1
|
Kim M, López-Cano M, Zhang K, Wang Y, Gómez-Santacana X, Flores Á, Wu M, Li S, Zhang H, Wei Y, Li X, Good CH, Banks AR, Llebaria A, Hernando J, Sunwoo SH, Gu J, Huang Y, Ciruela F, Rogers JA. Wireless, battery-free, remote photoactivation of caged-morphine for photopharmacological pain modulation without side effects. Biosens Bioelectron 2025; 281:117440. [PMID: 40220492 DOI: 10.1016/j.bios.2025.117440] [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: 12/19/2024] [Revised: 02/26/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Chronic pain severely impairs physical, psychological, and cognitive functions. While opioid-based therapies can be effective, they are limited by tolerance, dependence, and adverse side effects, highlighting the need for safer alternatives. Recent advances in photopharmacology allow precise modulation of pain-related neuronal circuits, offering improved control and effectiveness. For delivery of light, fully implantable, wireless, battery-free optical systems in miniaturized forms offer attractive options relative to alternatives that use conventional bulk hardware and fiber optic tethers. This work presents a technology of this type, based on microscale light-emitting diodes (μ-ILEDs) and near-field communication (NFC) protocols, and optimized to activate photocaged morphine (pc-Mor) in targeted regions of the spinal cord. The unique flexible, lightweight designs ensure stable, minimally invasive operation in small animal model behavioral studies, with efficient power consumption and minimized thermal load on fragile tissues. Experimental results demonstrate effective pain suppression and reduced opioid-related side effects in an animal model of pain, thereby establishing this platform as a promising solution for chronic pain management.
Collapse
Affiliation(s)
- Minsung Kim
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Neurolux Inc., Northfield, IL, 60093, USA
| | - Marc López-Cano
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907, Spain
| | - Kaiqing Zhang
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA; State Key Laboratory of Structural Analysis, Optimization and CAE Software for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Yue Wang
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xavier Gómez-Santacana
- MCS - Medicinal Chemistry & Synthesis, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - África Flores
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907, Spain
| | - Mingzheng Wu
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Shupeng Li
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Haohui Zhang
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yuanting Wei
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xiuyuan Li
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA; State Key Laboratory of Mechanical System and Vibration, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cameron H Good
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Neurolux Inc., Northfield, IL, 60093, USA; Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, 60208, USA
| | - Anthony R Banks
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Neurolux Inc., Northfield, IL, 60093, USA; Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, 60208, USA
| | - Amadeu Llebaria
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Jordi Hernando
- Departament de Química, Universitat Autònoma de Barcelona, Edifici C/n, Campus UAB, Cerdanyola del Vallès, 08193, Spain
| | - Sung-Hyuk Sunwoo
- Department of Chemical Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea
| | - Jianyu Gu
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA
| | - Yonggang Huang
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA.
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, 08907, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Bellvitge Biomedical Research Institute, L'Hospitalet de Llobregat, 08907, Spain.
| | - John A Rogers
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA; Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, 60208, USA; Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA; Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
2
|
Li L, Menendez-Lustri DM, Hartzler A, Pogharian A, Zaorski B, Chen A, Palen J, Traylor B, Quill E, Pawlowski CL, Bruckman MA, Gupta AS, Capadona JR, Shoffstall AJ. Systemically administered platelet-inspired nanoparticles to reduce inflammation surrounding intracortical microelectrodes. Biomaterials 2025; 317:123082. [PMID: 39787896 DOI: 10.1016/j.biomaterials.2025.123082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/19/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025]
Abstract
Intracortical microelectrodes (IMEs) are essential for neural signal acquisition in neuroscience and brain-machine interface (BMI) systems, aiding patients with neurological disorders, paralysis, and amputations. However, IMEs often fail to maintain robust signal quality over time, partly due to neuroinflammation caused by vascular damage during insertion. Platelet-inspired nanoparticles (PIN), which possess injury-targeting functions, mimic the adhesion and aggregation of active platelets through conjugated collagen-binding peptides (CBP), von Willebrand Factor-binding peptides (VBP), and fibrinogen-mimetic peptides (FMP). Systemically administered PINs can potentially enhance hemostasis and promote the resealing of IME insertion-induced leaky blood-brain barrier (BBB), thereby attenuating the influx of blood-derived proteins into the brain parenchyma that trigger neuroinflammation. This study explores the potential of PINs to mitigate neuroinflammation at implant sites. Male Sprague Dawley rats underwent craniotomies and IME implantations, followed by a single dose of Cy5 labeled PINs (2 mg/kg). Rats were sacrificed at intervals from 0 to 4 days post-implantation (DPI) for biodistribution analysis using an in vivo live imaging system (IVIS) and immunohistochemistry (IHC) to assess neuroinflammation, BBB permeability, and active platelet distribution. Another cohort of rats received weekly PINs, trehalose buffer (TH, diluent control), or control nanoparticles (CP, PEG-coated liposomes) for 4 weeks, with similar endpoint analyses. Results indicated that PIN concentrations were significantly elevated near IME interfaces acutely (0-4 DPI) and after 4 weeks of repeated dosing. At 3 DPI, peak intensities of active platelets (CD62P), activated microglia/macrophages (CD68), and PINs were observed. Immunoglobulin G (IgG) was upregulated during the first 24 h near implant sites but declined thereafter. At 4 weeks, the PINs group exhibited higher intensities of active platelets and PINs, and reduced CD68 and IgG levels compared to controls. PINs effectively targeted the IME-tissue interface, alongside endogenous activated platelets, resulting in reduced neuroinflammatory and BBB-leakage markers compared to the diluent-only-infused control group. Repeated dosing of PINs presents a promising approach for enhancing the quality of neural recordings in future studies.
Collapse
Affiliation(s)
- Longshun Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Dhariyat M Menendez-Lustri
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Aniya Hartzler
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Anna Pogharian
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Brett Zaorski
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Alex Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Jaquelynn Palen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | | | - Emma Quill
- Haima Therapeutics LLC, Cleveland, OH, United States
| | | | | | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, United States
| | - Andrew J Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, United States.
| |
Collapse
|
3
|
Szlak L, Shen J, Zohar E, Karavani E, Rotroff D, Vegh D, Punia V, Rosen-Zvi M, Shimoni Y, Jehi L. Peri-operative anti-inflammatory drug use and seizure recurrence after resective epilepsy surgery: Target trials emulation. iScience 2025; 28:112124. [PMID: 40241751 PMCID: PMC12003005 DOI: 10.1016/j.isci.2025.112124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/23/2025] [Accepted: 02/25/2025] [Indexed: 04/18/2025] Open
Abstract
We conducted a retrospective observational study to examine whether anti-inflammatory medications prescribed peri-operatively of resective brain surgery can reduce long-term seizure recurrence for individuals with drug-resistant focal epilepsy. We used insurance-claims data from across the United States to screen medications prescribed to 1,993 individuals undergoing epilepsy. We then validated the results in a well-characterized cohort of 671 epilepsy patients from a major surgical center. Twelve medications met the screening criteria and were evaluated, identifying dexamethasone and zonisamide as potentially beneficial. Dexamethasone reduced seizure recurrence by 42% over 9 years of follow-up (hazard-ratio = 0.742; 95% CI = 0.662, 0.831), and zonisamide reduced recurrence by 33% (HR = 0.782; 95% CI = 0.667, 0.917). While dexamethasone could not be validated, analysis of zonisamide in the clinical cohort corroborated the beneficial effect (HR = 0.828; 95% CI = 0.706, 0.971). If prospectively validated, this study suggests surgeons could improve long-term outcomes of epilepsy surgery by medically reducing neuro-inflammation in the surgical bed.
Collapse
Affiliation(s)
| | - Jingdi Shen
- Center for Computational Life Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | - Daniel Rotroff
- Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Deborah Vegh
- Center for Computational Life Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Vineet Punia
- Epilepsy Center, Cleveland Clinic, Cleveland, OH, USA
| | - Michal Rosen-Zvi
- IBM Research, Haifa, Israel
- Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | | | - Lara Jehi
- Center for Computational Life Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Epilepsy Center, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
4
|
Dalrymple AN, Jones ST, Fallon JB, Shepherd RK, Weber DJ. Overcoming failure: improving acceptance and success of implanted neural interfaces. Bioelectron Med 2025; 11:6. [PMID: 40083033 PMCID: PMC11907899 DOI: 10.1186/s42234-025-00168-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Implanted neural interfaces are electronic devices that stimulate or record from neurons with the purpose of improving the quality of life of people who suffer from neural injury or disease. Devices have been designed to interact with neurons throughout the body to treat a growing variety of conditions. The development and use of implanted neural interfaces is increasing steadily and has shown great success, with implants lasting for years to decades and improving the health and quality of life of many patient populations. Despite these successes, implanted neural interfaces face a multitude of challenges to remain effective for the lifetime of their users. The devices are comprised of several electronic and mechanical components that each may be susceptible to failure. Furthermore, implanted neural interfaces, like any foreign body, will evoke an immune response. The immune response will differ for implants in the central nervous system and peripheral nervous system, as well as over time, ultimately resulting in encapsulation of the device. This review describes the challenges faced by developers of neural interface systems, particularly devices already in use in humans. The mechanical and technological failure modes of each component of an implant system is described. The acute and chronic reactions to devices in the peripheral and central nervous system and how they affect system performance are depicted. Further, physical challenges such as micro and macro movements are reviewed. The clinical implications of device failures are summarized and a guide for determining the severity of complication was developed and provided. Common methods to diagnose and examine mechanical, technological, and biological failure modes at various stages of development and testing are outlined, with an emphasis on chronic in vivo characterization of implant systems. Finally, this review concludes with an overview of some of the innovative solutions developed to reduce or resolve the challenges faced by implanted neural interface systems.
Collapse
Affiliation(s)
- Ashley N Dalrymple
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
- Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, USA.
- NERVES Lab, University of Utah, Salt Lake City, UT, USA.
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Sonny T Jones
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- NERVES Lab, University of Utah, Salt Lake City, UT, USA
| | - James B Fallon
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
- Medical Bionics Department, University of Melbourne, Melbourne, VIC, Australia
| | - Robert K Shepherd
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Boufidis D, Garg R, Angelopoulos E, Cullen DK, Vitale F. Bio-inspired electronics: Soft, biohybrid, and "living" neural interfaces. Nat Commun 2025; 16:1861. [PMID: 39984447 PMCID: PMC11845577 DOI: 10.1038/s41467-025-57016-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/04/2025] [Indexed: 02/23/2025] Open
Abstract
Neural interface technologies are increasingly evolving towards bio-inspired approaches to enhance integration and long-term functionality. Recent strategies merge soft materials with tissue engineering to realize biologically-active and/or cell-containing living layers at the tissue-device interface that enable seamless biointegration and novel cell-mediated therapeutic opportunities. This review maps the field of bio-inspired electronics and discusses key recent developments in tissue-like and regenerative bioelectronics, from soft biomaterials and surface-functionalized bioactive coatings to cell-containing 'biohybrid' and 'all-living' interfaces. We define and contextualize key terminology in this emerging field and highlight how biological and living components can bridge the gap to clinical translation.
Collapse
Affiliation(s)
- Dimitris Boufidis
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raghav Garg
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eugenia Angelopoulos
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - D Kacy Cullen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA.
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Flavia Vitale
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA.
- Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
6
|
Shoffstall A, Li L, Hartzler A, Menendez-Lustri D, Zhang J, Chen A, Lam D, Traylor B, Quill E, Hoeferlin G, Pawlowski C, Bruckman M, Gupta SA, Capadona J. Dexamethasone-loaded platelet-inspired nanoparticles improve intracortical microelectrode recording performance. RESEARCH SQUARE 2025:rs.3.rs-6018202. [PMID: 39989959 PMCID: PMC11844648 DOI: 10.21203/rs.3.rs-6018202/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Long-term robust intracortical microelectrode (IME) neural recording quality is negatively affected by the neuroinflammatory response following microelectrode insertion. This adversely impacts brain-machine interface (BMI) performance for patients with neurological disorders or amputations. Recent studies suggest that the leakage of blood-brain barrier (BBB) and microhemorrhage caused by the IME insertions lead to the increased neuroinflammation and reduced neural recording performance. Additionally, a sustained presence of activated platelets and coagulation factors is found near the insertion site. Thus, we hypothesized that the systemic administration of dexamethasone sodium phosphate-loaded platelet-inspired nanoparticle (SPPINDEX) can improve the neural recording performance of intracortical microelectrodes (IMEs) by promoting hemostasis, facilitating blood-brain barrier (BBB) healing, and achieving implant-targeted drug delivery. Leveraging the hemostatic and coagulation factor-binding properties of the platelet-inspired nanoparticle (PIN) drug delivery platform, SPPINDEX treatment can initially attenuate the invasion of neuroinflammatory triggers into the brain parenchyma caused by insertion-induced microhemorrhages or a compromised BBB. Furthermore, targeted delivery of the anti-inflammatory drug dexamethasone sodium phosphate (DEXSP) to the implant site via these nanoparticles can attenuate ongoing neuroinflammation, enhancing overall therapeutic efficacy. Weekly treatment with SPPINDEX for 8 weeks significantly improved the recording capabilities of IMEs compared to platelet-inspired nanoparticles alone (PIN), free dexamethasone sodium phosphate (Free DEXSP), and a diluent control trehalose buffer (TH), as assessed through extracellular single-unit recordings. Immunohistochemical analyses of neuron density, activated microglia/macrophage density, astrocyte density, and BBB permeability suggest that the improved neural recording performance may be attributed to reduced neuron degeneration, activated microglia and astrocytes at the implant interface caused by the decreased infiltration of blood-derived proteins that trigger neuroinflammation and the therapeutic effects from DEXSP. Overall, SPPINDEX treatment promotes an anti-inflammatory environment that improves neuronal density and enhances recording performance.
Collapse
|
7
|
Rodríguez-Meana B, del Valle J, Navarro X. A Combinatory Therapy of Metformin and Dexamethasone Reduces the Foreign Body Reaction to Intraneural Electrodes. Cells 2024; 13:2112. [PMID: 39768202 PMCID: PMC11726768 DOI: 10.3390/cells13242112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Neural electrodes used for bidirectional communication between the nervous system and external devices like prosthetic limbs have advanced in neuroprosthetic applications. However, their effectiveness is hindered by the foreign body reaction, a natural immune response causing inflammation and fibrosis around the implanted device. This process involves protein adsorption, immune cell recruitment, cytokine release, and fibroblast activation, leading to a fibrous capsule formation and a decrease in electrode functionality. Anti-inflammatory and antifibrotic strategies have the potential to diminish the impact of the foreign body response. In this work, we have evaluated long-term metformin administration and short-term dexamethasone administration as a combined therapy to modulate the foreign body reaction induced by a polyimide intraneural implant in the sciatic nerve of rats. After a 12-week implant, the foreign body reaction was significantly reduced only in the group administered both drugs.
Collapse
Affiliation(s)
- Bruno Rodríguez-Meana
- Institute of Neurosciences, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Jaume del Valle
- Secció de Fisiologia, Departament de Bioquímica i Fisiologia, Universitat de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| | - Xavier Navarro
- Institute of Neurosciences, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
8
|
Ahmed Taha B, Addie AJ, Saeed AQ, Haider AJ, Chaudhary V, Arsad N. Nanostructured Photonics Probes: A Transformative Approach in Neurotherapeutics and Brain Circuitry. Neuroscience 2024; 562:106-124. [PMID: 39490518 DOI: 10.1016/j.neuroscience.2024.10.046] [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: 07/11/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Neuroprobes that use nanostructured photonic interfaces are capable of multimodal sensing, stimulation, and imaging with unprecedented spatio-temporal resolution. In addition to electrical recording, optogenetic modulation, high-resolution optical imaging, and molecular sensing, these advanced probes combine nanophotonic waveguides, optical transducers, nanostructured electrodes, and biochemical sensors. The potential of this technology lies in unraveling the mysteries of neural coding principles, mapping functional connectivity in complex brain circuits, and developing new therapeutic interventions for neurological disorders. Nevertheless, achieving the full potential of nanostructured photonic neural probes requires overcoming challenges such as ensuring long-term biocompatibility, integrating nanoscale components at high density, and developing robust data-analysis pipelines. In this review, we summarize and discuss the role of photonics in neural probes, trends in electrode diameter for neural interface technologies, nanophotonic technologies using nanostructured materials, advances in nanofabrication photonics interface engineering, and challenges and opportunities. Finally, interdisciplinary efforts are required to unlock the transformative potential of next-generation neuroscience therapies.
Collapse
Affiliation(s)
- Bakr Ahmed Taha
- UKM-Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, UKM Bangi 43600, Malaysia.
| | - Ali J Addie
- Center of Industrial Applications and Materials Technology, Scientific Research Commission, Iraq
| | - Ali Q Saeed
- Computer Center / Northern Technical University, Iraq
| | - Adawiya J Haider
- Applied Sciences Department/Laser Science and Technology Branch, University of Technology, Iraq.
| | - Vishal Chaudhary
- Research Cell & Department of Physics, Bhagini Nivedita College, University of Delhi, New Delhi 110045, India; Centre for Research Impact & Outcome, Chitkara University, Punjab, 140401 India
| | - Norhana Arsad
- UKM-Department of Electrical, Electronic and Systems Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, UKM Bangi 43600, Malaysia.
| |
Collapse
|
9
|
Oldroyd P, Hadwe SE, Barone DG, Malliaras GG. Thin-film implants for bioelectronic medicine. MRS BULLETIN 2024; 49:1045-1058. [PMID: 39397879 PMCID: PMC11469980 DOI: 10.1557/s43577-024-00786-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 10/15/2024]
Abstract
This article is based on the MRS Mid-Career Researcher Award "for outstanding contributions to the fundamentals and development of organic electronic materials and their application in biology and medicine" presentation given by George G. Malliaras, University of Cambridge, at the 2023 MRS Spring Meeting in San Francisco, Calif.Bioelectronic medicine offers a revolutionary approach to treating disease by stimulating the body with electricity. While current devices show safety and efficacy, limitations, including bulkiness, invasiveness, and scalability, hinder their wider application. Thin-film implants promise to overcome these limitations. Made using microfabrication technologies, these implants conform better to neural tissues, reduce tissue damage and foreign body response, and provide high-density, multimodal interfaces with the body. This article explores how thin-film implants using organic materials and novel designs may contribute to disease management, intraoperative monitoring, and brain mapping applications. Additionally, the technical challenges to be addressed for this technology to succeed are discussed. Graphical abstract
Collapse
Affiliation(s)
- Poppy Oldroyd
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| | - Salim El Hadwe
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Damiano G. Barone
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - George G. Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Aladev SD, Sokolov DK, Strokotova AV, Kazanskaya GM, Volkov AM, Aidagulova SV, Grigorieva EV. Multiple Administration of Dexamethasone Possesses a Deferred Long-Term Effect to Glycosylated Components of Mouse Brain. Neurol Int 2024; 16:790-803. [PMID: 39051219 PMCID: PMC11270268 DOI: 10.3390/neurolint16040058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/13/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Glucocorticoids are used during glioblastoma treatment to prevent the cerebral edema effect surrounding normal brain tissue. The aim of our study was to investigate the long-term effects of multiple administrations of glucocorticoids onto the glycosylated components (proteoglycans and glycosaminoglycans) of normal brain extracellular matrix and the glucocorticoid receptor (GR, Nr3c1) in an experimental model in vivo. Two-month-old male C57Bl/6 mice (n = 90) were injected intraperitoneally with various doses of dexamethasone (DXM) (1; 2.5 mg/kg) for 10 days. The mRNA levels of the GR, proteoglycans core proteins, and heparan sulfate metabolism-involved genes were determined at the 15th, 30th, 60th, and 90th days by a real-time RT-PCR. The glycosaminoglycans content was studied using dot blot and staining with Alcian blue. A DXM treatment increased total GAG content (2-fold), whereas the content of highly sulfated glycosaminoglycans decreased (1.5-2-fold). The mRNA level of the heparan sulfate metabolism-involved gene Hs3St2 increased 5-fold, the mRNA level of Hs6St2 increased6-7-fold, and the mRNA level of proteoglycan aggrecan increased 2-fold. A correlation analysis revealed an association between the mRNA level of the GR and the mRNA level of 8 of the 14 proteoglycans-coding and 4 of the 13 heparan sulfate metabolism-involved genes supporting GR involvement in the DXM regulation of the expression of these genes. In summary, multiple DXM administrations led to an increase in the total GAG content and reorganized the brain extracellular matrix in terms of its glycosylation pattern.
Collapse
Affiliation(s)
- Stanislav D. Aladev
- Institute of Molecular Biology and Biophysics FRC FTM, Novosibirsk 630117, Russia; (D.K.S.); (A.V.S.); (G.M.K.); (S.V.A.); (E.V.G.)
| | - Dmitry K. Sokolov
- Institute of Molecular Biology and Biophysics FRC FTM, Novosibirsk 630117, Russia; (D.K.S.); (A.V.S.); (G.M.K.); (S.V.A.); (E.V.G.)
| | - Anastasia V. Strokotova
- Institute of Molecular Biology and Biophysics FRC FTM, Novosibirsk 630117, Russia; (D.K.S.); (A.V.S.); (G.M.K.); (S.V.A.); (E.V.G.)
| | - Galina M. Kazanskaya
- Institute of Molecular Biology and Biophysics FRC FTM, Novosibirsk 630117, Russia; (D.K.S.); (A.V.S.); (G.M.K.); (S.V.A.); (E.V.G.)
- E.N. Meshalkin National Medical Research Center, Novosibirsk 630055, Russia;
| | - Alexander M. Volkov
- E.N. Meshalkin National Medical Research Center, Novosibirsk 630055, Russia;
| | - Svetlana V. Aidagulova
- Institute of Molecular Biology and Biophysics FRC FTM, Novosibirsk 630117, Russia; (D.K.S.); (A.V.S.); (G.M.K.); (S.V.A.); (E.V.G.)
- Laboratory of Cellular Biology, Novosibirsk State Medical University, Novosibirsk 630091, Russia
| | - Elvira V. Grigorieva
- Institute of Molecular Biology and Biophysics FRC FTM, Novosibirsk 630117, Russia; (D.K.S.); (A.V.S.); (G.M.K.); (S.V.A.); (E.V.G.)
| |
Collapse
|
11
|
Nolta NF, Christensen MB, Tresco PA. Advanced age is not a barrier to chronic intracortical single-unit recording in rat cortex. Front Neurosci 2024; 18:1389556. [PMID: 38817909 PMCID: PMC11138162 DOI: 10.3389/fnins.2024.1389556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Available evidence suggests that as we age, our brain and immune system undergo changes that increase our susceptibility to injury, inflammation, and neurodegeneration. Since a significant portion of the potential patients treated with a microelectrode-based implant may be older, it is important to understand the recording performance of such devices in an aged population. Methods We studied the chronic recording performance and the foreign body response (FBR) to a clinically used microelectrode array implanted in the cortex of 18-month-old Sprague Dawley rats. Results and discussion To the best of our knowledge, this is the first preclinical study of its type in the older mammalian brain. Here, we show that single-unit recording performance was initially robust then gradually declined over a 12-week period, similar to what has been previously reported using younger adult rats and in clinical trials. In addition, we show that FBR biomarker distribution was similar to what has been previously described for younger adult rats implanted with multi-shank recording arrays in the motor cortex. Using a quantitative immunohistochemcal approach, we observed that the extent of astrogliosis and tissue loss near the recording zone was inversely related to recording performance. A comparison of recording performance with a younger cohort supports the notion that aging, in and of itself, is not a limiting factor for the clinical use of penetrating microelectrode recording arrays for the treatment of certain CNS disorders.
Collapse
Affiliation(s)
- Nicholas F. Nolta
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Michael B. Christensen
- Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
- Department of Otolaryngology – Head & Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Patrick A. Tresco
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
12
|
Song SS, Druschel LN, Conard JH, Wang JJ, Kasthuri NM, Ricky Chan E, Capadona JR. Depletion of complement factor 3 delays the neuroinflammatory response to intracortical microelectrodes. Brain Behav Immun 2024; 118:221-235. [PMID: 38458498 DOI: 10.1016/j.bbi.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/26/2024] [Accepted: 03/02/2024] [Indexed: 03/10/2024] Open
Abstract
The neuroinflammatory response to intracortical microelectrodes (IMEs) used with brain-machine interfacing (BMI) applications is regarded as the primary contributor to poor chronic performance. Recent developments in high-plex gene expression technologies have allowed for an evolution in the investigation of individual proteins or genes to be able to identify specific pathways of upregulated genes that may contribute to the neuroinflammatory response. Several key pathways that are upregulated following IME implantation are involved with the complement system. The complement system is part of the innate immune system involved in recognizing and eliminating pathogens - a significant contributor to the foreign body response against biomaterials. Specifically, we have identified Complement 3 (C3) as a gene of interest because it is the intersection of several key complement pathways. In this study, we investigated the role of C3 in the IME inflammatory response by comparing the neuroinflammatory gene expression at the microelectrode implant site between C3 knockout (C3-/-) and wild-type (WT) mice. We have found that, like in WT mice, implantation of intracortical microelectrodes in C3-/- mice yields a dramatic increase in the neuroinflammatory gene expression at all post-surgery time points investigated. However, compared to WT mice, C3 depletion showed reduced expression of many neuroinflammatory genes pre-surgery and 4 weeks post-surgery. Conversely, depletion of C3 increased the expression of many neuroinflammatory genes at 8 weeks and 16 weeks post-surgery, compared to WT mice. Our results suggest that C3 depletion may be a promising therapeutic target for acute, but not chronic, relief of the neuroinflammatory response to IME implantation. Additional compensatory targets may also be required for comprehensive long-term reduction of the neuroinflammatory response for improved intracortical microelectrode performance.
Collapse
Affiliation(s)
- Sydney S 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.
| | - Jacob H Conard
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States.
| | - Jaime J Wang
- 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.
| | - Niveda M Kasthuri
- 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
|
13
|
Waldron OP, El-Mallah JC, Lochan D, Wen C, Landmesser ME, Asgardoon M, Dawes J, Horchler SN, Schlidt K, Agrawal S, Wang Y, Ravnic DJ. Ushering in the era of regenerative surgery. Minerva Surg 2024; 79:166-182. [PMID: 38088753 DOI: 10.23736/s2724-5691.23.10113-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Tissue loss, irrespective of etiology, often requires extensive reconstruction. In many instances, the need exceeds what current treatments and technologies modern medicine can offer. Tissue engineering has made immense strides within the past few decades due to advances in biologics, biomaterials, and manufacturing. The convergence of these three domains has created limitless potential for future surgical care. Unfortunately, there still exists a disconnect on how to best implant these 'replacement parts' and care for the patient. It is therefore vital to develop paradigms for the integration of advanced surgical and tissue engineering technologies. This paper explores the convergence between tissue engineering and reconstructive surgery. We will describe the clinical problem of tissue loss, discuss currently available solutions, address limitations, and propose processes for integrating surgery and tissue engineering, thereby ushering in the era of regenerative surgery.
Collapse
Affiliation(s)
- Olivia P Waldron
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Jessica C El-Mallah
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Dev Lochan
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Mary E Landmesser
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Mohammadhossein Asgardoon
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jazzmyn Dawes
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Summer N Horchler
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Kevin Schlidt
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
| | - Shailaja Agrawal
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Dino J Ravnic
- Irvin S. Zubar Plastic Surgery Research Laboratory, Penn State College of Medicine, Hershey, PA, USA -
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
14
|
Chae U, Chun H, Lim J, Shin H, Smith WC, Choi JW, Park KD, Lee CJ, Cho IJ. KDS2010, a reversible MAO-B inhibitor, extends the lifetime of neural probes by preventing glial scar formation. Glia 2024; 72:748-758. [PMID: 38200694 DOI: 10.1002/glia.24500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
Implantable neural probes have been extensively utilized in the fields of neurocircuitry, systems neuroscience, and brain-computer interface. However, the long-term functionality of these devices is hampered by the formation of glial scar and astrogliosis at the surface of electrodes. In this study, we administered KDS2010, a recently developed reversible MAO-B inhibitor, to mice through ad libitum drinking in order to prevent glial scar formation and astrogliosis. The administration of KDS2010 allowed long-term recordings of neural signals with implantable devices, which remained stable over a period of 6 months and even restored diminished neural signals after probe implantation. KDS2010 effectively prevented the formation of glial scar, which consists of reactive astrocytes and activated microglia around the implant. Furthermore, it restored neural activity by disinhibiting astrocytic MAO-B dependent tonic GABA inhibition induced by astrogliosis. We suggest that the use of KDS2010 is a promising approach to prevent glial scar formation around the implant, thereby enabling long-term functionality of neural devices.
Collapse
Affiliation(s)
- Uikyu Chae
- Department of Convergence Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Heejung Chun
- Yonsei-SLBigen Research Institute, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Jiwoon Lim
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hyogeun Shin
- Department of Convergence Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Wesley Charles Smith
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Ji Won Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science (IBS), Daejeon, Republic of Korea
- IBS School, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Il-Joo Cho
- Department of Convergence Medicine, College of Medicine, Korea University, Seoul, Republic of Korea
- Department of Anatomy, College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
15
|
Cho M, Han JK, Suh J, Kim JJ, Ryu JR, Min IS, Sang M, Lim S, Kim TS, Kim K, Kang K, Hwang K, Kim K, Hong EB, Nam MH, Kim J, Song YM, Lee GJ, Cho IJ, Yu KJ. Fully bioresorbable hybrid opto-electronic neural implant system for simultaneous electrophysiological recording and optogenetic stimulation. Nat Commun 2024; 15:2000. [PMID: 38448437 PMCID: PMC10917781 DOI: 10.1038/s41467-024-45803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Bioresorbable neural implants based on emerging classes of biodegradable materials offer a promising solution to the challenges of secondary surgeries for removal of implanted devices required for existing neural implants. In this study, we introduce a fully bioresorbable flexible hybrid opto-electronic system for simultaneous electrophysiological recording and optogenetic stimulation. The flexible and soft device, composed of biodegradable materials, has a direct optical and electrical interface with the curved cerebral cortex surface while exhibiting excellent biocompatibility. Optimized to minimize light transmission losses and photoelectric artifact interference, the device was chronically implanted in the brain of transgenic mice and performed to photo-stimulate the somatosensory area while recording local field potentials. Thus, the presented hybrid neural implant system, comprising biodegradable materials, promises to provide monitoring and therapy modalities for versatile applications in biomedicine.
Collapse
Affiliation(s)
- Myeongki Cho
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong-Kyu Han
- Brain Science Institute, Korea Institute of Science and Technology, 5. Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jungmin Suh
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong Jin Kim
- Department of Electronics Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| | - Jae Ryun Ryu
- Department of Anatomy, College of Medicine, Korea University, 17-gil Koryodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - In Sik Min
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Mingyu Sang
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Selin Lim
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Tae Soo Kim
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyubeen Kim
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyowon Kang
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyuhyun Hwang
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kanghwan Kim
- Brain Science Institute, Korea Institute of Science and Technology, 5. Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Eun-Bin Hong
- Center for Brain Function, Korea Institute of Science and Technology 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Min-Ho Nam
- Center for Brain Function, Korea Institute of Science and Technology 5, Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jongbaeg Kim
- School of Mechanical Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Min Song
- School of Electrical Engineering and Computer Science (EECS), Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Gil Ju Lee
- Department of Electronics Engineering, Pusan National University, 2, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.
| | - Il-Joo Cho
- Department of Convergence Medicine, College of Medicine, Korea University, 17-gil Koryodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
- Department of Anatomy, College of Medicine, Korea University, 7-gil Koryodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Ki Jun Yu
- Functional Bio-integrated Electronics and Energy Management Lab, School of Electrical and Electronic Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Department of Electrical and Electronic Engineering, YU-Korea Institute of Science and Technology (KIST) Institute, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
16
|
Sturgill B, Hernandez-Reynoso AG, Druschel LN, Smith TJ, Boucher PE, Hoeferlin GF, Thai TTD, Jiang MS, Hess JL, Alam NN, Menendez DM, Duncan JL, Cogan SF, Pancrazio JJ, Capadona JR. Reactive Amine Functionalized Microelectrode Arrays Provide Short-Term Benefit but Long-Term Detriment to In Vivo Recording Performance. ACS APPLIED BIO MATERIALS 2024; 7:1052-1063. [PMID: 38290529 PMCID: PMC10880090 DOI: 10.1021/acsabm.3c01014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Intracortical microelectrode arrays (MEAs) are used for recording neural signals. However, indwelling devices result in chronic neuroinflammation, which leads to decreased recording performance through degradation of the device and surrounding tissue. Coating the MEAs with bioactive molecules is being explored to mitigate neuroinflammation. Such approaches often require an intermediate functionalization step such as (3-aminopropyl)triethoxysilane (APTES), which serves as a linker. However, the standalone effect of this intermediate step has not been previously characterized. Here, we investigated the effect of coating MEAs with APTES by comparing APTES-coated to uncoated controls in vivo and ex vivo. First, we measured water contact angles between silicon uncoated and APTES-coated substrates to verify the hydrophilic characteristics of the APTES coating. Next, we implanted MEAs in the motor cortex (M1) of Sprague-Dawley rats with uncoated or APTES-coated devices. We assessed changes in the electrochemical impedance and neural recording performance over a chronic implantation period of 16 weeks. Additionally, histology and bulk gene expression were analyzed to understand further the reactive tissue changes arising from the coating. Results showed that APTES increased the hydrophilicity of the devices and decreased electrochemical impedance at 1 kHz. APTES coatings proved detrimental to the recording performance, as shown by a constant decay up to 16 weeks postimplantation. Bulk gene analysis showed differential changes in gene expression between groups that were inconclusive with regard to the long-term effect on neuronal tissue. Together, these results suggest that APTES coatings are ultimately detrimental to chronic neural recordings. Furthermore, interpretations of studies using APTES as a functionalization step should consider the potential consequences if the final functionalization step is incomplete.
Collapse
Affiliation(s)
- Brandon
S. Sturgill
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Ana G. Hernandez-Reynoso
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Lindsey N. Druschel
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Thomas J. Smith
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Pierce E. Boucher
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - George F. Hoeferlin
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Teresa Thuc Doan Thai
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Madison S. Jiang
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Jordan L. Hess
- School
of Behavioral and BrainSciences, The University
of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Neeha N. Alam
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Dhariyat M. Menendez
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Jonathan L. Duncan
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| | - Stuart F. Cogan
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Joseph J. Pancrazio
- Department
of Bioengineering, The University of Texas
at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Jeffrey R. Capadona
- Department
of Biomedical Engineering, Case Western
Reserve University. 10900 Euclid Ave, Cleveland, Ohio 44106, United States
- Advanced
Platform Technology Center, Louis Stokes Cleveland Veterans Affairs
Medical Center, Cleveland, Ohio 44106, United States
| |
Collapse
|
17
|
Aktas B, Ozgun A, Kilickap BD, Garipcan B. Cell adhesion molecule immobilized gold surfaces for enhanced neuron-electrode interfaces. J Biomed Mater Res B Appl Biomater 2024; 112:e35310. [PMID: 37950592 DOI: 10.1002/jbm.b.35310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 11/12/2023]
Abstract
To provide a long-term solution for increasing the biocompatibility of neuroprosthetics, approaches to reduce the side effects of invasive neuro-implantable devices are still in need of improvement. Physical, chemical, and bioactive design aspects of the biomaterials are proven to be important for providing proper cell-to-cell, cell-to-material interactions. Particularly, modification of implant surfaces with bioactive cues, especially cell adhesion molecules (CAMs) that capitalize on native neural adhesion mechanisms, are promising candidates in favor of providing efficient interfaces. Within this concept, this study utilized specific CAMs, namely N-Cadherin (Neural cadherin, N-Cad) and neural cell adhesion molecule (NCAM), to enhance neuron-electrode contact by mimicking the cell-to-ECM interactions for improving the survival of cells and promoting neurite outgrowth. For this purpose, representative gold electrode surfaces were modified with N-Cadherin, NCAM, and the mixture (1:1) of these molecules. Modifications were characterized, and the effect of surface modification on both differentiated and undifferentiated neuroblastoma SH-SY5Y cell lines were compared. The findings demonstrated the successful modification of these molecules which subsequently exhibited biocompatible properties as evidenced by the cell viability results. In cell culture experiments, the CAMs displayed promising results in promoting neurite outgrowth compared to conventional poly-l-lysine coated surfaces, especially NCAM and N-Cad/NCAM modified surfaces clearly showed significant improvement. Overall, this optimized approach is expected to provide an insight into the action mechanisms of cells against the local environment and advance processes for the fabrication of alternative neural interfaces.
Collapse
Affiliation(s)
- Bengu Aktas
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Alp Ozgun
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Bora Garipcan
- Institute of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| |
Collapse
|
18
|
Moslehi S, Rowland C, Smith JH, Watterson WJ, Griffiths W, Montgomery RD, Philliber S, Marlow CA, Perez MT, Taylor RP. Fractal Electronics for Stimulating and Sensing Neural Networks: Enhanced Electrical, Optical, and Cell Interaction Properties. ADVANCES IN NEUROBIOLOGY 2024; 36:849-875. [PMID: 38468067 DOI: 10.1007/978-3-031-47606-8_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Imagine a world in which damaged parts of the body - an arm, an eye, and ultimately a region of the brain - can be replaced by artificial implants capable of restoring or even enhancing human performance. The associated improvements in the quality of human life would revolutionize the medical world and produce sweeping changes across society. In this chapter, we discuss several approaches to the fabrication of fractal electronics designed to interface with neural networks. We consider two fundamental functions - stimulating electrical signals in the neural networks and sensing the location of the signals as they pass through the network. Using experiments and simulations, we discuss the favorable electrical performances that arise from adopting fractal rather than traditional Euclidean architectures. We also demonstrate how the fractal architecture induces favorable physical interactions with the cells they interact with, including the ability to direct the growth of neurons and glia to specific regions of the neural-electronic interface.
Collapse
Affiliation(s)
- S Moslehi
- Physics Department, University of Oregon, Eugene, OR, USA
| | - C Rowland
- Physics Department, University of Oregon, Eugene, OR, USA
| | - J H Smith
- Physics Department, University of Oregon, Eugene, OR, USA
| | - W J Watterson
- Physics Department, University of Oregon, Eugene, OR, USA
| | - W Griffiths
- Physics Department, University of Oregon, Eugene, OR, USA
| | - R D Montgomery
- Physics Department, University of Oregon, Eugene, OR, USA
| | - S Philliber
- Physics Department, University of Oregon, Eugene, OR, USA
| | - C A Marlow
- Physics Department, California Polytechnic State University, San Luis Obispo, CA, USA
| | - M-T Perez
- Department of Clinical Sciences Lund, Division of Ophthalmology, Lund University, Lund, Sweden
| | - R P Taylor
- Physics Department, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
19
|
Chan AH, Moore MJ, Grant AJ, Lam YTM, Darnell MV, Michael PL, Wise SG, Tan RP. Selective Immunosuppression Targeting the NLRP3 Inflammasome Mitigates the Foreign Body Response to Implanted Biomaterials While Preserving Angiogenesis. Adv Healthc Mater 2023; 12:e2301571. [PMID: 37846971 PMCID: PMC11469290 DOI: 10.1002/adhm.202301571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/04/2023] [Indexed: 10/18/2023]
Abstract
Medical devices are a mainstay of the healthcare industry, providing clinicians with innovative tools to diagnose, monitor, and treat a range of medical conditions. For implantable devices, it is widely regarded that chronic inflammation during the foreign body response (FBR) is detrimental to device performance, but also required for tissue regeneration and host integration. Current strategies to mitigate the FBR rely on broad acting anti-inflammatory drugs, most commonly, dexamethasone (DEX), which can inhibit angiogenesis and compromise long-term device function. This study challenges prevailing assumptions by suggesting that FBR inflammation is multifaceted, and selectively targeting its individual pathways can stop implant fibrosis while preserving beneficial repair pathways linked to improved device performance. MCC950, an anti-inflammatory drug that selectively inhibits the NLRP3 inflammasome, targets pathological inflammation without compromising global immune function. The effects of MCC950 and DEX on the FBR are compared using implanted polycaprolactone (PCL) scaffolds. The results demonstrate that both DEX and MCC950 halt immune cell recruitment and cytokine release, leading to reduced FBR. However, MCC950 achieves this while supporting capillary growth and enhancing tissue angiogenesis. These findings support selective immunosuppression approaches as a potential future direction for treating the FBR and enhancing the longevity and safety of implantable devices.
Collapse
Affiliation(s)
- Alex H.P. Chan
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Matthew J. Moore
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Angus J. Grant
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Yuen Ting Monica Lam
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Matthew V. Darnell
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Praveesuda L. Michael
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Steven G. Wise
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| | - Richard P. Tan
- School of Medical SciencesFaculty of Health and MedicineUniversity of SydneySydneyNSW2006Australia
- Charles Perkins CentreUniversity of SydneySydneyNSW2006Australia
| |
Collapse
|
20
|
Perna A, Angotzi GN, Berdondini L, Ribeiro JF. Advancing the interfacing performances of chronically implantable neural probes in the era of CMOS neuroelectronics. Front Neurosci 2023; 17:1275908. [PMID: 38027514 PMCID: PMC10644322 DOI: 10.3389/fnins.2023.1275908] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Tissue penetrating microelectrode neural probes can record electrophysiological brain signals at resolutions down to single neurons, making them invaluable tools for neuroscience research and Brain-Computer-Interfaces (BCIs). The known gradual decrease of their electrical interfacing performances in chronic settings, however, remains a major challenge. A key factor leading to such decay is Foreign Body Reaction (FBR), which is the cascade of biological responses that occurs in the brain in the presence of a tissue damaging artificial device. Interestingly, the recent adoption of Complementary Metal Oxide Semiconductor (CMOS) technology to realize implantable neural probes capable of monitoring hundreds to thousands of neurons simultaneously, may open new opportunities to face the FBR challenge. Indeed, this shift from passive Micro Electro-Mechanical Systems (MEMS) to active CMOS neural probe technologies creates important, yet unexplored, opportunities to tune probe features such as the mechanical properties of the probe, its layout, size, and surface physicochemical properties, to minimize tissue damage and consequently FBR. Here, we will first review relevant literature on FBR to provide a better understanding of the processes and sources underlying this tissue response. Methods to assess FBR will be described, including conventional approaches based on the imaging of biomarkers, and more recent transcriptomics technologies. Then, we will consider emerging opportunities offered by the features of CMOS probes. Finally, we will describe a prototypical neural probe that may meet the needs for advancing clinical BCIs, and we propose axial insertion force as a potential metric to assess the influence of probe features on acute tissue damage and to control the implantation procedure to minimize iatrogenic injury and subsequent FBR.
Collapse
Affiliation(s)
- Alberto Perna
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
- The Open University Affiliated Research Centre at Istituto Italiano di Tecnologia (ARC@IIT), Istituto Italiano di Tecnologia, Genova, Italy
| | - Gian Nicola Angotzi
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| | - Luca Berdondini
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| | - João Filipe Ribeiro
- Microtechnology for Neuroelectronics Lab, Fondazione Istituto Italiano di Tecnologia, Neuroscience and Brain Technologies, Genova, Italy
| |
Collapse
|
21
|
Song S, Druschel LN, Chan ER, Capadona JR. Differential expression of genes involved in the chronic response to intracortical microelectrodes. Acta Biomater 2023; 169:348-362. [PMID: 37507031 PMCID: PMC10528922 DOI: 10.1016/j.actbio.2023.07.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023]
Abstract
Brain-Machine Interface systems (BMIs) are clinically valuable devices that can provide functional restoration for patients with spinal cord injury or improved integration for patients requiring prostheses. Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for precisely controlling BMIs. However, intracortical microelectrodes have a demonstrated history of progressive decline in the recording performance with time, inhibiting their usefulness. One major contributor to decreased performance is the neuroinflammatory response to the implanted microelectrodes. The neuroinflammatory response can lead to neurodegeneration and the formation of a glial scar at the implant site. Historically, histological imaging of relatively few known cellular and protein markers has characterized the neuroinflammatory response to implanted microelectrode arrays. However, neuroinflammation requires many molecular players to coordinate the response - meaning traditional methods could result in an incomplete understanding. Taking advantage of recent advancements in tools to characterize the relative or absolute DNA/RNA expression levels, a few groups have begun to explore gene expression at the microelectrode-tissue interface. We have utilized a custom panel of ∼813 neuroinflammatory-specific genes developed with NanoString for bulk tissue analysis at the microelectrode-tissue interface. Our previous studies characterized the acute innate immune response to intracortical microelectrodes. Here we investigated the gene expression at the microelectrode-tissue interface in wild-type (WT) mice chronically implanted with nonfunctioning probes. We found 28 differentially expressed genes at chronic time points (4WK, 8WK, and 16WK), many in the complement and extracellular matrix system. Further, the expression levels were relatively stable over time. Genes identified here represent chronic molecular players at the microelectrode implant sites and potential therapeutic targets for the long-term integration of microelectrodes. STATEMENT OF SIGNIFICANCE: Intracortical microelectrodes can record neuronal action potentials at a resolution necessary for the precise control of Brain-Machine Interface systems (BMIs). However, intracortical microelectrodes have a demonstrated history of progressive declines in the recording performance with time, inhibiting their usefulness. One major contributor to the decline in these devices is the neuroinflammatory response against the implanted microelectrodes. Historically, neuroinflammation to implanted microelectrode arrays has been characterized by histological imaging of relatively few known cellular and protein markers. Few studies have begun to develop a more in-depth understanding of the molecular pathways facilitating device-mediated neuroinflammation. Here, we are among the first to identify genetic pathways that could represent targets to improve the host response to intracortical microelectrodes, and ultimately device performance.
Collapse
Affiliation(s)
- Sydney Song
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - Lindsey N Druschel
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States
| | - E Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, United States; Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, United States.
| |
Collapse
|
22
|
Sokolov DK, Shevelev OB, Khotskina AS, Tsidulko AY, Strokotova AV, Kazanskaya GM, Volkov AM, Kliver EE, Aidagulova SV, Zavjalov EL, Grigorieva EV. Dexamethasone Inhibits Heparan Sulfate Biosynthetic System and Decreases Heparan Sulfate Content in Orthotopic Glioblastoma Tumors in Mice. Int J Mol Sci 2023; 24:10243. [PMID: 37373391 DOI: 10.3390/ijms241210243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma (GB) is an aggressive cancer with a high probability of recurrence, despite active chemoradiotherapy with temozolomide (TMZ) and dexamethasone (DXM). These systemic drugs affect the glycosylated components of brain tissue involved in GB development; however, their effects on heparan sulfate (HS) remain unknown. Here, we used an animal model of GB relapse in which SCID mice first received TMZ and/or DXM (simulating postoperative treatment) with a subsequent inoculation of U87 human GB cells. Control, peritumor and U87 xenograft tissues were investigated for HS content, HS biosynthetic system and glucocorticoid receptor (GR, Nr3c1). In normal and peritumor brain tissues, TMZ/DXM administration decreased HS content (5-6-fold) but did not affect HS biosynthetic system or GR expression. However, the xenograft GB tumors grown in the pre-treated animals demonstrated a number of molecular changes, despite the fact that they were not directly exposed to TMZ/DXM. The tumors from DXM pre-treated animals possessed decreased HS content (1.5-2-fold), the inhibition of HS biosynthetic system mainly due to the -3-3.5-fold down-regulation of N-deacetylase/N-sulfotransferases (Ndst1 and Ndst2) and sulfatase 2 (Sulf2) expression and a tendency toward a decreased expression of the GRalpha but not the GRbeta isoform. The GRalpha expression levels in tumors from DXM or TMZ pre-treated mice were positively correlated with the expression of a number of HS biosynthesis-involved genes (Ext1/2, Ndst1/2, Glce, Hs2st1, Hs6st1/2), unlike tumors that have grown in intact SCID mice. The obtained data show that DXM affects HS content in mouse brain tissues, and GB xenografts grown in DXM pre-treated animals demonstrate attenuated HS biosynthesis and decreased HS content.
Collapse
Affiliation(s)
- Dmitry K Sokolov
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
| | - Oleg B Shevelev
- Institute of Cytology and Genetics, Novosibirsk 630090, Russia
| | | | - Alexandra Y Tsidulko
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
| | - Anastasia V Strokotova
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
| | - Galina M Kazanskaya
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
| | - Alexander M Volkov
- E.N. Meshalkin National Medical Research Center, Novosibirsk 630055, Russia
| | - Evgenii E Kliver
- E.N. Meshalkin National Medical Research Center, Novosibirsk 630055, Russia
| | - Svetlana V Aidagulova
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
- Laboratory of Cell Biology, Novosibirsk State Medical University, Novosibirsk 630091, Russia
| | | | - Elvira V Grigorieva
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine, Novosibirsk 630117, Russia
| |
Collapse
|
23
|
Abstract
Advances in bioelectronic implants have been offering valuable chances to interface and modulate neural systems. Potential mismatches between bioelectronics and targeted neural tissues require devices to exhibit "tissue-like" properties for better implant-bio integration. In particular, mechanical mismatches pose a significant challenge. In the past years, efforts were made in both materials synthesis and device design to achieve bioelectronics mechanically and biochemically mimicking biological tissues. In this perspective, we mainly summarized recent progress of developing "tissue-like" bioelectronics and categorized them into different strategies. We also discussed how these "tissue-like" bioelectronics were utilized for modulating in vivo nervous systems and neural organoids. We concluded the perspective by proposing further directions including personalized bioelectronics, novel materials design and the involvement of artificial intelligence and robotic techniques.
Collapse
Affiliation(s)
- Changxu Sun
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Zhe Cheng
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Jj Abu-Halimah
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
| | - Bozhi Tian
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA
- The James Franck Institute, University of Chicago, Chicago, IL 60637, USA
- The Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
24
|
Dal-Fabbro R, Swanson WB, Capalbo LC, Sasaki H, Bottino MC. Next-generation biomaterials for dental pulp tissue immunomodulation. Dent Mater 2023; 39:333-349. [PMID: 36894414 PMCID: PMC11034777 DOI: 10.1016/j.dental.2023.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
OBJECTIVES The current standard for treating irreversibly damaged dental pulp is root canal therapy, which involves complete removal and debridement of the pulp space and filling with an inert biomaterial. A regenerative approach to treating diseased dental pulp may allow for complete healing of the native tooth structure and enhance the long-term outcome of once-necrotic teeth. The aim of this paper is, therefore, to highlight the current state of dental pulp tissue engineering and immunomodulatory biomaterials properties, identifying exciting opportunities for their synergy in developing next-generation biomaterials-driven technologies. METHODS An overview of the inflammatory process focusing on immune responses of the dental pulp, followed by periapical and periodontal tissue inflammation are elaborated. Then, the most recent advances in treating infection-induced inflammatory oral diseases, focusing on biocompatible materials with immunomodulatory properties are discussed. Of note, we highlight some of the most used modifications in biomaterials' surface, or content/drug incorporation focused on immunomodulation based on an extensive literature search over the last decade. RESULTS We provide the readers with a critical summary of recent advances in immunomodulation related to pulpal, periapical, and periodontal diseases while bringing light to tissue engineering strategies focusing on healing and regenerating multiple tissue types. SIGNIFICANCE Significant advances have been made in developing biomaterials that take advantage of the host's immune system to guide a specific regenerative outcome. Biomaterials that efficiently and predictably modulate cells in the dental pulp complex hold significant clinical promise for improving standards of care compared to endodontic root canal therapy.
Collapse
Affiliation(s)
- Renan Dal-Fabbro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - W Benton Swanson
- Department of Biologic and Materials Science, Division of Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Leticia C Capalbo
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Hajime Sasaki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Aladev SD, Sokolov DK, Strokotova AV, Kazanskaya GM, Volkov AM, Politko MO, Shahmuradova AI, Kliver EE, Tsidulko AY, Aidagulova SV, Grigorieva EV. Dexamethasone effects on the expression and content of glycosylated components of mouse brain tissue. ADVANCES IN MOLECULAR ONCOLOGY 2023. [DOI: 10.17650/2313-805x-2023-10-1-25-39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Introduction. Glucocorticoids are actively used in the treatment of various diseases, however their long-term use leads to numerous negative side-effects, the molecular mechanisms of which remain poorly understood.Aim. Study of the short-term (1–10 days) effects of various doses of dexamethasone (Dex) (0,1–10 mg/kg) on the expression of the glucocorticoid receptor (GR, Nr3c1), core proteins of main proteoglycans and heparan sulfate metabolism-involved genes, as well as the content of carbohydrate macromolecules of glycosaminoglycans in the brain tissue of experimental animals.Materials and methods. In the study, C57Bl/6 mice were used. The expression of GR, proteoglycan core proteins and heparan sulfate metabolism-involved genes was determined by real-time polymerase chain reaction with reverse transcription. The content and localization of GR protein molecule were studied by Western blot and immunohistochemical analysis, and the glycosaminoglycan content was determined by dot-blot analysis and Alcian Blue staining.Results. It was shown that a single Dex administration leads to fast (1–3 days) short-term activation of GR expression (+1.5 times, p <0.05), proteoglycan’s genes (syndecan-3, Sdc3; perlecan, Hspg2; phosphacan, Ptprz1; neurocan, Ncan; +2–3-fold; p <0.05) and heparan sulfate-metabolism-involved genes (Ndst1, Glce, Hs2st1, Hs6st1, Sulf1 / 2; +1.5–2-fold; p <0.05) in the mouse brain, with a return to control values by 7–10 days after Dex administration. At the same time, the effect of Dex on carbohydrate macromolecules of glycosaminoglycans was more delayed and stable, increasing the content of low-sulfated glycosaminoglycans in the brain tissue in a dose-dependent manner starting from day 1 after Dex administration. Highly-sulfated glycosaminoglycans showed more delayed response to Dex administration, and an increase in their content was observed only at higher doses (2.5 and 10 mg/kg) and only on 7–10 days after its administration, apparently, mainly due to an increase in heparan sulfate content.Conclusion. In general, the effect of a single injection of Dex on the transcriptional activity of GR, proteoglycan core proteins and heparan sulfate metabolism-involved genes were short-termed, and the genes expression quickly returned to the normal levels. However, even a single use of Dex significantly increased the content of total as well as highly sulfated glycosaminoglycans in the mouse brain tissue, which can lead to the changes in the composition and structure of the brain tissue, as well as its functional characteristics.
Collapse
Affiliation(s)
- S. D. Aladev
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | - D. K. Sokolov
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | - A. V. Strokotova
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | - G. M. Kazanskaya
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | | | - M. O. Politko
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | - A. I. Shahmuradova
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | | | - A. Y. Tsidulko
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| | - S. V. Aidagulova
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation; Novosibirsk State Medical University
| | - E. V. Grigorieva
- Institute of Molecular Biology and Biophysics of the Federal Research Center for Fundamental and Translational Medicine, Ministry of Science and Higher Education of the Russian Federation
| |
Collapse
|
26
|
Sharon A, Jankowski MM, Shmoel N, Erez H, Spira ME. Significantly reduced inflammatory foreign-body-response to neuroimplants and improved recording performance in young compared to adult rats. Acta Biomater 2023; 158:292-307. [PMID: 36632879 DOI: 10.1016/j.actbio.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/11/2023]
Abstract
The multicellular inflammatory encapsulation of implanted intracortical multielectrode arrays (MEA) is associated with severe deterioration of their field potentials' (FP) recording performance, which thus limits the use of brain implants in basic research and clinical applications. Therefore, extensive efforts have been made to identify the conditions in which the inflammatory foreign body response (FBR) is alleviated, or to develop methods to mitigate the formation of the inflammatory barrier. Here, for the first time, we show that (1) in young rats (74±8 gr, 4 weeks old at the onset of the experiments), cortical tissue recovery following MEA implantation proceeds with ameliorated inflammatory scar as compared to adult rats (242 ± 18 gr, 9 weeks old at the experimental onset); (2) in contrast to adult rats in which the Colony Stimulating factor 1 Receptor (CSF1R) antagonist chow eliminated ∼95% of the cortical microglia but not microglia adhering to the implant surfaces, in young rats the microglia adhering to the implant were eliminated along with the parenchymal microglia population. The removal of microglia adhering to the implant surfaces was correlated with improved recording performance by in-house fabricated Perforated Polyimide MEA Platforms (PPMP). These results support the hypothesis that microglia adhering to the surface of the electrodes, rather than the multicellular inflammatory scar, is the major underlying mechanism that deteriorates implant recording performance, and that young rats provide an advantageous model to study months-long, multisite electrophysiology in freely behaving rats. STATEMENT OF SIGNIFICANCE: Multisite electrophysiological recordings and stimulation devices play central roles in basic brain research and medical applications. The insertion of multielectrode-array platforms into the brain's parenchyma unavoidably injures the tissue, and initiates a multicellular inflammatory cascade culminating in the formation of an encapsulating scar tissue (the foreign body response-FBR). The dominant view, which directs most current research efforts to mitigate the FBR, holds that the FBR is the major hurdle to effective electrophysiological use of neuroprobes. By contrast, this report demonstrates that microglia adhering to the surface of a neuroimplants, rather than the multicellular FBR, underlie the performance deterioration of neuroimplants. These findings pave the way to the development of novel and focused strategies to overcome the functional deterioration of neuroimplants.
Collapse
Affiliation(s)
- Aviv Sharon
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maciej M Jankowski
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel; Edmond and Lily Safra Center for Brain Sciences, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nava Shmoel
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadas Erez
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Micha E Spira
- Department of Neurobiology, the Alexander Silberman Institute of Life Science, the Hebrew University of Jerusalem, Jerusalem, Israel; The Charles E. Smith Family and Prof. Joel Elkes Laboratory for Collaborative Research in Psychobiology, the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
27
|
Ahnood A, Chambers A, Gelmi A, Yong KT, Kavehei O. Semiconducting electrodes for neural interfacing: a review. Chem Soc Rev 2023; 52:1491-1518. [PMID: 36734845 DOI: 10.1039/d2cs00830k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In the past 50 years, the advent of electronic technology to directly interface with neural tissue has transformed the fields of medicine and biology. Devices that restore or even replace impaired bodily functions, such as deep brain stimulators and cochlear implants, have ushered in a new treatment era for previously intractable conditions. Meanwhile, electrodes for recording and stimulating neural activity have allowed researchers to unravel the vast complexities of the human nervous system. Recent advances in semiconducting materials have allowed effective interfaces between electrodes and neuronal tissue through novel devices and structures. Often these are unattainable using conventional metallic electrodes. These have translated into advances in research and treatment. The development of semiconducting materials opens new avenues in neural interfacing. This review considers this emerging class of electrodes and how it can facilitate electrical, optical, and chemical sensing and modulation with high spatial and temporal precision. Semiconducting electrodes have advanced electrically based neural interfacing technologies owing to their unique electrochemical and photo-electrochemical attributes. Key operation modalities, namely sensing and stimulation in electrical, biochemical, and optical domains, are discussed, highlighting their contrast to metallic electrodes from the application and characterization perspective.
Collapse
Affiliation(s)
- Arman Ahnood
- School of Engineering, RMIT University, VIC 3000, Australia
| | - Andre Chambers
- School of Physics, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Amy Gelmi
- School of Science, RMIT University, VIC 3000, Australia
| | - Ken-Tye Yong
- School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia.,The University of Sydney Nano Institute, Sydney, NSW 2006, Australia.
| | - Omid Kavehei
- School of Biomedical Engineering, University of Sydney, Sydney, NSW 2006, Australia.,The University of Sydney Nano Institute, Sydney, NSW 2006, Australia.
| |
Collapse
|
28
|
Ziemba AM, Woodson MCC, Funnell JL, Wich D, Balouch B, Rende D, Amato DN, Bao J, Oprea I, Cao D, Bajalo N, Ereifej ES, Capadona JR, Palermo EF, Gilbert RJ. Development of a Slow-Degrading Polymerized Curcumin Coating for Intracortical Microelectrodes. ACS APPLIED BIO MATERIALS 2023; 6:806-818. [PMID: 36749645 PMCID: PMC11366415 DOI: 10.1021/acsabm.2c00969] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intracortical microelectrodes are used with brain-computer interfaces to restore lost limb function following nervous system injury. While promising, recording ability of intracortical microelectrodes diminishes over time due, in part, to neuroinflammation. As curcumin has demonstrated neuroprotection through anti-inflammatory activity, we fabricated a 300 nm-thick intracortical microelectrode coating consisting of a polyurethane copolymer of curcumin and polyethylene glycol (PEG), denoted as poly(curcumin-PEG1000 carbamate) (PCPC). The uniform PCPC coating reduced silicon wafer hardness by two orders of magnitude and readily absorbed water within minutes, demonstrating that the coating is soft and hydrophilic in nature. Using an in vitro release model, curcumin eluted from the PCPC coating into the supernatant over 1 week; the majority of the coating was intact after an 8-week incubation in buffer, demonstrating potential for longer term curcumin release and softness. Assessing the efficacy of PCPC within a rat intracortical microelectrode model in vivo, there were no significant differences in tissue inflammation, scarring, neuron viability, and myelin damage between the uncoated and PCPC-coated probes. As the first study to implant nonfunctional probes with a polymerized curcumin coating, we have demonstrated the biocompatibility of a PCPC coating and presented a starting point in the design of poly(pro-curcumin) polymers as coating materials for intracortical electrodes.
Collapse
Affiliation(s)
- Alexis M Ziemba
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Neuroscience Program, Department of Biological Sciences, Smith College, Northampton 01063, Massachusetts, United States
| | - Mary Clare Crochiere Woodson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Jessica L Funnell
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Douglas Wich
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Bailey Balouch
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Deniz Rende
- Center for Materials, Devices, and Integrated Systems, Rensselaer Polytechnic Institute, 110 8th Street, Troy 12180-3590, New York, United States
| | - Dahlia N Amato
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Jonathan Bao
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Ingrid Oprea
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Dominica Cao
- Neuroscience Program, Department of Biological Sciences, Smith College, Northampton 01063, Massachusetts, United States
| | - Neda Bajalo
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Evon S Ereifej
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor 48104, Michigan, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor 48104, Michigan, United States
- Department of Neurology, University of Michigan, Ann Arbor 48104, Michigan, United States
- United States Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland 44106, Ohio, United States
| | - Jeffrey R Capadona
- United States Advanced Platform Technology Center, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland 44106, Ohio, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland 44106, Ohio, United States
| | - Edmund F Palermo
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
- Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy 12180-3590, New York, United States
| |
Collapse
|
29
|
Strokotova AV, Grigorieva EV. Glucocorticoid Effects on Proteoglycans and Glycosaminoglycans. Int J Mol Sci 2022; 23:ijms232415678. [PMID: 36555315 PMCID: PMC9778983 DOI: 10.3390/ijms232415678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids are steroid hormones that play diverse roles in numerous normal and pathological processes. They are actively used to treat a wide variety of diseases, including neurodegenerative and inflammatory diseases, cancers, and COVID-19, among others. However, the long-term use of glucocorticoids is associated with numerous side effects. Molecular mechanisms of these negative side effects are not completely understood. Recently, arguments have been made that one such mechanisms may be related to the influence of glucocorticoids on O-glycosylated components of the cell surface and extracellular matrix, in particular on proteoglycans and glycosaminoglycans. The potential toxic effects of glucocorticoids on these glycosylated macromolecules are particularly meaningful for brain physiology because proteoglycans/glycosaminoglycans are the main extracellular components of brain tissue. Here, we aim to review the known effects of glucocorticoids on proteoglycan expression and glycosaminoglycan content in different tissues, with a specific focus on the brain.
Collapse
|
30
|
Walton F, Cerezo-Sanchez M, McGlynn E, Das R, Heidari H. Cleanroom strategies for micro- and nano-fabricating flexible implantable neural electronics. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2022; 380:20210009. [PMID: 35658678 PMCID: PMC9168450 DOI: 10.1098/rsta.2021.0009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Implantable electronic neural interfaces typically take the form of probes and are made with rigid materials such as silicon and metals. These have advantages such as compatibility with integrated microchips, simple implantation and high-density nanofabrication but tend to be lacking in terms of biointegration, biocompatibility and durability due to their mechanical rigidity. This leads to damage to the device or, more importantly, the brain tissue surrounding the implant. Flexible polymer-based probes offer superior biocompatibility in terms of surface biochemistry and better matched mechanical properties. Research which aims to bring the fabrication of electronics on flexible polymer substrates to the nano-regime is remarkably sparse, despite the push for flexible consumer electronics in the last decade or so. Cleanroom-based nanofabrication techniques such as photolithography have been used as pattern transfer methods by the semiconductor industry to produce single nanometre scale devices and are now also used for making flexible circuit boards. There is still much scope for miniaturizing flexible electronics further using photolithography, bringing the possibility of nanoscale, non-invasive, high-density flexible neural interfacing. This work explores the fabrication challenges of using photolithography and complementary techniques in a cleanroom for producing flexible electronic neural probes with nanometre-scale features. This article is part of the theme issue 'Advanced neurotechnologies: translating innovation for health and well-being'.
Collapse
Affiliation(s)
- Finlay Walton
- Microelectronics Lab, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Maria Cerezo-Sanchez
- Microelectronics Lab, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Eve McGlynn
- Microelectronics Lab, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Rupam Das
- Microelectronics Lab, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Hadi Heidari
- Microelectronics Lab, James Watt School of Engineering, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
31
|
Abstract
Neuroprosthetic devices that record and modulate neural activities have demonstrated immense potential for bypassing or restoring lost neurological functions due to neural injuries and disorders. However, implantable electrical devices interfacing with brain tissue are susceptible to a series of inflammatory tissue responses along with mechanical or electrical failures which can affect the device performance over time. Several biomaterial strategies have been implemented to improve device-tissue integration for high quality and stable performance. Ranging from developing smaller, softer, and more flexible electrode designs to introducing bioactive coatings and drug-eluting layers on the electrode surface, such strategies have shown different degrees of success but with limitations. With their hydrophilic properties and specific bioactivities, carbohydrates offer a potential solution for addressing some of the limitations of the existing biomolecular approaches. In this review, we summarize the role of polysaccharides in the central nervous system, with a primary focus on glycoproteins and proteoglycans, to shed light on their untapped potential as biomaterials for neural implants. Utilization of glycosaminoglycans for neural interface and tissue regeneration applications is comprehensively reviewed to provide the current state of carbohydrate-based biomaterials for neural implants. Finally, we will discuss the challenges and opportunities of applying carbohydrate-based biomaterials for neural tissue interfaces.
Collapse
Affiliation(s)
- Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Neural Basis of Cognition, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
32
|
Gurke J, Naegele TE, Hilton S, Pezone R, Curto VF, Barone DG, List-Kratochvil EJW, Carnicer-Lombarte A, Malliaras GG. Hybrid fabrication of multimodal intracranial implants for electrophysiology and local drug delivery. MATERIALS HORIZONS 2022; 9:1727-1734. [PMID: 35474130 PMCID: PMC9169700 DOI: 10.1039/d1mh01855h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/21/2022] [Indexed: 05/31/2023]
Abstract
New fabrication approaches for mechanically flexible implants hold the key to advancing the applications of neuroengineering in fundamental neuroscience and clinic. By combining the high precision of thin film microfabrication with the versatility of additive manufacturing, we demonstrate a straight-forward approach for the prototyping of intracranial implants with electrode arrays and microfluidic channels. We show that the implant can modulate neuronal activity in the hippocampus through localized drug delivery, while simultaneously recording brain activity by its electrodes. Moreover, good implant stability and minimal tissue response are seen one-week post-implantation. Our work shows the potential of hybrid fabrication combining different manufacturing techniques in neurotechnology and paves the way for a new approach to the development of multimodal implants.
Collapse
Affiliation(s)
- Johannes Gurke
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
| | - Tobias E Naegele
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
| | - Sam Hilton
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
| | - Roberto Pezone
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
| | - Vincenzo F Curto
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
| | - Damiano G Barone
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
- University of Cambridge, School of Clinical Medicine, Department of Clinical Neurosciences, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Emil J W List-Kratochvil
- Humboldt-Universität zu Berlin, Department of Chemistry and of Physics and IRIS Adlershof, Hybrid Devices Group, Zum Großen Windkanal 2, 12489 Berlin, Germany
- Helmholtz-Zentrum für Materialien und Energie GmbH, Hahn-Meitner-Platz 1, 14109, Berlin, Germany
| | | | - George G Malliaras
- University of Cambridge, Electrical Engineering Division, 9 JJ Thomson Ave, Cambridge, CB3 0FA, UK.
| |
Collapse
|
33
|
Moslehi S, Rowland C, Smith JH, Watterson WJ, Miller D, Niell CM, Alemán BJ, Perez MT, Taylor RP. Controlled assembly of retinal cells on fractal and Euclidean electrodes. PLoS One 2022; 17:e0265685. [PMID: 35385490 PMCID: PMC8985931 DOI: 10.1371/journal.pone.0265685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/04/2022] [Indexed: 11/25/2022] Open
Abstract
Controlled assembly of retinal cells on artificial surfaces is important for fundamental cell research and medical applications. We investigate fractal electrodes with branches of vertically-aligned carbon nanotubes and silicon dioxide gaps between the branches that form repeating patterns spanning from micro- to milli-meters, along with single-scaled Euclidean electrodes. Fluorescence and electron microscopy show neurons adhere in large numbers to branches while glial cells cover the gaps. This ensures neurons will be close to the electrodes’ stimulating electric fields in applications. Furthermore, glia won’t hinder neuron-branch interactions but will be sufficiently close for neurons to benefit from the glia’s life-supporting functions. This cell ‘herding’ is adjusted using the fractal electrode’s dimension and number of repeating levels. We explain how this tuning facilitates substantial glial coverage in the gaps which fuels neural networks with small-world structural characteristics. The large branch-gap interface then allows these networks to connect to the neuron-rich branches.
Collapse
Affiliation(s)
- Saba Moslehi
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
| | - Conor Rowland
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
| | - Julian H. Smith
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
| | - William J. Watterson
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
| | - David Miller
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
- Oregon Center for Optical, Molecular and Quantum Science, University of Oregon, Eugene, Oregon, United States of America
| | - Cristopher M. Niell
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Benjamín J. Alemán
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
- Oregon Center for Optical, Molecular and Quantum Science, University of Oregon, Eugene, Oregon, United States of America
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon, United States of America
| | - Maria-Thereza Perez
- Division of Ophthalmology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
- * E-mail: (RPT); (MTP)
| | - Richard P. Taylor
- Physics Department, University of Oregon, Eugene, Oregon, United States of America
- Materials Science Institute, University of Oregon, Eugene, Oregon, United States of America
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, Oregon, United States of America
- * E-mail: (RPT); (MTP)
| |
Collapse
|
34
|
Bianchi M, De Salvo A, Asplund M, Carli S, Di Lauro M, Schulze‐Bonhage A, Stieglitz T, Fadiga L, Biscarini F. Poly(3,4-ethylenedioxythiophene)-Based Neural Interfaces for Recording and Stimulation: Fundamental Aspects and In Vivo Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104701. [PMID: 35191224 PMCID: PMC9036021 DOI: 10.1002/advs.202104701] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/04/2022] [Indexed: 05/29/2023]
Abstract
Next-generation neural interfaces for bidirectional communication with the central nervous system aim to achieve the intimate integration with the neural tissue with minimal neuroinflammatory response, high spatio-temporal resolution, very high sensitivity, and readout stability. The design and manufacturing of devices for low power/low noise neural recording and safe and energy-efficient stimulation that are, at the same time, conformable to the brain, with matched mechanical properties and biocompatibility, is a convergence area of research where neuroscientists, materials scientists, and nanotechnologists operate synergically. The biotic-abiotic neural interface, however, remains a formidable challenge that prompts for new materials platforms and innovation in device layouts. Conductive polymers (CP) are attractive materials to be interfaced with the neural tissue and to be used as sensing/stimulating electrodes because of their mixed ionic-electronic conductivity, their low contact impedance, high charge storage capacitance, chemical versatility, and biocompatibility. This manuscript reviews the state-of-the-art of poly(3,4-ethylenedioxythiophene)-based neural interfaces for extracellular recording and stimulation, focusing on those technological approaches that are successfully demonstrated in vivo. The aim is to highlight the most reliable and ready-for-clinical-use solutions, in terms of materials technology and recording performance, other than spot major limitations and identify future trends in this field.
Collapse
Affiliation(s)
- Michele Bianchi
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
| | - Anna De Salvo
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Sezione di FisiologiaUniversità di Ferraravia Fossato di Mortara 17Ferrara44121Italy
| | - Maria Asplund
- Division of Nursing and Medical TechnologyLuleå University of TechnologyLuleå971 87Sweden
- Department of Microsystems Engineering‐IMTEKUniversity of FreiburgFreiburg79110Germany
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburg79110Germany
| | - Stefano Carli
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Present address:
Department of Environmental and Prevention SciencesUniversità di FerraraFerrara44121Italy
| | - Michele Di Lauro
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
| | - Andreas Schulze‐Bonhage
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburg79110Germany
- Epilepsy CenterFaculty of MedicineUniversity of FreiburgFreiburg79110Germany
| | - Thomas Stieglitz
- Department of Microsystems Engineering‐IMTEKUniversity of FreiburgFreiburg79110Germany
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburg79110Germany
| | - Luciano Fadiga
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Sezione di FisiologiaUniversità di Ferraravia Fossato di Mortara 17Ferrara44121Italy
| | - Fabio Biscarini
- Center for Translational Neurophysiology of Speech and CommunicationFondazione Istituto Italiano di Tecnologiavia Fossato di Mortara 17Ferrara44121Italy
- Life Science DepartmentUniversità di Modena e Reggio EmiliaVia Campi 103Modena41125Italy
| |
Collapse
|
35
|
Ning S, Jorfi M, Patel SR, Kim DY, Tanzi RE. Neurotechnological Approaches to the Diagnosis and Treatment of Alzheimer’s Disease. Front Neurosci 2022; 16:854992. [PMID: 35401082 PMCID: PMC8989850 DOI: 10.3389/fnins.2022.854992] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in the elderly, clinically defined by progressive cognitive decline and pathologically, by brain atrophy, neuroinflammation, and accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles. Neurotechnological approaches, including optogenetics and deep brain stimulation, have exploded as new tools for not only the study of the brain but also for application in the treatment of neurological diseases. Here, we review the current state of AD therapeutics and recent advancements in both invasive and non-invasive neurotechnologies that can be used to ameliorate AD pathology, including neurostimulation via optogenetics, photobiomodulation, electrical stimulation, ultrasound stimulation, and magnetic neurostimulation, as well as nanotechnologies employing nanovectors, magnetic nanoparticles, and quantum dots. We also discuss the current challenges in developing these neurotechnological tools and the prospects for implementing them in the treatment of AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Shen Ning
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Graduate Program for Neuroscience, Boston University School of Medicine, Boston, MA, United States
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Mehdi Jorfi,
| | - Shaun R. Patel
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Rudolph E. Tanzi,
| |
Collapse
|
36
|
Zaidi SMT, Kocatürk S, Baykaş T, Kocatürk M. A behavioral paradigm for cortical control of a robotic actuator by freely moving rats in a one-dimensional two-target reaching task. J Neurosci Methods 2022; 373:109555. [PMID: 35271875 DOI: 10.1016/j.jneumeth.2022.109555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/01/2022] [Accepted: 03/04/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Controlling the trajectory of a neuroprosthesis to reach distant targets is a commonly used brain-machine interface (BMI) task in primates and has not been available for rodents yet. NEW METHOD Here, we describe a novel, fine-tuned behavioral paradigm and setup which enables this task for rats in one-dimensional space for reaching two distant targets depending on their limited cognitive and visual capabilities compared to those of primates. An online transform was used to convert the activity of a pair of primary motor cortex (M1) units into two robotic actions. The rats were shaped to adapt to the transform and direct the robotic actuator toward the selected target by modulating the activity of the M1 neurons. RESULTS All three rats involved in the study were capable of achieving randomly selected targets with at least 78% accuracy. A total of 9 out of 16 pairs of units examined were eligible for exceeding this success criterion. Two out of three rats were capable of reversal learning, where the mapping between the activity of the M1 units and the robotic actions were reversed. COMPARISON WITH EXISTING METHODS The present work is the first demonstration of trajectory-based control of a neuroprosthetic device by rodents to reach two distant targets using visual feedback. CONCLUSION The behavioral paradigm and setup introduced here can be used as a cost-effective platform for elucidating the information processing principles in the neural circuits related to neuroprosthetic control and for studying the performance of novel BMI technologies using freely moving rats.
Collapse
Affiliation(s)
| | - Samet Kocatürk
- Department of Biomedical Engineering, Istanbul Medipol University, Istanbul, Turkey
| | - Tunçer Baykaş
- Department of Computer Engineering, Istanbul Medipol University, Istanbul, Turkey
| | - Mehmet Kocatürk
- Department of Biomedical Engineering, Istanbul Medipol University, Istanbul, Turkey; Research Institute for Health Sciences and Technology, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
37
|
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: 0.7] [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
|
38
|
Gifford EK, Robbins EM, Jaquins-Gerstl A, Rerick MT, Nwachuku EL, Weber SG, Boutelle MG, Okonkwo DO, Puccio AM, Michael AC. Validation of Dexamethasone-Enhanced Continuous-Online Microdialysis for Monitoring Glucose for 10 Days after Brain Injury. ACS Chem Neurosci 2021; 12:3588-3597. [PMID: 34506125 DOI: 10.1021/acschemneuro.1c00231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) induces a pathophysiologic state that can be worsened by secondary injury. Monitoring brain metabolism with intracranial microdialysis can provide clinical insights to limit secondary injury in the days following TBI. Recent enhancements to microdialysis include the implementation of continuously operating electrochemical biosensors for monitoring the dialysate sample stream in real time and dexamethasone retrodialysis to mitigate the tissue response to probe insertion. Dexamethasone-enhanced continuous-online microdialysis (Dex-enhanced coMD) records long-lasting declines of glucose after controlled cortical impact in rats and TBI in patients. The present study employed retrodialysis and fluorescence microscopy to investigate the mechanism responsible for the decline of dialysate glucose after injury of the rat cortex. Findings confirm the long-term functionality of Dex-enhanced coMD for monitoring brain glucose after injury, demonstrate that intracranial glucose microdialysis is coupled to glucose utilization in the tissues surrounding the probes, and validate the conclusion that aberrant glucose utilization drives the postinjury glucose decline.
Collapse
Affiliation(s)
- Emily K. Gifford
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Elaine M. Robbins
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Andrea Jaquins-Gerstl
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Michael T. Rerick
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Enyinna L. Nwachuku
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Stephen G. Weber
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Martyn G. Boutelle
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - David O. Okonkwo
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Ava M. Puccio
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, United States
| | - Adrian C. Michael
- Department of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
39
|
Intracortical Microelectrode Array Unit Yield under Chronic Conditions: A Comparative Evaluation. MICROMACHINES 2021; 12:mi12080972. [PMID: 34442594 PMCID: PMC8400387 DOI: 10.3390/mi12080972] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 01/01/2023]
Abstract
While microelectrode arrays (MEAs) offer the promise of elucidating functional neural circuitry and serve as the basis for a cortical neuroprosthesis, the challenge of designing and demonstrating chronically reliable technology remains. Numerous studies report “chronic” data but the actual time spans and performance measures corresponding to the experimental work vary. In this study, we reviewed the experimental durations that constitute chronic studies across a range of MEA types and animal species to gain an understanding of the widespread variability in reported study duration. For rodents, which are the most commonly used animal model in chronic studies, we examined active electrode yield (AEY) for different array types as a means to contextualize the study duration variance, as well as investigate and interpret the performance of custom devices in comparison to conventional MEAs. We observed wide-spread variance within species for the chronic implantation period and an AEY that decayed linearly in rodent models that implanted commercially-available devices. These observations provide a benchmark for comparing the performance of new technologies and highlight the need for consistency in chronic MEA studies. Additionally, to fully derive performance under chronic conditions, the duration of abiotic failure modes, biological processes induced by indwelling probes, and intended application of the device are key determinants.
Collapse
|
40
|
Wang D, Tan J, Zhu H, Mei Y, Liu X. Biomedical Implants with Charge-Transfer Monitoring and Regulating Abilities. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004393. [PMID: 34166584 PMCID: PMC8373130 DOI: 10.1002/advs.202004393] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/12/2021] [Indexed: 05/06/2023]
Abstract
Transmembrane charge (ion/electron) transfer is essential for maintaining cellular homeostasis and is involved in many biological processes, from protein synthesis to embryonic development in organisms. Designing implant devices that can detect or regulate cellular transmembrane charge transfer is expected to sense and modulate the behaviors of host cells and tissues. Thus, charge transfer can be regarded as a bridge connecting living systems and human-made implantable devices. This review describes the mode and mechanism of charge transfer between organisms and nonliving materials, and summarizes the strategies to endow implants with charge-transfer regulating or monitoring abilities. Furthermore, three major charge-transfer controlling systems, including wired, self-activated, and stimuli-responsive biomedical implants, as well as the design principles and pivotal materials are systematically elaborated. The clinical challenges and the prospects for future development of these implant devices are also discussed.
Collapse
Affiliation(s)
- Donghui Wang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- School of Materials Science and EngineeringHebei University of TechnologyTianjin300130China
| | - Ji Tan
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
| | - Hongqin Zhu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Yongfeng Mei
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institutes of CeramicsChinese Academy of SciencesShanghai200050China
- School of Chemistry and Materials ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhou310024China
| |
Collapse
|
41
|
Woeppel KM, Cui XT. Nanoparticle and Biomolecule Surface Modification Synergistically Increases Neural Electrode Recording Yield and Minimizes Inflammatory Host Response. Adv Healthc Mater 2021; 10:e2002150. [PMID: 34190425 DOI: 10.1002/adhm.202002150] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/08/2021] [Indexed: 11/08/2022]
Abstract
Due to their ability to interface with neural tissues, neural electrodes are the key tool used for neurophysiological studies, electrochemical detection, brain computer interfacing, and countless neuromodulation therapies and diagnostic procedures. However, the long-term applications of neural electrodes are limited by the inflammatory host tissue response, decreasing detectable electrical signals, and insulating the device from the native environment. Surface modification methods are proposed to limit these detrimental responses but each has their own limitations. Here, a combinatorial approach is presented toward creating a stable interface between the electrode and host tissues. First, a thiolated nanoparticle (TNP) coating is utilized to increase the surface area and roughness. Next, the neural adhesion molecule L1 is immobilized to the nanoparticle modified substrate. In vitro, the combined nanotopographical and bioactive modifications (TNP+L1) elevate the bioactivity of L1, which is maintained for 28 d. In vivo, TNP+L1 modification improves the recording performance of the neural electrode arrays compared to TNP or L1 modification alone. Postmortem histology reveals greater neural cell density around the TNP+L1 coating while eliminating any inflammatory microglial encapsulation after 4 weeks. These results demonstrate that nanotopographical and bioactive modifications synergistically produce a seamless neural tissue interface for chronic neural implants.
Collapse
Affiliation(s)
- Kevin M. Woeppel
- Department of Bioengineering University of Pittsburgh Pittsburgh PA 15260 USA
- Center for the Neural basis of Cognition Pittsburgh PA 15260 USA
| | - Xinyan Tracy Cui
- Department of Bioengineering University of Pittsburgh Pittsburgh PA 15260 USA
- Center for the Neural basis of Cognition Pittsburgh PA 15260 USA
- McGowan Institute for Regenerative Medicine Pittsburgh PA 15260 USA
| |
Collapse
|
42
|
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: 17] [Impact Index Per Article: 4.3] [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
|
43
|
Bouadi O, Tay TL. More Than Cell Markers: Understanding Heterogeneous Glial Responses to Implantable Neural Devices. Front Cell Neurosci 2021; 15:658992. [PMID: 33912015 PMCID: PMC8071943 DOI: 10.3389/fncel.2021.658992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/17/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ouzéna Bouadi
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Faculty of Life Sciences, University of Strasbourg, Strasbourg, France
| | - Tuan Leng Tay
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,BrainLinks-BrainTools Centre, University of Freiburg, Freiburg, Germany.,Freiburg Institute of Advanced Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Costanza A, Radomska M, Bondolfi G, Zenga F, Amerio A, Aguglia A, Serafini G, Amore M, Berardelli I, Pompili M, Nguyen KD. Suicidality Associated With Deep Brain Stimulation in Extrapyramidal Diseases: A Critical Review and Hypotheses on Neuroanatomical and Neuroimmune Mechanisms. Front Integr Neurosci 2021; 15:632249. [PMID: 33897384 PMCID: PMC8060445 DOI: 10.3389/fnint.2021.632249] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 03/15/2021] [Indexed: 12/13/2022] Open
Abstract
Deep brain stimulation (DBS) is a very well-established and effective treatment for patients with extrapyramidal diseases. Despite its generally favorable clinical efficacy, some undesirable outcomes associated with DBS have been reported. Among such complications are incidences of suicidal ideation (SI) and behavior (SB) in patients undergoing this neurosurgical procedure. However, causal associations between DBS and increased suicide risk are not demonstrated and they constitute a debated issue. In light of these observations, the main objective of this work is to provide a comprehensive and unbiased overview of the literature on suicide risk in patients who received subthalamic nucleus (STN) and internal part of globus pallidum (GPi) DBS treatment. Additionally, putative mechanisms that might be involved in the development of SI and SB in these patients as well as caveats associated with these hypotheses are introduced. Finally, we briefly propose some clinical implications, including therapeutic strategies addressing these potential disease mechanisms. While a mechanistic connection between DBS and suicidality remains a controversial topic that requires further investigation, it is of critical importance to consider suicide risk as an integral component of candidate selection and post-operative care in DBS.
Collapse
Affiliation(s)
- Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland.,Department of Psychiatry, ASO Santi Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Italy
| | - Michalina Radomska
- Faculty of Psychology, University of Geneva (UNIGE), Geneva, Switzerland
| | - Guido Bondolfi
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), Geneva, Switzerland.,Department of Psychiatry, Service of Liaison Psychiatry and Crisis Intervention (SPLIC), Geneva University Hospitals (HUG), Geneva, Switzerland
| | - Francesco Zenga
- Department of Neurosurgery, University and City of Health and Science Hospital, Turin, Italy
| | - Andrea Amerio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Mood Disorders Program, Tufts Medical Center, Boston, MA, United States
| | - Andrea Aguglia
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianluca Serafini
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, Genova, Italy.,Department of Psychiatry, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Isabella Berardelli
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Khoa D Nguyen
- Department of Microbiology and Immunology, Stanford University, Palo Alto, CA, United States.,Tranquis Therapeutics, Palo Alto, CA, United States.,Hong Kong University of Science and Technology, Hong Kong, China
| |
Collapse
|
45
|
Wu P, Liang Y, Sun G. Engineering immune-responsive biomaterials for skin regeneration. BIOMATERIALS TRANSLATIONAL 2021; 2:61-71. [PMID: 35837252 PMCID: PMC9255827 DOI: 10.3877/cma.j.issn.2096-112x.2021.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/10/2020] [Accepted: 12/29/2020] [Indexed: 01/17/2023]
Abstract
The progress of biomaterials and tissue engineering has led to significant advances in wound healing, but the clinical therapy to regenerate perfect skin remains a great challenge. The implantation of biomaterial scaffolds to heal wounds inevitably leads to a host immune response. Many recent studies revealed that the immune system plays a significant role in both the healing process and the outcome. Immunomodulation or immuno-engineering has thus become a promising approach to develop pro-regenerative scaffolds for perfect skin regeneration. In this paper, we will review recent advancements in immunomodulating biomaterials in the field of skin repair and regeneration, and discuss strategies to modulate the immune response by tailoring the chemical, physical and biological properties of the biomaterials. Understanding the important role of immune responses and manipulating the inherent properties of biomaterials to regulate the immune reaction are approaches to overcome the current bottleneck of skin repair and regeneration.
Collapse
Affiliation(s)
- Pingli Wu
- College of Chemistry and Environmental Science, Hebei University, Baoding, Hebei Province, China
| | - Yangyang Liang
- College of Chemistry and Environmental Science, Hebei University, Baoding, Hebei Province, China
| | - Guoming Sun
- Affiliated Hospital of Hebei University, College of Chemistry and Environmental Science, Hebei University, Baoding, Hebei Province, China,Corresponding author: Guoming Sun,
| |
Collapse
|
46
|
Dubaniewicz M, Eles JR, Lam S, Song S, Cambi F, Sun D, Wellman SM, Kozai TDY. Inhibition of Na +/H +exchanger modulates microglial activation and scar formation following microelectrode implantation. J Neural Eng 2021; 18. [PMID: 33621208 DOI: 10.1088/1741-2552/abe8f1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/23/2021] [Indexed: 11/12/2022]
Abstract
Objective.Intracortical microelectrodes are an important tool for neuroscience research and have great potential for clinical use. However, the use of microelectrode arrays to treat neurological disorders and control prosthetics is limited by biological challenges such as glial scarring, which can impair chronic recording performance. Microglia activation is an early and prominent contributor to glial scarring. After insertion of an intracortical microelectrode, nearby microglia transition into a state of activation, migrate, and encapsulate the device. Na+/H+exchanger isoform-1 (NHE-1) is involved in various microglial functions, including their polarity and motility, and has been implicated in pro-inflammatory responses to tissue injury. HOE-642 (cariporide) is an inhibitor of NHE-1 and has been shown to depress microglial activation and inflammatory response in brain injury models.Approach.In this study, the effects of HOE-642 treatment on microglial interactions to intracortical microelectrodes was evaluated using two-photon microscopyin vivo.Main results.The rate at which microglia processes and soma migrate in response to electrode implantation was unaffected by HOE-642 administration. However, HOE-642 administration effectively reduced the radius of microglia activation at 72 h post-implantation from 222.2µm to 177.9µm. Furthermore, treatment with HOE-642 significantly reduced microglial encapsulation of implanted devices at 5 h post-insertion from 50.7 ± 6.0% to 8.9 ± 6.1%, which suggests an NHE-1-specific mechanism mediating microglia reactivity and gliosis during implantation injury.Significance.This study implicates NHE-1 as a potential target of interest in microglial reactivity and HOE-642 as a potential treatment to attenuate the glial response and scar formation around implanted intracortical microelectrodes.
Collapse
Affiliation(s)
- Mitchell Dubaniewicz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Stephanie Lam
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, United States of America.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Dandan Sun
- Veterans Administration Pittsburgh, Pittsburgh, PA, United States of America.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.,NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
47
|
Elyahoodayan S, Jiang W, Lee CD, Shao X, Weiland G, Whalen JJ, Petrossians A, Song D. Stimulation and Recording of the Hippocampus Using the Same Pt-Ir Coated Microelectrodes. Front Neurosci 2021; 15:616063. [PMID: 33716647 PMCID: PMC7943859 DOI: 10.3389/fnins.2021.616063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/28/2021] [Indexed: 01/11/2023] Open
Abstract
Same-electrode stimulation and recording with high spatial resolution, signal quality, and power efficiency is highly desirable in neuroscience and neural engineering. High spatial resolution and signal-to-noise ratio is necessary for obtaining unitary activities and delivering focal stimulations. Power efficiency is critical for battery-operated implantable neural interfaces. This study demonstrates the capability of recording single units as well as evoked potentials in response to a wide range of electrochemically safe stimulation pulses through high-resolution microelectrodes coated with co-deposition of Pt-Ir. It also compares signal-to-noise ratio, single unit activity, and power efficiencies between Pt-Ir coated and uncoated microelectrodes. To enable stimulation and recording with the same microelectrodes, microelectrode arrays were treated with electrodeposited platinum-iridium coating (EPIC) and tested in the CA1 cell body layer of rat hippocampi. The electrodes' ability to (1) inject a large range of electrochemically reversable stimulation pulses to the tissue, and (2) record evoked potentials and single unit activities were quantitively assessed over an acute time period. Compared to uncoated electrodes, EPIC electrodes recorded signals with higher signal-to-noise ratios (coated: 9.77 ± 1.95 dB; uncoated: 1.95 ± 0.40 dB) and generated lower voltages (coated: 100 mV; uncoated: 650 mV) for a given stimulus (5 μA). The improved performance corresponded to lower energy consumptions and electrochemically safe stimulation above 5 μA (>0.38 mC/cm2), which enabled elicitation of field excitatory post synaptic potentials and population spikes. Spontaneous single unit activities were also modulated by varying stimulation intensities and monitored through the same electrodes. This work represents an example of stimulation and recording single unit activities from the same microelectrode, which provides a powerful tool for monitoring and manipulating neural circuits at the single neuron level.
Collapse
Affiliation(s)
- Sahar Elyahoodayan
- Department of Biomedical Engineering, Center for Neural Engineering, University of Southern California, Los Angeles, CA, United States
| | - Wenxuan Jiang
- Department of Biomedical Engineering, Center for Neural Engineering, University of Southern California, Los Angeles, CA, United States
| | | | - Xiecheng Shao
- Department of Biomedical Engineering, Center for Neural Engineering, University of Southern California, Los Angeles, CA, United States
| | | | | | | | - Dong Song
- Department of Biomedical Engineering, Center for Neural Engineering, University of Southern California, Los Angeles, CA, United States
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
48
|
Zaidi SMT, Kocatürk S, Baykaş T, Kocatürk M. A behavioral paradigm for cortical control of a robotic actuator by freely moving rats in a one-dimensional two-target reaching task.. [DOI: 10.1101/2021.01.24.427325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
AbstractControlling the trajectory of a neuroprosthetic device to reach multiple targets is a commonly used brain-machine interface (BMI) task in primates and has not been available for rodents yet. Here, we describe a novel experimental paradigm which enables this task for rats in one-dimensional space for reaching two distant targets depending on their limited cognitive and visual capabilities compared to primates. An online transform was used to convert the activity of a pair of primary motor cortex (M1) units into two robotic actions. The rats were shaped to adapt to the transform and direct the robotic actuator toward the selected target by modulating the activity of the M1 neurons. All three rats involved in the study were capable of achieving randomly selected targets with at least 78% accuracy. A total of 9 out of 16 pairs of units examined were eligible for exceeding this success criterion. Two out of three rats were capable of reversal learning, where the mapping between the activity of the unit pairs and the robotic actions were reversed. The present work is the first demonstration of trajectory-based control of a neuroprosthetic device by rodents to reach two distant targets using visual feedback. The paradigm introduced here may be used as a cost-effective platform for elucidating the information processing principles in the neural circuits related to neuroprosthetic control and for studying the performance of novel BMI technologies using freely moving rats.
Collapse
|
49
|
Page DM, George JA, Wendelken SM, Davis TS, Kluger DT, Hutchinson DT, Clark GA. Discriminability of multiple cutaneous and proprioceptive hand percepts evoked by intraneural stimulation with Utah slanted electrode arrays in human amputees. J Neuroeng Rehabil 2021; 18:12. [PMID: 33478534 PMCID: PMC7819250 DOI: 10.1186/s12984-021-00808-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Electrical stimulation of residual afferent nerve fibers can evoke sensations from a missing limb after amputation, and bionic arms endowed with artificial sensory feedback have been shown to confer functional and psychological benefits. Here we explore the extent to which artificial sensations can be discriminated based on location, quality, and intensity. METHODS We implanted Utah Slanted Electrode Arrays (USEAs) in the arm nerves of three transradial amputees and delivered electrical stimulation via different electrodes and frequencies to produce sensations on the missing hand with various locations, qualities, and intensities. Participants performed blind discrimination trials to discriminate among these artificial sensations. RESULTS Participants successfully discriminated cutaneous and proprioceptive sensations ranging in location, quality and intensity. Performance was significantly greater than chance for all discrimination tasks, including discrimination among up to ten different cutaneous location-intensity combinations (15/30 successes, p < 0.0001) and seven different proprioceptive location-intensity combinations (21/40 successes, p < 0.0001). Variations in the site of stimulation within the nerve, via electrode selection, enabled discrimination among up to five locations and qualities (35/35 successes, p < 0.0001). Variations in the stimulation frequency enabled discrimination among four different intensities at the same location (13/20 successes, p < 0.0005). One participant also discriminated among individual stimulation of two different USEA electrodes, simultaneous stimulation on both electrodes, and interleaved stimulation on both electrodes (20/24 successes, p < 0.0001). CONCLUSION Electrode location, stimulation frequency, and stimulation pattern can be modulated to evoke functionally discriminable sensations with a range of locations, qualities, and intensities. This rich source of artificial sensory feedback may enhance functional performance and embodiment of bionic arms endowed with a sense of touch.
Collapse
Affiliation(s)
| | - Jacob A George
- Division of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Suzanne M Wendelken
- Department of Anesthesiology, Maine Medical Center, Portland, ME, 04102, USA
| | - Tyler S Davis
- Department of Neurosurgery, University of Utah, Salt Lake City, UT, 84112, USA
| | | | | | - Gregory A Clark
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| |
Collapse
|
50
|
Maiolo L, Guarino V, Saracino E, Convertino A, Melucci M, Muccini M, Ambrosio L, Zamboni R, Benfenati V. Glial Interfaces: Advanced Materials and Devices to Uncover the Role of Astroglial Cells in Brain Function and Dysfunction. Adv Healthc Mater 2021; 10:e2001268. [PMID: 33103375 DOI: 10.1002/adhm.202001268] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/06/2020] [Indexed: 12/13/2022]
Abstract
Research over the past four decades has highlighted the importance of certain brain cells, called glial cells, and has moved the neurocentric vision of structure, function, and pathology of the nervous system toward a more holistic perspective. In this view, the demand for technologies that are able to target and both selectively monitor and control glial cells is emerging as a challenge across neuroscience, engineering, chemistry, and material science. Frequently neglected or marginally considered as a barrier to be overcome between neural implants and neuronal targets, glial cells, and in particular astrocytes, are increasingly considered as active players in determining the outcomes of device implantation. This review provides a concise overview not only of the previously established but also of the emerging physiological and pathological roles of astrocytes. It also critically discusses the most recent advances in biomaterial interfaces and devices that interact with glial cells and thus have enabled scientists to reach unprecedented insights into the role of astroglial cells in brain function and dysfunction. This work proposes glial interfaces and glial engineering as multidisciplinary fields that have the potential to enable significant advancement of knowledge surrounding cognitive function and acute and chronic neuropathologies.
Collapse
Affiliation(s)
- Luca Maiolo
- Consiglio Nazionale delle Ricerche Istituto per la Microelettronica e i Microsistemi Via del Fosso del Cavaliere n.100 Roma 00133 Italy
| | - Vincenzo Guarino
- Consiglio Nazionale delle Ricerche Istituto per i Polimeri Compositi e Biomateriali Viale J.F. Kennedy 54, Mostra d'Oltremare, Pad 20 Napoli 80125 Italy
| | - Emanuela Saracino
- Consiglio Nazionale delle Ricerche Istituto per la Sintesi Organica e la Fotoreattività via P. Gobetti 101 Bologna 40129 Italy
| | - Annalisa Convertino
- Consiglio Nazionale delle Ricerche Istituto per la Microelettronica e i Microsistemi Via del Fosso del Cavaliere n.100 Roma 00133 Italy
| | - Manuela Melucci
- Consiglio Nazionale delle Ricerche Istituto per la Sintesi Organica e la Fotoreattività via P. Gobetti 101 Bologna 40129 Italy
| | - Michele Muccini
- Consiglio Nazionale delle Ricerche Istituto per la Studio dei Materiali Nanostrutturati via P. Gobetti 101 Bologna 40129 Italy
| | - Luigi Ambrosio
- Consiglio Nazionale delle Ricerche Istituto per i Polimeri Compositi e Biomateriali Viale J.F. Kennedy 54, Mostra d'Oltremare, Pad 20 Napoli 80125 Italy
| | - Roberto Zamboni
- Consiglio Nazionale delle Ricerche Istituto per la Sintesi Organica e la Fotoreattività via P. Gobetti 101 Bologna 40129 Italy
| | - Valentina Benfenati
- Consiglio Nazionale delle Ricerche Istituto per la Sintesi Organica e la Fotoreattività via P. Gobetti 101 Bologna 40129 Italy
| |
Collapse
|