1
|
Kwon YW, Kim E, Koh CS, Park YG, Hong YM, Lee S, Lee J, Kim TJ, Mun W, Min SH, Kim S, Lim JA, Jung HH, Park JU. Implantable Soft Neural Electrodes of Liquid Metals for Deep Brain Stimulation. ACS NANO 2025; 19:7337-7349. [PMID: 39957079 DOI: 10.1021/acsnano.4c18030] [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: 02/18/2025]
Abstract
Stimulating large volumes of neural networks using macroelectrodes can modulate disorder-associated brain circuits effectively. However, conventional solid-metal electrodes often cause unwanted brain damage due to their high mechanical stiffness. In contrast, low-modulus liquid metals provide tissue-like stiffness while maintaining macroscale electrode dimensions. Here, we present implantable soft macroelectrodes made from biocompatible liquid metals for brain stimulation. These probes can be easily fabricated by simply filling polymeric tubes with a liquid metal, offering a straightforward method for creating brain stimulation devices. They can be customized in various lengths and diameters and also serve as recording microelectrodes. The electrode tips are enhanced with platinum nanoclusters, resulting in low impedance and effective charge injection while preventing liquid metal leakage into brain tissue. In vivo experiments in neuropathic pain rat models demonstrate the stability and effectiveness of these probes for simultaneous neural stimulation and recording, demonstrating their potential for pain alleviation and behavioral control.
Collapse
Affiliation(s)
- Yong Won Kwon
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Enji Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Chin Su Koh
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young-Geun Park
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Yeon-Mi Hong
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Sanghoon Lee
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Jakyoung Lee
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Tae Jun Kim
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Wonki Mun
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Hyun Min
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sumin Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Jung Ah Lim
- Yonsei-KIST Convergence Research Institute, Seoul 03722, Republic of Korea
- Soft Hybrid Materials Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Nanoscience and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Hyun Ho Jung
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, South Korea
- Yonsei-KIST Convergence Research Institute, Seoul 03722, Republic of Korea
| |
Collapse
|
2
|
Kim J, Lee JS, Noh S, Seo E, Lee J, Kim T, Cho SW, Kim G, Kim SS, Park J. Cellular level cryo-neuromodulation using rapid and localized cooling device combined with microelectrode array. Biosens Bioelectron 2025; 277:117257. [PMID: 39978154 DOI: 10.1016/j.bios.2025.117257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Cryotherapy, a rapid and effective medical treatment utilizing low temperatures, has not been widely adopted in clinical practice due to a limited understanding of its mechanisms and efficacy. This challenge stems from the absence of methods for fast, precise, and localized spatiotemporal temperature control, as well as the lack of reliable real-time quantitative techniques for measuring and analyzing the effects of cooling. To address these limitations, this study introduces a cryo-neuromodulation platform that integrates a high-speed precision cooling device with a microelectrode array (MEA) system. This platform enables the investigation of cellular-level cryo-modulation of neuronal activity and its effects on surrounding cells, providing a novel framework for advancing research in cryotherapy and neuromodulation. Experiments show that neurons recovered fully within 1 min of cooling with a fast-cooling rate (-20 °C/s at cooling) and that silenced neurons can influence distant cells via a well-organized network. Extended cooling durations (e.g., 10 min) resulted in altered neuronal dynamics, including delayed recovery and reduced burst activity, highlighting the importance of precise control over cooling parameters. This device offers reversible neural control, with potential applications in both research and clinical settings, such as anesthesia, pain management and treatment of neurological disorders like neocortical seizures.
Collapse
Affiliation(s)
- Jaehyun Kim
- Department of Mechanical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Jong Seung Lee
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Soyeon Noh
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Eonyang-eup, Ulju-gun, Ulsan, 44919, Republic of Korea
| | - Eunseok Seo
- Department of Mechanical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Jungchul Lee
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Taesung Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Eonyang-eup, Ulju-gun, Ulsan, 44919, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Gunho Kim
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology (UNIST), 50 UNIST-gil, Eonyang-eup, Ulju-gun, Ulsan, 44919, Republic of Korea.
| | - Sung Soo Kim
- Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA.
| | - Jungyul Park
- Department of Mechanical Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
3
|
Zou H, Jiang Z, Bian E, Zhou J, Li S, Yang Y, Guo H, Liu Y, Wu W, Deng C. Nanoencapsulated Optical Fiber-Based PEC Microelectrode: Highly Sensitive and Specific Detection of NT-proBNP and Its Implantable Performance. Anal Chem 2025; 97:2724-2735. [PMID: 39868898 DOI: 10.1021/acs.analchem.4c04757] [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: 01/28/2025]
Abstract
Microelectrodes offer exceptional sensitivity, rapid response, and versatility, making them ideal for real-time detection and monitoring applications. Photoelectrochemical (PEC) sensors have shown great value in many fields due to their high sensitivity, fast response, and ease of operation. Nevertheless, conventional PEC sensing relies on cumbersome external light sources and bulky electrodes, hindering its miniaturization and implantation, thereby limiting its application in real-time disease monitoring. To overcome these limitations, we developed a nanoencapsulated optical fiber (OF)-based PEC microelectrode. The microelectrode features TiO2/CdS nanocrystals and bis (2,2'-bipyridine) (10-methylphenanthroline [3,2-a:2'3'-c] pyridine ruthenium(II) dichloride ([Ru(bpy)2dppz]2+) @dsDNA/Au@epigallocatechin gallate nanoparticle (EGCG NP) layers. And its application for the detection of N-terminal pro-brain natriuretic peptide (NT-proBNP) as a biomarker of cardiovascular diseases was explored. An extensive linear range of 1-5000 pg mL-1 combined with a low detection limit of 0.36 pg mL-1 was achieved. This range covers not only the recommended threshold for excluding cardiovascular diseases in the clinical diagnosis of individuals across all age groups but also the prognostic target value. The sensor exhibited excellent selectivity and stability and notable labeling recovery capability in serum tests. Critically, the sensor successfully discriminated the alterations in NT-proBNP secretion levels within human smooth muscle cells, comparing pre- and poststimulation by platelet-derived growth factor-BB. Even more significantly, the skin puncture experiment conducted in mice demonstrated the remarkable implantability and biological compatibility of the OF-PEC microelectrode. This addresses critical challenges commonly faced by microelectrodes when used as implanted devices, such as minimizing invasive trauma, mitigating inflammation, and preventing biofouling, thereby firmly establishing their suitability for the development of advanced implantable sensing devices. Therefore, the present OF microelectrode PEC biosensor is not only cost-effective, easy to operate, and miniaturized but also holds significant potential for enabling more precise, more minimally invasive, and continuous monitoring of biological markers without causing inflammation. This capability is crucial for early disease detection, tracking disease progression, and facilitating personalized treatment strategies, which expands the practical application of PEC sensors.
Collapse
Affiliation(s)
- HuiYu Zou
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Zhizhong Jiang
- School of Electronic Science and Engineering, Hunan Institute of Information Technology, Changsha, Hunan 410151, China
| | - ErKang Bian
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Jingjing Zhou
- Beijing Key Laboratory of Maternal-Fetal Medicine and Fetal Heart Disease & Echocardiography Department, Beijing Anzhen Hospital, Capital Medical University, Beijing 100069, China
| | - Shengqing Li
- School of Electronic Science and Engineering, Hunan Institute of Information Technology, Changsha, Hunan 410151, China
| | - Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai HospitalRINGGOLD, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 102308, China
| | - HuiPing Guo
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| | - Yuancheng Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410078, China
| | - WuMing Wu
- School of Electronic Science and Engineering, Hunan Institute of Information Technology, Changsha, Hunan 410151, China
| | - ChunYan Deng
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
4
|
Xu Q, Wang L, Xi Y, Ruan T, Cao J, Xu M, Zheng K, Du Z, Wei N, Wang X, Yang B, Liu J. An Efficient MEMS Microelectrode Array with Reliable Interelectrode Insulation Processes for In Vivo Neural Recording. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2407950. [PMID: 39935127 DOI: 10.1002/smll.202407950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/18/2025] [Indexed: 02/13/2025]
Abstract
Microelectrode arrays, particularly Utah arrays, offer irreplaceable advantages in clinical applications and play a crucial role in advancing brain-computer interactions. However, the glass-fused monolithic structure of Utah arrays limits functional expansion, and the glass insulation process is complex, costly, and time-intensive. This paper presents a microelectrode array with a simple and time-saving fabrication process, utilizing low-resistance silicon and borosilicate glass wafers as electrodes and insulation substrates, respectively. The utilization of the anodic bonding process improves production efficiency and enhances process compatibility. A one-step static wet etching process is used to form microneedle morphology to further simplify the fabrication process. Sputtered iridium oxide, as the electrode interface material, significantly reduces electrochemical impedance, and cellular experiments have confirmed its non-cytotoxicity. Moreover, the implantation into the primary visual cortex of mice has demonstrated the ability of the electrode to record in vivo electrical signals within 15 days. Movement trajectory experiments demonstrate that the mice exhibit good behavior activities following electrode implantation. The bonded microelectrode array (BMEA) presented in this work provides a universal and effective tool for neural recording, with prospective applications in multi-physiological monitoring and microelectromechanical system integration.
Collapse
Affiliation(s)
- Qingda Xu
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Longchun Wang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ye Xi
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tao Ruan
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiawei Cao
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Mengfei Xu
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Kunyu Zheng
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhiyuan Du
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ning Wei
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaolin Wang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bin Yang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jingquan Liu
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
5
|
Kim DH, Choi SJ, Liu Z, Yang F, Wang H, George D, Gracias D. 3D Spatiotemporal Activation Mapping of Cardiac Organoids Using Conformal Shell Microelectrode Arrays (MEAs). RESEARCH SQUARE 2025:rs.3.rs-5939602. [PMID: 39975924 PMCID: PMC11838751 DOI: 10.21203/rs.3.rs-5939602/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cardiac organoids have emerged as transformative models for investigating cardiogenesis and cardiac diseases. While traditional 2D microelectrode arrays (MEAs) have been used to assess the functionality of cardiac organoids, they are limited to electrophysiological measurements from a single plane and do not capture the 3D propagation of electrical signals. Here, we present a programmable, shape-adaptive shell MEA designed to map the electrical activity across the entire surface of cardiac organoids. These shell MEAs are fabricated on-chip, with tunable dimensions and electrode layout, enabling precise encapsulation of spherical organoids. Using shell MEAs, we generated 3D isochrone maps with conduction velocity vectors, revealing the speed and trajectory of electrical signal propagation in spontaneously beating cardiac organoids. The optical transparency of the shell MEAs allowed for simultaneous calcium imaging, validating the electrophysiological propagation pattern. To demonstrate their utility in cardiotoxicity screening, we monitored the electrophysiological changes of organoids treated with isoproterenol and E-4031 over nine days. We anticipate that shell MEAs, combined with spatiotemporal mapping, can significantly advance the development of spatially organized cardiac organoids, structural disease models, and high-throughput drug screening platforms.
Collapse
|
6
|
Schellberg BG, Koppes RA, Koppes AN. Recent Advances in Integrated Organ-Chip Sensing Toward Robust and User-Friendly Systems. J Biomed Mater Res A 2025; 113:e37876. [PMID: 39893559 DOI: 10.1002/jbm.a.37876] [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: 10/18/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025]
Abstract
Organs-on-a-chip (OOC) are an emergent technology that bridge the gap between current in vitro and in vivo models used to inform drug discovery and investigate disease pathophysiology. These systems offer improved bio-relevance and controlled complexity through the integration of physical and/or chemical stimuli matched to physiologically relevant conditions. Although significant advancements have been made toward recreating organ-specific physiology on chip, the methods available to study structure and function of the cell microenvironment are still limited. Established analysis approaches, including fluorescence microscopy, rely on laborious offline workflows that yield limited time-point data. As the OOC field continues to evolve, there is a unique opportunity to engineer improved characterization methods into organ-chip devices. This review provides an overview of current integrated sensing approaches that address current limitations and enable real-time readout of relevant physiological parameters in OOC.
Collapse
Affiliation(s)
- Bryan G Schellberg
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
7
|
Alghannam F, Alayed M, Alfihed S, Sakr MA, Almutairi D, Alshamrani N, Al Fayez N. Recent Progress in PDMS-Based Microfluidics Toward Integrated Organ-on-a-Chip Biosensors and Personalized Medicine. BIOSENSORS 2025; 15:76. [PMID: 39996978 DOI: 10.3390/bios15020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 02/26/2025]
Abstract
The organ-on-a-chip (OoC) technology holds significant promise for biosensors and personalized medicine by enabling the creation of miniature, patient-specific models of human organs. This review studies the recent advancements in the application of polydimethylsiloxane (PDMS) microfluidics for OoC purposes. It underscores the main fabrication technologies of PDMS microfluidic systems, such as photolithography, injection molding, hot embossing, and 3D printing. The review also highlights the crucial role of integrated biosensors within OoC platforms. These electrochemical, electrical, and optical sensors, integrated within the microfluidic environment, provide valuable insights into cellular behavior and drug response. Furthermore, the review explores the exciting potential of PDMS-based OoC technology for personalized medicine. OoC devices can forecast drug effectiveness and tailor therapeutic strategies for patients by incorporating patient-derived cells and replicating individual physiological variations, helping the healing process and accelerating recovery. This personalized approach can revolutionize healthcare by offering more precise and efficient treatment options. Understanding OoC fabrication and its applications in biosensors and personalized medicine can play a pivotal role in future implementations of multifunctional OoC biosensors.
Collapse
Affiliation(s)
- Fahad Alghannam
- Microelectronics and Semiconductors Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Mrwan Alayed
- Microelectronics and Semiconductors Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Salman Alfihed
- Microelectronics and Semiconductors Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Mahmoud A Sakr
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Dhaifallah Almutairi
- Microelectronics and Semiconductors Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Naif Alshamrani
- Microelectronics and Semiconductors Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Nojoud Al Fayez
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| |
Collapse
|
8
|
Costa BNL, Marote A, Barbosa C, Campos J, Salgado AJ, Nieder JB. Smart Polymeric 3D Microscaffolds Hosting Spheroids for Neuronal Research via Quantum Metrology. Adv Healthc Mater 2025:e2403875. [PMID: 39815162 DOI: 10.1002/adhm.202403875] [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: 10/07/2024] [Revised: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Toward the aim of reducing animal testing, innovative in vitro models are required. Here, this study proposes a novel smart polymeric microscaffold to establish an advanced 3D model of dopaminergic neurons. These scaffolds are fabricated with Ormocomp via Two-Photon Polymerization. The scaffolds are further enhanced by functionalization with fluorescent nanodiamonds (FNDs), which can serve as quantum nanosensors for both magnetic and temperature sensing. The material biocompatibility is tested using two different cell lines, SH-SY5Y and A431, with cell viability over 98%. A total of 69% of the FNDs are grafted on the structure compared to those that remained on the glass surface. Cells are tested with the scaffolds in several microenvironments, and the final assembly required for 3D quantum metrology experiments achieved 91% biocompatibility. Subsequently, embryoid bodies containing dopaminergic neurons, the cell type affected by Parkinson's disease (PD), are integrated with FND-functionalized scaffolds. This 3D model is successfully established, demonstrated by strong interactions between dopaminergic neurons and the scaffold, with the directional growth of neurites along the 3D scaffold. Ultimately, this study have developed a 3D platform that enables the readout of signaling in a model that holds great potential for future PD research.
Collapse
Affiliation(s)
- Beatriz N L Costa
- INL - International Iberian Nanotechnology Laboratory, Ultrafast Bio- and Nanophotonics group, Av. Mestre José Veiga s/n, Braga, 4715-330, Portugal
- Escola de Enxeñaría de Minas e Enerxía, University of Vigo, Vigo, Pontevedra, 36310, Spain
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Catarina Barbosa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, 4710-057, Portugal
- ICVS/3B's - Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jana B Nieder
- INL - International Iberian Nanotechnology Laboratory, Ultrafast Bio- and Nanophotonics group, Av. Mestre José Veiga s/n, Braga, 4715-330, Portugal
| |
Collapse
|
9
|
Gou X, Xing Z, Zhang Z, Jin R, Xu Q, Sojic N, Zhu JJ, Ma C. Designable Electrochemiluminescence Patterning for Renewable and Enhanced Bioimaging. Angew Chem Int Ed Engl 2025; 64:e202410825. [PMID: 39536295 DOI: 10.1002/anie.202410825] [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: 06/08/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024]
Abstract
Electrochemical imaging enables an in-depth analysis of the interface heterogeneity and reaction kinetics of single entities. However, electrode passivation during electrochemical reactions decreases the active sites and harms the long-term stability. Here, we introduce a method using laser-induced photothermal effects to restore the electrochemical activity, which is particularly displayed as enhanced micrometric patterns in electrochemiluminescence (ECL) microscopy. By co-localization characterization and X-ray photoelectron spectroscopy (XPS), the mechanism of active site regeneration is validated as the removal of the oxide film for restoring the local surface ECL reactivity under laser irradiation. The surface-confined and voltage-dependent features of ECL allows for easy pattern erasure and rewriting, and it shows good reversibility and anti-counterfeiting potential. This approach overcomes the passivation processes, evidently improves the image quality of single biological entities including Shewanella bacteria and cells, and makes the subtle contour structures more distinct. The renewable electrode interface also enhances the ECL signal of model bead-based bioassays. This approach not only showcases precise control in fabricating micron patterns but also holds promise for enhancing the sensitivity in electrochemical immunoassays and bioimaging.
Collapse
Affiliation(s)
- Xiaodan Gou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
- University of Bordeaux, Bordeaux INP, ISM, UMR, CNRS 5255, 33607, Pessac, France
| | - Zejing Xing
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Zhichen Zhang
- School of Chemistry and Chemical Engineering, Yangzhou University, 225002, Yangzhou, P. R. China
| | - Rong Jin
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Qin Xu
- School of Chemistry and Chemical Engineering, Yangzhou University, 225002, Yangzhou, P. R. China
| | - Neso Sojic
- University of Bordeaux, Bordeaux INP, ISM, UMR, CNRS 5255, 33607, Pessac, France
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Cheng Ma
- School of Chemistry and Chemical Engineering, Yangzhou University, 225002, Yangzhou, P. R. China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| |
Collapse
|
10
|
Mottet G, Grassart A, Barthélemy P, Antignac C, Arrabal S, Bourdin A, Descroix S, De Vos J, Doutriaux A, Fabrega Q, Galaup A, Graff-Dubois S, Illiano S, Legallais C, Maisonneuve B, Piwnica D, Quéméneur E, Salentey V, Rozenberg J, Sotiropoulos A, Tomasi R, Vergnolle N, Devillier P. Organoïdes, organes sur puce, complex in vitro model : définitions, applications, validation, éthique. Therapie 2025; 80:1-16. [PMID: 39710544 DOI: 10.1016/j.therap.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024]
Affiliation(s)
- Guillaume Mottet
- Large Molecule Research Platform, Microfluidic Team, Sanofi R&D, 94400 Vitry-sur-Seine, France
| | - Alexandre Grassart
- CNRS, Inserm, institut Pasteur de Lille, U1019, UMR 9017, Center for Infection and Immunity of Lille (CIIL), CHU de Lille, université de Lille, 59000 Lille, France.
| | | | - Corinne Antignac
- Laboratoire des maladies rénales héréditaires, Inserm UMR1153, institut Imagine, université Paris Cité, 75000 Paris, France
| | | | - Arnaud Bourdin
- Département de pneumologie et addictologie, PhyMedExp, Inserm U1046, CNRS UMR 9214, University of Montpellier, 34000 Montpellier, France; Hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 34000 Montpellier, France
| | - Stéphanie Descroix
- Laboratoire physique des cellules et cancer, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75000 Paris, France
| | - John De Vos
- Ingénierie cellulaire et tissulaire, unité de thérapie cellulaire, CHU de Montpellier, 34000 Montpellier, France
| | | | - Quentin Fabrega
- Direction des filières industrielles, Bpifrance, 75000 Paris, France
| | | | - Stéphanie Graff-Dubois
- UMRS 959, laboratoire i3, groupe hospitalier Pitié-Salpêtrière, Sorbonne université, 75000 Paris, France
| | | | - Cécile Legallais
- UMR CNRS 7338 biomécanique et bioingénierie, université de technologie de Compiègne, 60200 Compiègne, France
| | | | - David Piwnica
- Institut de R&D Servier Paris-Saclay, 91190 Gif-sur-Yvette, France
| | | | - Valérie Salentey
- Regulatory Affairs and Quality Assurance, Sensorion, 34000 Montpellier, France
| | | | | | | | - Nathalie Vergnolle
- IRSD, Inserm, INRAE, ENVT, université Toulouse III - Paul-Sabatier, université de Toulouse, 31000 Toulouse, France
| | - Philippe Devillier
- VIM Suresnes, UMR_0892, hôpital Foch, université Paris-Saclay, 92150 Suresnes, France
| |
Collapse
|
11
|
Mottet G, Grassart A, Barthélemy P, Antignac C, Arrabal S, Bourdin A, Descroix S, De Vos J, Doutriaux A, Fabrega Q, Galaup A, Graff-Dubois S, Illiano S, Legallais C, Maisonneuve B, Piwnica D, Quéméneur E, Salentey V, Rozenberg J, Sotiropoulos A, Tomasi R, Vergnolle N, Devillier P. Organoids, organs-on-chips, complex in vitro model: Definitions, applications, validation, ethics. Therapie 2025; 80:17-31. [PMID: 39809611 DOI: 10.1016/j.therap.2024.12.002] [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: 10/11/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
Over the past decade, new in vitro biological models have emerged which can reproduce certain characteristics of human physiology and pathologies. From organoids to organs-on-chips, these new technologies are currently revolutionizing the entire chain of research and development in pharmacology. All stakeholders are thus involved, from academic laboratories to pharmaceutical companies, start-ups, and assessment agencies. By providing better predictability, these new human biomimetic models also help address societal and ethical challenges regarding the place and use of animals in biomedical research. In this context, participants at the Ateliers de Giens (Giens Workshops) roundtable wished to examine the issues related to these new technologies with their various expertise and, given the stakes involved in the medicine and pharmacology of the future, formulate several recommendations aimed at strengthening the national structuring of the academic and industrial sector, therefore accelerating the development and validation of these new models.
Collapse
Affiliation(s)
- Guillaume Mottet
- Large Molecule Research Platform, Microfluidic Team, Sanofi R&D, 94400 Vitry-sur-Seine, France
| | - Alexandre Grassart
- CNRS, Inserm, institut Pasteur de Lille, U1019, UMR 9017, Center for Infection and Immunity of Lille (CIIL), CHU de Lille, université de Lille, 59000 Lille, France.
| | | | - Corinne Antignac
- Laboratoire des maladies rénales héréditaires, Inserm UMR1153, institut Imagine, université Paris Cité, 75000 Paris, France
| | | | - Arnaud Bourdin
- Département de pneumologie et addictologie, PhyMedExp, Inserm U1046, CNRS UMR 9214, University of Montpellier, 34000 Montpellier, France; Hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 34000 Montpellier, France
| | - Stéphanie Descroix
- Laboratoire physique des cellules et cancer, PCC, CNRS UMR168, institut Curie, Sorbonne University, PSL University, 75000 Paris, France
| | - John De Vos
- Ingénierie cellulaire et tissulaire, unité de thérapie cellulaire, CHU de Montpellier, 34000 Montpellier, France
| | | | - Quentin Fabrega
- Direction des filières industrielles, Bpifrance, 75000 Paris, France
| | | | - Stéphanie Graff-Dubois
- UMRS 959, laboratoire i3, groupe hospitalier Pitié-Salpêtrière, Sorbonne université, 75000 Paris, France
| | | | - Cécile Legallais
- UMR CNRS 7338 biomécanique et bioingénierie, université de technologie de Compiègne, 60200 Compiègne, France
| | | | - David Piwnica
- Institut de R&D Servier Paris-Saclay, 91190 Gif-sur-Yvette, France
| | | | - Valérie Salentey
- Regulatory Affairs and Quality Assurance, Sensorion, 34000 Montpellier, France
| | | | | | | | - Nathalie Vergnolle
- IRSD, Inserm, INRAE, ENVT, université Toulouse III - Paul-Sabatier, université de Toulouse, 31000 Toulouse, France
| | - Philippe Devillier
- VIM Suresnes, UMR_0892, hôpital Foch, université Paris-Saclay, 92150 Suresnes, France
| |
Collapse
|
12
|
Geng J, Voitiuk K, Parks DF, Robbins A, Spaeth A, Sevetson JL, Hernandez S, Schweiger HE, Andrews JP, Seiler ST, Elliott MA, Chang EF, Nowakowski TJ, Currie R, Mostajo-Radji MA, Haussler D, Sharf T, Salama SR, Teodorescu M. Multiscale Cloud-Based Pipeline for Neuronal Electrophysiology Analysis and Visualization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623530. [PMID: 39605518 PMCID: PMC11601321 DOI: 10.1101/2024.11.14.623530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Electrophysiology offers a high-resolution method for real-time measurement of neural activity. Longitudinal recordings from high-density microelectrode arrays (HD-MEAs) can be of considerable size for local storage and of substantial complexity for extracting neural features and network dynamics. Analysis is often demanding due to the need for multiple software tools with different runtime dependencies. To address these challenges, we developed an open-source cloud-based pipeline to store, analyze, and visualize neuronal electrophysiology recordings from HD-MEAs. This pipeline is dependency agnostic by utilizing cloud storage, cloud computing resources, and an Internet of Things messaging protocol. We containerized the services and algorithms to serve as scalable and flexible building blocks within the pipeline. In this paper, we applied this pipeline on two types of cultures, cortical organoids and ex vivo brain slice recordings to show that this pipeline simplifies the data analysis process and facilitates understanding neuronal activity.
Collapse
Affiliation(s)
- Jinghui Geng
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kateryna Voitiuk
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - David F. Parks
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ash Robbins
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Alex Spaeth
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jessica L. Sevetson
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sebastian Hernandez
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - John P. Andrews
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Spencer T. Seiler
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Matthew A.T. Elliott
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Edward F. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J. Nowakowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Rob Currie
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - David Haussler
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tal Sharf
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sofie R. Salama
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mircea Teodorescu
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Lead Contact
| |
Collapse
|
13
|
Zimina TM, Sitkov NO, Gareev KG, Mikhailova NV, Combs SE, Shevtsov MA. Hybrid-integrated devices for mimicking malignant brain tumors ("tumor-on-a-chip") for in vitro development of targeted drug delivery and personalized therapy approaches. Front Med (Lausanne) 2024; 11:1452298. [PMID: 39629230 PMCID: PMC11611596 DOI: 10.3389/fmed.2024.1452298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/14/2024] [Indexed: 12/07/2024] Open
Abstract
Acute and requiring attention problem of oncotheranostics is a necessity for the urgent development of operative and precise diagnostics methods, followed by efficient therapy, to significantly reduce disability and mortality of citizens. A perspective way to achieve efficient personalized treatment is to use methods for operative evaluation of the individual drug load, properties of specific tumors and the effectiveness of selected therapy, and other actual features of pathology. Among the vast diversity of tumor types-brain tumors are the most invasive and malignant in humans with poor survival after diagnosis. Among brain tumors glioblastoma shows exceptionally high mortality. More studies are urgently needed to understand the risk factors and improve therapy approaches. One of the actively developing approaches is the tumor-on-a-chip (ToC) concept. This review examines the achievements of recent years in the field of ToC system developments. The basics of microfluidic chips technologies are considered in the context of their applications in solving oncological problems. Then the basic principles of tumors cultivation are considered to evaluate the main challengers in implementation of microfluidic devices, for growing cell cultures and possibilities of their treatment and observation. The main achievements in the culture types diversity approaches and their advantages are being analyzed. The modeling of angiogenesis and blood-brain barrier (BBB) on a chip, being a principally important elements of the life system, were considered in detail. The most interesting examples and achievements in the field of tumor-on-a-chip developments have been presented.
Collapse
Affiliation(s)
- Tatiana M. Zimina
- Department of Micro and Nanoelectronics, St. Petersburg Electrotechnical University “LETI” (ETU), Saint Petersburg, Russia
| | - Nikita O. Sitkov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Kamil G. Gareev
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Natalia V. Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Stephanie E. Combs
- Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maxim A. Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
- Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
14
|
Costa BL, Camarneiro F, Marote A, Barbosa C, Vedor C, Tomé D, Costa FJ, Dias MS, Correia J, Pires J, Chícharo A, Almeida RD, Salgado A, Nieder JB. Functionalized Nanodiamonds for Targeted Neuronal Electromagnetic Signal Detection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:60828-60841. [PMID: 39445729 PMCID: PMC11551900 DOI: 10.1021/acsami.4c12462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Intracellular sensing technologies necessitate a delicate balance of spatial resolution, sensitivity, biocompatibility, and stability. While existing methods partially fulfill these criteria, none offer a comprehensive solution. Nanodiamonds (NDs) harboring nitrogen-vacancy (NV) centers have emerged as promising candidates due to their sensing capabilities under biological conditions and their ability to meet all aforementioned requirements. This study focuses on expanding the application of NDs and NV center-based sensing to neuronal contexts by investigating their functionalization and subsequent effects on three distinct cell lines relevant to neurodegenerative disease research. Our study concentrates on positioning fluorescent NDs (FNDs) with NV center point defects onto neuronal cell surfaces. Achieving this through specific antibody attachment enhances the proximity of FND to neurites, facilitating the detection of local action potentials. Targeting voltage-dependent calcium channels (Cav2.2) with biotin-streptavidin-bound antibodies enables the precise positioning of FNDs. The functionalized FNDs (f-FNDs) show increased size and zeta potential, confirming the antibody presence without compromising cell viability. Two-color confocal imaging and co-localization algorithms are employed to further attest to the success of the functionalization. The f-FNDs are applied to cell cultures of three cell lines: SH-SY5Y, differentiated dopaminergic neurons, and hippocampal rat neurons; their biocompatibility and effects on synaptic activity are explored. Moreover, preliminary total internal reflection fluorescence - optically detected magnetic resonance (TIRF-ODMR) experiments across cellular sites demonstrate the magnetic field sensitivity of our sensor network. The successful establishment of this sensor network provides a platform for characterizing neuronal signaling in healthy models and conditions mimicking Parkinson's disease.
Collapse
Affiliation(s)
- Beatriz
N. L. Costa
- INL
- International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
- Escola
de Enxeñaría de Minas e Enerxía, University of Vigo, 36310 Vigo, Pontevedra, Spain
| | - Filipe Camarneiro
- INL
- International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Ana Marote
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s
– PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Catarina Barbosa
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s
– PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carlos Vedor
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s
– PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Diogo Tomé
- iBiMED-
Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CNC,
Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filipa J. Costa
- iBiMED-
Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marta S. Dias
- iBiMED-
Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joana Correia
- iBiMED-
Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joel Pires
- iBiMED-
Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CNC,
Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Alexandre Chícharo
- INL
- International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Ramiro D. Almeida
- iBiMED-
Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- CNC,
Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - António Salgado
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s
– PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Jana B. Nieder
- INL
- International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| |
Collapse
|
15
|
Della Rosa G, Gostynska N, Ephraim JW, Marras S, Moroni M, Tirelli N, Panuccio G, Palazzolo G. Magnesium vs. sodium alginate as precursors of calcium alginate: Mechanical differences and advantages in the development of functional neuronal networks. Carbohydr Polym 2024; 342:122375. [PMID: 39048194 DOI: 10.1016/j.carbpol.2024.122375] [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: 02/27/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/27/2024]
Abstract
Calcium alginate is one of the most widely employed matrices in regenerative medicine. A downside is its heterogeneity, due to the poorly controllable character of the gelation of sodium alginate (NaAlg), i.e. the commonly used alginate salt, with calcium. Here, we have used magnesium alginate (MgAlg) as an alternative precursor of calcium alginate. MgAlg coils, more compact and thus less entangled than those of NaAlg, allow for an easier diffusion of calcium ions, whereas Mg is exchanged with calcium more slowly than Na; this allows for the formation of a material (Ca(Mg)Alg) with a more reversible creep behaviour than Ca(Na)Alg, due to a more homogeneous - albeit lower - density of elastically active cross-links. We also show that Ca(Mg)Alg supports better than Ca(Na)Alg the network development and function of embedded (rat cortical) neurons: they show greater neurite extension and branching at 7 and 21 days (Tubb3 and Map2 immunofluorescence) and better neuronal network functional maturation / more robust and longer-lasting activity, probed by calcium imaging and microelectrode array electrophysiology. Overall, our results unveil the potential of MgAlg as bioactive biomaterial for enabling the formation of functional neuron-based tissue analogues.
Collapse
Affiliation(s)
- Giulia Della Rosa
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy; University of Pavia, Department of Molecular Medicine, Pavia, Italy.
| | - Natalia Gostynska
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| | - John W Ephraim
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| | - Sergio Marras
- Istituto Italiano di Tecnologia, Materials Characterization Facility, Genova, Italy.
| | | | - Nicola Tirelli
- Istituto Italiano di Tecnologia, Laboratory for Polymers and Biomaterials, Genova, Italy.
| | - Gabriella Panuccio
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| | - Gemma Palazzolo
- Istituto Italiano di Tecnologia, Laboratory for Enhanced Regenerative Medicine, Genova, Italy.
| |
Collapse
|
16
|
Xu W, Wang M, Yang G, Mo F, Liu Y, Shan J, Jing L, Li M, Liu J, Lv S, Duan Y, Han M, Xu Z, Song Y, Cai X. Neuronal activity in the ventral tegmental area during goal-directed navigation recorded by low-curvature microelectrode arrays. MICROSYSTEMS & NANOENGINEERING 2024; 10:145. [PMID: 39396959 PMCID: PMC11471829 DOI: 10.1038/s41378-024-00778-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 10/15/2024]
Abstract
Navigating toward destinations with rewards is a common behavior among animals. The ventral tegmental area (VTA) has been shown to be responsible for reward coding and reward cue learning, and its response to other variables, such as kinematics, has also been increasingly studied. These findings suggest a potential relationship between animal navigation behavior and VTA activity. However, the deep location and small volume of the VTA pose significant challenges to the precision of electrode implantation, increasing the uncertainty of measurement results during animal navigation and thus limiting research on the role of the VTA in goal-directed navigation. To address this gap, we innovatively designed and fabricated low-curvature microelectrode arrays (MEAs) via a novel backside dry etching technique to release residual stress. Histological verification confirmed that low-curvature MEAs indeed improved electrode implantation precision. These low-curvature MEAs were subsequently implanted into the VTA of the rats to observe their electrophysiological activity in a freely chosen modified T-maze. The results of the behavioral experiments revealed that the rats could quickly learn the reward probability corresponding to the left and right paths and that VTA neurons were deeply involved in goal-directed navigation. Compared with those in no-reward trials, VTA neurons in reward trials presented a significantly greater firing rate and larger local field potential (LFP) amplitude during the reward-consuming period. Notably, we discovered place fields mapped by VTA neurons, which disappeared or were reconstructed with changes in the path-outcome relationship. These results provide new insights into the VTA and its role in goal-directed navigation. Our designed and fabricated low-curvature microelectrode arrays can serve as a new device for precise deep brain implantation in the future.
Collapse
Affiliation(s)
- Wei Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mixia Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Gucheng Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fan Mo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaoyao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jin Shan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Luyi Jing
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ming Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiya Lv
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiming Duan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meiqi Han
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaojie Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
17
|
Yoon D, Nam Y. A 3D neuronal network read-out interface with high recording performance using a neuronal cluster patterning on a microelectrode array. Biosens Bioelectron 2024; 261:116507. [PMID: 38905857 DOI: 10.1016/j.bios.2024.116507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/27/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024]
Abstract
In recent years, in vitro three-dimensional (3D) neuronal network models utilizing extracellular matrices have been advancing. To understand the network activity from these models, attempts have been made to measure activity in multiple regions simultaneously using a microelectrode array (MEA). Although there hve been many attempts to measure the activity of 3D networks using 2-dimensional (2D) MEAs, the physical coupling between the 3D network and the microelectrodes was not stable and needed to be improved. In this study, we proposed a neuronal cluster interface that improves the active channel ratio of commercial 2D MEAs, enabling reliable measurement of 3D network activity. To achieve this, neuronal clusters, which consist of a small number of neurons, were patterned on microelectrodes and used as mediators to transmit the signal between the 3D network and the microelectrodes. We confirmed that the patterned neuronal clusters enhanced the active channel ratio and SNR(signal-to-noise-ratio) about 3D network recording and stimulation for a month. Our interface was able to functionally connect with 3D networks and measure the 3D network activity without significant alternation of activity characteristics. Finally, we demonstrated that our interface can be used to analyze the differences in the dynamics of 3D and 2D networks and to construct the 3D clustered network. This method is expected to be useful for studying the functional activity of various 3D neuronal network models, offering broad applications for the use of these models.
Collapse
Affiliation(s)
- Dongjo Yoon
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yoonkey Nam
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
18
|
Habibey R, Striebel J, Meinert M, Latiftikhereshki R, Schmieder F, Nasiri R, Latifi S. Engineered modular neuronal networks-on-chip represent structure-function relationship. Biosens Bioelectron 2024; 261:116518. [PMID: 38924816 DOI: 10.1016/j.bios.2024.116518] [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: 02/08/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Brain function is substantially linked to the highly organized modular structure of neuronal networks. However, the structure of in vitro assembled neuronal circuits often exhibits variability, complicating the consistent recording of network functional output and its correlation to network structure. Therefore, engineering neuronal structures with predefined geometry and reproducible functional features is essential to precisely model in vivo neuronal circuits. Here, we engineered microchannel devices to assemble 2D and 3D modular networks. The microchannel devices were coupled with a multi-electrode array (MEA) electrophysiology system to enable recordings from circuits. Each network consisted of 64 modules connected to their adjacent modules by micron-sized channels. Modular circuits within microchannel devices showed enhanced activity and functional connectivity traits. This includes metrics such as connection weights, clustering coefficient, global efficiency, and the number of hub neurons with higher betweenness centrality. In addition, modular networks demonstrated an increased functional modularity score compared to the randomly formed circuits. Neurons within individual modules displayed uniform network characteristics and predominantly participated in their respective functional communities within the same or neighboring physical modules. These observations highlight that the modular network structure promotes the development of segregated functional connectivity traits while simultaneously enhancing the efficiency of overall network connectivity. Our findings emphasize the significant impact of physical constraints on the activity patterns and functional organization within engineered modular networks. These circuits, characterized by stable modular architecture and intricate functional dynamics-key features of the brain networks-offer a robust in vitro model for advancing neuroscience research.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany; CRTD - Center for Regenerative Therapies TU Dresden, 01307, Dresden, Germany; Dept. Neuroscience, Italian Institute of Technology. Genova, Italy.
| | - Johannes Striebel
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Melissa Meinert
- Department of Ophthalmology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Roshanak Latiftikhereshki
- Department of Computer Engineering, Faculty of Engineering, Kermanshah Branch, Azad University, Kermanshah, Iran
| | - Felix Schmieder
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Helmholtzstraße 18, 01069, Dresden, Germany
| | - Rohollah Nasiri
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden; AIMES, Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, Solna, Sweden
| | - Shahrzad Latifi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Neuroscience, Rockefeller Neuroscience Institute West Virginia University, Morgantown, WV, 26506, USA
| |
Collapse
|
19
|
Yi D, Yao Y, Wang Y, Chen L. Design, Fabrication, and Implantation of Invasive Microelectrode Arrays as in vivo Brain Machine Interfaces: A Comprehensive Review. JOURNAL OF MANUFACTURING PROCESSES 2024; 126:185-207. [PMID: 39185373 PMCID: PMC11340637 DOI: 10.1016/j.jmapro.2024.07.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Invasive Microelectrode Arrays (MEAs) have been a significant and useful tool for us to gain a fundamental understanding of how the brain works through high spatiotemporal resolution neuron-level recordings and/or stimulations. Through decades of research, various types of microwire, silicon, and flexible substrate-based MEAs have been developed using the evolving new materials, novel design concepts, and cutting-edge advanced manufacturing capabilities. Surgical implantation of the latest minimal damaging flexible MEAs through the hard-to-penetrate brain membranes introduces new challenges and thus the development of implantation strategies and instruments for the latest MEAs. In this paper, studies on the design considerations and enabling manufacturing processes of various invasive MEAs as in vivo brain-machine interfaces have been reviewed to facilitate the development as well as the state-of-art of such brain-machine interfaces from an engineering perspective. The challenges and solution strategies developed for surgically implanting such interfaces into the brain have also been evaluated and summarized. Finally, the research gaps have been identified in the design, manufacturing, and implantation perspectives, and future research prospects in invasive MEA development have been proposed.
Collapse
Affiliation(s)
- Dongyang Yi
- Department of Mechanical and Industrial Engineering, University of Massachusetts Lowell, Lowell, MA 01854
| | - Yao Yao
- Department of Industrial and Systems Engineering, University of Missouri, Columbia, MO 65211
| | - Yi Wang
- Department of Industrial and Systems Engineering, University of Missouri, Columbia, MO 65211
| | - Lei Chen
- Department of Mechanical and Industrial Engineering, University of Massachusetts Lowell, Lowell, MA 01854
| |
Collapse
|
20
|
Xu Q, Xi Y, Wang L, Du Z, Xu M, Ruan T, Cao J, Zheng K, Wang X, Yang B, Liu J. An Opto-electrophysiology Neural Probe with Photoelectric Artifact-Free for Advanced Single-Neuron Analysis. ACS NANO 2024; 18:25193-25204. [PMID: 39193830 DOI: 10.1021/acsnano.4c07379] [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: 08/29/2024]
Abstract
Opto-electrophysiology neural probes targeting single-cell levels offer an important avenue for elucidating the intrinsic mechanisms of the nervous system using different physical quantities, representing a significant future direction for brain-computer interface (BCI) devices. However, the highly integrated structure poses significant challenges to fabrication processes and the presence of photoelectric artifacts complicates the extraction and analysis of target signals. Here, we propose a highly miniaturized and integrated opto-electrophysiology neural probe for electrical recording and optical stimulation at the single-cell/subcellular level. The design of a total internal reflection layer addresses the photoelectric artifacts that are more pronounced in single-cell devices compared to conventional implantable BCI devices. Finite element simulations and electrical signal tests demonstrate that the opto-electrophysiology neural probe eliminates the photoelectric artifacts in the time domain, which represents a significant breakthrough for optoelectrical integrated BCI devices. Our proposed opto-electrophysiology neural probe holds substantial potential for promoting the development of in vivo BCI devices and developing advanced therapeutic strategies for neurological disorders.
Collapse
Affiliation(s)
- Qingda Xu
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ye Xi
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Longchun Wang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhiyuan Du
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mengfei Xu
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tao Ruan
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiawei Cao
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kunyu Zheng
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
- DCI Joint Team, Collaborative Innovation Center of IFSA, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaolin Wang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bin Yang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jingquan Liu
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
21
|
Lee S, Chung WG, Jeong H, Cui G, Kim E, Lim JA, Seo H, Kwon YW, Byeon SH, Lee J, Park JU. Electrophysiological Analysis of Retinal Organoid Development Using 3D Microelectrodes of Liquid Metals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404428. [PMID: 38896876 DOI: 10.1002/adma.202404428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/17/2024] [Indexed: 06/21/2024]
Abstract
Despite of the substantial potential of human-derived retinal organoids, the degeneration of retinal ganglion cells (RGCs) during maturation limits their utility in assessing the functionality of later-born retinal cell subtypes. Additionally, conventional analyses primarily rely on fluorescent emissions, which limits the detection of actual cell functionality while risking damage to the 3D cytoarchitecture of organoids. Here, an electrophysiological analysis is presented to monitor RGC development in early to mid-stage retinal organoids, and compare distinct features with fully-mature mouse retina. This approach utilizes high-resolution 3D printing of liquid-metal microelectrodes, enabling precise targeting of specific inner retinal layers within organoids. The adaptable distribution and softness of these microelectrodes facilitate the spatiotemporal recording of inner retinal signals. This study not only demonstrates the functional properties of RGCs in retinal organoid development but also provides insights into their synaptic connectivity, reminiscent of fetal native retinas. Further comparison with fully-mature mouse retina in vivo verifies the organoid features, highlighting the potential of early-stage retinal organoids in biomedical research.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Won Gi Chung
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Han Jeong
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Gang Cui
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Enji Kim
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jeong Ah Lim
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hunkyu Seo
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yong Won Kwon
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suk Ho Byeon
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Junwon Lee
- Institute of Vision Research, Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
22
|
Nahon DM, Moerkens R, Aydogmus H, Lendemeijer B, Martínez-Silgado A, Stein JM, Dostanić M, Frimat JP, Gontan C, de Graaf MNS, Hu M, Kasi DG, Koch LS, Le KTT, Lim S, Middelkamp HHT, Mooiweer J, Motreuil-Ragot P, Niggl E, Pleguezuelos-Manzano C, Puschhof J, Revyn N, Rivera-Arbelaez JM, Slager J, Windt LM, Zakharova M, van Meer BJ, Orlova VV, de Vrij FMS, Withoff S, Mastrangeli M, van der Meer AD, Mummery CL. Standardizing designed and emergent quantitative features in microphysiological systems. Nat Biomed Eng 2024; 8:941-962. [PMID: 39187664 DOI: 10.1038/s41551-024-01236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/06/2024] [Indexed: 08/28/2024]
Abstract
Microphysiological systems (MPSs) are cellular models that replicate aspects of organ and tissue functions in vitro. In contrast with conventional cell cultures, MPSs often provide physiological mechanical cues to cells, include fluid flow and can be interlinked (hence, they are often referred to as microfluidic tissue chips or organs-on-chips). Here, by means of examples of MPSs of the vascular system, intestine, brain and heart, we advocate for the development of standards that allow for comparisons of quantitative physiological features in MPSs and humans. Such standards should ensure that the in vivo relevance and predictive value of MPSs can be properly assessed as fit-for-purpose in specific applications, such as the assessment of drug toxicity, the identification of therapeutics or the understanding of human physiology or disease. Specifically, we distinguish designed features, which can be controlled via the design of the MPS, from emergent features, which describe cellular function, and propose methods for improving MPSs with readouts and sensors for the quantitative monitoring of complex physiology towards enabling wider end-user adoption and regulatory acceptance.
Collapse
Affiliation(s)
- Dennis M Nahon
- Leiden University Medical Center, Leiden, the Netherlands
| | - Renée Moerkens
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Bas Lendemeijer
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Adriana Martínez-Silgado
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands
| | - Jeroen M Stein
- Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Cristina Gontan
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Michel Hu
- Leiden University Medical Center, Leiden, the Netherlands
| | - Dhanesh G Kasi
- Leiden University Medical Center, Leiden, the Netherlands
| | - Lena S Koch
- University of Twente, Enschede, the Netherlands
| | - Kieu T T Le
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sangho Lim
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands
| | | | - Joram Mooiweer
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Eva Niggl
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Jens Puschhof
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands
| | - Nele Revyn
- Delft University of Technology, Delft, the Netherlands
| | | | - Jelle Slager
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Laura M Windt
- Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | - Sebo Withoff
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | |
Collapse
|
23
|
Choi MS, Park SM, Kim S, Jegal H, Lee HA, Han HY, Yoon S, Kim SK, Oh JH. Enhanced electrophysiological activity and neurotoxicity screening of environmental chemicals using 3D neurons from human neural precursor cells purified with PSA-NCAM. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116516. [PMID: 38820819 DOI: 10.1016/j.ecoenv.2024.116516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/02/2024]
Abstract
The assessment of neurotoxicity for environmental chemicals is of utmost importance in ensuring public health and environmental safety. Multielectrode array (MEA) technology has emerged as a powerful tool for assessing disturbances in the electrophysiological activity. Although human embryonic stem cell (hESC)-derived neurons have been used in MEA for neurotoxicity screening, obtaining a substantial and sufficiently active population of neurons from hESCs remains challenging. In this study, we successfully differentiated neurons from a large population of human neuronal precursor cells (hNPC) purified using a polysialylated neural cell adhesion molecule (PSA-NCAM), referred to as hNPCPSA-NCAM+. The functional characterization demonstrated that hNPCPSA-NCAM+-derived neurons improve functionality by enhancing electrophysiological activity compared to total hNPC-derived neurons. Furthermore, three-dimensional (3D) neurons derived from hNPCPSA-NCAM+ exhibited reduced maturation time and enhanced electrophysiological activity on MEA. We employed subdivided population analysis of active mean firing rate (MFR) based on electrophysiological intensity to characterize the electrophysiological properties of hNPCPSA-NCAM+-3D neurons. Based on electrophysiological activity including MFR and burst parameters, we evaluated the sensitivity of hNPCPSA-NCAM+-3D neurons on MEA to screen both inhibitory and excitatory neuroactive environmental chemicals. Intriguingly, electrophysiologically active hNPCPSA-NCAM+-3D neurons demonstrated good sensitivity to evaluate neuroactive chemicals, particularly in discriminating excitatory chemicals. Our findings highlight the effectiveness of MEA approaches using hNPCPSA-NCAM+-3D neurons in the assessment of neurotoxicity associated with environmental chemicals. Furthermore, we emphasize the importance of selecting appropriate signal intensity thresholds to enhance neurotoxicity prediction and screening of environmental chemicals.
Collapse
Affiliation(s)
- Mi-Sun Choi
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; College of Pharmacy, Chungnam National University, Daejeon, the Republic of Korea
| | - Se-Myo Park
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea
| | - Soojin Kim
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea
| | - Hyun Jegal
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea
| | - Hyang-Ae Lee
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea
| | - Hyoung-Yun Han
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea
| | - Seokjoo Yoon
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea
| | - Sang-Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, the Republic of Korea.
| | - Jung-Hwa Oh
- Department of predictive toxicology, Korea Institute of Toxicology (KIT), Daejeon, the Republic of Korea; Department of Human and Environmental Toxicology, University of Science & Technology, Daejeon, the Republic of Korea.
| |
Collapse
|
24
|
Sandoval SO, Cappuccio G, Kruth K, Osenberg S, Khalil SM, Méndez-Albelo NM, Padmanabhan K, Wang D, Niciu MJ, Bhattacharyya A, Stein JL, Sousa AMM, Waxman EA, Buttermore ED, Whye D, Sirois CL, Williams A, Maletic-Savatic M, Zhao X. Rigor and reproducibility in human brain organoid research: Where we are and where we need to go. Stem Cell Reports 2024; 19:796-816. [PMID: 38759644 PMCID: PMC11297560 DOI: 10.1016/j.stemcr.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/19/2024] Open
Abstract
Human brain organoid models have emerged as a promising tool for studying human brain development and function. These models preserve human genetics and recapitulate some aspects of human brain development, while facilitating manipulation in an in vitro setting. Despite their potential to transform biology and medicine, concerns persist about their fidelity. To fully harness their potential, it is imperative to establish reliable analytic methods, ensuring rigor and reproducibility. Here, we review current analytical platforms used to characterize human forebrain cortical organoids, highlight challenges, and propose recommendations for future studies to achieve greater precision and uniformity across laboratories.
Collapse
Affiliation(s)
- Soraya O Sandoval
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Gerarda Cappuccio
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Karina Kruth
- Department of Psychiatry, University of Iowa Health Care, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Sivan Osenberg
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Saleh M Khalil
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Natasha M Méndez-Albelo
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Molecular Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Krishnan Padmanabhan
- Department of Neuroscience, Center for Visual Science, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Departments of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mark J Niciu
- Department of Psychiatry, University of Iowa Health Care, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - André M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Elisa A Waxman
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Epilepsy and NeuroDevelopmental Disorders (ENDD), The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth D Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Carissa L Sirois
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Aislinn Williams
- Department of Psychiatry, University of Iowa Health Care, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Health Care, Iowa City, IA 52242, USA.
| | - Mirjana Maletic-Savatic
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA; Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
25
|
Kwak T, Park SH, Lee S, Shin Y, Yoon KJ, Cho SW, Park JC, Yang SH, Cho H, Im HI, Ahn SJ, Sun W, Yang JH. Guidelines for Manufacturing and Application of Organoids: Brain. Int J Stem Cells 2024; 17:158-181. [PMID: 38777830 PMCID: PMC11170118 DOI: 10.15283/ijsc24056] [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: 04/24/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
This study offers a comprehensive overview of brain organoids for researchers. It combines expert opinions with technical summaries on organoid definitions, characteristics, culture methods, and quality control. This approach aims to enhance the utilization of brain organoids in research. Brain organoids, as three-dimensional human cell models mimicking the nervous system, hold immense promise for studying the human brain. They offer advantages over traditional methods, replicating anatomical structures, physiological features, and complex neuronal networks. Additionally, brain organoids can model nervous system development and interactions between cell types and the microenvironment. By providing a foundation for utilizing the most human-relevant tissue models, this work empowers researchers to overcome limitations of two-dimensional cultures and conduct advanced disease modeling research.
Collapse
Affiliation(s)
| | - Si-Hyung Park
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
| | | | | | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
- Organoid Standards Initiative
| | - Seung-Woo Cho
- Organoid Standards Initiative
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Jong-Chan Park
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Seung-Ho Yang
- Organoid Standards Initiative
- Department of Neurosurgery, St. Vincent’s Hospital, The Catholic University of Korea, Suwon, Korea
| | - Heeyeong Cho
- Organoid Standards Initiative
- Center for Rare Disease Therapeutic Technology, Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Heh-In Im
- Organoid Standards Initiative
- Behavioral and Molecular Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Sun-Ju Ahn
- Organoid Standards Initiative
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea
- Organoid Standards Initiative
| | - Ji Hun Yang
- Next & Bio Inc., Seoul, Korea
- Organoid Standards Initiative
| |
Collapse
|
26
|
Xiang Y, Zhao Y, Cheng T, Sun S, Wang J, Pei R. Implantable Neural Microelectrodes: How to Reduce Immune Response. ACS Biomater Sci Eng 2024; 10:2762-2783. [PMID: 38591141 DOI: 10.1021/acsbiomaterials.4c00238] [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] [Indexed: 04/10/2024]
Abstract
Implantable neural microelectrodes exhibit the great ability to accurately capture the electrophysiological signals from individual neurons with exceptional submillisecond precision, holding tremendous potential for advancing brain science research, as well as offering promising avenues for neurological disease therapy. Although significant advancements have been made in the channel and density of implantable neural microelectrodes, challenges persist in extending the stable recording duration of these microelectrodes. The enduring stability of implanted electrode signals is primarily influenced by the chronic immune response triggered by the slight movement of the electrode within the neural tissue. The intensity of this immune response increases with a higher bending stiffness of the electrode. This Review thoroughly analyzes the sequential reactions evoked by implanted electrodes in the brain and highlights strategies aimed at mitigating chronic immune responses. Minimizing immune response mainly includes designing the microelectrode structure, selecting flexible materials, surface modification, and controlling drug release. The purpose of this paper is to provide valuable references and ideas for reducing the immune response of implantable neural microelectrodes and stimulate their further exploration in the field of brain science.
Collapse
Affiliation(s)
- Ying Xiang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tingting Cheng
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Shengkai Sun
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Jine Wang
- Jiangxi Institute of Nanotechnology, Nanchang 330200, China
- College of Medicine and Nursing, Shandong Provincial Engineering Laboratory of Novel Pharmaceutical Excipients, Sustained and Controlled Release Preparations, Dezhou University, Dezhou 253023, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
27
|
Stoppini L, Heuschkel MO, Loussert-Fonta C, Gomez Baisac L, Roux A. Versatile micro-electrode array to monitor human iPSC derived 3D neural tissues at air-liquid interface. Front Cell Neurosci 2024; 18:1389580. [PMID: 38784710 PMCID: PMC11112036 DOI: 10.3389/fncel.2024.1389580] [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: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
Engineered 3D neural tissues made of neurons and glial cells derived from human induced pluripotent stem cells (hiPSC) are among the most promising tools in drug discovery and neurotoxicology. They represent a cheaper, faster, and more ethical alternative to in vivo animal testing that will likely close the gap between in vitro animal models and human clinical trials. Micro-Electrode Array (MEA) technology is known to provide an assessment of compound effects on neural 2D cell cultures and acute tissue preparations by real-time, non-invasive, and long-lasting electrophysiological monitoring of spontaneous and evoked neuronal activity. Nevertheless, the use of engineered 3D neural tissues in combination with MEA biochips still involves series of constraints, such as drastically limited diffusion of oxygen and nutrients within tissues mainly due to the lack of vascularization. Therefore, 3D neural tissues are extremely sensitive to experimental conditions and require an adequately designed interface that provides optimal tissue survival conditions. A well-suited technique to overcome this issue is the combination of the Air-Liquid Interface (ALI) tissue culture method with the MEA technology. We have developed a full 3D neural tissue culture process and a data acquisition system composed of high-end electronics and novel MEA biochips based on porous, flexible, thin-film membranes integrating recording electrodes, named as "Strip-MEA," to allow the maintenance of an ALI around the 3D neural tissues. The main motivation of the porous MEA biochips development was the possibility to monitor and to study the electrical activity of 3D neural tissues under different recording configurations, (i) the Strip-MEA can be placed below a tissue, (ii) or by taking advantage of the ALI, be directly placed on top of the tissue, or finally, (iii) it can be embedded into a larger neural tissue generated by the fusion of two (or more) tissues placed on both sides of the Strip-MEA allowing the recording from its inner part. This paper presents the recording and analyses of spontaneous activity from the three positioning configurations of the Strip-MEAs. Obtained results are discussed with the perspective of developing in vitro models of brain diseases and/or impairment of neural network functioning.
Collapse
Affiliation(s)
| | | | | | | | - Adrien Roux
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| |
Collapse
|
28
|
Zhao Q, Gribkova E, Shen Y, Cui J, Naughton N, Liu L, Seo J, Tong B, Gazzola M, Gillette R, Zhao H. Highly stretchable and customizable microneedle electrode arrays for intramuscular electromyography. SCIENCE ADVANCES 2024; 10:eadn7202. [PMID: 38691612 PMCID: PMC11062587 DOI: 10.1126/sciadv.adn7202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/29/2024] [Indexed: 05/03/2024]
Abstract
Stretchable three-dimensional (3D) penetrating microelectrode arrays have potential utility in various fields, including neuroscience, tissue engineering, and wearable bioelectronics. These 3D microelectrode arrays can penetrate and conform to dynamically deforming tissues, thereby facilitating targeted sensing and stimulation of interior regions in a minimally invasive manner. However, fabricating custom stretchable 3D microelectrode arrays presents material integration and patterning challenges. In this study, we present the design, fabrication, and applications of stretchable microneedle electrode arrays (SMNEAs) for sensing local intramuscular electromyography signals ex vivo. We use a unique hybrid fabrication scheme based on laser micromachining, microfabrication, and transfer printing to enable scalable fabrication of individually addressable SMNEA with high device stretchability (60 to 90%). The electrode geometries and recording regions, impedance, array layout, and length distribution are highly customizable. We demonstrate the use of SMNEAs as bioelectronic interfaces in recording intramuscular electromyography from various muscle groups in the buccal mass of Aplysia.
Collapse
Affiliation(s)
- Qinai Zhao
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
- Center for Advanced Manufacturing, University of Southern California, Los Angeles, CA, USA
| | - Ekaterina Gribkova
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Coordinated Science Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Yiyang Shen
- Center for Advanced Manufacturing, University of Southern California, Los Angeles, CA, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Jilai Cui
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Coordinated Science Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Noel Naughton
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Liangshu Liu
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
- Center for Advanced Manufacturing, University of Southern California, Los Angeles, CA, USA
| | - Jaemin Seo
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
- Center for Advanced Manufacturing, University of Southern California, Los Angeles, CA, USA
| | - Baixin Tong
- Center for Advanced Manufacturing, University of Southern California, Los Angeles, CA, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Mattia Gazzola
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Rhanor Gillette
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hangbo Zhao
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
- Center for Advanced Manufacturing, University of Southern California, Los Angeles, CA, USA
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
29
|
Spitz S, Schobesberger S, Brandauer K, Ertl P. Sensor-integrated brain-on-a-chip platforms: Improving the predictive validity in neurodegenerative research. Bioeng Transl Med 2024; 9:e10604. [PMID: 38818126 PMCID: PMC11135156 DOI: 10.1002/btm2.10604] [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: 07/05/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 06/01/2024] Open
Abstract
Affecting millions of individuals worldwide, neurodegenerative diseases (NDDs) pose a significant and growing health concern in people over the age of 60 years. Contributing to this trend are the steady increase in the aging population coupled with a persistent lack of disease-altering treatment strategies targeting NDDs. The absence of efficient therapeutics can be attributed to high failure rates in clinical trials and the ineptness of animal models in preceding preclinical studies. To that end, in recent years, significant research effort has been dedicated to the development of human cell-based preclinical disease models characterized by a higher degree of predictive validity. However, a key requirement of any in vitro model constitutes the precise knowledge and replication of the target tissues' (patho-)physiological microenvironment. Herein, microphysiological systems have demonstrated superiority over conventional static 2D/3D in vitro cell culture systems, as they allow for the emulation and continuous monitoring of the onset, progression, and remission of disease-associated phenotypes. This review provides an overview of recent advances in the field of NDD research using organ-on-a-chip platforms. Specific focus is directed toward non-invasive sensing strategies encompassing electrical, electrochemical, and optical sensors. Additionally, promising on- and integrable off-chip sensing strategies targeting key analytes in NDDs will be presented and discussed in detail.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
- Present address:
Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | | | - Peter Ertl
- Faculty of Technical ChemistryVienna University of TechnologyViennaAustria
| |
Collapse
|
30
|
Liu X, Gong Y, Jiang Z, Stevens T, Li W. Flexible high-density microelectrode arrays for closed-loop brain-machine interfaces: a review. Front Neurosci 2024; 18:1348434. [PMID: 38686330 PMCID: PMC11057246 DOI: 10.3389/fnins.2024.1348434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/12/2024] [Indexed: 05/02/2024] Open
Abstract
Flexible high-density microelectrode arrays (HDMEAs) are emerging as a key component in closed-loop brain-machine interfaces (BMIs), providing high-resolution functionality for recording, stimulation, or both. The flexibility of these arrays provides advantages over rigid ones, such as reduced mismatch between interface and tissue, resilience to micromotion, and sustained long-term performance. This review summarizes the recent developments and applications of flexible HDMEAs in closed-loop BMI systems. It delves into the various challenges encountered in the development of ideal flexible HDMEAs for closed-loop BMI systems and highlights the latest methodologies and breakthroughs to address these challenges. These insights could be instrumental in guiding the creation of future generations of flexible HDMEAs, specifically tailored for use in closed-loop BMIs. The review thoroughly explores both the current state and prospects of these advanced arrays, emphasizing their potential in enhancing BMI technology.
Collapse
Affiliation(s)
- Xiang Liu
- Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, United States
| | - Yan Gong
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, United States
| | - Zebin Jiang
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, United States
| | - Trevor Stevens
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, United States
| | - Wen Li
- Neuroscience Program, Department of Physiology, Michigan State University, East Lansing, MI, United States
- Institute for Quantitative Health Science and Engineering (IQ), East Lansing, MI, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
31
|
Heesen SH, Köhr G. GABAergic interneuron diversity and organization are crucial for the generation of human-specific functional neural networks in cerebral organoids. Front Cell Neurosci 2024; 18:1389335. [PMID: 38665372 PMCID: PMC11044699 DOI: 10.3389/fncel.2024.1389335] [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: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
This mini review investigates the importance of GABAergic interneurons for the network function of human-induced pluripotent stem cells (hiPSC)-derived brain organoids. The presented evidence suggests that the abundance, diversity and three-dimensional cortical organization of GABAergic interneurons are the primary elements responsible for the creation of synchronous neuronal firing patterns. Without intricate inhibition, coupled oscillatory patterns cannot reach a sufficient complexity to transfer spatiotemporal information constituting physiological network function. Furthermore, human-specific brain network function seems to be mediated by a more complex and interconnected inhibitory structure that remains developmentally flexible for a longer period when compared to rodents. This suggests that several characteristics of human brain networks cannot be captured by rodent models, emphasizing the need for model systems like organoids that adequately mimic physiological human brain function in vitro.
Collapse
Affiliation(s)
- Sebastian H. Heesen
- Molecular and Behavioural Neurobiology, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Georg Köhr
- Department of Neurophysiology, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany
- Physiology of Neural Networks, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
32
|
Abu Shihada J, Jung M, Decke S, Koschinski L, Musall S, Rincón Montes V, Offenhäusser A. Highly Customizable 3D Microelectrode Arrays for In Vitro and In Vivo Neuronal Tissue Recordings. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305944. [PMID: 38240370 PMCID: PMC10987114 DOI: 10.1002/advs.202305944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/05/2023] [Indexed: 02/16/2024]
Abstract
Planar microelectrode arrays (MEAs) for - in vitro or in vivo - neuronal signal recordings lack the spatial resolution and sufficient signal-to-noise ratio (SNR) required for a detailed understanding of neural network function and synaptic plasticity. To overcome these limitations, a highly customizable three-dimensional (3D) printing process is used in combination with thin film technology and a self-aligned template-assisted electrochemical deposition process to fabricate 3D-printed-based MEAs on stiff or flexible substrates. Devices with design flexibility and physical robustness are shown for recording neural activity in different in vitro and in vivo applications, achieving high-aspect ratio 3D microelectrodes of up to 33:1. Here, MEAs successfully record neural activity in 3D neuronal cultures, retinal explants, and the cortex of living mice, thereby demonstrating the versatility of the 3D MEA while maintaining high-quality neural recordings. Customizable 3D MEAs provide unique opportunities to study neural activity under regular or various pathological conditions, both in vitro and in vivo, and contribute to the development of drug screening and neuromodulation systems that can accurately monitor the activity of large neural networks over time.
Collapse
Affiliation(s)
- J. Abu Shihada
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
- RWTH Aachen University52062AachenGermany
| | - M. Jung
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
- RWTH Aachen University52062AachenGermany
| | - S. Decke
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
| | - L. Koschinski
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
- RWTH Aachen University52062AachenGermany
- Helmholtz Nano Facility (HNF)Forschungszentrum Jülich52428JülichGermany
| | - S. Musall
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
- RWTH Aachen University52062AachenGermany
- Faculty of MedicineInstitute of Experimental Epileptology and Cognition ResearchUniversity of Bonn53127BonnGermany
- University Hospital Bonn53127BonnGermany
| | - V. Rincón Montes
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
| | - A. Offenhäusser
- Institute of Biological Information Processing (IBI‐3) – BioelectronicsForschungszentrum52428JülichGermany
| |
Collapse
|
33
|
Jones PD, Molina-Martínez B, Niedworok A, Cesare P. A microphysiological system for parallelized morphological and electrophysiological read-out of 3D neuronal cell culture. LAB ON A CHIP 2024; 24:1750-1761. [PMID: 38348692 DOI: 10.1039/d3lc00963g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Three-dimensional in vitro models in microfluidic systems are promising tools for studying cell biology, with complex models using multiple cell types combined with high resolution imaging. Neuronal models demand electrical readout of the activity of networks of single neurons, yet classical planar microelectrode arrays struggle to capture extracellular action potentials when neural soma are suspended distant from the microelectrodes. This study introduces sophisticated microfluidic microelectrode arrays, specifically tailored for electrophysiology of 3D neuronal cultures. Using multilayer photolithography of permanent epoxy photoresists, we developed devices having 12 independent culture modules in a convenient format. Each module has two adjacent compartments for hydrogel-based 3D cell culture, with tunnels allowing projection of neurites between compartments. Microelectrodes integrated in the tunnels record action potentials as they pass between the compartments. Mesh ceilings separate the compartments from overlying wells, allowing for simple cell seeding and later nutrient, gas and waste exchange and application of test substances. Using these devices, we have demonstrated 3D neuronal culture, including electrophysiological recording and live imaging. This microphysiological platform will enable high-throughput investigation of neuronal networks for investigation of neurological disorders, neural pharmacology and basic neuroscience. Further models could include cocultures representing multiple brain regions or innervation models of other organs.
Collapse
Affiliation(s)
- Peter D Jones
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany.
| | - Beatriz Molina-Martínez
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany.
| | - Anita Niedworok
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany.
| | - Paolo Cesare
- NMI Natural and Medical Sciences Institute at the University of Tübingen, 72770 Reutlingen, Germany.
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| |
Collapse
|
34
|
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
|
35
|
Zhang X, Dou Z, Kim SH, Upadhyay G, Havert D, Kang S, Kazemi K, Huang K, Aydin O, Huang R, Rahman S, Ellis‐Mohr A, Noblet HA, Lim KH, Chung HJ, Gritton HJ, Saif MTA, Kong HJ, Beggs JM, Gazzola M. Mind In Vitro Platforms: Versatile, Scalable, Robust, and Open Solutions to Interfacing with Living Neurons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306826. [PMID: 38161217 PMCID: PMC10953569 DOI: 10.1002/advs.202306826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Motivated by the unexplored potential of in vitro neural systems for computing and by the corresponding need of versatile, scalable interfaces for multimodal interaction, an accurate, modular, fully customizable, and portable recording/stimulation solution that can be easily fabricated, robustly operated, and broadly disseminated is presented. This approach entails a reconfigurable platform that works across multiple industry standards and that enables a complete signal chain, from neural substrates sampled through micro-electrode arrays (MEAs) to data acquisition, downstream analysis, and cloud storage. Built-in modularity supports the seamless integration of electrical/optical stimulation and fluidic interfaces. Custom MEA fabrication leverages maskless photolithography, favoring the rapid prototyping of a variety of configurations, spatial topologies, and constitutive materials. Through a dedicated analysis and management software suite, the utility and robustness of this system are demonstrated across neural cultures and applications, including embryonic stem cell-derived and primary neurons, organotypic brain slices, 3D engineered tissue mimics, concurrent calcium imaging, and long-term recording. Overall, this technology, termed "mind in vitro" to underscore the computing inspiration, provides an end-to-end solution that can be widely deployed due to its affordable (>10× cost reduction) and open-source nature, catering to the expanding needs of both conventional and unconventional electrophysiology.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Zhi Dou
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Seung Hyun Kim
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Gaurav Upadhyay
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Daniel Havert
- Department of PhysicsIndiana University BloomingtonBloomingtonIN47405USA
| | - Sehong Kang
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Kimia Kazemi
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Kai‐Yu Huang
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Onur Aydin
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Raymond Huang
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Saeedur Rahman
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Austin Ellis‐Mohr
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Hayden A. Noblet
- Molecular and Integrative PhysiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Neuroscience ProgramUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Ki H. Lim
- Molecular and Integrative PhysiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Hee Jung Chung
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Molecular and Integrative PhysiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Neuroscience ProgramUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Howard J. Gritton
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Department of Comparative BiosciencesUniversity of Illinois at Urbana–ChampaignUrbanaIL61802USA
| | - M. Taher A. Saif
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - Hyun Joon Kong
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| | - John M. Beggs
- Department of PhysicsIndiana University BloomingtonBloomingtonIN47405USA
| | - Mattia Gazzola
- Carl R. Woese Institute for Genomic BiologyUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
- Department of Mechanical Science and EngineeringUniversity of Illinois at Urbana–ChampaignUrbanaIL61801USA
| |
Collapse
|
36
|
Park YG, Kwon YW, Koh CS, Kim E, Lee DH, Kim S, Mun J, Hong YM, Lee S, Kim JY, Lee JH, Jung HH, Cheon J, Chang JW, Park JU. In-vivo integration of soft neural probes through high-resolution printing of liquid electronics on the cranium. Nat Commun 2024; 15:1772. [PMID: 38413568 PMCID: PMC10899244 DOI: 10.1038/s41467-024-45768-0] [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: 12/07/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Current soft neural probes are still operated by bulky, rigid electronics mounted to a body, which deteriorate the integrity of the device to biological systems and restrict the free behavior of a subject. We report a soft, conformable neural interface system that can monitor the single-unit activities of neurons with long-term stability. The system implements soft neural probes in the brain, and their subsidiary electronics which are directly printed on the cranial surface. The high-resolution printing of liquid metals forms soft neural probes with a cellular-scale diameter and adaptable lengths. Also, the printing of liquid metal-based circuits and interconnections along the curvature of the cranium enables the conformal integration of electronics to the body, and the cranial circuit delivers neural signals to a smartphone wirelessly. In the in-vivo studies using mice, the system demonstrates long-term recording (33 weeks) of neural activities in arbitrary brain regions. In T-maze behavioral tests, the system shows the behavior-induced activation of neurons in multiple brain regions.
Collapse
Affiliation(s)
- Young-Geun Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Yong Won Kwon
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Chin Su Koh
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Enji Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Dong Ha Lee
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Sumin Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Jongmin Mun
- Department of Statistics and Data Science, Yonsei University, Seoul, 03722, South Korea
| | - Yeon-Mi Hong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Sanghoon Lee
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
| | - Ju-Young Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, South Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, South Korea
| | - Hyun Ho Jung
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, South Korea.
- Department of Chemistry, Yonsei University, Seoul, 03722, South Korea.
| | - Jin Woo Chang
- Department of Neurosurgery, Korea University Anam Hospital, Seoul, 02841, South Korea.
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, South Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, South Korea.
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, South Korea.
| |
Collapse
|
37
|
Wang Y, Chen Z, Davis B, Lipman W, Xing S, Zhang L, Wang T, Hafiz P, Xie W, Yan Z, Huang Z, Song J, Bai W. Digital automation of transdermal drug delivery with high spatiotemporal resolution. Nat Commun 2024; 15:511. [PMID: 38218967 PMCID: PMC10787768 DOI: 10.1038/s41467-023-44532-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/18/2023] [Indexed: 01/15/2024] Open
Abstract
Transdermal drug delivery is of vital importance for medical treatments. However, user adherence to long-term repetitive drug delivery poses a grand challenge. Furthermore, the dynamic and unpredictable disease progression demands a pharmaceutical treatment that can be actively controlled in real-time to ensure medical precision and personalization. Here, we report a spatiotemporal on-demand patch (SOP) that integrates drug-loaded microneedles with biocompatible metallic membranes to enable electrically triggered active control of drug release. Precise control of drug release to targeted locations (<1 mm2), rapid drug release response to electrical triggers (<30 s), and multi-modal operation involving both drug release and electrical stimulation highlight the novelty. Solution-based fabrication ensures high customizability and scalability to tailor the SOP for various pharmaceutical needs. The wireless-powered and digital-controlled SOP demonstrates great promise in achieving full automation of drug delivery, improving user adherence while ensuring medical precision. Based on these characteristics, we utilized SOPs in sleep studies. We revealed that programmed release of exogenous melatonin from SOPs improve sleep of mice, indicating potential values for basic research and clinical treatments.
Collapse
Affiliation(s)
- Yihang Wang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zeka Chen
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brayden Davis
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Will Lipman
- Department of Psychology and Neuroscience, University of North Carolina at chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sicheng Xing
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Lin Zhang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tian Wang
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Priyash Hafiz
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Wanrong Xie
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zijie Yan
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhili Huang
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Wubin Bai
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
38
|
Buentello DC, Garcia-Corral M, Trujillo-de Santiago G, Alvarez MM. Neuron(s)-on-a-Chip: A Review of the Design and Use of Microfluidic Systems for Neural Tissue Culture. IEEE Rev Biomed Eng 2024; 17:243-263. [PMID: 36301779 DOI: 10.1109/rbme.2022.3217486] [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: 11/06/2022]
Abstract
Neuron-on-chip (NoC) systems-microfluidic devices in which neurons are cultured-have become a promising alternative to replace or minimize the use of animal models and have greatly facilitated in vitro research. Here, we review and discuss current developments in neuron-on-chip platforms, with a particular emphasis on existing biological models, culturing techniques, biomaterials, and topologies. We also discuss how the architecture, flow, and gradients affect neuronal growth, differentiation, and development. Finally, we discuss some of the most recent applications of NoCs in fundamental research (i.e., studies on the effects of electrical, mechanical/topological, or chemical stimuli) and in disease modeling.
Collapse
|
39
|
Lam D, Enright HA, Cadena J, George VK, Soscia DA, Tooker AC, Triplett M, Peters SKG, Karande P, Ladd A, Bogguri C, Wheeler EK, Fischer NO. Spatiotemporal analysis of 3D human iPSC-derived neural networks using a 3D multi-electrode array. Front Cell Neurosci 2023; 17:1287089. [PMID: 38026689 PMCID: PMC10679684 DOI: 10.3389/fncel.2023.1287089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
While there is a growing appreciation of three-dimensional (3D) neural tissues (i.e., hydrogel-based, organoids, and spheroids), shown to improve cellular health and network activity to mirror brain-like activity in vivo, functional assessment using current electrophysiology techniques (e.g., planar multi-electrode arrays or patch clamp) has been technically challenging and limited to surface measurements at the bottom or top of the 3D tissue. As next-generation MEAs, specifically 3D MEAs, are being developed to increase the spatial precision across all three dimensions (X, Y, Z), development of improved computational analytical tools to discern region-specific changes within the Z dimension of the 3D tissue is needed. In the present study, we introduce a novel computational analytical pipeline to analyze 3D neural network activity recorded from a "bottom-up" 3D MEA integrated with a 3D hydrogel-based tissue containing human iPSC-derived neurons and primary astrocytes. Over a period of ~6.5 weeks, we describe the development and maturation of 3D neural activity (i.e., features of spiking and bursting activity) within cross sections of the 3D tissue, based on the vertical position of the electrode on the 3D MEA probe, in addition to network activity (identified using synchrony analysis) within and between cross sections. Then, using the sequential addition of postsynaptic receptor antagonists, bicuculline (BIC), 2-amino-5-phosphonovaleric acid (AP-5), and 6-cyano-5-nitroquinoxaline-2,3-dione (CNQX), we demonstrate that networks within and between cross sections of the 3D hydrogel-based tissue show a preference for GABA and/or glutamate synaptic transmission, suggesting differences in the network composition throughout the neural tissue. The ability to monitor the functional dynamics of the entire 3D reconstructed neural tissue is a critical bottleneck; here we demonstrate a computational pipeline that can be implemented in studies to better interpret network activity within an engineered 3D neural tissue and have a better understanding of the modeled organ tissue.
Collapse
Affiliation(s)
- Doris Lam
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Heather A. Enright
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jose Cadena
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Vivek Kurien George
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - David A. Soscia
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Angela C. Tooker
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Michael Triplett
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Sandra K. G. Peters
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Piyush Karande
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Alexander Ladd
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Chandrakumar Bogguri
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Elizabeth K. Wheeler
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Nicholas O. Fischer
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| |
Collapse
|
40
|
Lee JM, Pyo YW, Kim YJ, Hong JH, Jo Y, Choi W, Lin D, Park HG. The ultra-thin, minimally invasive surface electrode array NeuroWeb for probing neural activity. Nat Commun 2023; 14:7088. [PMID: 37925553 PMCID: PMC10625630 DOI: 10.1038/s41467-023-42860-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/24/2023] [Indexed: 11/06/2023] Open
Abstract
Electrophysiological recording technologies can provide valuable insights into the functioning of the central and peripheral nervous systems. Surface electrode arrays made of soft materials or implantable multi-electrode arrays with high electrode density have been widely utilized as neural probes. However, neither of these probe types can simultaneously achieve minimal invasiveness and robust neural signal detection. Here, we present an ultra-thin, minimally invasive neural probe (the "NeuroWeb") consisting of hexagonal boron nitride and graphene, which leverages the strengths of both surface electrode array and implantable multi-electrode array. The NeuroWeb open lattice structure with a total thickness of 100 nm demonstrates high flexibility and strong adhesion, establishing a conformal and tight interface with the uneven mouse brain surface. In vivo electrophysiological recordings show that NeuroWeb detects stable single-unit activity of neurons with high signal-to-noise ratios. Furthermore, we investigate neural interactions between the somatosensory cortex and the cerebellum using transparent dual NeuroWebs and optical stimulation, and measure the times of neural signal transmission between the brain regions depending on the pathway. Therefore, NeuroWeb can be expected to pave the way for understanding complex brain networks with optical and electrophysiological mapping of the brain.
Collapse
Affiliation(s)
- Jung Min Lee
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young-Woo Pyo
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Physics, Korea University, Seoul, 02841, Republic of Korea
| | - Yeon Jun Kim
- Department of Physics, Korea University, Seoul, 02841, Republic of Korea
| | - Jin Hee Hong
- Department of Physics, Korea University, Seoul, 02841, Republic of Korea
- Center for Molecular Spectroscopy and Dynamics, Institute for Basic Science, Seoul, 02841, Republic of Korea
| | - Yonghyeon Jo
- Department of Physics, Korea University, Seoul, 02841, Republic of Korea
- Center for Molecular Spectroscopy and Dynamics, Institute for Basic Science, Seoul, 02841, Republic of Korea
| | - Wonshik Choi
- Department of Physics, Korea University, Seoul, 02841, Republic of Korea
- Center for Molecular Spectroscopy and Dynamics, Institute for Basic Science, Seoul, 02841, Republic of Korea
| | - Dingchang Lin
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Hong-Gyu Park
- Department of Physics and Astronomy, and Institute of Applied Physics, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
41
|
Marino A, Battaglini M, Lefevre MC, Ceccarelli MC, Ziaja K, Ciofani G. Sensorization of microfluidic brain-on-a-chip devices: Towards a new generation of integrated drug screening systems. Trends Analyt Chem 2023; 168:117319. [PMID: 37915756 PMCID: PMC7615229 DOI: 10.1016/j.trac.2023.117319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Brain-on-a-chip (BoC) devices show typical characteristics of brain complexity, including the presence of different cell types, separation in different compartments, tissue-like three-dimensionality, and inclusion of the extracellular matrix components. Moreover, the incorporation of a vascular system mimicking the blood-brain barrier (BBB) makes BoC particularly attractive, since they can be exploited to test the brain delivery of different drugs and nanoformulations. In this review, we introduce the main innovations in BoC and BBB-on-a-chip models, especially focusing sensorization: electrical, electrochemical, and optical biosensors permit the real-time monitoring of different biological phenomena and markers, such as the release of growth factors, the expression of specific receptors/biomarkers, the activation of immune cells, cell viability, cell-cell interactions, and BBB crossing of drugs and nanoparticles. The recent improvements in signal amplification, miniaturization, and multiplication of the sensors are discussed in an effort to highlight their benefits versus limitations and delineate future challenges in this field.
Collapse
Affiliation(s)
- Attilio Marino
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Matteo Battaglini
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Marie Celine Lefevre
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Maria Cristina Ceccarelli
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
- Scuola Superiore Sant’Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| | - Kamil Ziaja
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
- Scuola Superiore Sant’Anna, The Biorobotics Institute, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
- University of Aveiro, Department of Chemistry, CICECO-Aveiro Institute of Materials, Rua de Calouste Gulbenkian 1, 3810-074, Aveiro, Portugal
| | - Gianni Ciofani
- Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, 56025, Pontedera, Italy
| |
Collapse
|
42
|
Yao Y, Coleman HA, Meagher L, Forsythe JS, Parkington HC. 3D Functional Neuronal Networks in Free-Standing Bioprinted Hydrogel Constructs. Adv Healthc Mater 2023; 12:e2300801. [PMID: 37369123 DOI: 10.1002/adhm.202300801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 06/29/2023]
Abstract
The composition, elasticity, and organization of the extracellular matrix within the central nervous system contribute to the architecture and function of the brain. From an in vitro modeling perspective, soft biomaterials are needed to mimic the 3D neural microenvironments. While many studies have investigated 3D culture and neural network formation in bulk hydrogel systems, these approaches have limited ability to position cells to mimic sophisticated brain architectures. In this study, cortical neurons and astrocytes acutely isolated from the brains of rats are bioprinted in a hydrogel to form 3D neuronal constructs. Successful bioprinting of cellular and acellular strands in a multi-bioink approach allows the subsequent formation of gray- and white-matter tracts reminiscent of cortical structures. Immunohistochemistry shows the formation of dense, 3D axon networks. Calcium signaling and extracellular electrophysiology in these 3D neuronal networks confirm spontaneous activity in addition to evoked activities under pharmacological and electrical stimulation. The system and bioprinting approaches are capable of fabricating soft, free-standing neuronal structures of different bioink and cell types with high resolution and throughput, which provide a promising platform for understanding fundamental questions of neural networks, engineering neuromorphic circuits, and for in vitro drug screening.
Collapse
Affiliation(s)
- Yue Yao
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- School of Physics, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Harold A Coleman
- Department of Physiology, Monash University, Clayton, VIC, 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | - John S Forsythe
- Department of Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC, 3800, Australia
| | | |
Collapse
|
43
|
Bartlett M, He M, Ranke D, Wang Y, Cohen-Karni T. A snapshot review on materials enabled multimodal bioelectronics for neurological and cardiac research. MRS ADVANCES 2023; 8:1047-1060. [PMID: 38283671 PMCID: PMC10812139 DOI: 10.1557/s43580-023-00645-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/08/2023] [Indexed: 01/30/2024]
Abstract
Seamless integration of the body and electronics toward the understanding, quantification, and control of disease states remains one of the grand scientific challenges of this era. As such, research efforts have been dedicated to developing bioelectronic devices for chemical, mechanical, and electrical sensing, and cellular and tissue functionality modulation. The technologies developed to achieve these capabilities cross a wide range of materials and scale (and dimensionality), e.g., from micrometer to centimeters (from 2-dimensional (2D) to 3-dimensional (3D) assemblies). The integration into multimodal systems which allow greater insight and control into intrinsically multifaceted biological systems requires careful design and selection. This snapshot review will highlight the state-of-the-art in cellular recording and modulation as well as the material considerations for the design and manufacturing of devices integrating their capabilities.
Collapse
Affiliation(s)
- Mabel Bartlett
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Mengdi He
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Ranke
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Yingqiao Wang
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Tzahi Cohen-Karni
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
44
|
Yao X, Kang JH, Kim KP, Shin H, Jin ZL, Guo H, Xu YN, Li YH, Hali S, Kwon J, La H, Park C, Kim YJ, Wang L, Hong K, Cao Q, Cho IJ, Kim NH, Han DW. Production of Highly Uniform Midbrain Organoids from Human Pluripotent Stem Cells. Stem Cells Int 2023; 2023:3320211. [PMID: 37810631 PMCID: PMC10558263 DOI: 10.1155/2023/3320211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/05/2023] [Accepted: 08/24/2023] [Indexed: 10/10/2023] Open
Abstract
Brain organoids have been considered as an advanced platform for in vitro disease modeling and drug screening, but numerous roadblocks exist, such as lack of large-scale production technology and lengthy protocols with multiple manipulation steps, impeding the industrial translation of brain organoid technology. Here, we describe the high-speed and large-scale production of midbrain organoids using a high-throughput screening-compatible platform within 30 days. Micro midbrain organoids (µMOs) exhibit a highly uniform morphology and gene expression pattern with minimal variability. Notably, µMOs show dramatically accelerated maturation, resulting in the generation of functional µMOs within only 30 days of differentiation. Furthermore, individual µMOs display highly consistent responsiveness to neurotoxin, suggesting their usefulness as an in vitro high-throughput drug toxicity screening platform. Collectively, our data indicate that µMO technology could represent an advanced and robust platform for in vitro disease modeling and drug screening for human neuronal diseases.
Collapse
Affiliation(s)
- Xuerui Yao
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development Department, Qingdao Haier Biotech Co. Ltd., Qingdao, China
| | - Ji Hyun Kang
- Laboratory of Stem Cells and Organoids, OrganFactory Co. Ltd., Cheongju 28864, Republic of Korea
| | - Kee-Pyo Kim
- Department of Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Zhe-Long Jin
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Hao Guo
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Sai Hali
- Institute of Ophthalmology, University College London, London, UK
| | - Jeongwoo Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Hyeonwoo La
- Department of Stem Cell and Regenerative Biotechnology, The Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029, Republic of Korea
| | - Chanhyeok Park
- Department of Stem Cell and Regenerative Biotechnology, The Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029, Republic of Korea
| | - Yong-June Kim
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Department of Urology, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Lin Wang
- Research and Development Department, Qingdao Haier Biotech Co. Ltd., Qingdao, China
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, The Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029, Republic of Korea
| | - Qilong Cao
- Research and Development Department, Qingdao Haier Biotech Co. Ltd., Qingdao, China
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development Department, Qingdao Haier Biotech Co. Ltd., Qingdao, China
- Laboratory of Stem Cells and Organoids, OrganFactory Co. Ltd., Cheongju 28864, Republic of Korea
| | - Dong Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development Department, Qingdao Haier Biotech Co. Ltd., Qingdao, China
- Laboratory of Stem Cells and Organoids, OrganFactory Co. Ltd., Cheongju 28864, Republic of Korea
| |
Collapse
|
45
|
Shang X, Ling W, Chen Y, Li C, Huang X. Construction of a Flexible Optogenetic Device for Multisite and Multiregional Optical Stimulation Through Flexible µ-LED Displays on the Cerebral Cortex. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302241. [PMID: 37260144 DOI: 10.1002/smll.202302241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/14/2023] [Indexed: 06/02/2023]
Abstract
Precisely delivering light to multiple locations in biological tissue is crucial for advancing multiregional optogenetics in neuroscience research. However, conventional implantable devices typically have rigid geometries and limited light sources, allowing only single or dual probe placement with fixed spacing. Here, a fully flexible optogenetic device with multiple thin-film microscale light-emitting diode (µ-LED) displays scattering from a central controller is presented. Each display is heterogeneously integrated with thin-film 5 × 10 µ-LEDs and five optical fibers 125 µm in diameter to achieve cellular-scale spatial resolution. Meanwhile, the device boasts a compact, flexible circuit capable of multichannel configuration and wireless transmission, with an overall weight of 1.31 g, enabling wireless, real-time neuromodulation of freely moving rats. Characterization results and finite element analysis have demonstrated excellent optical properties and mechanical stability, while cytotoxicity tests further ensure the biocompatibility of the device for implantable applications. Behavior studies under optogenetic modulation indicate great promise for wirelessly modulating neural functions in freely moving animals. The device with multisite and multiregional optogenetic modulation capability offers a comprehensive platform to advance both fundamental neuroscience studies and potential applications in brain-computer interfaces.
Collapse
Affiliation(s)
- Xue Shang
- Department of Biomedical Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, China
| | - Wei Ling
- Department of Biomedical Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, China
- Research Center for Augmented Intelligence, Research Institute of Artificial Intelligence, Zhejiang Laboratory, Hangzhou, 311100, China
| | - Ying Chen
- Institute of Flexible Electronic Technology of Tsinghua, Jiaxing, 314006, China
- Jiaxing Key Laboratory of Flexible Electronics based Intelligent Sensing and Advanced Manufacturing Technology, Jiaxing, 314000, China
| | - Chenxi Li
- State Key Laboratory of Precision Measuring Technology and Instruments, Tianjin University, 92 Weijin Road, Tianjin, 300072, China
| | - Xian Huang
- Department of Biomedical Engineering, Tianjin University, 92 Weijin Road, Tianjin, 300072, China
- Institute of Wearable Technology and Bioelectronics, Qiantang Science and Technology Innovation Center, 1002 23rd Street, Hangzhou, 310018, China
| |
Collapse
|
46
|
Phouphetlinthong O, Partiot E, Bernou C, Sebban A, Gaudin R, Charlot B. Protruding cantilever microelectrode array to monitor the inner electrical activity of cerebral organoids. LAB ON A CHIP 2023; 23:3603-3614. [PMID: 37489118 DOI: 10.1039/d3lc00294b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Stem cell-derived cerebral organoids are artificially grown miniature organ-like structures mimicking embryonic brain architecture. They are composed of multiple neural cell types with 3D cell layer organization exhibiting local field potential. Measuring the extracellular electrical activity by means of conventional planar microelectrode arrays is particularly challenging due to the 3D architecture of organoids. In order to monitor the intra-organoid electrical activity of thick spheroid-shaped samples, we developed long protruding microelectrode arrays able to penetrate the inner regions of cerebral organoids to measure the local potential of neurons within the organoids. A new microfabrication process has been developed which, thanks to the relaxation of internal stresses of a stack of materials deposited over a sacrificial layer, allows one to build a protruding cantilever microelectrode array placed at the apex of beams which rise vertically, over two hundred microns. These slender beams inserted deeply into the organoids give access to the recording of local field potential from neurons buried inside the organoid. This novel device shall provide valuable tools to study neural functions in greater detail.
Collapse
Affiliation(s)
- Oramany Phouphetlinthong
- IES, Institut d'Electronique et des Systèmes, UMR 5214 CNRS, Montpellier, France
- University of Montpellier, Montpellier, France.
| | - Emma Partiot
- IRIM, Institut de Recherche en Infectiologie de Montpellier, UMR 9004 CNRS, Montpellier, France
- University of Montpellier, Montpellier, France.
| | - Corentin Bernou
- IRIM, Institut de Recherche en Infectiologie de Montpellier, UMR 9004 CNRS, Montpellier, France
- University of Montpellier, Montpellier, France.
| | - Audrey Sebban
- IES, Institut d'Electronique et des Systèmes, UMR 5214 CNRS, Montpellier, France
- University of Montpellier, Montpellier, France.
| | - Raphael Gaudin
- IRIM, Institut de Recherche en Infectiologie de Montpellier, UMR 9004 CNRS, Montpellier, France
- University of Montpellier, Montpellier, France.
| | - Benoit Charlot
- IES, Institut d'Electronique et des Systèmes, UMR 5214 CNRS, Montpellier, France
- University of Montpellier, Montpellier, France.
| |
Collapse
|
47
|
Dupuit V, Briançon-Marjollet A, Delacour C. Portrait of intense communications within microfluidic neural networks. Sci Rep 2023; 13:12306. [PMID: 37516789 PMCID: PMC10387102 DOI: 10.1038/s41598-023-39477-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023] Open
Abstract
In vitro model networks could provide cellular models of physiological relevance to reproduce and investigate the basic function of neural circuits on a chip in the laboratory. Several tools and methods have been developed since the past decade to build neural networks on a chip; among them, microfluidic circuits appear to be a highly promising approach. One of the numerous advantages of this approach is that it preserves stable somatic and axonal compartments over time due to physical barriers that prevent the soma from exploring undesired areas and guide neurites along defined pathways. As a result, neuron compartments can be identified and isolated, and their interconnectivity can be modulated to build a topological neural network (NN). Here, we have assessed the extent to which the confinement imposed by the microfluidic environment can impact cell development and shape NN activity. Toward that aim, microelectrode arrays have enabled the monitoring of the short- and mid-term evolution of neuron activation over the culture period at specific locations in organized (microfluidic) and random (control) networks. In particular, we have assessed the spike and burst rate, as well as the correlations between the extracted spike trains over the first stages of maturation. This study enabled us to observe intense neurite communications that would have been weaker and more delayed within random networks; the spiking rate, burst and correlations being reinforced over time in terms of number and amplitude, exceeding the electrophysiological features of standard cultures. Beyond the enhanced detection efficiency that was expected from the microfluidic channels, the confinement of cells seems to reinforce neural communications and cell development throughout the network.
Collapse
Affiliation(s)
- Victor Dupuit
- Institut Néel, University Grenoble Alpes, CNRS, Grenoble INP, 38000, Grenoble, France
| | - Anne Briançon-Marjollet
- HP2 Laboratory, University Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1300, Grenoble, France
| | - Cécile Delacour
- Institut Néel, University Grenoble Alpes, CNRS, Grenoble INP, 38000, Grenoble, France.
| |
Collapse
|
48
|
Zips S, Huang B, Hotte S, Hiendlmeier L, Wang C, Rajamani K, Buriez O, Al Boustani G, Chen Y, Wolfrum B, Yamada A. Aerosol Jet-Printed High-Aspect Ratio Micro-Needle Electrode Arrays Applied for Human Cerebral Organoids and 3D Neurospheroid Networks. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37469180 DOI: 10.1021/acsami.3c06210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
The human brain is a complex and poorly accessible organ. Thus, new tools are required for studying the neural function in a controllable environment that preserves multicellular interaction and neuronal wiring. In particular, high-throughput methods that alleviate the need for animal experiments are essential for future studies. Recent developments of induced pluripotent stem cell technologies have enabled in vitro modeling of the human brain by creating three-dimensional brain tissue mimic structures. To leverage these new technologies, a systematic and versatile approach for evaluating neuronal activity at larger tissue depths within the regime of tens to hundreds of micrometers is required. Here, we present an aerosol-jet- and inkjet-printing-based method to fabricate microelectrode arrays, equipped with high-aspect ratio μ-needle electrodes that penetrate 3D neural network assemblies. The arrays have been successfully applied for electrophysiological recordings on interconnected neurospheroids formed on an engineered substrate and on cerebral organoids, both derived from human induced pluripotent stem cells.
Collapse
Affiliation(s)
- Sabine Zips
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Boxin Huang
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Salammbô Hotte
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Lukas Hiendlmeier
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Chen Wang
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Karthyayani Rajamani
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Olivier Buriez
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - George Al Boustani
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Yong Chen
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Bernhard Wolfrum
- Neuroelectronics─Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 11, 85748 Garching, Germany
| | - Ayako Yamada
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| |
Collapse
|
49
|
Mullis AS, Kaplan DL. Functional bioengineered tissue models of neurodegenerative diseases. Biomaterials 2023; 298:122143. [PMID: 37146365 PMCID: PMC10209845 DOI: 10.1016/j.biomaterials.2023.122143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 04/27/2023] [Accepted: 05/01/2023] [Indexed: 05/07/2023]
Abstract
Aging-associated neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases remain poorly understood and no disease-modifying treatments exist despite decades of investigation. Predominant in vitro (e.g., 2D cell culture, organoids) and in vivo (e.g., mouse) models of these diseases are insufficient mimics of human brain tissue structure and function and of human neurodegenerative pathobiology, and have thus contributed to this collective translational failure. This has been a longstanding challenge in the field, and new strategies are required to address both fundamental and translational needs. Bioengineered tissue culture models constitute a class of promising alternatives, as they can overcome the low cell density, poor nutrient exchange, and long term culturability limitations of existing in vitro models. Further, they can reconstruct the structural, mechanical, and biochemical cues of native brain tissue, providing a better mimic of human brain tissues for in vitro pathobiological investigation and drug development. We discuss bioengineering techniques for the generation of these neurodegenerative tissue models, including biomaterials-, organoid-, and microfluidics-based approaches, and design considerations for their construction. To aid the development of the next generation of functional neurodegenerative disease models, we discuss approaches to incorporate greater cellular diversity and simulate aging processes within bioengineered brain tissues.
Collapse
Affiliation(s)
- Adam S Mullis
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA.
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA; Allen Discovery Center, Tufts University, Medford, MA, 02155, USA.
| |
Collapse
|
50
|
Lu B, Fan P, Li M, Wang Y, Liang W, Yang G, Mo F, Xu Z, Shan J, Song Y, Liu J, Wu Y, Cai X. Detection of neuronal defensive discharge information transmission and characteristics in periaqueductal gray double-subregions using PtNP/PEDOT:PSS modified microelectrode arrays. MICROSYSTEMS & NANOENGINEERING 2023; 9:70. [PMID: 37275263 PMCID: PMC10232427 DOI: 10.1038/s41378-023-00546-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/14/2023] [Accepted: 03/23/2023] [Indexed: 06/07/2023]
Abstract
Threatened animals respond with appropriate defensive behaviors to survive. It has been accepted that midbrain periaqueductal gray (PAG) plays an essential role in the circuitry system and organizes defensive behavioral responses. However, the role and correlation of different PAG subregions in the expression of different defensive behaviors remain largely unexplored. Here, we designed and manufactured a microelectrode array (MEA) to simultaneously detect the activities of dPAG and vPAG neurons in freely behaving rats. To improve the detection performance of the MEAs, PtNP/PEDOT:PSS nanocomposites were modified onto the MEAs. Subsequently, the predator odor was used to induce the rat's innate fear, and the changes and information transmission in neuronal activities were detected in the dPAG and vPAG. Our results showed that the dPAG and vPAG participated in innate fear, but the activation degree was distinct in different defense behaviors. During flight, neuronal responses were stronger and earlier in the dPAG than the vPAG, while vPAG neurons responded more strongly during freezing. By applying high-performance MEA, it was revealed that neural information spread from the activated dPAG to the weakly activated vPAG. Our research also revealed that dPAG and vPAG neurons exhibited different defensive discharge characteristics, and dPAG neurons participated in the regulation of defense responses with burst-firing patterns. The slow activation and continuous firing of vPAG neurons cooresponded with the regulation of long-term freezing responses. The results demonstrated the important role of PAG neuronal activities in controlling different aspects of defensive behaviors and provided novel insights for investigating defense from the electrophysiological perspective.
Collapse
Affiliation(s)
- Botao Lu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Penghui Fan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ming Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yiding Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Wei Liang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
| | - Gucheng Yang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Fan Mo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhaojie Xu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jin Shan
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yilin Song
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Yirong Wu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, 100190 China
- University of Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|