1
|
Yin J, Lees JG, Gong S, Nguyen JT, Phang RJ, Shi Q, Huang Y, Kong AM, Dyson JM, Lim SY, Cheng W. Real-time electro-mechanical profiling of dynamically beating human cardiac organoids by coupling resistive skins with microelectrode arrays. Biosens Bioelectron 2025; 267:116752. [PMID: 39276439 DOI: 10.1016/j.bios.2024.116752] [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: 06/05/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Cardiac organoids differentiated from induced pluripotent stem cells are emerging as a promising platform for pre-clinical drug screening, assessing cardiotoxicity, and disease modelling. However, it is challenging to simultaneously measure mechanical contractile forces and electrophysiological signals of cardiac organoids in real-time and in-situ with the existing methods. Here, we present a biting-inspired sensory system based on a resistive skin sensor and a microelectrode array. The bite-like contact can be established with a micromanipulator to precisely position the resistive skin sensor on the top of the cardiac organoid while the 3D microneedle electrode array probes from underneath. Such reliable contact is key to achieving simultaneous electro-mechanical measurements. We demonstrate the use of our system for modelling cardiotoxicity with the anti-cancer drug doxorubicin. The electro-mechanical parameters described here elucidate the acute cardiotoxic effects induced by doxorubicin. This integrated electro-mechanical system enables a suite of new diagnostic options for assessing cardiac organoids and tissues.
Collapse
Affiliation(s)
- Jialiang Yin
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Jarmon G Lees
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia; O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia
| | - Shu Gong
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - John Tan Nguyen
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Ren Jie Phang
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia
| | - Qianqian Shi
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Yifeng Huang
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Anne M Kong
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia
| | - Jennifer M Dyson
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Clayton, Victoria, 3800, Australia; Faculty of Engineering, Monash Institute of Medical Engineering (MIME), Monash University, Clayton, Victoria, 3800, Australia
| | - Shiang Y Lim
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia; O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia; Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Victoria, Monash University, Australia; National Heart Research Institute Singapore, National Heart Centre, Singapore
| | - Wenlong Cheng
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia; The Melbourne Centre for Nanofabrication, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
2
|
Chea M, Bouvier S, Gris JC. The hemostatic system in chronic brain diseases: A new challenging frontier? Thromb Res 2024; 243:109154. [PMID: 39305718 DOI: 10.1016/j.thromres.2024.109154] [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: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Neurological diseases (ND), including neurodegenerative diseases (NDD) and psychiatric disorders (PD), present a significant public health challenge, ranking third in Europe for disability and premature death, following cardiovascular diseases and cancers. In 2017, approximately 540 million cases of ND were reported among Europe's 925 million people, with strokes, dementia, and headaches being most prevalent. Nowadays, more and more evidence highlight the hemostasis critical role in cerebral homeostasis and vascular events. Indeed, hemostasis, thrombosis, and brain abnormalities contributing to ND form a complex and poorly understood equilibrium. Alterations in vascular biology, particularly involving the blood-brain barrier, are implicated in ND, especially dementia, and PD. While the roles of key coagulation players such as thrombin and fibrinogen are established, the roles of other hemostasis components are less clear. Moreover, the involvement of these elements in psychiatric disease pathogenesis is virtually unstudied, except in specific pathological models such as antiphospholipid syndrome. Advanced imaging techniques, primarily functional magnetic resonance imaging and its derivatives like diffusion tensor imaging, have been developed to study brain areas affected by ND and to improve our understanding of the pathophysiology of these diseases. This literature review aims to clarify the current understanding of the connections between hemostasis, thrombosis, and neurological diseases, as well as explore potential future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mathias Chea
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France.
| | - Sylvie Bouvier
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Jean-Christophe Gris
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France; I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
3
|
Cheng JL, Cook AL, Talbot J, Perry S. How is Excitotoxicity Being Modelled in iPSC-Derived Neurons? Neurotox Res 2024; 42:43. [PMID: 39405005 PMCID: PMC11480214 DOI: 10.1007/s12640-024-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Excitotoxicity linked either to environmental causes (pesticide and cyanotoxin exposure), excitatory neurotransmitter imbalance, or to intrinsic neuronal hyperexcitability, is a pathological mechanism central to neurodegeneration in amyotrophic lateral sclerosis (ALS). Investigation of excitotoxic mechanisms using in vitro and in vivo animal models has been central to understanding ALS mechanisms of disease. In particular, advances in induced pluripotent stem cell (iPSC) technologies now provide human cell-based models that are readily amenable to environmental and network-based excitotoxic manipulations. The cell-type specific differentiation of iPSC, combined with approaches to modelling excitotoxicity that include editing of disease-associated gene variants, chemogenetics, and environmental risk-associated exposures make iPSC primed to examine gene-environment interactions and disease-associated excitotoxic mechanisms. Critical to this is knowledge of which neurotransmitter receptor subunits are expressed by iPSC-derived neuronal cultures being studied, how their activity responds to antagonists and agonists of these receptors, and how to interpret data derived from multi-parameter electrophysiological recordings. This review explores how iPSC-based studies have contributed to our understanding of ALS-linked excitotoxicity and highlights novel approaches to inducing excitotoxicity in iPSC-derived neurons to further our understanding of its pathological pathways.
Collapse
Affiliation(s)
- Jan L Cheng
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Anthony L Cook
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Jana Talbot
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Bogguri C, George VK, Amiri B, Ladd A, Hum NR, Sebastian A, Enright HA, Valdez CA, Mundhenk TN, Cadena J, Lam D. Biphasic response of human iPSC-derived neural network activity following exposure to a sarin-surrogate nerve agent. Front Cell Neurosci 2024; 18:1378579. [PMID: 39301218 PMCID: PMC11410629 DOI: 10.3389/fncel.2024.1378579] [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: 01/29/2024] [Accepted: 08/16/2024] [Indexed: 09/22/2024] Open
Abstract
Organophosphorus nerve agents (OPNA) are hazardous environmental exposures to the civilian population and have been historically weaponized as chemical warfare agents (CWA). OPNA exposure can lead to several neurological, sensory, and motor symptoms that can manifest into chronic neurological illnesses later in life. There is still a large need for technological advancement to better understand changes in brain function following OPNA exposure. The human-relevant in vitro multi-electrode array (MEA) system, which combines the MEA technology with human stem cell technology, has the potential to monitor the acute, sub-chronic, and chronic consequences of OPNA exposure on brain activity. However, the application of this system to assess OPNA hazards and risks to human brain function remains to be investigated. In a concentration-response study, we have employed a human-relevant MEA system to monitor and detect changes in the electrical activity of engineered neural networks to increasing concentrations of the sarin surrogate 4-nitrophenyl isopropyl methylphosphonate (NIMP). We report a biphasic response in the spiking (but not bursting) activity of neurons exposed to low (i.e., 0.4 and 4 μM) versus high concentrations (i.e., 40 and 100 μM) of NIMP, which was monitored during the exposure period and up to 6 days post-exposure. Regardless of the NIMP concentration, at a network level, communication or coordination of neuronal activity decreased as early as 60 min and persisted at 24 h of NIMP exposure. Once NIMP was removed, coordinated activity was no different than control (0 μM of NIMP). Interestingly, only in the high concentration of NIMP did coordination of activity at a network level begin to decrease again at 2 days post-exposure and persisted on day 6 post-exposure. Notably, cell viability was not affected during or after NIMP exposure. Also, while the catalytic activity of AChE decreased during NIMP exposure, its activity recovered once NIMP was removed. Gene expression analysis suggests that human iPSC-derived neurons and primary human astrocytes resulted in altered genes related to the cell's interaction with the extracellular environment, its intracellular calcium signaling pathways, and inflammation, which could have contributed to how neurons communicated at a network level.
Collapse
Affiliation(s)
- Chandrakumar Bogguri
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Vivek Kurien George
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Beheshta Amiri
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Alexander Ladd
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Nicholas R Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Aimy Sebastian
- 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
| | - Carlos A Valdez
- Global Security Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - T Nathan Mundhenk
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jose Cadena
- Engineering Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Doris Lam
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| |
Collapse
|
7
|
Yuan S, Yuan H, Hay DC, Hu H, Wang C. Revolutionizing Drug Discovery: The Impact of Distinct Designs and Biosensor Integration in Microfluidics-Based Organ-on-a-Chip Technology. BIOSENSORS 2024; 14:425. [PMID: 39329800 PMCID: PMC11430660 DOI: 10.3390/bios14090425] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
Traditional drug development is a long and expensive process with high rates of failure. This has prompted the pharmaceutical industry to seek more efficient drug development frameworks, driving the emergence of organ-on-a-chip (OOC) based on microfluidic technologies. Unlike traditional animal experiments, OOC systems provide a more accurate simulation of human organ microenvironments and physiological responses, therefore offering a cost-effective and efficient platform for biomedical research, particularly in the development of new medicines. Additionally, OOC systems enable quick and real-time analysis, high-throughput experimentation, and automation. These advantages have shown significant promise in enhancing the drug development process. The success of an OOC system hinges on the integration of specific designs, manufacturing techniques, and biosensors to meet the need for integrated multiparameter datasets. This review focuses on the manufacturing, design, sensing systems, and applications of OOC systems, highlighting their design and sensing capabilities, as well as the technical challenges they currently face.
Collapse
Affiliation(s)
- Sheng Yuan
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
| | - Huipu Yuan
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - David C. Hay
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK;
| | - Huan Hu
- Zhejiang University-University of Illinois Urbana-Champaign Institute (ZJU-UIUC Institute), International Campus, Zhejiang University, Haining 314400, China
| | - Chaochen Wang
- Centre of Biomedical Systems and Informatics, Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), School of Medicine, International Campus, Zhejiang University, Haining 314400, China
- Department of Gynecology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| |
Collapse
|
8
|
Ren K, Wang Q, Jiang D, Liu E, Alsmaan J, Jiang R, Rutkove SB, Tian F. A comprehensive review of electrophysiological techniques in amyotrophic lateral sclerosis research. Front Cell Neurosci 2024; 18:1435619. [PMID: 39280794 PMCID: PMC11393746 DOI: 10.3389/fncel.2024.1435619] [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: 05/20/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease, is characterized by progressive motor neuron degeneration, leading to widespread weakness and respiratory failure. While a variety of mechanisms have been proposed as causes of this disease, a full understanding remains elusive. Electrophysiological alterations, including increased motor axon excitability, likely play an important role in disease progression. There remains a critical need for non-animal disease models that can integrate electrophysiological tools to better understand underlying mechanisms, track disease progression, and evaluate potential therapeutic interventions. This review explores the integration of electrophysiological technologies with ALS disease models. It covers cellular and clinical electrophysiological tools and their applications in ALS research. Additionally, we examine conventional animal models and highlight advancements in humanized models and 3D organoid technologies. By bridging the gap between these models, we aim to enhance our understanding of ALS pathogenesis and facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Keyuan Ren
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Qinglong Wang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Douglas Jiang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Scripps Institution of Oceanography, San Diego, CA, United States
| | - Ethan Liu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Julie Alsmaan
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- School of Arts and Science, Harvard College, Cambridge, MA, United States
| | - Rui Jiang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- School of Arts and Science, Harvard College, Cambridge, MA, United States
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Feng Tian
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
9
|
Peng H, Kopic I, Potfode SR, Teshima TF, Boustani GA, Hiendlmeier L, Wang C, Hussain MZ, Özkale B, Fischer RA, Wolfrum B. Laser-patterned epoxy-based 3D microelectrode arrays for extracellular recording. NANOSCALE 2024; 16:14295-14301. [PMID: 39011647 DOI: 10.1039/d4nr01727g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Microelectrode arrays are commonly used to study the electrophysiological behavior of cells. Recently, there has been a growing interest in fabricating three-dimensional microelectrode arrays. Here, we present a novel process for the fast fabrication of epoxy-based 3D microelectrode array platforms with the assistance of laser-patterning technology. To this end, we photopatterned 3D pillars as scaffolds using epoxy-based dry films. Electrodes and conductor traces were fabricated by laser patterning of sputtered platinum films on top of the 3D structures, followed by deposition of parylene-C for insulation. Microelectrodes at the tip of the 3D structures were exposed using a vertical laser ablation process. The final electrodes demonstrated a low impedance of ∼10 kΩ at 1 kHz in electrochemical impedance spectroscopy measurements under physiological conditions. We investigated the maximum compression force of the 3D structures, which could withstand approximately 0.6 N per pillar. The 3D microelectrode arrays were used to record extracellular signals from HL-1 cells in culture as a proof of principle. Our results show regular firing of action potentials recorded at the tip of the 3D structures, demonstrating the possibility of recording cell signals in non-planar environments.
Collapse
Affiliation(s)
- Hu Peng
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
| | - Inola Kopic
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
| | - Shivani Ratnakar Potfode
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
| | - Tetsuhiko F Teshima
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
- Medical & Health Informatics Laboratories NTT Research Incorporated, 940 Stewart Dr, Sunnyvale, CA 94085, USA
| | - George Al Boustani
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
| | - Lukas Hiendlmeier
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
| | - Chen Wang
- Microrobotic Bioengineering Lab (MRBL), Department of Electrical Engineering, TUM School of Computation, Information, and Technology, Technical University of Munich, Hans-Piloty-Str. 1, Garching 85748, Germany
| | - Mian Zahid Hussain
- Chair of Inorganic and Metal-Organic Chemistry, School of Natural Sciences and Catalysis Research Centre, Department of Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| | - Berna Özkale
- Microrobotic Bioengineering Lab (MRBL), Department of Electrical Engineering, TUM School of Computation, Information, and Technology, Technical University of Munich, Hans-Piloty-Str. 1, Garching 85748, Germany
| | - Roland A Fischer
- Chair of Inorganic and Metal-Organic Chemistry, School of Natural Sciences and Catalysis Research Centre, Department of Chemistry, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching, Germany
| | - Bernhard Wolfrum
- Neuroelectronics, Munich Institute of Biomedical Engineering, Department of Electrical Engineering, TUM School of Computation, Information and Technology, Technical University of Munich, Hans-Piloty-Str. 1, 85748, Garching, Germany
- Medical & Health Informatics Laboratories NTT Research Incorporated, 940 Stewart Dr, Sunnyvale, CA 94085, USA
| |
Collapse
|
10
|
Patel D, Shetty S, Acha C, Pantoja IEM, Zhao A, George D, Gracias DH. Microinstrumentation for Brain Organoids. Adv Healthc Mater 2024; 13:e2302456. [PMID: 38217546 DOI: 10.1002/adhm.202302456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/10/2023] [Indexed: 01/15/2024]
Abstract
Brain organoids are three-dimensional aggregates of self-organized differentiated stem cells that mimic the structure and function of human brain regions. Organoids bridge the gaps between conventional drug screening models such as planar mammalian cell culture, animal studies, and clinical trials. They can revolutionize the fields of developmental biology, neuroscience, toxicology, and computer engineering. Conventional microinstrumentation for conventional cellular engineering, such as planar microfluidic chips; microelectrode arrays (MEAs); and optical, magnetic, and acoustic techniques, has limitations when applied to three-dimensional (3D) organoids, primarily due to their limits with inherently two-dimensional geometry and interfacing. Hence, there is an urgent need to develop new instrumentation compatible with live cell culture techniques and with scalable 3D formats relevant to organoids. This review discusses conventional planar approaches and emerging 3D microinstrumentation necessary for advanced organoid-machine interfaces. Specifically, this article surveys recently developed microinstrumentation, including 3D printed and curved microfluidics, 3D and fast-scan optical techniques, buckling and self-folding MEAs, 3D interfaces for electrochemical measurements, and 3D spatially controllable magnetic and acoustic technologies relevant to two-way information transfer with brain organoids. This article highlights key challenges that must be addressed for robust organoid culture and reliable 3D spatiotemporal information transfer.
Collapse
Affiliation(s)
- Devan Patel
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Saniya Shetty
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Chris Acha
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Itzy E Morales Pantoja
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Alice Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Derosh George
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - David H Gracias
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD, 21218, USA
- Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Center for MicroPhysiological Systems (MPS), Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Zhang K, Deng Y, Liu Y, Luo J, Glidle A, Cooper JM, Xu S, Yang Y, Lv S, Xu Z, Wu Y, Sha L, Xu Q, Yin H, Cai X. Investigating Communication Dynamics in Neuronal Network using 3D Gold Microelectrode Arrays. ACS NANO 2024; 18:17162-17174. [PMID: 38902594 PMCID: PMC11349149 DOI: 10.1021/acsnano.4c03983] [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: 03/25/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024]
Abstract
Although in vitro neuronal network models hold great potential for advancing neuroscience research, with the capacity to provide fundamental insights into mechanisms underlying neuronal functions, the dynamics of cell communication within such networks remain poorly understood. Here, we develop a customizable, polymer modified three-dimensional gold microelectrode array with sufficient stability for high signal-to-noise, long-term, neuronal recording of cultured networks. By using directed spatial and temporal patterns of electrical stimulation of cells to explore synaptic-based communication, we monitored cell network dynamics over 3 weeks, quantifying communication capability using correlation heatmaps and mutual information networks. Analysis of synaptic delay and signal speed between cells enabled us to establish a communication connectivity model. We anticipate that our discoveries of the dynamic changes in communication across the neuronal network will provide a valuable tool for future studies in understanding health and disease as well as in developing effective platforms for evaluating therapies.
Collapse
Affiliation(s)
- Kui Zhang
- 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
| | - Yu Deng
- State
Key Laboratory of Medical Molecular Biology, Institute of Basic Medical
Sciences, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing 100005, 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
| | - Jinping Luo
- 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
| | - Andrew Glidle
- James
Watt School of Engineering, University of
Glasgow, Glasgow G12 8LT, United Kingdom
| | - Jonathan M. Cooper
- James
Watt School of Engineering, University of
Glasgow, Glasgow G12 8LT, United Kingdom
| | - Shihong 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
| | - Yan 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
| | - 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
| | - 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
| | - Yirong Wu
- 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
| | - Longzhe Sha
- State
Key Laboratory of Medical Molecular Biology, Institute of Basic Medical
Sciences, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing 100005, China
| | - Qi Xu
- State
Key Laboratory of Medical Molecular Biology, Institute of Basic Medical
Sciences, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing 100005, China
| | - Huabing Yin
- James
Watt School of Engineering, University of
Glasgow, Glasgow G12 8LT, United Kingdom
| | - 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
|
14
|
Meng L, Akhoundian M, Al Azawi A, Shoja Y, Chi PY, Meinander K, Suihkonen S, Franssila S. Ultrasensitive Monolithic Dopamine Microsensors Employing Vertically Aligned Carbon Nanofibers. Adv Healthc Mater 2024; 13:e2303872. [PMID: 38837670 DOI: 10.1002/adhm.202303872] [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: 11/07/2023] [Revised: 03/19/2024] [Indexed: 06/07/2024]
Abstract
Brain-on-Chip devices, which facilitate on-chip cultures of neurons to simulate brain functions, are receiving tremendous attention from both fundamental and clinical research. Consequently, microsensors are being developed to accomplish real-time monitoring of neurotransmitters, which are the benchmarks for neuron network operation. Among these, electrochemical sensors have emerged as promising candidates for detecting a critical neurotransmitter, dopamine. However, current state-of-the-art electrochemical dopamine sensors are suffering from issues like limited sensitivity and cumbersome fabrication. Here, a novel route in monolithically microfabricating vertically aligned carbon nanofiber electrochemical dopamine microsensors is reported with an anti-blistering slow cooling process. Thanks to the microfabrication process, microsensors is created with complete insulation and large surface areas. The champion device shows extremely high sensitivity of 4.52× 104 µAµM-1·cm-2, which is two-orders-of-magnitude higher than current devices, and a highly competitive limit of detection of 0.243 nM. These remarkable figures-of-merit will open new windows for applications such as electrochemical recording from a single neuron.
Collapse
Affiliation(s)
- Lingju Meng
- Department of Chemistry and Materials Science, Aalto University, Espoo, 02150, Finland
- Micronova Nanofabrication Centre, Aalto University, Espoo, 02150, Finland
| | - Maedeh Akhoundian
- Department of Electrical Engineering and Automation, Aalto University, Espoo, 02150, Finland
| | - Anas Al Azawi
- Department of Chemistry and Materials Science, Aalto University, Espoo, 02150, Finland
- Micronova Nanofabrication Centre, Aalto University, Espoo, 02150, Finland
| | - Yalda Shoja
- Department of Chemistry and Materials Science, Aalto University, Espoo, 02150, Finland
- Micronova Nanofabrication Centre, Aalto University, Espoo, 02150, Finland
| | - Pei-Yin Chi
- Department of Chemistry and Materials Science, Aalto University, Espoo, 02150, Finland
- Micronova Nanofabrication Centre, Aalto University, Espoo, 02150, Finland
| | - Kristoffer Meinander
- Department of Bioproducts and Biosystems, Aalto University, Espoo, 02150, Finland
| | - Sami Suihkonen
- Department of Electronics and Nanoengineering, Aalto University, Espoo, 02150, Finland
| | - Sami Franssila
- Department of Chemistry and Materials Science, Aalto University, Espoo, 02150, Finland
- Micronova Nanofabrication Centre, Aalto University, Espoo, 02150, Finland
| |
Collapse
|
15
|
Kang R, Park S, Shin S, Bak G, Park JC. Electrophysiological insights with brain organoid models: a brief review. BMB Rep 2024; 57:311-317. [PMID: 38919012 PMCID: PMC11289503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Brain organoid is a three-dimensional (3D) tissue derived from stem cells such as induced pluripotent stem cells (iPSCs) embryonic stem cells (ESCs) that reflect real human brain structure. It replicates the complexity and development of the human brain, enabling studies of the human brain in vitro. With emerging technologies, its application is various, including disease modeling and drug screening. A variety of experimental methods have been used to study structural and molecular characteristics of brain organoids. However, electrophysiological analysis is necessary to understand their functional characteristics and complexity. Although electrophysiological approaches have rapidly advanced for monolayered cells, there are some limitations in studying electrophysiological and neural network characteristics due to the lack of 3D characteristics. Herein, electrophysiological measurement and analytical methods related to neural complexity and 3D characteristics of brain organoids are reviewed. Overall, electrophysiological understanding of brain organoids allows us to overcome limitations of monolayer in vitro cell culture models, providing deep insights into the neural network complex of the real human brain and new ways of disease modeling. [BMB Reports 2024; 57(7): 311-317].
Collapse
Affiliation(s)
- Rian Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Metabiohealth, Sungkyunkwan University, Suwon 16419, Korea
| | - Soomin Park
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Saewoon Shin
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Gyusoo Bak
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Metabiohealth, Sungkyunkwan University, Suwon 16419, Korea
| | - Jong-Chan Park
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Korea
- Department of Metabiohealth, Sungkyunkwan University, Suwon 16419, Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
16
|
Rahav N, Marrero D, Soffer A, Glickman E, Beldjilali‐Labro M, Yaffe Y, Tadmor K, Leichtmann‐Bardoogo Y, Ashery U, Maoz BM. Multi-Sensor Origami Platform: A Customizable System for Obtaining Spatiotemporally Precise Functional Readouts in 3D Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305555. [PMID: 38634605 PMCID: PMC11200086 DOI: 10.1002/advs.202305555] [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: 08/09/2023] [Revised: 03/14/2024] [Indexed: 04/19/2024]
Abstract
Bioprinting technology offers unprecedented opportunities to construct in vitro tissue models that recapitulate the 3D morphology and functionality of native tissue. Yet, it remains difficult to obtain adequate functional readouts from such models. In particular, it is challenging to position sensors in desired locations within pre-fabricated 3D bioprinted structures. At the same time, bioprinting tissue directly onto a sensing device is not feasible due to interference with the printer head. As such, a multi-sensing platform inspired by origami that overcomes these challenges by "folding" around a separately fabricated 3D tissue structure is proposed, allowing for the insertion of electrodes into precise locations, which are custom-defined using computer-aided-design software. The multi-sensing origami platform (MSOP) can be connected to a commercial multi-electrode array (MEA) system for data-acquisition and processing. To demonstrate the platform, how integrated 3D MEA electrodes can record neuronal electrical activity in a 3D model of a neurovascular unit is shown. The MSOP also enables a microvascular endothelial network to be cultured separately and integrated with the 3D tissue structure. Accordingly, how impedance-based sensors in the platform can measure endothelial barrier function is shown. It is further demonstrated the device's versatility by using it to measure neuronal activity in brain organoids.
Collapse
Affiliation(s)
- Noam Rahav
- School of Neurobiology, Biochemistry and BiophysicsThe George S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv69978Israel
| | - Denise Marrero
- Instituto de Microelectrónica de Barcelona (IMB‐CNM, CSIC)Campus UABBellaterraBarcelona08193Spain
- Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y NanomedicinaMadrid50018Spain
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Adi Soffer
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Emma Glickman
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | | | - Yakey Yaffe
- Sagol Center for Regenerative MedicineTel Aviv UniversityTel Aviv69978Israel
| | - Keshet Tadmor
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | | | - Uri Ashery
- School of Neurobiology, Biochemistry and BiophysicsThe George S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv69978Israel
- Sagol Center for Regenerative MedicineTel Aviv UniversityTel Aviv69978Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | - Ben M. Maoz
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
- Sagol Center for Regenerative MedicineTel Aviv UniversityTel Aviv69978Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
- The Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv69978Israel
| |
Collapse
|
17
|
Wu Y, Cheng J, Qi J, Hang C, Dong R, Low BC, Yu H, Jiang X. Three-dimensional liquid metal-based neuro-interfaces for human hippocampal organoids. Nat Commun 2024; 15:4047. [PMID: 38744873 PMCID: PMC11094048 DOI: 10.1038/s41467-024-48452-5] [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: 05/07/2023] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
Human hippocampal organoids (hHOs) derived from human induced pluripotent stem cells (hiPSCs) have emerged as promising models for investigating neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. However, obtaining the electrical information of these free-floating organoids in a noninvasive manner remains a challenge using commercial multi-electrode arrays (MEAs). The three-dimensional (3D) MEAs developed recently acquired only a few neural signals due to limited channel numbers. Here, we report a hippocampal cyborg organoid (cyb-organoid) platform coupling a liquid metal-polymer conductor (MPC)-based mesh neuro-interface with hHOs. The mesh MPC (mMPC) integrates 128-channel multielectrode arrays distributed on a small surface area (~2*2 mm). Stretchability (up to 500%) and flexibility of the mMPC enable its attachment to hHOs. Furthermore, we show that under Wnt3a and SHH activator induction, hHOs produce HOPX+ and PAX6+ progenitors and ZBTB20+PROX1+ dentate gyrus (DG) granule neurons. The transcriptomic signatures of hHOs reveal high similarity to the developing human hippocampus. We successfully detect neural activities from hHOs via the mMPC from this cyb-organoid. Compared with traditional planar devices, our non-invasive coupling offers an adaptor for recording neural signals from 3D models.
Collapse
Affiliation(s)
- Yan Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Jinhao Cheng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jie Qi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chen Hang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Ruihua Dong
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Yadav N, Giacomozzi F, Cian A, Giubertoni D, Lorenzelli L. Enhancing the Deposition Rate and Uniformity in 3D Gold Microelectrode Arrays via Ultrasonic-Enhanced Template-Assisted Electrodeposition. SENSORS (BASEL, SWITZERLAND) 2024; 24:1251. [PMID: 38400408 PMCID: PMC10893058 DOI: 10.3390/s24041251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/22/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
In the pursuit of refining the fabrication of three-dimensional (3D) microelectrode arrays (MEAs), this study investigates the application of ultrasonic vibrations in template-assisted electrodeposition. This was driven by the need to overcome limitations in the deposition rate and the height uniformity of microstructures developed using conventional electrodeposition methods, particularly in the field of in vitro electrophysiological investigations. This study employs a template-assisted electrodeposition approach coupled with ultrasonic vibrations to enhance the deposition process. The method involves utilizing a polymeric hard mask to define the shape of electrodeposited microstructures (i.e., micro-pillars). The results show that the integration of ultrasonic vibrations significantly increases the deposition rate by up to 5 times and substantially improves the uniformity in 3D MEAs. The key conclusion drawn is that ultrasonic-enhanced template-assisted electrodeposition emerges as a powerful technique and enables the development of 3D MEAs at a higher rate and with a superior uniformity. This advancement holds promising implications for the precision of selective electrodeposition applications and signifies a significant stride in developing micro- and nanofabrication methodologies for biomedical applications.
Collapse
Affiliation(s)
- Neeraj Yadav
- Department of Industrial Engineering, University of Trento, 38123 Trento, Italy
- Center for Sensors & Devices (SD), FBK—Foundation Bruno Kessler, 38123 Trento, Italy; (F.G.); (A.C.); (L.L.)
| | - Flavio Giacomozzi
- Center for Sensors & Devices (SD), FBK—Foundation Bruno Kessler, 38123 Trento, Italy; (F.G.); (A.C.); (L.L.)
| | - Alessandro Cian
- Center for Sensors & Devices (SD), FBK—Foundation Bruno Kessler, 38123 Trento, Italy; (F.G.); (A.C.); (L.L.)
| | - Damiano Giubertoni
- Center for Sensors & Devices (SD), FBK—Foundation Bruno Kessler, 38123 Trento, Italy; (F.G.); (A.C.); (L.L.)
| | - Leandro Lorenzelli
- Center for Sensors & Devices (SD), FBK—Foundation Bruno Kessler, 38123 Trento, Italy; (F.G.); (A.C.); (L.L.)
| |
Collapse
|
24
|
Cho Y, Choi Y, Seong H. Nanoscale surface coatings and topographies for neural interfaces. Acta Biomater 2024; 175:55-75. [PMID: 38141934 DOI: 10.1016/j.actbio.2023.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
With the lack of minimally invasive tools for probing neuronal systems across spatiotemporal scales, understanding the working mechanism of the nervous system and limited assessments available are imperative to prevent or treat neurological disorders. In particular, nanoengineered neural interfaces can provide a solution to this technological barrier. This review covers recent surface engineering approaches, including nanoscale surface coatings, and a range of topographies from the microscale to the nanoscale, primarily focusing on neural-interfaced biosystems. Specifically, the immobilization of bioactive molecules to fertilize the neural cell lineage, topographical engineering to induce mechanotransduction in neural cells, and enhanced cell-chip coupling using three-dimensional structured surfaces are highlighted. Advances in neural interface design will help us understand the nervous system, thereby achieving the effective treatments for neurological disorders. STATEMENT OF SIGNIFICANCE: • This review focuses on designing bioactive neural interface with a nanoscale chemical modification and topographical engineering at multiscale perspective. • Versatile nanoscale surface coatings and topographies for neural interface are summarized. • Recent advances in bioactive materials applicable for neural cell culture, electrophysiological sensing, and neural implants are reviewed.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yunyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyejeong Seong
- 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), Seoul, Republic of Korea.
| |
Collapse
|
25
|
Gong S, Lu Y, Yin J, Levin A, Cheng W. Materials-Driven Soft Wearable Bioelectronics for Connected Healthcare. Chem Rev 2024; 124:455-553. [PMID: 38174868 DOI: 10.1021/acs.chemrev.3c00502] [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: 01/05/2024]
Abstract
In the era of Internet-of-things, many things can stay connected; however, biological systems, including those necessary for human health, remain unable to stay connected to the global Internet due to the lack of soft conformal biosensors. The fundamental challenge lies in the fact that electronics and biology are distinct and incompatible, as they are based on different materials via different functioning principles. In particular, the human body is soft and curvilinear, yet electronics are typically rigid and planar. Recent advances in materials and materials design have generated tremendous opportunities to design soft wearable bioelectronics, which may bridge the gap, enabling the ultimate dream of connected healthcare for anyone, anytime, and anywhere. We begin with a review of the historical development of healthcare, indicating the significant trend of connected healthcare. This is followed by the focal point of discussion about new materials and materials design, particularly low-dimensional nanomaterials. We summarize material types and their attributes for designing soft bioelectronic sensors; we also cover their synthesis and fabrication methods, including top-down, bottom-up, and their combined approaches. Next, we discuss the wearable energy challenges and progress made to date. In addition to front-end wearable devices, we also describe back-end machine learning algorithms, artificial intelligence, telecommunication, and software. Afterward, we describe the integration of soft wearable bioelectronic systems which have been applied in various testbeds in real-world settings, including laboratories that are preclinical and clinical environments. Finally, we narrate the remaining challenges and opportunities in conjunction with our perspectives.
Collapse
Affiliation(s)
- Shu Gong
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Yan Lu
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Jialiang Yin
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Arie Levin
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Wenlong Cheng
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
26
|
Shukla S, Khanna S, Sahoo S, Joshi N, Narayan R. Nanomaterial-Coated Carbon-Fiber-Based Multicontact Array Sensors for In Vitro Monitoring of Serotonin Levels. ACS APPLIED BIO MATERIALS 2024; 7:472-484. [PMID: 38166214 DOI: 10.1021/acsabm.3c01089] [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: 01/04/2024]
Abstract
In this study, we demonstrated the fabrication of multicontact hierarchical probes for the in vitro detection of serotonin levels. The basic three-dimensional (3D) bendable prototypes with 3 (C1), 6 (C2), or 9 (C3) contact surfaces were printed from polymeric resin via the digital light processing (DLP) technique. We chose ultrasonicated carbon fiber strands to transform these designs into multicontact carbon fiber electrodes (MCCFEs). The exposed carbon fiber (CF) surfaces were modified with aminopropyl alkoxysilane (APTMS), followed by the subsequent loading of palladium nanoclusters (PdNPs) to build active recording sites. CF functionalization with PdNPs was achieved by the wet chemical reduction of Pd(II) to Pd(0). The MCCFE configurations demonstrated an enhancement in the electroactive surface area and an improved voltammetric response toward 5-HT oxidation by increasing the points of the contacts (i.e., from C1 to C3). These MCCFEs are comparable to 3D-protruding electrodes as they can enable multipoint analyte detection. Along with the electrode patterns, morphological irregularities associated with both Pd-doped and undoped CFs supported the creation of proximal diffusion layers for facile mass transfer. Low detection limits of 0.8-10 nM over a wide concentration range, from 0.005 nM to 1 mM, were demonstrated. The MCCFE sensors had a relatively low standard deviation value of ∼2%. This type of sensitive and cost-effective electrochemical sensor may prove useful for collecting electrical impulses and long-term monitoring of 5-HT in vivo in addition to in vitro testing.
Collapse
Affiliation(s)
- Shubhangi Shukla
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina 27695-7907, United States
| | - Sumeer Khanna
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina 27695-7907, United States
| | - Siba Sahoo
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina 27695-7907, United States
| | - Naveen Joshi
- Department of Materials Science and Engineering, North Carolina State University, Raleigh, North Carolina 27695-7907, United States
| | - Roger Narayan
- Joint Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina 27695-7907, United States
| |
Collapse
|
27
|
Saglam-Metiner P, Yildirim E, Dincer C, Basak O, Yesil-Celiktas O. Humanized brain organoids-on-chip integrated with sensors for screening neuronal activity and neurotoxicity. Mikrochim Acta 2024; 191:71. [PMID: 38168828 DOI: 10.1007/s00604-023-06165-4] [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: 10/10/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
The complex structure and function of the human central nervous system that develops from the neural tube made in vitro modeling quite challenging until the discovery of brain organoids. Human-induced pluripotent stem cells-derived brain organoids offer recapitulation of the features of early human neurodevelopment in vitro, including the generation, proliferation, and differentiation into mature neurons and micro-macroglial cells, as well as the complex interactions among these diverse cell types of the developing brain. Recent advancements in brain organoids, microfluidic systems, real-time sensing technologies, and their cutting-edge integrated use provide excellent models and tools for emulation of fundamental neurodevelopmental processes, the pathology of neurological disorders, personalized transplantation therapy, and high-throughput neurotoxicity testing by bridging the gap between two-dimensional models and the complex three-dimensional environment in vivo. In this review, we summarize how bioengineering approaches are applied to mitigate the limitations of brain organoids for biomedical and clinical research. We further provide an extensive overview and future perspectives of the humanized brain organoids-on-chip platforms with integrated sensors toward brain organoid intelligence and biocomputing studies. Such approaches might pave the way for increasing approvable clinical applications by solving their current limitations.
Collapse
Affiliation(s)
- Pelin Saglam-Metiner
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey
- Department of Translational Neuroscience, Division of Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ender Yildirim
- Department of Mechanical Engineering, Middle East Technical University, Ankara, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| | - Can Dincer
- Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg, Germany
- FIT Freiburg Center for Interactive Materials and Bioinspired Technologies, University of Freiburg, Freiburg, Germany
| | - Onur Basak
- Department of Translational Neuroscience, Division of Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Turkey.
| |
Collapse
|
28
|
Lojek NM, Williams VA, Rogers AM, Sajo E, Black BJ, Ghezzi CE. A 3D In Vitro Cortical Tissue Model Based on Dense Collagen to Study the Effects of Gamma Radiation on Neuronal Function. Adv Healthc Mater 2024; 13:e2301123. [PMID: 37921265 PMCID: PMC11468710 DOI: 10.1002/adhm.202301123] [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: 04/10/2023] [Revised: 10/14/2023] [Indexed: 11/04/2023]
Abstract
Studies on gamma radiation-induced injury have long been focused on hematopoietic, gastrointestinal, and cardiovascular systems, yet little is known about the effects of gamma radiation on the function of human cortical tissue. The challenge in studying radiation-induced cortical injury is, in part, due to a lack of human tissue models and physiologically relevant readouts. Here, a physiologically relevant 3D collagen-based cortical tissue model (CTM) is developed for studying the functional response of human iPSC-derived neurons and astrocytes to a sub-lethal radiation exposure (5 Gy). Cytotoxicity, DNA damage, morphology, and extracellular electrophysiology are quantified. It is reported that 5 Gy exposure significantly increases cytotoxicity, DNA damage, and astrocyte reactivity while significantly decreasing neurite length and neuronal network activity. Additionally, it is found that clinically deployed radioprotectant amifostine ameliorates the DNA damage, cytotoxicity, and astrocyte reactivity. The CTM provides a critical experimental platform to understand cell-level mechanisms by which gamma radiation (GR) affects human cortical tissue and to screen prospective radioprotectant compounds.
Collapse
Affiliation(s)
- Neal M. Lojek
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Victoria A. Williams
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Andrew M. Rogers
- Department of Physics and Applied PhysicsUniversity of Massachusetts LowellLowellMA01854USA
| | - Erno Sajo
- Department of Physics and Applied PhysicsUniversity of Massachusetts LowellLowellMA01854USA
| | - Bryan J. Black
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Chiara E. Ghezzi
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| |
Collapse
|
29
|
Alahi MEE, Rizu MI, Tina FW, Huang Z, Nag A, Afsarimanesh N. Recent Advancements in Graphene-Based Implantable Electrodes for Neural Recording/Stimulation. SENSORS (BASEL, SWITZERLAND) 2023; 23:9911. [PMID: 38139756 PMCID: PMC10747868 DOI: 10.3390/s23249911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
Implantable electrodes represent a groundbreaking advancement in nervous system research, providing a pivotal tool for recording and stimulating human neural activity. This capability is integral for unraveling the intricacies of the nervous system's functionality and for devising innovative treatments for various neurological disorders. Implantable electrodes offer distinct advantages compared to conventional recording and stimulating neural activity methods. They deliver heightened precision, fewer associated side effects, and the ability to gather data from diverse neural sources. Crucially, the development of implantable electrodes necessitates key attributes: flexibility, stability, and high resolution. Graphene emerges as a highly promising material for fabricating such electrodes due to its exceptional properties. It boasts remarkable flexibility, ensuring seamless integration with the complex and contoured surfaces of neural tissues. Additionally, graphene exhibits low electrical resistance, enabling efficient transmission of neural signals. Its transparency further extends its utility, facilitating compatibility with various imaging techniques and optogenetics. This paper showcases noteworthy endeavors in utilizing graphene in its pure form and as composites to create and deploy implantable devices tailored for neural recordings and stimulations. It underscores the potential for significant advancements in this field. Furthermore, this paper delves into prospective avenues for refining existing graphene-based electrodes, enhancing their suitability for neural recording applications in in vitro and in vivo settings. These future steps promise to revolutionize further our capacity to understand and interact with the neural research landscape.
Collapse
Affiliation(s)
- Md Eshrat E. Alahi
- School of Engineering and Technology, Walailak University, 222 Thaiburi, Thasala District, Nakhon Si Thammarat 80160, Thailand
| | - Mubdiul Islam Rizu
- Microsystems Nanotechnologies for Chemical Analysis (MINOS), Universitat Rovira I Virgili, Avinguda Països Catalans, 26—Campus Sescelades, 43007 Tarragona, Spain;
| | - Fahmida Wazed Tina
- Creative Innovation in Science and Technology Program, Faculty of Science and Technology, Nakhon Si Thammarat Rajabhat University, Nakhon Si Thammarat 80280, Thailand;
| | - Zhaoling Huang
- School of Mechanical and Electrical Engineering, Guilin University of Electronic Technology, Guilin 541004, China;
| | - Anindya Nag
- Faculty of Electrical and Computer Engineering, Technische Universität Dresden, 01062 Dresden, Germany;
- Centre for Tactile Internet with Human-in-the-Loop (CeTI), Technische Universität Dresden, 01069 Dresden, Germany
| | - Nasrin Afsarimanesh
- School of Civil and Mechanical Engineering, Curtin University, Perth, WA 6102, Australia;
| |
Collapse
|
30
|
Huang A, Krueger R, Moeller KD. Microelectrode Arrays, Electrocatalysis, and the Need for Proper Characterization. ChemElectroChem 2023; 10:e202300457. [PMID: 38450252 PMCID: PMC10914339 DOI: 10.1002/celc.202300457] [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: 09/08/2023] [Indexed: 03/08/2024]
Abstract
Indirect electrochemical methods are a powerful tool for synthetic chemistry because they allow for the optimization of chemical selectivity in a reaction while maintaining the advantages of electrochemistry in terms of sustainability. Recently, we have found that such methods provide a handle for not only the synthesis of complex molecules, but also the construction of complex, addressable molecular surfaces. In this effort, the indirect electrochemical methods enable the placement or synthesis of molecules by any electrode or set of electrodes in a microelectrode array. The success of these surface-based reactions are typically evaluated with the use of fluorescence labelling studies. However, these fluorescence-based evaluations can be misleading. While they are excellent for determining that a reaction has occurred in a site-selective fashion on an array, they do not provide information on whether that reaction is the one desired or how well it worked. We describe here how the use of a "safety-catch" linker strategy allows for a more accurate assessment of reaction quality on an array, and then use that capability to illustrate how the use of transition metal mediated cross-coupling reactions on an array prevent unwanted background reactions that can occur on a polymer-coated electrode surface. The method enables a unique level of quality control for array-based transformations.
Collapse
Affiliation(s)
- Albert Huang
- Washington University in Saint Louis, Saint Louis, Missouri 63130, United States
| | - Ruby Krueger
- Washington University in Saint Louis, Saint Louis, Missouri 63130, United States
| | - Kevin D. Moeller
- Washington University in Saint Louis, Saint Louis, Missouri 63130, United States
| |
Collapse
|
31
|
Dong K, Liu WC, Su Y, Lyu Y, Huang H, Zheng N, Rogers JA, Nan K. Scalable Electrophysiology of Millimeter-Scale Animals with Electrode Devices. BME FRONTIERS 2023; 4:0034. [PMID: 38435343 PMCID: PMC10907027 DOI: 10.34133/bmef.0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/08/2023] [Indexed: 03/05/2024] Open
Abstract
Millimeter-scale animals such as Caenorhabditis elegans, Drosophila larvae, zebrafish, and bees serve as powerful model organisms in the fields of neurobiology and neuroethology. Various methods exist for recording large-scale electrophysiological signals from these animals. Existing approaches often lack, however, real-time, uninterrupted investigations due to their rigid constructs, geometric constraints, and mechanical mismatch in integration with soft organisms. The recent research establishes the foundations for 3-dimensional flexible bioelectronic interfaces that incorporate microfabricated components and nanoelectronic function with adjustable mechanical properties and multidimensional variability, offering unique capabilities for chronic, stable interrogation and stimulation of millimeter-scale animals and miniature tissue constructs. This review summarizes the most advanced technologies for electrophysiological studies, based on methods of 3-dimensional flexible bioelectronics. A concluding section addresses the challenges of these devices in achieving freestanding, robust, and multifunctional biointerfaces.
Collapse
Affiliation(s)
- Kairu Dong
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems,
Zhejiang University, Hangzhou 310058, China
- College of Biomedical Engineering & Instrument Science,
Zhejiang University, Hangzhou, 310027, China
| | - Wen-Che Liu
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems,
Zhejiang University, Hangzhou 310058, China
| | - Yuyan Su
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, China
- Department of Gastroenterology, Brigham and Women’s Hospital,
Harvard Medical School, Boston, MA 02115, USA
| | - Yidan Lyu
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, China
| | - Hao Huang
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, China
- College of Chemical and Biological Engineering,
Zhejiang University, Hangzhou 310058, China
| | - Nenggan Zheng
- Qiushi Academy for Advanced Studies,
Zhejiang University, Hangzhou 310027, China
- College of Computer Science and Technology,
Zhejiang University, Hangzhou 310027, China
- State Key Lab of Brain-Machine Intelligence,
Zhejiang University, Hangzhou 310058, China
- CCAI by MOE and Zhejiang Provincial Government (ZJU), Hangzhou 310027, China
| | - John A. Rogers
- Querrey Simpson Institute for Bioelectronics,
Northwestern University, Evanston, IL 60208, USA
- Department of Biomedical Engineering,
Northwestern University, Evanston, IL 60208, USA
- Department of Materials Science and Engineering,
Northwestern University, Evanston, IL 60208, USA
- Department of Mechanical Engineering,
Northwestern University, Evanston, IL 60208, USA
| | - Kewang Nan
- College of Pharmaceutical Sciences,
Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems,
Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
32
|
Gu Y, Zhang W, Wu X, Zhang Y, Xu K, Su J. Organoid assessment technologies. Clin Transl Med 2023; 13:e1499. [PMID: 38115706 PMCID: PMC10731122 DOI: 10.1002/ctm2.1499] [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: 06/14/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2023] Open
Abstract
Despite enormous advances in the generation of organoids, robust and stable protocols of organoids are still a major challenge to researchers. Research for assessing structures of organoids and the evaluations of their functions on in vitro or in vivo is often limited by precision strategies. A growing interest in assessing organoids has arisen, aimed at standardizing the process of obtaining organoids to accurately resemble human-derived tissue. The complex microenvironment of organoids, intricate cellular crosstalk, organ-specific architectures and further complicate functions urgently quest for high-through schemes. By utilizing multi-omics analysis and single-cell analysis, cell-cell interaction mechanisms can be deciphered, and their structures can be investigated in a detailed view by histological analysis. In this review, we will conclude the novel approaches to study the molecular mechanism and cell heterogeneity of organoids and discuss the histological and morphological similarity of organoids in comparison to the human body. Future perspectives on functional analysis will be developed and the organoids will become mature models.
Collapse
Affiliation(s)
- Yuyuan Gu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- School of MedicineShanghai UniversityShanghaiChina
| | - Wencai Zhang
- Department of OrthopedicsFirst Affiliated HospitalJinan UniversityGuangzhouChina
| | - Xianmin Wu
- Department of OrthopedicsShanghai Zhongye HospitalShanghaiChina
| | - Yuanwei Zhang
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Department of OrthopaedicsXinhua Hospital Affiliated to Shanghai JiaoTong University School of MedicineShanghaiChina
| | - Ke Xu
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Wenzhou Institute of Shanghai UniversityWenzhouChina
| | - Jiacan Su
- Institute of Translational MedicineShanghai UniversityShanghaiChina
- Organoid Research CenterShanghai UniversityShanghaiChina
- Department of OrthopaedicsXinhua Hospital Affiliated to Shanghai JiaoTong University School of MedicineShanghaiChina
| |
Collapse
|
33
|
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
|
34
|
Lv S, He E, Luo J, Liu Y, Liang W, Xu S, Zhang K, Yang Y, Wang M, Song Y, Wu Y, Cai X. Using Human-Induced Pluripotent Stem Cell Derived Neurons on Microelectrode Arrays to Model Neurological Disease: A Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301828. [PMID: 37863819 PMCID: PMC10667858 DOI: 10.1002/advs.202301828] [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: 03/22/2023] [Revised: 09/04/2023] [Indexed: 10/22/2023]
Abstract
In situ physiological signals of in vitro neural disease models are essential for studying pathogenesis and drug screening. Currently, an increasing number of in vitro neural disease models are established using human-induced pluripotent stem cell (hiPSC) derived neurons (hiPSC-DNs) to overcome interspecific gene expression differences. Microelectrode arrays (MEAs) can be readily interfaced with two-dimensional (2D), and more recently, three-dimensional (3D) neural stem cell-derived in vitro models of the human brain to monitor their physiological activity in real time. Therefore, MEAs are emerging and useful tools to model neurological disorders and disease in vitro using human iPSCs. This is enabling a real-time window into neuronal signaling at the network scale from patient derived. This paper provides a comprehensive review of MEA's role in analyzing neural disease models established by hiPSC-DNs. It covers the significance of MEA fabrication, surface structure and modification schemes for hiPSC-DNs culturing and signal detection. Additionally, this review discusses advances in the development and use of MEA technology to study in vitro neural disease models, including epilepsy, autism spectrum developmental disorder (ASD), and others established using hiPSC-DNs. The paper also highlights the application of MEAs combined with hiPSC-DNs in detecting in vitro neurotoxic substances. Finally, the future development and outlook of multifunctional and integrated devices for in vitro medical diagnostics and treatment are discussed.
Collapse
Affiliation(s)
- Shiya Lv
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Enhui He
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
- The State Key Lab of Brain‐Machine IntelligenceZhejiang UniversityHangzhou321100China
| | - Jinping Luo
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaoyao Liu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Liang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shihong Xu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Kui Zhang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yan Yang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mixia Wang
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yilin Song
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yirong Wu
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xinxia Cai
- State Key Laboratory of Transducer TechnologyAerospace Information Research InstituteChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
35
|
Liu Y, Yao X, Fan C, Zhang G, Luo X, Qian Y. Microfabrication and lab-on-a-chip devices promote in vitromodeling of neural interfaces for neuroscience researches and preclinical applications. Biofabrication 2023; 16:012002. [PMID: 37832555 DOI: 10.1088/1758-5090/ad032a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 10/13/2023] [Indexed: 10/15/2023]
Abstract
Neural tissues react to injuries through the orchestration of cellular reprogramming, generating specialized cells and activating gene expression that helps with tissue remodeling and homeostasis. Simplified biomimetic models are encouraged to amplify the physiological and morphological changes during neural regeneration at cellular and molecular levels. Recent years have witnessed growing interest in lab-on-a-chip technologies for the fabrication of neural interfaces. Neural system-on-a-chip devices are promisingin vitromicrophysiological platforms that replicate the key structural and functional characteristics of neural tissues. Microfluidics and microelectrode arrays are two fundamental techniques that are leveraged to address the need for microfabricated neural devices. In this review, we explore the innovative fabrication, mechano-physiological parameters, spatiotemporal control of neural cell cultures and chip-based neurogenesis. Although the high variability in different constructs, and the restriction in experimental and analytical access limit the real-life applications of microphysiological models, neural system-on-a-chip devices have gained considerable translatability for modeling neuropathies, drug screening and personalized therapy.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Guifeng Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xi Luo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Yun Qian
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| |
Collapse
|
36
|
Zhao C, Wang Z, Tang X, Qin J, Jiang Z. Recent advances in sensor-integrated brain-on-a-chip devices for real-time brain monitoring. Colloids Surf B Biointerfaces 2023; 229:113431. [PMID: 37473652 DOI: 10.1016/j.colsurfb.2023.113431] [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: 02/21/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023]
Abstract
Brain science has remained in the global spotlight as an important field of scientific and technological discovery. Numerous in vitro and in vivo animal studies have been performed to understand the pathological processes involved in brain diseases and develop strategies for their diagnosis and treatment. However, owing to species differences between animals and humans, several drugs have shown high rates of treatment failure in clinical settings, hindering the development of diagnostic and treatment modalities for brain diseases. In this scenario, microfluidic brain-on-a-chip (BOC) devices, which allow the direct use of human tissues for experiments, have emerged as novel tools for effectively avoiding species differences and performing screening for new drugs. Although microfluidic BOC technology has achieved significant progress in recent years, monitoring slight changes in neurochemicals, neurotransmitters, and environmental states in the brain has remained challenging owing to the brain's complex environment. Hence, the integration of BOC with new sensors that have high sensitivity and high selectivity is urgently required for the real-time dynamic monitoring of BOC parameters. As sensor-based technologies for BOC have not been summarized, here, we review the principle, fabrication process, and application-based classification of sensor-integrated BOC, and then summarize the opportunities and challenges for their development. Generally, sensor-integrated BOC enables real-time monitoring and dynamic analysis, accurately measuring minute changes in the brain and thus enabling the realization of in vivo brain analysis and drug development.
Collapse
Affiliation(s)
- Chen Zhao
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zihao Wang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoying Tang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China.
| | - Jieling Qin
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Zhenqi Jiang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
37
|
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
|
38
|
Forró C, Musall S, Montes VR, Linkhorst J, Walter P, Wessling M, Offenhäusser A, Ingebrandt S, Weber Y, Lampert A, Santoro F. Toward the Next Generation of Neural Iontronic Interfaces. Adv Healthc Mater 2023; 12:e2301055. [PMID: 37434349 PMCID: PMC11468917 DOI: 10.1002/adhm.202301055] [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: 04/03/2023] [Revised: 05/23/2023] [Indexed: 07/13/2023]
Abstract
Neural interfaces are evolving at a rapid pace owing to advances in material science and fabrication, reduced cost of scalable complementary metal oxide semiconductor (CMOS) technologies, and highly interdisciplinary teams of researchers and engineers that span a large range from basic to applied and clinical sciences. This study outlines currently established technologies, defined as instruments and biological study systems that are routinely used in neuroscientific research. After identifying the shortcomings of current technologies, such as a lack of biocompatibility, topological optimization, low bandwidth, and lack of transparency, it maps out promising directions along which progress should be made to achieve the next generation of symbiotic and intelligent neural interfaces. Lastly, it proposes novel applications that can be achieved by these developments, ranging from the understanding and reproduction of synaptic learning to live-long multimodal measurements to monitor and treat various neuronal disorders.
Collapse
Affiliation(s)
- Csaba Forró
- Institute for Biological Information Processing ‐ Bioelectronics IBI‐3Wilhelm‐Johnen‐Straße52428JülichGermany
- Institute of Materials in Electrical Engineering 1RWTH AachenSommerfeldstr. 2452074AachenGermany
| | - Simon Musall
- Institute for Biological Information Processing ‐ Bioelectronics IBI‐3Wilhelm‐Johnen‐Straße52428JülichGermany
- Institute for ZoologyRWTH Aachen UniversityWorringerweg 352074AachenGermany
| | - Viviana Rincón Montes
- Institute for Biological Information Processing ‐ Bioelectronics IBI‐3Wilhelm‐Johnen‐Straße52428JülichGermany
| | - John Linkhorst
- Chemical Process EngineeringRWTH AachenForckenbeckstr. 5152074AachenGermany
| | - Peter Walter
- Department of OphthalmologyUniversity Hospital RWTH AachenPauwelsstr. 3052074AachenGermany
| | - Matthias Wessling
- Chemical Process EngineeringRWTH AachenForckenbeckstr. 5152074AachenGermany
- DWI Leibniz Institute for Interactive MaterialsRWTH AachenForckenbeckstr. 5052074AachenGermany
| | - Andreas Offenhäusser
- Institute for Biological Information Processing ‐ Bioelectronics IBI‐3Wilhelm‐Johnen‐Straße52428JülichGermany
| | - Sven Ingebrandt
- Institute of Materials in Electrical Engineering 1RWTH AachenSommerfeldstr. 2452074AachenGermany
| | - Yvonne Weber
- Department of EpileptologyNeurology, RWTH AachenPauwelsstr. 3052074AachenGermany
| | - Angelika Lampert
- Institute of NeurophysiologyUniklinik RWTH AachenPauwelsstrasse 3052074AachenGermany
| | - Francesca Santoro
- Institute for Biological Information Processing ‐ Bioelectronics IBI‐3Wilhelm‐Johnen‐Straße52428JülichGermany
- Institute of Materials in Electrical Engineering 1RWTH AachenSommerfeldstr. 2452074AachenGermany
| |
Collapse
|
39
|
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
|
40
|
Brown MA, Zappitelli KM, Singh L, Yuan RC, Bemrose M, Brogden V, Miller DJ, Smear MC, Cogan SF, Gardner TJ. Direct laser writing of 3D electrodes on flexible substrates. Nat Commun 2023; 14:3610. [PMID: 37330565 PMCID: PMC10276853 DOI: 10.1038/s41467-023-39152-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/31/2023] [Indexed: 06/19/2023] Open
Abstract
This report describes a 3D microelectrode array integrated on a thin-film flexible cable for neural recording in small animals. The fabrication process combines traditional silicon thin-film processing techniques and direct laser writing of 3D structures at micron resolution via two-photon lithography. Direct laser-writing of 3D-printed electrodes has been described before, but this report is the first to provide a method for producing high-aspect-ratio structures. One prototype, a 16-channel array with 300 µm pitch, demonstrates successful electrophysiological signal capture from bird and mouse brains. Additional devices include 90 µm pitch arrays, biomimetic mosquito needles that penetrate through the dura of birds, and porous electrodes with enhanced surface area. The rapid 3D printing and wafer-scale methods described here will enable efficient device fabrication and new studies examining the relationship between electrode geometry and electrode performance. Applications include small animal models, nerve interfaces, retinal implants, and other devices requiring compact, high-density 3D electrodes.
Collapse
Affiliation(s)
- Morgan A Brown
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Kara M Zappitelli
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Loveprit Singh
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Rachel C Yuan
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Melissa Bemrose
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Valerie Brogden
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - David J Miller
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Matthew C Smear
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Stuart F Cogan
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Timothy J Gardner
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
41
|
Hollow ring-like flexible electrode architecture enabling subcellular multi-directional neural interfacing. Biosens Bioelectron 2023; 227:115182. [PMID: 36870146 DOI: 10.1016/j.bios.2023.115182] [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: 01/10/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 02/27/2023]
Abstract
Implantable neural microelectrodes for recording and stimulating neural activity are critical for research in neuroscience and clinical neuroprosthetic applications. A current need exists for developing new technological solutions for obtaining highly selective and stealthy electrodes that provide reliable neural integration and maintain neuronal viability. This paper reports a novel Hollow Ring-like type electrode to sense and/or stimulate neural activity from three-dimensional neural networks. Due to its unique design, the ring electrode architecture enables easy and reliable access of the electrode to three-dimensional neural networks with reduced mechanical contact on the biological tissue, while providing improved electrical interface with cells. The Hollow Ring electrodes, particularly when coated with the conducting polymer poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS), show improved electrical properties with extremely low impedance (7 MΩ μm2) and high charge injection capabilities (15 mC/cm2), when compared to traditional planar disk-type electrodes. The ring design also serves as an optimal architecture for cell growth to create an optimal subcellular electrical-neural interface. In addition, we showed that neural signals recorded by the ring electrode were better resolved than recordings from a traditional disk-type electrode improving the signal-to-noise ratio (SNR) and the burst detection from 3D neuronal networks in vitro. Overall, our results suggest the great potential of the hollow ring design for developing next-generation microelectrodes for applications in neural interfaces used in physiological studies and neuromodulation applications.
Collapse
|
42
|
Yu J, Yin Y, Leng Y, Zhang J, Wang C, Chen Y, Li X, Wang X, Liu H, Liao Y, Jin Y, Zhang Y, Lu K, Wang K, Wang X, Wang L, Zheng F, Gu Z, Li Y, Fan Y. Emerging strategies of engineering retinal organoids and organoid-on-a-chip in modeling intraocular drug delivery: current progress and future perspectives. Adv Drug Deliv Rev 2023; 197:114842. [PMID: 37105398 DOI: 10.1016/j.addr.2023.114842] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Retinal diseases are a rising concern as major causes of blindness in an aging society; therapeutic options are limited, and the precise pathogenesis of these diseases remains largely unknown. Intraocular drug delivery and nanomedicines offering targeted, sustained, and controllable delivery are the most challenging and popular topics in ocular drug development and toxicological evaluation. Retinal organoids (ROs) and organoid-on-a-chip (ROoC) are both emerging as promising in-vitro models to faithfully recapitulate human eyes for retinal research in the replacement of experimental animals and primary cells. In this study, we review the generation and application of ROs resembling the human retina in cell subtypes and laminated structures and introduce the emerging engineered ROoC as a technological opportunity to address critical issues. On-chip vascularization, perfusion, and close inter-tissue interactions recreate physiological environments in vitro, whilst integrating with biosensors facilitates real-time analysis and monitoring during organogenesis of the retina representing engineering efforts in ROoC models. We also emphasize that ROs and ROoCs hold the potential for applications in modeling intraocular drug delivery in vitro and developing next-generation retinal drug delivery strategies.
Collapse
Affiliation(s)
- Jiaheng Yu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yuqi Yin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubing Leng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jingcheng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yanyun Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaorui Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xudong Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yishan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yihan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Keyu Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Kehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Xiaofei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Lizhen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Yubo Fan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| |
Collapse
|
43
|
Liu S, Kumari S, He H, Mishra P, Singh BN, Singh D, Liu S, Srivastava P, Li C. Biosensors integrated 3D organoid/organ-on-a-chip system: A real-time biomechanical, biophysical, and biochemical monitoring and characterization. Biosens Bioelectron 2023; 231:115285. [PMID: 37058958 DOI: 10.1016/j.bios.2023.115285] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/16/2023]
Abstract
As a full-fidelity simulation of human cells, tissues, organs, and even systems at the microscopic scale, Organ-on-a-Chip (OOC) has significant ethical advantages and development potential compared to animal experiments. The need for the design of new drug high-throughput screening platforms and the mechanistic study of human tissues/organs under pathological conditions, the evolving advances in 3D cell biology and engineering, etc., have promoted the updating of technologies in this field, such as the iteration of chip materials and 3D printing, which in turn facilitate the connection of complex multi-organs-on-chips for simulation and the further development of technology-composite new drug high-throughput screening platforms. As the most critical part of organ-on-a-chip design and practical application, verifying the success of organ model modeling, i.e., evaluating various biochemical and physical parameters in OOC devices, is crucial. Therefore, this paper provides a logical and comprehensive review and discussion of the advances in organ-on-a-chip detection and evaluation technologies from a broad perspective, covering the directions of tissue engineering scaffolds, microenvironment, single/multi-organ function, and stimulus-based evaluation, and provides a more comprehensive review of the progress in the significant organ-on-a-chip research areas in the physiological state.
Collapse
Affiliation(s)
- Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Shikha Kumari
- School of Biochemical Engineering, IIT BHU, Varanasi, Uttar Pradesh, India
| | - Hongyi He
- West China School of Medicine & West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Parichita Mishra
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Bhisham Narayan Singh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Divakar Singh
- School of Biochemical Engineering, IIT BHU, Varanasi, Uttar Pradesh, India
| | - Sutong Liu
- Juxing College of Digital Economics, Haikou University of Economics, Haikou, 570100, China
| | - Pradeep Srivastava
- School of Biochemical Engineering, IIT BHU, Varanasi, Uttar Pradesh, India.
| | - Chenzhong Li
- Biomedical Engineering, School of Medicine, The Chinese University of Hong Kong(Shenzhen), Shenzhen, 518172, China.
| |
Collapse
|
44
|
Biosensor integrated brain-on-a-chip platforms: Progress and prospects in clinical translation. Biosens Bioelectron 2023; 225:115100. [PMID: 36709589 DOI: 10.1016/j.bios.2023.115100] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/07/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
Because of the brain's complexity, developing effective treatments for neurological disorders is a formidable challenge. Research efforts to this end are advancing as in vitro systems have reached the point that they can imitate critical components of the brain's structure and function. Brain-on-a-chip (BoC) was first used for microfluidics-based systems with small synthetic tissues but has expanded recently to include in vitro simulation of the central nervous system (CNS). Defining the system's qualifying parameters may improve the BoC for the next generation of in vitro platforms. These parameters show how well a given platform solves the problems unique to in vitro CNS modeling (like recreating the brain's microenvironment and including essential parts like the blood-brain barrier (BBB)) and how much more value it offers than traditional cell culture systems. This review provides an overview of the practical concerns of creating and deploying BoC systems and elaborates on how these technologies might be used. Not only how advanced biosensing technologies could be integrated with BoC system but also how novel approaches will automate assays and improve point-of-care (PoC) diagnostics and accurate quantitative analyses are discussed. Key challenges providing opportunities for clinical translation of BoC in neurodegenerative disorders are also addressed.
Collapse
|
45
|
Goshi N, Kim H, Girardi G, Gardner A, Seker E. Electrophysiological Activity of Primary Cortical Neuron-Glia Mixed Cultures. Cells 2023; 12:cells12050821. [PMID: 36899957 PMCID: PMC10001406 DOI: 10.3390/cells12050821] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Neuroinflammation plays a central role in many neurological disorders, ranging from traumatic brain injuries to neurodegeneration. Electrophysiological activity is an essential measure of neuronal function, which is influenced by neuroinflammation. In order to study neuroinflammation and its electrophysiological fingerprints, there is a need for in vitro models that accurately capture the in vivo phenomena. In this study, we employed a new tri-culture of primary rat neurons, astrocytes, and microglia in combination with extracellular electrophysiological recording techniques using multiple electrode arrays (MEAs) to determine the effect of microglia on neural function and the response to neuroinflammatory stimuli. Specifically, we established the tri-culture and its corresponding neuron-astrocyte co-culture (lacking microglia) counterpart on custom MEAs and monitored their electrophysiological activity for 21 days to assess culture maturation and network formation. As a complementary assessment, we quantified synaptic puncta and averaged spike waveforms to determine the difference in excitatory to inhibitory neuron ratio (E/I ratio) of the neurons. The results demonstrate that the microglia in the tri-culture do not disrupt neural network formation and stability and may be a better representation of the in vivo rat cortex due to its more similar E/I ratio as compared to more traditional isolated neuron and neuron-astrocyte co-cultures. In addition, only the tri-culture displayed a significant decrease in both the number of active channels and spike frequency following pro-inflammatory lipopolysaccharide exposure, highlighting the critical role of microglia in capturing electrophysiological manifestations of a representative neuroinflammatory insult. We expect the demonstrated technology to assist in studying various brain disease mechanisms.
Collapse
Affiliation(s)
- Noah Goshi
- Department of Biomedical Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Hyehyun Kim
- Department of Biomedical Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Gregory Girardi
- Department of Biomedical Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Alexander Gardner
- Department of Electrical and Computer Engineering, University of California—Davis, Davis, CA 95616, USA
| | - Erkin Seker
- Department of Electrical and Computer Engineering, University of California—Davis, Davis, CA 95616, USA
- Correspondence:
| |
Collapse
|
46
|
Callegari F, Brofiga M, Massobrio P. Modeling the three-dimensional connectivity of in vitro cortical ensembles coupled to Micro-Electrode Arrays. PLoS Comput Biol 2023; 19:e1010825. [PMID: 36780570 PMCID: PMC9956882 DOI: 10.1371/journal.pcbi.1010825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 02/24/2023] [Accepted: 12/17/2022] [Indexed: 02/15/2023] Open
Abstract
Nowadays, in vitro three-dimensional (3D) neuronal networks are becoming a consolidated experimental model to overcome most of the intrinsic limitations of bi-dimensional (2D) assemblies. In the 3D environment, experimental evidence revealed a wider repertoire of activity patterns, characterized by a modulation of the bursting features, than the one observed in 2D cultures. However, it is not totally clear and understood what pushes the neuronal networks towards different dynamical regimes. One possible explanation could be the underlying connectivity, which could involve a larger number of neurons in a 3D rather than a 2D space and could organize following well-defined topological schemes. Driven by experimental findings, achieved by recording 3D cortical networks organized in multi-layered structures coupled to Micro-Electrode Arrays (MEAs), in the present work we developed a large-scale computational network model made up of leaky integrate-and-fire (LIF) neurons to investigate possible structural configurations able to sustain the emerging patterns of electrophysiological activity. In particular, we investigated the role of the number of layers defining a 3D assembly and the spatial distribution of the connections within and among the layers. These configurations give rise to different patterns of activity that could be compared to the ones emerging from real in vitro 3D neuronal populations. Our results suggest that the introduction of three-dimensionality induced a global reduction in both firing and bursting rates with respect to 2D models. In addition, we found that there is a minimum number of layers necessary to obtain a change in the dynamics of the network. However, the effects produced by a 3D organization of the cells is somewhat mitigated if a scale-free connectivity is implemented in either one or all the layers of the network. Finally, the best matching of the experimental data is achieved supposing a 3D connectivity organized in structured bundles of links located in different areas of the 2D network.
Collapse
Affiliation(s)
- Francesca Callegari
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | - Martina Brofiga
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- ScreenNeuroPharm, Sanremo, Italy
| | - Paolo Massobrio
- Department of Informatics, Bioengineering, Robotics and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
- National Institute for Nuclear Physics (INFN), Genova, Italy
- * E-mail:
| |
Collapse
|
47
|
Liu Y, Xu S, Yang Y, Zhang K, He E, Liang W, Luo J, Wu Y, Cai X. Nanomaterial-based microelectrode arrays for in vitro bidirectional brain-computer interfaces: a review. MICROSYSTEMS & NANOENGINEERING 2023; 9:13. [PMID: 36726940 PMCID: PMC9884667 DOI: 10.1038/s41378-022-00479-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/04/2022] [Accepted: 10/21/2022] [Indexed: 06/18/2023]
Abstract
A bidirectional in vitro brain-computer interface (BCI) directly connects isolated brain cells with the surrounding environment, reads neural signals and inputs modulatory instructions. As a noninvasive BCI, it has clear advantages in understanding and exploiting advanced brain function due to the simplified structure and high controllability of ex vivo neural networks. However, the core of ex vivo BCIs, microelectrode arrays (MEAs), urgently need improvements in the strength of signal detection, precision of neural modulation and biocompatibility. Notably, nanomaterial-based MEAs cater to all the requirements by converging the multilevel neural signals and simultaneously applying stimuli at an excellent spatiotemporal resolution, as well as supporting long-term cultivation of neurons. This is enabled by the advantageous electrochemical characteristics of nanomaterials, such as their active atomic reactivity and outstanding charge conduction efficiency, improving the performance of MEAs. Here, we review the fabrication of nanomaterial-based MEAs applied to bidirectional in vitro BCIs from an interdisciplinary perspective. We also consider the decoding and coding of neural activity through the interface and highlight the various usages of MEAs coupled with the dissociated neural cultures to benefit future developments of BCIs.
Collapse
Affiliation(s)
- 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 PR China
| | - Shihong 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 PR China
| | - Yan 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 PR China
| | - Kui Zhang
- 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 PR China
| | - Enhui He
- 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 PR China
| | - Wei Liang
- 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 PR China
| | - Jinping Luo
- 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 PR China
| | - Yirong Wu
- 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 PR 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 PR China
| |
Collapse
|
48
|
Chen Z, Liang Q, Wei Z, Chen X, Shi Q, Yu Z, Sun T. An Overview of In Vitro Biological Neural Networks for Robot Intelligence. CYBORG AND BIONIC SYSTEMS 2023; 4:0001. [PMID: 37040493 PMCID: PMC10076061 DOI: 10.34133/cbsystems.0001] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/17/2022] [Indexed: 01/12/2023] Open
Abstract
In vitro biological neural networks (BNNs) interconnected with robots, so-called BNN-based neurorobotic systems, can interact with the external world, so that they can present some preliminary intelligent behaviors, including learning, memory, robot control, etc. This work aims to provide a comprehensive overview of the intelligent behaviors presented by the BNN-based neurorobotic systems, with a particular focus on those related to robot intelligence. In this work, we first introduce the necessary biological background to understand the 2 characteristics of the BNNs: nonlinear computing capacity and network plasticity. Then, we describe the typical architecture of the BNN-based neurorobotic systems and outline the mainstream techniques to realize such an architecture from 2 aspects: from robots to BNNs and from BNNs to robots. Next, we separate the intelligent behaviors into 2 parts according to whether they rely solely on the computing capacity (computing capacity-dependent) or depend also on the network plasticity (network plasticity-dependent), which are then expounded respectively, with a focus on those related to the realization of robot intelligence. Finally, the development trends and challenges of the BNN-based neurorobotic systems are discussed.
Collapse
Affiliation(s)
- Zhe Chen
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
| | - Qian Liang
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zihou Wei
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xie Chen
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Qing Shi
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zhiqiang Yu
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Tao Sun
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 10081, China
- Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing 100081, China
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
49
|
Chung WG, Kim E, Song H, Lee J, Lee S, Lim K, Jeong I, Park JU. Recent Advances in Electrophysiological Recording Platforms for Brain and Heart Organoids. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Won Gi Chung
- Department of Materials Science and Engineering Yonsei University Seoul 03722 Republic of Korea
- Center for Nanomedicine Institute for Basic Science (IBS) Yonsei University 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) Yonsei University Seoul 03722 Republic of Korea
| | - Hayoung Song
- Department of Materials Science and Engineering Yonsei University Seoul 03722 Republic of Korea
- Center for Nanomedicine Institute for Basic Science (IBS) Yonsei University 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) Yonsei University 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) Yonsei University Seoul 03722 Republic of Korea
| | - Kyeonghee Lim
- Department of Materials Science and Engineering Yonsei University Seoul 03722 Republic of Korea
- Center for Nanomedicine Institute for Basic Science (IBS) Yonsei University Seoul 03722 Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering Yonsei University Seoul 03722 Republic of Korea
- Center for Nanomedicine Institute for Basic Science (IBS) Yonsei University 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) Yonsei University Seoul 03722 Republic of Korea
- KIURI Institute Yonsei University Seoul 03722 Republic of Korea
| |
Collapse
|
50
|
Drayton-White K, Liu S, Chang YC, Uppal S, Moeller KD. Microelectrode arrays, electrosynthesis, and the optimization of signaling on an inert, stable surface. Beilstein J Org Chem 2022; 18:1488-1498. [DOI: 10.3762/bjoc.18.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/10/2022] [Indexed: 11/23/2022] Open
Abstract
Microelectrode arrays are powerful tools for monitoring binding interactions between small molecules and biological targets. In most cases, molecules to be studied using such devices are attached directly to the electrodes in the array. Strategies are in place for calibrating signaling studies utilizing the modified electrodes so that they can be quantified relative to a positive control. In this way, the relative binding constants for multiple ligands for a receptor can potentially be determined in the same experiment. However, there are applications of microelectrode arrays that require stable, tunable, and chemically inert surfaces on the electrodes. The use of those surfaces dictate the use of indirect detection methods that are dependent on the nature of the stable surface used and the amount of the binding partner that is placed on the surface. If one wants to do a quantitative study of binding events that involve molecules on such a stable surface, then once again a method for calibrating the signal from a positive control is needed. Fortunately, the electrodes in an array are excellent handles for conducting synthetic reactions on the surface of an array, and those reactions can be used to tune the surface above the electrodes and calibrate the signal from a positive control. Here, we describe how available Cu-based electrosynthetic reactions can be used to calibrate electrochemical signals on a polymer-coated electrode array and delineate the factors to be considered when choosing a polymer surface for such a study.
Collapse
|