- Mijail D Serruya
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - James P Harris
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Dayo O Adewole
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laura A Struzyna
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin C Burrell
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Ashley Nemes
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Dmitriy Petrov
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - Reuben H Kraft
- Computational Biomechanics Group, Department of Mechanical & Nuclear Engineering, Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16801, USA
| | - H Isaac Chen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - John A Wolf
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| | - D Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Ham TR, Leipzig ND. Biomaterial strategies for limiting the impact of secondary events following spinal cord injury. Biomed Mater 2018; 13:024105. [PMID: 29155409 PMCID: PMC5824690 DOI: 10.1088/1748-605x/aa9bbb] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The nature of traumatic spinal cord injury (SCI) often involves limited recovery and long-term quality of life complications. The initial injury sets off a variety of secondary cascades, which result in an expanded lesion area. Ultimately, the native tissue fails to regenerate. As treatments are developed in the laboratory, the management of this secondary cascade is an important first step in achieving recovery of normal function. Current literature identifies four broad targets for intervention: inflammation, oxidative stress, disruption of the blood-spinal cord barrier, and formation of an inhibitory glial scar. Because of the complex and interconnected nature of these events, strategies that combine multiple therapies together show much promise. Specifically, approaches that rely on biomaterials to perform a variety of functions are generating intense research interest. In this review, we examine each target and discuss how biomaterials are currently used to address them. Overall, we show that there are an impressive amount of biomaterials and combinatorial treatments which show good promise for slowing secondary events and improving outcomes. If more emphasis is placed on growing our understanding of how materials can manage secondary events, treatments for SCI can be designed in an increasingly rational manner, ultimately improving their potential for translation to the clinic.
Collapse
Affiliation(s)
- Trevor R Ham
- Department of Biomedical Engineering, Auburn Science and Engineering Center 275, West Tower, University of Akron, Akron, OH 44325-3908, United States of America
| | | |
Collapse
|
25
|
Prospects for a Robust Cortical Recording Interface. Neuromodulation 2018. [DOI: 10.1016/b978-0-12-805353-9.00028-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
26
|
Woeppel K, Yang Q, Cui XT. Recent Advances in Neural Electrode-Tissue Interfaces. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 4:21-31. [PMID: 29423457 PMCID: PMC5798641 DOI: 10.1016/j.cobme.2017.09.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurotechnology is facing an exponential growth in the recent decades. Neural electrode-tissue interface research has been well recognized as an instrumental component of neurotechnology development. While satisfactory long-term performance was demonstrated in some applications, such as cochlear implants and deep brain stimulators, more advanced neural electrode devices requiring higher resolution for single unit recording or microstimulation still face significant challenges in reliability and longevity. In this article, we review the most recent findings that contribute to our current understanding of the sources of poor reliability and longevity in neural recording or stimulation, including the material failure, biological tissue response and the interplay between the two. The newly developed characterization tools are introduced from electrophysiology models, molecular and biochemical analysis, material characterization to live imaging. The effective strategies that have been applied to improve the interface are also highlighted. Finally, we discuss the challenges and opportunities in improving the interface and achieving seamless integration between the implanted electrodes and neural tissue both anatomically and functionally.
Collapse
Affiliation(s)
- Kevin Woeppel
- Bioengineering, University of Pittsburgh
- Center for the Neural Basis of Cognition, University of Pittsburgh
| | - Qianru Yang
- Bioengineering, University of Pittsburgh
- Center for the Neural Basis of Cognition, University of Pittsburgh
| | - Xinyan Tracy Cui
- Bioengineering, University of Pittsburgh
- Center for the Neural Basis of Cognition, University of Pittsburgh
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
| |
Collapse
|
27
|
Lee HC, Gaire J, Currlin SW, McDermott MD, Park K, Otto KJ. Foreign Body Response to Intracortical Microelectrodes Is Not Altered with Dip-Coating of Polyethylene Glycol (PEG). Front Neurosci 2017; 11:513. [PMID: 28959183 PMCID: PMC5603673 DOI: 10.3389/fnins.2017.00513] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/31/2017] [Indexed: 11/24/2022] Open
Abstract
Poly(ethylene glycol) (PEG) is a frequently used polymer for neural implants due to its biocompatible property. As a follow-up to our recent study that used PEG for stiffening flexible neural probes, we have evaluated the biological implications of using devices dip-coated with PEG for chronic neural implants. Mice (wild-type and CX3CR1-GFP) received bilateral implants within the sensorimotor cortex, one hemisphere with a PEG-coated probe and the other with a non-coated probe for 4 weeks. Quantitative analyses were performed using biomarkers for activated microglia/macrophages, astrocytes, blood-brain barrier leakage, and neuronal nuclei to determine the degree of foreign body response (FBR) resulting from the implanted microelectrodes. Despite its well-known acute anti-biofouling property, we observed that PEG-coated devices caused no significantly different FBR compared to non-coated controls at 4 weeks. A repetition using CX3CR1-GFP mice cohort showed similar results. Our histological findings suggest that there is no significant impact of acute delivery of PEG on the FBR in the long-term, and that temporary increase in the device footprint due to the coating of PEG also does not have a significant impact. Large variability seen within the same treatment group also implies that avoiding large superficial vasculature during implantation is not sufficient to minimize inter-animal variability.
Collapse
Affiliation(s)
- Heui C Lee
- Weldon School of Biomedical Engineering, Purdue UniversityWest Lafayette, IN, United States.,J. Crayton Pruitt Family Department of Biomedical Engineering, University of FloridaGainesville, FL, United States
| | - Janak Gaire
- Department of Neuroscience, University of FloridaGainesville, FL, United States
| | - Seth W Currlin
- Department of Neuroscience, University of FloridaGainesville, FL, United States
| | - Matthew D McDermott
- Weldon School of Biomedical Engineering, Purdue UniversityWest Lafayette, IN, United States
| | - Kinam Park
- Weldon School of Biomedical Engineering, Purdue UniversityWest Lafayette, IN, United States.,Department of Industrial and Physical Pharmacy, Purdue UniversityWest Lafayette, IN, United States
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of FloridaGainesville, FL, United States.,Department of Neuroscience, University of FloridaGainesville, FL, United States.,Department of Materials Science and Engineering, University of FloridaGainesville, FL, United States.,Department of Neurology, University of FloridaGainesville, FL, United States.,Nanoscience Institute for Medical and Engineering Technology, University of FloridaGainesville, FL, United States
| |
Collapse
|
28
|
Nyström L, Malmsten M. Surface-bound microgels - From physicochemical properties to biomedical applications. Adv Colloid Interface Sci 2016; 238:88-104. [PMID: 27865424 DOI: 10.1016/j.cis.2016.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 12/18/2022]
Abstract
Microgels offer robust and facile approaches for surface modification, as well as opportunities to introduce biological functionality by loading such structures with bioactive agents, e.g., in the context of drug delivery, functional biomaterials, and biosensors. As such, they provide a versatile approach for the design of surfaces with pre-determined characteristics compared to more elaborate bottom-up approaches, such as layer-by-layer deposition and surface-initiated polymerization. In the present overview, properties of surface-bound microgels are discussed, ranging from physical adsorption and covalent grafting in dilute systems, to directed self-assembly, multilayer structures, and composites, as well as loading an release of drugs and other cargo molecules into/from such systems, and biomedical applications of these.
Collapse
|
29
|
Hara SA, Kim BJ, Kuo JTW, Lee CD, Meng E, Pikov V. Long-term stability of intracortical recordings using perforated and arrayed Parylene sheath electrodes. J Neural Eng 2016; 13:066020. [DOI: 10.1088/1741-2560/13/6/066020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Capeletti LB, Cardoso MB, Dos Santos JHZ, He W. Hybrid Thin Film Organosilica Sol-Gel Coatings To Support Neuronal Growth and Limit Astrocyte Growth. ACS APPLIED MATERIALS & INTERFACES 2016; 8:27553-27563. [PMID: 27715001 DOI: 10.1021/acsami.6b09393] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Thin films of silica prepared by a sol-gel process are becoming a feasible coating option for surface modification of implantable neural sensors without imposing adverse effects on the devices' electrical properties. In order to advance the application of such silica-based coatings in the context of neural interfacing, the characteristics of silica sol-gel are further tailored to gain active control of interactions between cells and the coating materials. By incorporating various readily available organotrialkoxysilanes carrying distinct organic functional groups during the sol-gel process, a library of hybrid organosilica coatings is developed and investigated. In vitro neural cultures using PC12 cells and primary cortical neurons both reveal that, among these different types of hybrid organosilica, the introduction of aminopropyl groups drastically transforms the silica into robust neural permissive substrate, supporting neuron adhesion and neurite outgrowth. Moreover, when this organosilica is cultured with astrocytes, a key type of glial cells responsible for glial scar response toward neural implants, such cell growth promoting effect is not observed. These findings highlight the potential of organo-group-bearing silica sol-gel to function as advanced coating materials to selectively modulate cell response and promote neural integration with implantable sensing devices.
Collapse
Affiliation(s)
- Larissa Brentano Capeletti
- LNLS - Laboratório Nacional de Luz Síncrotron, Caixa Postal 6192, CEP 13083-970 Campinas, SP, Brazil
- Chemistry Institute, Universidade Federal do Rio Grande do Sul , CEP 91501-970, Porto Alegre, RS, Brazil
| | - Mateus Borba Cardoso
- LNLS - Laboratório Nacional de Luz Síncrotron, Caixa Postal 6192, CEP 13083-970 Campinas, SP, Brazil
| | | | | |
Collapse
|
31
|
Methods for Generating Hydrogel Particles for Protein Delivery. Ann Biomed Eng 2016; 44:1946-58. [PMID: 27160672 DOI: 10.1007/s10439-016-1637-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/03/2016] [Indexed: 10/21/2022]
Abstract
Proteins represent a major class of therapeutic molecules with vast potential for the treatment of acute and chronic diseases and regenerative medicine applications. Hydrogels have long been investigated for their potential in carrying and delivering proteins. As compared to bulk hydrogels, hydrogel microparticles (microgels) hold promise in improving aspects of delivery owing to their less traumatic route of entry into the body and improved versatility. This review discusses common methods of fabricating microgels, including emulsion polymerization, microfluidic techniques, and lithographic techniques. Microgels synthesized from both natural and synthetic polymers are discussed, as are a series of microgels fashioned from environment-responsive materials.
Collapse
|
32
|
Pan J, Lin XJ, Ling ZH, Cai YZ. Effect of down-regulation of voltage-gated sodium channel Nav1.7 on activation of astrocytes and microglia in DRG in rats with cancer pain. ASIAN PAC J TROP MED 2015; 8:405-11. [PMID: 26003602 DOI: 10.1016/s1995-7645(14)60352-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To evaluate the effect of down-regulation of Nav1.7 on the activation of astrocytes and microglia in DRG of rats with cancer pain, and explore the transmission of the nociceptive information. METHODS Lentiviral vector harboring RNAi sequence targeting the Nav1.7 gene was constructed, and Walker 256 breast cancer cell and morphine was injected to build the bone cancer pain model and morphine tolerance model in rats. Lentiviral vector was injected. Rats in each model were divided into 4 groups: model group, PBS group, vehicle group and LV-Nav1.7 group. The expression levels of GFAP and OX42 in dorsal root ganglia (DRG) were measured. RESULTS After the animal model was built, the level of Nav1.7, GFAP and OX42 was improved obviously with the time prolonged, which was statistically significant (P<0.05). The expression level of GFAP and OX42 in the DRG in the LV-Nav1.7 group declined obviously compared to the model group, PBS group and vehicle group (P<0.05). CONCLUSIONS Intrathecal injection of Navl.7 shRNA lentiviral vector can reduce the expression of Nav1.7 and inhibit the activation of astrocytes and microglia in DRG. The effort is also effective in morphine tolerance bone cancer pain model rats.
Collapse
Affiliation(s)
- Jun Pan
- Department of Orthopaedics, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou 310003, China
| | - Xiang-Jin Lin
- Department of Orthopaedics, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou 310003, China.
| | - Zhi-Heng Ling
- Department of Orthopaedics, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou 310003, China
| | - You-Zhi Cai
- Department of Orthopaedics, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
33
|
Potter-Baker KA, Capadona JR. Reducing the "Stress": Antioxidative Therapeutic and Material Approaches May Prevent Intracortical Microelectrode Failure. ACS Macro Lett 2015; 4:275-279. [PMID: 35596335 DOI: 10.1021/mz500743a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite the promising potential of intracortical microelectrodes, current designs suffer from short functional lifetimes, due in large part to the neuroinflammatory response to the implanted devices. An increasing body of literature is beginning to link neuroinflammatory-mediated oxidative damage to both the loss of neuronal structures around the implanted microelectrodes, and the degradation/corrosion of electrode materials. The goal of this viewpoint paper was to summarize the current progress toward understanding the role of oxidative damage to neurons and microelectrodes. Further, we seek to highlight the initial antioxidative approaches to mitigate oxidative damage, as well as suggest how current advances in macromolecular science for various applications may play a distinct role in enabling intracortical microelectrodes as reliable choices for long-term neuroprosthetic applications.
Collapse
Affiliation(s)
- Kelsey A. Potter-Baker
- Department of Biomedical
Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Jeffrey R. Capadona
- Department of Biomedical
Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
34
|
Wellert S, Kesal D, Schön S, von Klitzing R, Gawlitza K. Ethylene glycol-based microgels at solid surfaces: swelling behavior and control of particle number density. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:2202-2210. [PMID: 25654206 DOI: 10.1021/la504556m] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The adsorption of ethylene glycol (EG)-based microgel particles at silicon surfaces was investigated. Monodisperse p-MeO2MA-co-OEGMA microgel particles were synthesized by precipitation polymerization. Particle size and the volume phase transition temperature (VPTT) can be tailored by changing the amount of comonomer. The effect of geometrical confinement on the microgel particles was studied at the solid/liquid interface. Therefore, layer formation, particle number density, and swelling/deswelling at the surface were studied in dependence on the spin-coating preparation parameters and characterized by means of AFM against ambient conditions. The deswelling/swelling behavior was investigated by AFM in the water-swollen state.
Collapse
Affiliation(s)
- Stefan Wellert
- Stranski-Laboratory for Physical and Theoretical Chemistry, Technische Universität Berlin , Straße des 17. Juni 124, 10623 Berlin, Germany
| | | | | | | | | |
Collapse
|
35
|
Gutowski SM, Shoemaker JT, Templeman KL, Wei Y, Latour RA, Bellamkonda RV, LaPlaca MC, García AJ. Protease-degradable PEG-maleimide coating with on-demand release of IL-1Ra to improve tissue response to neural electrodes. Biomaterials 2015; 44:55-70. [PMID: 25617126 DOI: 10.1016/j.biomaterials.2014.12.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 12/01/2014] [Accepted: 12/16/2014] [Indexed: 01/18/2023]
Abstract
Neural electrodes are an important part of brain-machine interface devices that can restore functionality to patients with sensory and movement disorders. Chronically implanted neural electrodes induce an unfavorable tissue response which includes inflammation, scar formation, and neuronal cell death, eventually causing loss of electrode function. We developed a poly(ethylene glycol) hydrogel coating for neural electrodes with non-fouling characteristics, incorporated an anti-inflammatory agent, and engineered a stimulus-responsive degradable portion for on-demand release of the anti-inflammatory agent in response to inflammatory stimuli. This coating reduces in vitro glial cell adhesion, cell spreading, and cytokine release compared to uncoated controls. We also analyzed the in vivo tissue response using immunohistochemistry and microarray qRT-PCR. Although no differences were observed among coated and uncoated electrodes for inflammatory cell markers, lower IgG penetration into the tissue around PEG+IL-1Ra coated electrodes indicates an improvement in blood-brain barrier integrity. Gene expression analysis showed higher expression of IL-6 and MMP-2 around PEG+IL-1Ra samples, as well as an increase in CNTF expression, an important marker for neuronal survival. Importantly, increased neuronal survival around coated electrodes compared to uncoated controls was observed. Collectively, these results indicate promising findings for an engineered coating to increase neuronal survival and improve tissue response around implanted neural electrodes.
Collapse
Affiliation(s)
- Stacie M Gutowski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - James T Shoemaker
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kellie L Templeman
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yang Wei
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Robert A Latour
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Ravi V Bellamkonda
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michelle C LaPlaca
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
36
|
Jorfi M, Skousen JL, Weder C, Capadona JR. Progress towards biocompatible intracortical microelectrodes for neural interfacing applications. J Neural Eng 2014; 12:011001. [PMID: 25460808 DOI: 10.1088/1741-2560/12/1/011001] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To ensure long-term consistent neural recordings, next-generation intracortical microelectrodes are being developed with an increased emphasis on reducing the neuro-inflammatory response. The increased emphasis stems from the improved understanding of the multifaceted role that inflammation may play in disrupting both biologic and abiologic components of the overall neural interface circuit. To combat neuro-inflammation and improve recording quality, the field is actively progressing from traditional inorganic materials towards approaches that either minimizes the microelectrode footprint or that incorporate compliant materials, bioactive molecules, conducting polymers or nanomaterials. However, the immune-privileged cortical tissue introduces an added complexity compared to other biomedical applications that remains to be fully understood. This review provides a comprehensive reflection on the current understanding of the key failure modes that may impact intracortical microelectrode performance. In addition, a detailed overview of the current status of various materials-based approaches that have gained interest for neural interfacing applications is presented, and key challenges that remain to be overcome are discussed. Finally, we present our vision on the future directions of materials-based treatments to improve intracortical microelectrodes for neural interfacing.
Collapse
Affiliation(s)
- Mehdi Jorfi
- Adolphe Merkle Institute, University of Fribourg, Rte de l'Ancienne Papeterie, CH-1723 Marly, Switzerland
| | | | | | | |
Collapse
|
37
|
A strategy to passively reduce neuroinflammation surrounding devices implanted chronically in brain tissue by manipulating device surface permeability. Biomaterials 2014; 36:33-43. [PMID: 25310936 DOI: 10.1016/j.biomaterials.2014.08.039] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/24/2014] [Indexed: 01/08/2023]
Abstract
Available evidence indicates that pro-inflammatory cytokines produced by immune cells are likely responsible for the negative sequela associated with the foreign body response (FBR) to chronic indwelling implants in brain tissue. In this study a computational modeling approach was used to design a diffusion sink placed at the device surface that would retain pro-inflammatory cytokines for sufficient time to passively antagonize their impact on the FBR. Using quantitative immunohistochemistry, we examined the FBR to such engineered devices after a 16-week implantation period in the cortex of adult male Sprague-Dawley rats. Our results indicate that thick permeable surface coatings, which served as diffusion sinks, significantly reduced the FBR compared to implants either with no coating or with a thinner coating. The results suggest that increasing surface permeability of solid implanted devices to create a diffusion sink can be used to reduce the FBR and improve biocompatibility of chronic indwelling devices in brain tissue.
Collapse
|
38
|
Sommakia S, Gaire J, Rickus JL, Otto KJ. Resistive and reactive changes to the impedance of intracortical microelectrodes can be mitigated with polyethylene glycol under acute in vitro and in vivo settings. FRONTIERS IN NEUROENGINEERING 2014; 7:33. [PMID: 25136315 PMCID: PMC4120760 DOI: 10.3389/fneng.2014.00033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/11/2014] [Indexed: 01/05/2023]
Abstract
The reactive response of brain tissue to implantable intracortical microelectrodes is thought to negatively affect their recordable signal quality and impedance, resulting in unreliable longitudinal performance. The relationship between the progression of the reactive tissue into a glial scar and the decline in device performance is unclear. We show that exposure to a model protein solution in vitro and acute implantation result in both resistive and capacitive changes to electrode impedance, rather than purely resistive changes. We also show that applying 4000 MW polyethylene glycol (PEG) prevents impedance increases in vitro, and reduces the percent change in impedance in vivo following implantation. Our results highlight the importance of considering the contributions of non-cellular components to the decline in neural microelectrode performance, and present a proof of concept for using a simple dip-coated PEG film to modulate changes in microelectrode impedance.
Collapse
Affiliation(s)
- Salah Sommakia
- Weldon School of Biomedical Engineering, Purdue University West Lafayette, IN, USA ; Physiological Sensing Facility at the Bindley Bioscience Center and Birck Nanotechnology Center, Purdue University West Lafayette, IN, USA
| | - Janak Gaire
- Department of Biological Sciences, Purdue University West Lafayette, IN, USA
| | - Jenna L Rickus
- Weldon School of Biomedical Engineering, Purdue University West Lafayette, IN, USA ; Physiological Sensing Facility at the Bindley Bioscience Center and Birck Nanotechnology Center, Purdue University West Lafayette, IN, USA ; Department of Agricultural and Biological Engineering, Purdue University West Lafayette, IN, USA
| | - Kevin J Otto
- Weldon School of Biomedical Engineering, Purdue University West Lafayette, IN, USA ; Department of Biological Sciences, Purdue University West Lafayette, IN, USA
| |
Collapse
|
39
|
Potter KA, Jorfi M, Householder KT, Foster EJ, Weder C, Capadona JR. Curcumin-releasing mechanically adaptive intracortical implants improve the proximal neuronal density and blood-brain barrier stability. Acta Biomater 2014; 10:2209-22. [PMID: 24468582 DOI: 10.1016/j.actbio.2014.01.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/09/2013] [Accepted: 01/15/2014] [Indexed: 10/25/2022]
Abstract
The cellular and molecular mechanisms by which neuroinflammatory pathways respond to and propagate the reactive tissue response to intracortical microelectrodes remain active areas of research. We previously demonstrated that both the mechanical mismatch between rigid implants and the much softer brain tissue, as well as oxidative stress, contribute to the neurodegenerative reactive tissue response to intracortical implants. In this study, we utilize physiologically responsive, mechanically adaptive polymer implants based on poly(vinyl alcohol) (PVA), with the capability to also locally administer the antioxidant curcumin. The goal of this study is to investigate if the combination of two independently effective mechanisms - softening of the implant and antioxidant release - leads to synergistic effects in vivo. Over the first 4weeks of the implantation, curcumin-releasing, mechanically adaptive implants were associated with higher neuron survival and a more stable blood-brain barrier at the implant-tissue interface than the neat PVA controls. 12weeks post-implantation, the benefits of the curcumin release were lost, and both sets of compliant materials (with and without curcumin) had no statistically significant differences in neuronal density distribution profiles. Overall, however, the curcumin-releasing softening polymer implants cause minimal implant-mediated neuroinflammation, and embody the new concept of localized drug delivery from mechanically adaptive intracortical implants.
Collapse
|