1
|
Vulić K, Amos G, Ruff T, Kasm R, Ihle SJ, Küchler J, Vörös J, Weaver S. Impact of microchannel width on axons for brain-on-chip applications. LAB ON A CHIP 2024; 24:5155-5166. [PMID: 39440578 PMCID: PMC11497309 DOI: 10.1039/d4lc00440j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024]
Abstract
Technologies for axon guidance for in vitro disease models and bottom up investigations are increasingly being used in neuroscience research. One of the most prevalent patterning methods is using polydimethylsiloxane (PDMS) microstructures due to compatibility with microscopy and electrophysiology which enables systematic tracking of axon development with precision and efficiency. Previous investigations of these guidance platforms have noted axons tend to follow edges and avoid sharp turns; however, the specific impact of spatial constraints remains only partially explored. We investigated the influence of microchannel width beyond a constriction point, as well as the number of available microchannels, on axon growth dynamics. Further, by manipulating the size of micron/submicron-sized PDMS tunnels we investigated the space restriction that prevents growth cone penetration showing that restrictions smaller than 350 nm were sufficient to exclude axons. This research offers insights into the interplay of spatial constraints, axon development, and neural behavior. The findings are important for designing in vitro platforms and in vivo neural interfaces for both fundamental neuroscience and translational applications in rapidly evolving neural implant technologies.
Collapse
Affiliation(s)
- Katarina Vulić
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - Giulia Amos
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - Tobias Ruff
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - Revan Kasm
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - Stephan J Ihle
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - Joël Küchler
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| | - Sean Weaver
- Laboratory of Biosensors and Bioelectronics (LBB), ETH Zürich, 8092 Zürich, Switzerland.
| |
Collapse
|
2
|
Beck C, Killeen CT, Johnson SC, Kunze A. Nanomagnetic Guidance Shapes the Structure-Function Relationship of Developing Cortical Networks. NANO LETTERS 2024; 24:13564-13573. [PMID: 39432086 PMCID: PMC11529602 DOI: 10.1021/acs.nanolett.4c03156] [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: 07/03/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
In this study, we implement large-scale nanomagnetic guidance on cortical neurons to guide dissociated neuronal networks during development. Cortical networks cultured over microelectrode arrays were exposed to functionalized magnetic nanoparticles, followed by magnetic field exposure to guide neurites over 14 days in vitro. Immunofluorescence of the axonal protein Tau revealed a greater number of neurites that were longer and aligned with the nanomagnetic force relative to nonguided networks. This was further confirmed through brightfield imaging on the microelectrode arrays during development. Spontaneous electrophysiological recordings revealed that the guided networks exhibited increased firing rates and frequency in force-aligned connectivity identified through Granger Causality. Applying this methodology across networks with nonuniform force directions increased local activity in target regions, identified as regions in the direction of the nanomagnetic force. Altogether, these results demonstrate that nanomagnetic forces guide the structure and function of dissociated cortical neuron networks at the millimeter scale.
Collapse
Affiliation(s)
- Connor
L. Beck
- Department
of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Conner T. Killeen
- Department
of Microbiology, Montana State University, Bozeman, Montana 59717, United States
| | - Sara C. Johnson
- Department
of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Anja Kunze
- Department
of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, United States
- Optical
Technology Center, Montana State University, Bozeman, Montana 59717, United States
- Montana
Nanotechnology Center, Montana State University, Bozeman, Montana 59717, United States
| |
Collapse
|
3
|
Guerra San Juan I, Brunner J, Eggan K, Toonen RF, Verhage M. KIF5A regulates axonal repair and time-dependent axonal transport of SFPQ granules and mitochondria in human motor neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611684. [PMID: 39314491 PMCID: PMC11418931 DOI: 10.1101/2024.09.06.611684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Mutations in the microtubule binding motor protein, kinesin family member 5A (KIF5A), cause the fatal motor neuron disease, Amyotrophic Lateral Sclerosis. While KIF5 family members transport a variety of cargos along axons, it is still unclear which cargos are affected by KIF5A mutations. We generated KIF5A null mutant human motor neurons to investigate the impact of KIF5A loss on the transport of various cargoes and its effect on motor neuron function at two different timepoints in vitro. The absence of KIF5A resulted in reduced neurite complexity in young motor neurons (DIV14) and significant defects in axonal regeneration capacity at all developmental stages. KIF5A loss did not affect neurofilament transport but resulted in decreased mitochondria motility and anterograde speed at DIV42. More prominently, KIF5A depletion strongly reduced anterograde transport of SFPQ-associated RNA granules in DIV42 motor neuron axons. We conclude that KIF5A most prominently functions in human motor neurons to promote axonal regrowth after injury as well as to anterogradely transport mitochondria and, to a larger extent, SFPQ-associated RNA granules in a time-dependent manner.
Collapse
Affiliation(s)
- Irune Guerra San Juan
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jessie Brunner
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ruud F. Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Amos G, Ihle SJ, Clément BF, Duru J, Girardin S, Maurer B, Delipinar T, Vörös J, Ruff T. Engineering an in vitro retinothalamic nerve model. Front Neurosci 2024; 18:1396966. [PMID: 38835836 PMCID: PMC11148348 DOI: 10.3389/fnins.2024.1396966] [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: 03/06/2024] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
Understanding the retinogeniculate pathway in vitro can offer insights into its development and potential for future therapeutic applications. This study presents a Polydimethylsiloxane-based two-chamber system with axon guidance channels, designed to replicate unidirectional retinogeniculate signal transmission in vitro. Using embryonic rat retinas, we developed a model where retinal spheroids innervate thalamic targets through up to 6 mm long microfluidic channels. Using a combination of electrical stimulation and functional calcium imaging we assessed how channel length and electrical stimulation frequency affects thalamic target response. In the presented model we integrated up to 20 identical functional retinothalamic neural networks aligned on a single transparent microelectrode array, enhancing the robustness and quality of recorded functional data. We found that network integrity depends on channel length, with 0.5-2 mm channels maintaining over 90% morphological and 50% functional integrity. A reduced network integrity was recorded in longer channels. The results indicate a notable reduction in forward spike propagation in channels longer than 4 mm. Additionally, spike conduction fidelity decreased with increasing channel length. Yet, stimulation-induced thalamic target activity remained unaffected by channel length. Finally, the study found that a sustained thalamic calcium response could be elicited with stimulation frequencies up to 31 Hz, with higher frequencies leading to transient responses. In conclusion, this study presents a high-throughput platform that demonstrates how channel length affects retina to brain network formation and signal transmission in vitro.
Collapse
Affiliation(s)
- Giulia Amos
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Stephan J Ihle
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Blandine F Clément
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Jens Duru
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Sophie Girardin
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Benedikt Maurer
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Tuğçe Delipinar
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Tobias Ruff
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Morais AS, Mendes M, Cordeiro MA, Sousa JJ, Pais AC, Mihăilă SM, Vitorino C. Organ-on-a-Chip: Ubi sumus? Fundamentals and Design Aspects. Pharmaceutics 2024; 16:615. [PMID: 38794277 PMCID: PMC11124787 DOI: 10.3390/pharmaceutics16050615] [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: 02/29/2024] [Revised: 04/08/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
This review outlines the evolutionary journey from traditional two-dimensional (2D) cell culture to the revolutionary field of organ-on-a-chip technology. Organ-on-a-chip technology integrates microfluidic systems to mimic the complex physiological environments of human organs, surpassing the limitations of conventional 2D cultures. This evolution has opened new possibilities for understanding cell-cell interactions, cellular responses, drug screening, and disease modeling. However, the design and manufacture of microchips significantly influence their functionality, reliability, and applicability to different biomedical applications. Therefore, it is important to carefully consider design parameters, including the number of channels (single, double, or multi-channels), the channel shape, and the biological context. Simultaneously, the selection of appropriate materials compatible with the cells and fabrication methods optimize the chips' capabilities for specific applications, mitigating some disadvantages associated with these systems. Furthermore, the success of organ-on-a-chip platforms greatly depends on the careful selection and utilization of cell resources. Advances in stem cell technology and tissue engineering have contributed to the availability of diverse cell sources, facilitating the development of more accurate and reliable organ-on-a-chip models. In conclusion, a holistic perspective of in vitro cellular modeling is provided, highlighting the integration of microfluidic technology and meticulous chip design, which play a pivotal role in replicating organ-specific microenvironments. At the same time, the sensible use of cell resources ensures the fidelity and applicability of these innovative platforms in several biomedical applications.
Collapse
Affiliation(s)
- Ana Sofia Morais
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - Marta Agostinho Cordeiro
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - João J. Sousa
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - Alberto Canelas Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| | - Silvia M. Mihăilă
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, The Netherlands;
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (A.S.M.); (M.M.); (M.A.C.); (J.J.S.)
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal;
| |
Collapse
|
6
|
Farhang Doost N, Srivastava SK. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. BIOSENSORS 2024; 14:225. [PMID: 38785699 PMCID: PMC11118005 DOI: 10.3390/bios14050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that simulates an artificial organ within a microfluidic cell culture chip. Current cell biology research focuses on in vitro cell cultures due to various limitations of in vivo testing. Unfortunately, in-vitro cell culturing fails to provide an accurate microenvironment, and in vivo cell culturing is expensive and has historically been a source of ethical controversy. OOC aims to overcome these shortcomings and provide the best of both in vivo and in vitro cell culture research. The critical component of the OOC design is utilizing microfluidics to ensure a stable concentration gradient, dynamic mechanical stress modeling, and accurate reconstruction of a cellular microenvironment. OOC also has the advantage of complete observation and control of the system, which is impossible to recreate in in-vivo research. Multiple throughputs, channels, membranes, and chambers are constructed in a polydimethylsiloxane (PDMS) array to simulate various organs on a chip. Various experiments can be performed utilizing OOC technology, including drug delivery research and toxicology. Current technological expansions involve multiple organ microenvironments on a single chip, allowing for studying inter-tissue interactions. Other developments in the OOC technology include finding a more suitable material as a replacement for PDMS and minimizing artefactual error and non-translatable differences.
Collapse
Affiliation(s)
| | - Soumya K. Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
7
|
Kantawala B, Shariff S, Ramadan N, Fawaz V, Hassan Y, Mugisha N, Yenkoyan K, Nazir A, Uwishema O. Revolutionizing neurotherapeutics: blood-brain barrier-on-a-chip technologies for precise drug delivery. Ann Med Surg (Lond) 2024; 86:2794-2804. [PMID: 38694300 PMCID: PMC11060226 DOI: 10.1097/ms9.0000000000001887] [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: 11/23/2023] [Accepted: 02/23/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction The blood-brain barrier (BBB) is a critical neurovascular unit regulating substances' passage from the bloodstream to the brain. Its selective permeability poses significant challenges in drug delivery for neurological disorders. Conventional methods often fail due to the BBB's complex structure. Aim The study aims to shed light on their pivotal role in revolutionizing neurotherapeutics and explores the transformative potential of BBB-on-a-Chip technologies in drug delivery research to comprehensively review BBB-on-a-chip technologies, focusing on their design, and substantiate advantages over traditional models. Methods A detailed analysis of existing literature and experimental data pertaining to BBB-on-a-Chip technologies was conducted. Various models, their physiological relevance, and innovative design considerations were examined through databases like Scopus, EbscoHost, PubMed Central, and Medline. Case studies demonstrating enhanced drug transport through BBB-on-a-Chip models were also reviewed, highlighting their potential impact on neurological disorders. Results BBB-on-a-Chip models offer a revolutionary approach, accurately replicating BBB properties. These microphysiological systems enable high-throughput screening, real-time monitoring of drug transport, and precise localization of drugs. Case studies demonstrate their efficacy in enhancing drug penetration, offering potential therapies for diseases like Parkinson's and Alzheimer's. Conclusion BBB-on-a-Chip models represent a transformative milestone in drug delivery research. Their ability to replicate BBB complexities, offer real-time monitoring, and enhance drug transport holds immense promise for neurological disorders. Continuous research and development are imperative to unlock BBB-on-a-Chip models' full potential, ushering in a new era of targeted, efficient, and safer drug therapies for challenging neurological conditions.
Collapse
Affiliation(s)
- Burhan Kantawala
- Oli Health Magazine Organization, Research and Education
- Neuroscience Laboratory, Cobrain Centre
| | - Sanobar Shariff
- Oli Health Magazine Organization, Research and Education
- Neuroscience Laboratory, Cobrain Centre
| | - Nagham Ramadan
- Oli Health Magazine Organization, Research and Education
- Faculty of Medicine
| | - Violette Fawaz
- Oli Health Magazine Organization, Research and Education
- Faculty of Pharmacy, Beirut Arab University, Beirut, Lebanon
| | - Youmna Hassan
- Oli Health Magazine Organization, Research and Education
- Faculty of Medicine and Surgery, Ahfad University for Women, Omdurman, Sudan
| | - Nadine Mugisha
- Oli Health Magazine Organization, Research and Education
- Faculty of Global Surgery, University of Global Health Equity, Kigali, Rwanda
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Centre
- Department of Biochemistry, Yerevan State Medical University named after Mkhitar Heratsi, Yerevan, Armenia
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | | |
Collapse
|
8
|
Mateus JC, Sousa MM, Burrone J, Aguiar P. Beyond a Transmission Cable-New Technologies to Reveal the Richness in Axonal Electrophysiology. J Neurosci 2024; 44:e1446232023. [PMID: 38479812 PMCID: PMC10941245 DOI: 10.1523/jneurosci.1446-23.2023] [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: 08/07/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 03/17/2024] Open
Abstract
The axon is a neuronal structure capable of processing, encoding, and transmitting information. This assessment contrasts with a limiting, but deeply rooted, perspective where the axon functions solely as a transmission cable of somatodendritic activity, sending signals in the form of stereotypical action potentials. This perspective arose, at least partially, because of the technical difficulties in probing axons: their extreme length-to-diameter ratio and intricate growth paths preclude the study of their dynamics through traditional techniques. Recent findings are challenging this view and revealing a much larger repertoire of axonal computations. Axons display complex signaling processes and structure-function relationships, which can be modulated via diverse activity-dependent mechanisms. Additionally, axons can exhibit patterns of activity that are dramatically different from those of their corresponding soma. Not surprisingly, many of these recent discoveries have been driven by novel technology developments, which allow for in vitro axon electrophysiology with unprecedented spatiotemporal resolution and signal-to-noise ratio. In this review, we outline the state-of-the-art in vitro toolset for axonal electrophysiology and summarize the recent discoveries in axon function it has enabled. We also review the increasing repertoire of microtechnologies for controlling axon guidance which, in combination with the available cutting-edge electrophysiology and imaging approaches, have the potential for more controlled and high-throughput in vitro studies. We anticipate that a larger adoption of these new technologies by the neuroscience community will drive a new era of experimental opportunities in the study of axon physiology and consequently, neuronal function.
Collapse
Affiliation(s)
- J C Mateus
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - M M Sousa
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - J Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom
| | - P Aguiar
- i3S- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
9
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [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: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|
10
|
Lefebvre C, Vreulx AC, Dumortier C, Bégard S, Gelle C, Siedlecki-Wullich D, Colin M, Kilinc D, Halliez S. Integration of Microfluidic Devices with Microelectrode Arrays to Functionally Assay Amyloid-β-Induced Synaptotoxicity. ACS Biomater Sci Eng 2024; 10:1856-1868. [PMID: 38385618 DOI: 10.1021/acsbiomaterials.3c00997] [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: 02/23/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and the most frequent cause of dementia. It is characterized by the accumulation in the brain of two pathological protein aggregates: amyloid-β peptides (Aβ) and abnormally phosphorylated tau. The progressive cognitive decline observed in patients strongly correlates with the synaptic loss. Many lines of evidence suggest that soluble forms of Aβ accumulate into the brain where they cause synapse degeneration. Stopping their spreading and/or targeting the pathophysiological mechanisms leading to synaptic loss would logically be beneficial for the patients. However, we are still far from understanding these processes. Our objective was therefore to develop a versatile model to assay and study Aβ-induced synaptotoxicity. We integrated a microfluidic device that physically isolates synapses from presynaptic and postsynaptic neurons with a microelectrode array. We seeded mouse primary cortical cells in the presynaptic and postsynaptic chambers. After functional synapses have formed in the synaptic chamber, we exposed them to concentrated conditioned media from cell lines overexpressing the wild-type or mutated amyloid precursor protein and thus secreting different levels of Aβ. We recorded the neuronal activity before and after exposition to Aβ and quantified Aβ's effects on the connectivity between presynaptic and postsynaptic neurons. We observed that the application of Aβ on the synapses for 48 h strongly decreased the interchamber connectivity without significantly affecting the neuronal activity in the presynaptic or postsynaptic chambers. Thus, through this model, we are able to functionally assay the impact of Aβ peptides (or other molecules) on synaptic connectivity and to use the latter as a proxy to study Aβ-induced synaptotoxicity. Moreover, since the presynaptic, postsynaptic, and synaptic chambers can be individually targeted, our assay provides a powerful tool to evaluate the involvement of candidate genes in synaptic vulnerability and/or test therapeutic strategies for AD.
Collapse
Affiliation(s)
- Camille Lefebvre
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Anaïs-Camille Vreulx
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, 59019Lille ,France
| | - Corentin Dumortier
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Séverine Bégard
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Carla Gelle
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, 59019Lille ,France
| | - Dolores Siedlecki-Wullich
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, 59019Lille ,France
| | - Morvane Colin
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Devrim Kilinc
- Université de Lille, Institut Pasteur de Lille, CHU Lille, INSERM U1167, LabEx DISTALZ, 59019Lille ,France
| | - Sophie Halliez
- Université de Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| |
Collapse
|
11
|
Buentello DC, Garcia-Corral M, Trujillo-de Santiago G, Alvarez MM. Neuron(s)-on-a-Chip: A Review of the Design and Use of Microfluidic Systems for Neural Tissue Culture. IEEE Rev Biomed Eng 2024; 17:243-263. [PMID: 36301779 DOI: 10.1109/rbme.2022.3217486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neuron-on-chip (NoC) systems-microfluidic devices in which neurons are cultured-have become a promising alternative to replace or minimize the use of animal models and have greatly facilitated in vitro research. Here, we review and discuss current developments in neuron-on-chip platforms, with a particular emphasis on existing biological models, culturing techniques, biomaterials, and topologies. We also discuss how the architecture, flow, and gradients affect neuronal growth, differentiation, and development. Finally, we discuss some of the most recent applications of NoCs in fundamental research (i.e., studies on the effects of electrical, mechanical/topological, or chemical stimuli) and in disease modeling.
Collapse
|
12
|
Hidalgo-Alvarez V, Madl CM. Leveraging Biomaterial Platforms to Study Aging-Related Neural and Muscular Degeneration. Biomolecules 2024; 14:69. [PMID: 38254669 PMCID: PMC10813704 DOI: 10.3390/biom14010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Aging is a complex multifactorial process that results in tissue function impairment across the whole organism. One of the common consequences of this process is the loss of muscle mass and the associated decline in muscle function, known as sarcopenia. Aging also presents with an increased risk of developing other pathological conditions such as neurodegeneration. Muscular and neuronal degeneration cause mobility issues and cognitive impairment, hence having a major impact on the quality of life of the older population. The development of novel therapies that can ameliorate the effects of aging is currently hindered by our limited knowledge of the underlying mechanisms and the use of models that fail to recapitulate the structure and composition of the cell microenvironment. The emergence of bioengineering techniques based on the use of biomimetic materials and biofabrication methods has opened the possibility of generating 3D models of muscular and nervous tissues that better mimic the native extracellular matrix. These platforms are particularly advantageous for drug testing and mechanistic studies. In this review, we discuss the developments made in the creation of 3D models of aging-related neuronal and muscular degeneration and we provide a perspective on the future directions for the field.
Collapse
Affiliation(s)
| | - Christopher M. Madl
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA;
| |
Collapse
|
13
|
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
|
14
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
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
|
16
|
Tran DM, Son JW, Ju TS, Hwang C, Park BH. Dopamine-Regulated Plasticity in MoO 3 Synaptic Transistors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:49329-49337. [PMID: 37819637 DOI: 10.1021/acsami.3c06866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Field-effect transistor-based biosensors have gained increasing interest due to their reactive surface to external stimuli and the adaptive feedback required for advanced sensing platforms in biohybrid neural interfaces. However, complex probing methods for surface functionalization remain a challenge that limits the industrial implementation of such devices. Herein, a simple, label-free biosensor based on molybdenum oxide (MoO3) with dopamine-regulated plasticity is demonstrated. Dopamine oxidation facilitated locally at the channel surface initiates a charge transfer mechanism between the molecule and the oxide, altering the channel conductance and successfully emulating the tunable synaptic weight by neurotransmitter activity. The oxygen level of the channel is shown to heavily affect the device's electrochemical properties, shifting from a nonreactive metallic characteristic to highly responsive semiconducting behavior. Controllable responsivity is achieved by optimizing the channel's dimension, which allows the devices to operate in wide ranges of dopamine concentration, from 100 nM to sub-mM levels, with excellent selectivity compared with K+, Na+, and Ca2+.
Collapse
Affiliation(s)
- Duc Minh Tran
- Division of Quantum Phases and Devices, Department of Physics, Konkuk University, Seoul 05029, Republic of Korea
| | - Jong Wan Son
- Division of Quantum Phases and Devices, Department of Physics, Konkuk University, Seoul 05029, Republic of Korea
- Quantum Spin Team, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Tae-Seong Ju
- Quantum Spin Team, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Chanyong Hwang
- Quantum Spin Team, Korea Research Institute of Standards and Science, Daejeon 34113, Republic of Korea
| | - Bae Ho Park
- Division of Quantum Phases and Devices, Department of Physics, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
17
|
Leng Y, Li X, Zheng F, Liu H, Wang C, Wang X, Liao Y, Liu J, Meng K, Yu J, Zhang J, Wang B, Tan Y, Liu M, Jia X, Li D, Li Y, Gu Z, Fan Y. Advances in In Vitro Models of Neuromuscular Junction: Focusing on Organ-on-a-Chip, Organoids, and Biohybrid Robotics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2211059. [PMID: 36934404 DOI: 10.1002/adma.202211059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/18/2023] [Indexed: 06/18/2023]
Abstract
The neuromuscular junction (NMJ) is a peripheral synaptic connection between presynaptic motor neurons and postsynaptic skeletal muscle fibers that enables muscle contraction and voluntary motor movement. Many traumatic, neurodegenerative, and neuroimmunological diseases are classically believed to mainly affect either the neuronal or the muscle side of the NMJ, and treatment options are lacking. Recent advances in novel techniques have helped develop in vitro physiological and pathophysiological models of the NMJ as well as enable precise control and evaluation of its functions. This paper reviews the recent developments in in vitro NMJ models with 2D or 3D cultures, from organ-on-a-chip and organoids to biohybrid robotics. Related derivative techniques are introduced for functional analysis of the NMJ, such as the patch-clamp technique, microelectrode arrays, calcium imaging, and stimulus methods, particularly optogenetic-mediated light stimulation, microelectrode-mediated electrical stimulation, and biochemical stimulation. Finally, the applications of the in vitro NMJ models as disease models or for drug screening related to suitable neuromuscular diseases are summarized and their future development trends and challenges are discussed.
Collapse
Affiliation(s)
- Yubing Leng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Xiaorui Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, 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
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xudong Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jiangyue Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Kaiqi Meng
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jiaheng Yu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Jingyi Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Binyu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Meili Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Xiaoling Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Deyu Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, and with the School of Engineering Medicine, Beihang University, Beijing, 100083, China
| |
Collapse
|
18
|
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: 5] [Impact Index Per Article: 2.5] [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
|
19
|
Kim J, Kim J, Jin Y, Cho SW. In situbiosensing technologies for an organ-on-a-chip. Biofabrication 2023; 15:042002. [PMID: 37587753 DOI: 10.1088/1758-5090/aceaae] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Thein vitrosimulation of organs resolves the accuracy, ethical, and cost challenges accompanyingin vivoexperiments. Organoids and organs-on-chips have been developed to model thein vitro, real-time biological and physiological features of organs. Numerous studies have deployed these systems to assess thein vitro, real-time responses of an organ to external stimuli. Particularly, organs-on-chips can be most efficiently employed in pharmaceutical drug development to predict the responses of organs before approving such drugs. Furthermore, multi-organ-on-a-chip systems facilitate the close representations of thein vivoenvironment. In this review, we discuss the biosensing technology that facilitates thein situ, real-time measurements of organ responses as readouts on organ-on-a-chip systems, including multi-organ models. Notably, a human-on-a-chip system integrated with automated multi-sensing will be established by further advancing the development of chips, as well as their assessment techniques.
Collapse
Affiliation(s)
- Jinyoung Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Junghoon Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Institute for Basic Science (IBS), Center for Nanomedicine, Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
20
|
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
|
21
|
Dupuit V, Briançon-Marjollet A, Delacour C. Portrait of intense communications within microfluidic neural networks. Sci Rep 2023; 13:12306. [PMID: 37516789 PMCID: PMC10387102 DOI: 10.1038/s41598-023-39477-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023] Open
Abstract
In vitro model networks could provide cellular models of physiological relevance to reproduce and investigate the basic function of neural circuits on a chip in the laboratory. Several tools and methods have been developed since the past decade to build neural networks on a chip; among them, microfluidic circuits appear to be a highly promising approach. One of the numerous advantages of this approach is that it preserves stable somatic and axonal compartments over time due to physical barriers that prevent the soma from exploring undesired areas and guide neurites along defined pathways. As a result, neuron compartments can be identified and isolated, and their interconnectivity can be modulated to build a topological neural network (NN). Here, we have assessed the extent to which the confinement imposed by the microfluidic environment can impact cell development and shape NN activity. Toward that aim, microelectrode arrays have enabled the monitoring of the short- and mid-term evolution of neuron activation over the culture period at specific locations in organized (microfluidic) and random (control) networks. In particular, we have assessed the spike and burst rate, as well as the correlations between the extracted spike trains over the first stages of maturation. This study enabled us to observe intense neurite communications that would have been weaker and more delayed within random networks; the spiking rate, burst and correlations being reinforced over time in terms of number and amplitude, exceeding the electrophysiological features of standard cultures. Beyond the enhanced detection efficiency that was expected from the microfluidic channels, the confinement of cells seems to reinforce neural communications and cell development throughout the network.
Collapse
Affiliation(s)
- Victor Dupuit
- Institut Néel, University Grenoble Alpes, CNRS, Grenoble INP, 38000, Grenoble, France
| | - Anne Briançon-Marjollet
- HP2 Laboratory, University Grenoble Alpes, Institut National de la Santé et de la Recherche Médicale U1300, Grenoble, France
| | - Cécile Delacour
- Institut Néel, University Grenoble Alpes, CNRS, Grenoble INP, 38000, Grenoble, France.
| |
Collapse
|
22
|
Pigareva Y, Gladkov A, Kolpakov V, Bukatin A, Li S, Kazantsev VB, Mukhina I, Pimashkin A. Microfluidic Bi-Layer Platform to Study Functional Interaction between Co-Cultured Neural Networks with Unidirectional Synaptic Connectivity. MICROMACHINES 2023; 14:835. [PMID: 37421068 DOI: 10.3390/mi14040835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/05/2023] [Accepted: 04/09/2023] [Indexed: 07/09/2023]
Abstract
The complex synaptic connectivity architecture of neuronal networks underlies cognition and brain function. However, studying the spiking activity propagation and processing in heterogeneous networks in vivo poses significant challenges. In this study, we present a novel two-layer PDMS chip that facilitates the culturing and examination of the functional interaction of two interconnected neural networks. We utilized cultures of hippocampal neurons grown in a two-chamber microfluidic chip combined with a microelectrode array. The asymmetric configuration of the microchannels between the chambers ensured the growth of axons predominantly in one direction from the Source chamber to the Target chamber, forming two neuronal networks with unidirectional synaptic connectivity. We showed that the local application of tetrodotoxin (TTX) to the Source network did not alter the spiking rate in the Target network. The results indicate that stable network activity in the Target network was maintained for at least 1-3 h after TTX application, demonstrating the feasibility of local chemical activity modulation and the influence of electrical activity from one network on the other. Additionally, suppression of synaptic activity in the Source network by the application of CPP and CNQX reorganized spatio-temporal characteristics of spontaneous and stimulus-evoked spiking activity in the Target network. The proposed methodology and results provide a more in-depth examination of the network-level functional interaction between neural circuits with heterogeneous synaptic connectivity.
Collapse
Affiliation(s)
- Yana Pigareva
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
- Central Research Laboratory, Cell Technology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Arseniy Gladkov
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
- Central Research Laboratory, Cell Technology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Vladimir Kolpakov
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
- Central Research Laboratory, Cell Technology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Anton Bukatin
- Department of Nanobiotechnology, Alferov Saint-Petersburg National Research Academic University of the Russian Academy of Sciences, Saint Petersburg 194021, Russia
- Institute for Analytical Instrumentation of the RAS, Saint Petersburg 198095, Russia
| | - Sergei Li
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
| | - Victor B Kazantsev
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
- Central Research Laboratory, Cell Technology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Irina Mukhina
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
- Central Research Laboratory, Cell Technology Department, Privolzhsky Research Medical University, Nizhny Novgorod 603005, Russia
| | - Alexey Pimashkin
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russia
| |
Collapse
|
23
|
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: 15] [Impact Index Per Article: 7.5] [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
|
24
|
Xu S, Liu Y, Yang Y, Zhang K, Liang W, Xu Z, Wu Y, Luo J, Zhuang C, Cai X. Recent Progress and Perspectives on Neural Chip Platforms Integrating PDMS-Based Microfluidic Devices and Microelectrode Arrays. MICROMACHINES 2023; 14:709. [PMID: 37420942 PMCID: PMC10145465 DOI: 10.3390/mi14040709] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/17/2023] [Accepted: 03/19/2023] [Indexed: 07/09/2023]
Abstract
Recent years have witnessed a spurt of progress in the application of the encoding and decoding of neural activities to drug screening, diseases diagnosis, and brain-computer interactions. To overcome the constraints of the complexity of the brain and the ethical considerations of in vivo research, neural chip platforms integrating microfluidic devices and microelectrode arrays have been raised, which can not only customize growth paths for neurons in vitro but also monitor and modulate the specialized neural networks grown on chips. Therefore, this article reviews the developmental history of chip platforms integrating microfluidic devices and microelectrode arrays. First, we review the design and application of advanced microelectrode arrays and microfluidic devices. After, we introduce the fabrication process of neural chip platforms. Finally, we highlight the recent progress on this type of chip platform as a research tool in the field of brain science and neuroscience, focusing on neuropharmacology, neurological diseases, and simplified brain models. This is a detailed and comprehensive review of neural chip platforms. This work aims to fulfill the following three goals: (1) summarize the latest design patterns and fabrication schemes of such platforms, providing a reference for the development of other new platforms; (2) generalize several important applications of chip platforms in the field of neurology, which will attract the attention of scientists in the field; and (3) propose the developmental direction of neural chip platforms integrating microfluidic devices and microelectrode arrays.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - 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
| | - 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, 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
| | - 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
| | - Chengyu Zhuang
- Department of Orthopaedics, Rujing Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
25
|
Ngo H, Amartumur S, Tran VTA, Tran M, Diep YN, Cho H, Lee LP. In Vitro Tumor Models on Chip and Integrated Microphysiological Analysis Platform (MAP) for Life Sciences and High-Throughput Drug Screening. BIOSENSORS 2023; 13:231. [PMID: 36831997 PMCID: PMC9954135 DOI: 10.3390/bios13020231] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 06/18/2023]
Abstract
The evolution of preclinical in vitro cancer models has led to the emergence of human cancer-on-chip or microphysiological analysis platforms (MAPs). Although it has numerous advantages compared to other models, cancer-on-chip technology still faces several challenges such as the complexity of the tumor microenvironment and integrating multiple organs to be widely accepted in cancer research and therapeutics. In this review, we highlight the advancements in cancer-on-chip technology in recapitulating the vital biological features of various cancer types and their applications in life sciences and high-throughput drug screening. We present advances in reconstituting the tumor microenvironment and modeling cancer stages in breast, brain, and other types of cancer. We also discuss the relevance of MAPs in cancer modeling and precision medicine such as effect of flow on cancer growth and the short culture period compared to clinics. The advanced MAPs provide high-throughput platforms with integrated biosensors to monitor real-time cellular responses applied in drug development. We envision that the integrated cancer MAPs has a promising future with regard to cancer research, including cancer biology, drug discovery, and personalized medicine.
Collapse
Affiliation(s)
- Huyen Ngo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sarnai Amartumur
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Van Thi Ai Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yen N. Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Luke P. Lee
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Bioengineering, University of California at Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
26
|
Habibey R. Incubator-independent perfusion system integrated with microfluidic device for continuous electrophysiology and microscopy readouts. Biofabrication 2023; 15. [PMID: 36652708 DOI: 10.1088/1758-5090/acb466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/18/2023] [Indexed: 01/20/2023]
Abstract
Advances in primary and stem cell derived neuronal cell culture techniques and abundance of available neuronal cell types have enabledin vitroneuroscience as a substantial approach to modelin vivoneuronal networks. Survival of the cultured neurons is inevitably dependent on the cell culture incubators to provide stable temperature and humidity and to supply required CO2levels for controlling the pH of culture medium. Therefore, imaging and electrophysiology recordings outside of the incubator are often limited to the short-term experimental sessions. This restricts our understanding of physiological events to the short snapshots of recorded data while the major part of temporal data is neglected. Multiple custom-made and commercially available platforms like integrated on-stage incubators have been designed to enable long-term microscopy. Nevertheless, long-term high-spatiotemporal electrophysiology recordings from developing neuronal networks needs to be addressed. In the present work an incubator-independent polydimethylsiloxane-based double-wall perfusion chamber was designed and integrated with multi-electrode arrays (MEAs) electrophysiology and compartmentalized microfluidic device to continuously record from engineered neuronal networks at sub-cellular resolution. Cell culture media underwent iterations of conditioning to the ambient CO2and adjusting its pH to physiological ranges to retain a stable pH for weeks outside of the incubator. Double-wall perfusion chamber and an integrated air bubble trapper reduced media evaporation and osmolality drifts of the conditioned media for two weeks. Aligned microchannel-microfluidic device on MEA electrodes allowed neurite growth on top of the planar electrodes and amplified their extracellular activity. This enabled continuous sub-cellular resolution imaging and electrophysiology recordings from developing networks and their growing neurites. The on-chip versatile and self-contained system provides long-term, continuous and high spatiotemporal access to the network data and offers a robustin vitroplatform with many potentials to be applied on advanced cell culture systems including organ-on-chip and organoid models.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany.,CRTD-Center for Regenerative Therapies TU Dresden, 01307 Dresden, Germany
| |
Collapse
|
27
|
Han S, Bang S, Kim HN, Choi N, Kim SH. Modulating and monitoring the functionality of corticostriatal circuits using an electrostimulable microfluidic device. Mol Brain 2023; 16:13. [PMID: 36670465 PMCID: PMC9863144 DOI: 10.1186/s13041-023-01007-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/14/2023] [Indexed: 01/22/2023] Open
Abstract
The central nervous system is organized into different neural circuits, each with particular functions and properties. Studying neural circuits is essential to understanding brain function and neuronal diseases. Microfluidic systems are widely used for reconstructing and studying neural circuits but still need improvement to allow modulation and monitoring of the physiological properties of circuits. In this study, we constructed an improved microfluidic device that supports the electrical modulation of neural circuits and proper reassembly. We demonstrated that our microfluidic device provides a platform for electrically modulating and monitoring the physiological function of neural circuits with genetic indicators for synaptic functionality in corticostriatal (CStr) circuits. In particular, our microfluidic device measures activity-driven Ca2+ dynamics using Ca2+ indicators (synaptophysin-GCaMP6f and Fluo5F-AM), as well as activity-driven synaptic transmission and retrieval using vGlut-pHluorin. Overall, our findings indicate that the improved microfluidic platform described here is an invaluable tool for studying the physiological properties of specific neural circuits.
Collapse
Affiliation(s)
- Sukmin Han
- grid.289247.20000 0001 2171 7818Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447 Republic of Korea
| | - Seokyoung Bang
- grid.35541.360000000121053345Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Hong Nam Kim
- grid.35541.360000000121053345Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Nakwon Choi
- grid.35541.360000000121053345Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 Republic of Korea
| | - Sung Hyun Kim
- grid.289247.20000 0001 2171 7818Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447 Republic of Korea ,grid.289247.20000 0001 2171 7818Department of Physiology, School of Medicine, Kyung Hee University, Seoul, 02447 Republic of Korea ,grid.289247.20000 0001 2171 7818Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Kyung Hee University, Seoul, 02447 South Korea
| |
Collapse
|
28
|
Hrynevich A, Li Y, Cedillo-Servin G, Malda J, Castilho M. (Bio)fabrication of microfluidic devices and organs-on-a-chip. 3D Print Med 2023. [DOI: 10.1016/b978-0-323-89831-7.00001-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
|
29
|
Morales IA, Boghdady CM, Campbell BE, Moraes C. Integrating mechanical sensor readouts into organ-on-a-chip platforms. Front Bioeng Biotechnol 2022; 10:1060895. [PMID: 36588933 PMCID: PMC9800895 DOI: 10.3389/fbioe.2022.1060895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Organs-on-a-chip have emerged as next-generation tissue engineered models to accurately capture realistic human tissue behaviour, thereby addressing many of the challenges associated with using animal models in research. Mechanical features of the culture environment have emerged as being critically important in designing organs-on-a-chip, as they play important roles in both stimulating realistic tissue formation and function, as well as capturing integrative elements of homeostasis, tissue function, and tissue degeneration in response to external insult and injury. Despite the demonstrated impact of incorporating mechanical cues in these models, strategies to measure these mechanical tissue features in microfluidically-compatible formats directly on-chip are relatively limited. In this review, we first describe general microfluidically-compatible Organs-on-a-chip sensing strategies, and categorize these advances based on the specific advantages of incorporating them on-chip. We then consider foundational and recent advances in mechanical analysis techniques spanning cellular to tissue length scales; and discuss their integration into Organs-on-a-chips for more effective drug screening, disease modeling, and characterization of biological dynamics.
Collapse
Affiliation(s)
| | | | | | - Christopher Moraes
- Division of Experimental Medicine, McGill University, Montreal, QC, Canada,Department of Chemical Engineering, McGill University, Montreal, QC, Canada,Department of Biomedical Engineering, McGill University, Montreal, QC, Canada,*Correspondence: Christopher Moraes,
| |
Collapse
|
30
|
Lenoir S, Lahaye RA, Vitet H, Scaramuzzino C, Virlogeux A, Capellano L, Genoux A, Gershoni-Emek N, Geva M, Hayden MR, Saudou F. Pridopidine rescues BDNF/TrkB trafficking dynamics and synapse homeostasis in a Huntington disease brain-on-a-chip model. Neurobiol Dis 2022; 173:105857. [PMID: 36075537 DOI: 10.1016/j.nbd.2022.105857] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by polyglutamine-encoding CAG repeat expansion in the huntingtin (HTT) gene. HTT is involved in the axonal transport of vesicles containing brain-derived neurotrophic factor (BDNF). In HD, diminished BDNF transport leads to reduced BDNF delivery to the striatum, contributing to striatal and cortical neuronal death. Pridopidine is a selective and potent sigma-1 receptor (S1R) agonist currently in clinical development for HD. The S1R is located at the endoplasmic reticulum (ER)-mitochondria interface, where it regulates key cellular pathways commonly impaired in neurodegenerative diseases. We used a microfluidic device that reconstitutes the corticostriatal network, allowing the investigation of presynaptic dynamics, synaptic morphology and transmission, and postsynaptic signaling. Culturing primary neurons from the HD mouse model HdhCAG140/+ provides a "disease-on-a-chip" platform ideal for investigating pathogenic mechanisms and drug activity. Pridopidine rescued the trafficking of BDNF and TrkB resulting in an increased neurotrophin signaling at the synapse. This increased the capacity of HD neurons to release glutamate and restored homeostasis at the corticostriatal synapse. These data suggest that pridopidine enhances the availability of corticostriatal BDNF via S1R activation, leading to neuroprotective effects.
Collapse
Affiliation(s)
- Sophie Lenoir
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Romane A Lahaye
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Hélène Vitet
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Chiara Scaramuzzino
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Amandine Virlogeux
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Laetitia Capellano
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Aurélie Genoux
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | | | | | - Michael R Hayden
- Prilenia Therapeutics, Herzliya, Israel; The Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frédéric Saudou
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France..
| |
Collapse
|
31
|
Habibey R, Rojo Arias JE, Striebel J, Busskamp V. Microfluidics for Neuronal Cell and Circuit Engineering. Chem Rev 2022; 122:14842-14880. [PMID: 36070858 PMCID: PMC9523714 DOI: 10.1021/acs.chemrev.2c00212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 02/07/2023]
Abstract
The widespread adoption of microfluidic devices among the neuroscience and neurobiology communities has enabled addressing a broad range of questions at the molecular, cellular, circuit, and system levels. Here, we review biomedical engineering approaches that harness the power of microfluidics for bottom-up generation of neuronal cell types and for the assembly and analysis of neural circuits. Microfluidics-based approaches are instrumental to generate the knowledge necessary for the derivation of diverse neuronal cell types from human pluripotent stem cells, as they enable the isolation and subsequent examination of individual neurons of interest. Moreover, microfluidic devices allow to engineer neural circuits with specific orientations and directionality by providing control over neuronal cell polarity and permitting the isolation of axons in individual microchannels. Similarly, the use of microfluidic chips enables the construction not only of 2D but also of 3D brain, retinal, and peripheral nervous system model circuits. Such brain-on-a-chip and organoid-on-a-chip technologies are promising platforms for studying these organs as they closely recapitulate some aspects of in vivo biological processes. Microfluidic 3D neuronal models, together with 2D in vitro systems, are widely used in many applications ranging from drug development and toxicology studies to neurological disease modeling and personalized medicine. Altogether, microfluidics provide researchers with powerful systems that complement and partially replace animal models.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Jesús Eduardo Rojo Arias
- Wellcome—MRC
Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge
Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Johannes Striebel
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Volker Busskamp
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| |
Collapse
|
32
|
Miny L, Maisonneuve BGC, Quadrio I, Honegger T. Modeling Neurodegenerative Diseases Using In Vitro Compartmentalized Microfluidic Devices. Front Bioeng Biotechnol 2022; 10:919646. [PMID: 35813998 PMCID: PMC9263267 DOI: 10.3389/fbioe.2022.919646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
The human brain is a complex organ composed of many different types of cells interconnected to create an organized system able to efficiently process information. Dysregulation of this delicately balanced system can lead to the development of neurological disorders, such as neurodegenerative diseases (NDD). To investigate the functionality of human brain physiology and pathophysiology, the scientific community has been generated various research models, from genetically modified animals to two- and three-dimensional cell culture for several decades. These models have, however, certain limitations that impede the precise study of pathophysiological features of neurodegeneration, thus hindering therapeutical research and drug development. Compartmentalized microfluidic devices provide in vitro minimalistic environments to accurately reproduce neural circuits allowing the characterization of the human central nervous system. Brain-on-chip (BoC) is allowing our capability to improve neurodegeneration models on the molecular and cellular mechanism aspects behind the progression of these troubles. This review aims to summarize and discuss the latest advancements of microfluidic models for the investigations of common neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Louise Miny
- NETRI, Lyon, France
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
| | | | - Isabelle Quadrio
- BIORAN Team, Lyon Neuroscience Research Center, CNRS UMR 5292, INSERM U1028, Lyon 1 University, Bron, France
- Laboratory of Neurobiology and Neurogenetics, Department of Biochemistry and Molecular Biology, Lyon University Hospital, Bron, France
| | | |
Collapse
|
33
|
Kim JW, Choi YY, Park SH, Ha JH, Lee HU, Kang T, Sun W, Chung BG. Microfluidic electrode array chip for electrical stimulation-mediated axonal regeneration. LAB ON A CHIP 2022; 22:2122-2130. [PMID: 35388823 DOI: 10.1039/d1lc01158h] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The precise manipulation of the neural stem cell (NSC)-derived neural differentiation is still challenging, and there is a technological barrier to regulate the axonal regeneration in a controlled manner. Here, we developed a microfluidic chip integrated with a microelectrode array as an axonal guidance platform. The microfluidic electrode array chip consisted of two compartments and a bridge microchannel that could isolate and guide the axons. We demonstrated that the NSCs were largely differentiated into neural cells as the electric field was applied to the microfluidic electrode array chip. We also confirmed the synergistic effects of the electrical stimulation (ES) and neurotrophic factor (NF) on axonal outgrowth. This microfluidic electrode array chip can serve as a central nervous system (CNS) model for axonal injury and regeneration. Therefore, it could be a potentially powerful tool for an in vitro model of the axonal regeneration.
Collapse
Affiliation(s)
- Ji Woon Kim
- Department of Biomedical Engineering, Sogang University, Seoul, Korea
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
| | - Si-Hyung Park
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea.
| | - Jang Ho Ha
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| | - Hee Uk Lee
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| | - Taewook Kang
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul, Korea.
| | - Bong Geun Chung
- Institute of Integrated Biotechnology, Sogang University, Seoul, Korea
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| |
Collapse
|
34
|
Mateus JC, Weaver S, van Swaay D, Renz AF, Hengsteler J, Aguiar P, Vörös J. Nanoscale Patterning of In Vitro Neuronal Circuits. ACS NANO 2022; 16:5731-5742. [PMID: 35404570 DOI: 10.1021/acsnano.1c10750] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Methods for patterning neurons in vitro have gradually improved and are used to investigate questions that are difficult to address in or ex vivo. Though these techniques guide axons between groups of neurons, multiscale control of neuronal connectivity, from circuits to synapses, is yet to be achieved in vitro. As studying neuronal circuits with synaptic resolution in vivo poses significant challenges, we present an in vitro alternative to validate biophysical and computational models. In this work we use a combination of electron beam lithography and photolithography to create polydimethylsiloxane (PDMS) structures with features ranging from 150 nm to a few millimeters. Leveraging the difference between average axon and dendritic spine diameters, we restrict axon growth while allowing spines to pass through nanochannels to guide synapse formation between small groups of neurons (i.e., nodes). We show this technique can be used to generate large numbers of isolated feed-forward circuits where connections between nodes are restricted to regions connected by nanochannels. Using a genetically encoded calcium indicator in combination with fluorescently tagged postsynaptic protein, PSD-95, we demonstrate functional synapses can form in this region.
Collapse
Affiliation(s)
- José C Mateus
- Neuroengineering and Computational Neuroscience Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Sean Weaver
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, 8092 Zürich, Switzerland
| | | | - Aline F Renz
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, 8092 Zürich, Switzerland
| | - Julian Hengsteler
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, 8092 Zürich, Switzerland
| | - Paulo Aguiar
- Neuroengineering and Computational Neuroscience Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - János Vörös
- Laboratory for Biosensors and Bioelectronics, ETH Zürich, 8092 Zürich, Switzerland
| |
Collapse
|
35
|
Amirifar L, Shamloo A, Nasiri R, de Barros NR, Wang ZZ, Unluturk BD, Libanori A, Ievglevskyi O, Diltemiz SE, Sances S, Balasingham I, Seidlits SK, Ashammakhi N. Brain-on-a-chip: Recent advances in design and techniques for microfluidic models of the brain in health and disease. Biomaterials 2022; 285:121531. [DOI: 10.1016/j.biomaterials.2022.121531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022]
|
36
|
Hong N, Nam Y. Neurons-on-a-Chip: In Vitro NeuroTools. Mol Cells 2022; 45:76-83. [PMID: 35236782 PMCID: PMC8906998 DOI: 10.14348/molcells.2022.2023] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/24/2021] [Accepted: 02/15/2022] [Indexed: 11/27/2022] Open
Abstract
Neurons-on-a-Chip technology has been developed to provide diverse in vitro neuro-tools to study neuritogenesis, synaptogensis, axon guidance, and network dynamics. The two core enabling technologies are soft-lithography and microelectrode array technology. Soft lithography technology made it possible to fabricate microstamps and microfluidic channel devices with a simple replica molding method in a biological laboratory and innovatively reduced the turn-around time from assay design to chip fabrication, facilitating various experimental designs. To control nerve cell behaviors at the single cell level via chemical cues, surface biofunctionalization methods and micropatterning techniques were developed. Microelectrode chip technology, which provides a functional readout by measuring the electrophysiological signals from individual neurons, has become a popular platform to investigate neural information processing in networks. Due to these key advances, it is possible to study the relationship between the network structure and functions, and they have opened a new era of neurobiology and will become standard tools in the near future.
Collapse
Affiliation(s)
- Nari Hong
- Department of Information and Communication Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Yoonkey Nam
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- KAIST Institute for Institute for Health Science and Technology, KAIST, Daejeon 34141, Korea
| |
Collapse
|
37
|
Wan J, Zhou S, Mea HJ, Guo Y, Ku H, Urbina BM. Emerging Roles of Microfluidics in Brain Research: From Cerebral Fluids Manipulation to Brain-on-a-Chip and Neuroelectronic Devices Engineering. Chem Rev 2022; 122:7142-7181. [PMID: 35080375 DOI: 10.1021/acs.chemrev.1c00480] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Remarkable progress made in the past few decades in brain research enables the manipulation of neuronal activity in single neurons and neural circuits and thus allows the decipherment of relations between nervous systems and behavior. The discovery of glymphatic and lymphatic systems in the brain and the recently unveiled tight relations between the gastrointestinal (GI) tract and the central nervous system (CNS) further revolutionize our understanding of brain structures and functions. Fundamental questions about how neurons conduct two-way communications with the gut to establish the gut-brain axis (GBA) and interact with essential brain components such as glial cells and blood vessels to regulate cerebral blood flow (CBF) and cerebrospinal fluid (CSF) in health and disease, however, remain. Microfluidics with unparalleled advantages in the control of fluids at microscale has emerged recently as an effective approach to address these critical questions in brain research. The dynamics of cerebral fluids (i.e., blood and CSF) and novel in vitro brain-on-a-chip models and microfluidic-integrated multifunctional neuroelectronic devices, for example, have been investigated. This review starts with a critical discussion of the current understanding of several key topics in brain research such as neurovascular coupling (NVC), glymphatic pathway, and GBA and then interrogates a wide range of microfluidic-based approaches that have been developed or can be improved to advance our fundamental understanding of brain functions. Last, emerging technologies for structuring microfluidic devices and their implications and future directions in brain research are discussed.
Collapse
Affiliation(s)
- Jiandi Wan
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Sitong Zhou
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Hing Jii Mea
- Department of Chemical Engineering, University of California, Davis, California 95616, United States
| | - Yaojun Guo
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Hansol Ku
- Department of Electrical and Computer Engineering, University of California, Davis, California 95616, United States
| | - Brianna M Urbina
- Biochemistry, Molecular, Cellular and Developmental Biology Program, University of California, Davis, California 95616, United States
| |
Collapse
|
38
|
Lopez-Muñoz GA, Mughal S, Ramón-Azcón J. Sensors and Biosensors in Organs-on-a-Chip Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:55-80. [DOI: 10.1007/978-3-031-04039-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
39
|
Gupta P, Shinde A, Illath K, Kar S, Nagai M, Tseng FG, Santra TS. Microfluidic platforms for single neuron analysis. Mater Today Bio 2022; 13:100222. [PMID: 35243297 PMCID: PMC8866890 DOI: 10.1016/j.mtbio.2022.100222] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 02/05/2022] [Accepted: 02/14/2022] [Indexed: 11/29/2022]
Abstract
Single-neuron actions are the basis of brain function, as clinical sequelae, neuronal dysfunction or failure for most of the central nervous system (CNS) diseases and injuries can be identified via tracing single-neurons. The bulk analysis methods tend to miscue critical information by assessing the population-averaged outcomes. However, its primary requisite in neuroscience to analyze single-neurons and to understand dynamic interplay of neurons and their environment. Microfluidic systems enable precise control over nano-to femto-liter volumes via adjusting device geometry, surface characteristics, and flow-dynamics, thus facilitating a well-defined micro-environment with spatio-temporal control for single-neuron analysis. The microfluidic platform not only offers a comprehensive landscape to study brain cell diversity at the level of transcriptome, genome, and/or epigenome of individual cells but also has a substantial role in deciphering complex dynamics of brain development and brain-related disorders. In this review, we highlight recent advances of microfluidic devices for single-neuron analysis, i.e., single-neuron trapping, single-neuron dynamics, single-neuron proteomics, single-neuron transcriptomics, drug delivery at the single-neuron level, single axon guidance, and single-neuron differentiation. Moreover, we also emphasize limitations and future challenges of single-neuron analysis by focusing on key performances of throughput and multiparametric activity analysis on microfluidic platforms.
Collapse
|
40
|
Functional Characterization of Human Pluripotent Stem Cell-Derived Models of the Brain with Microelectrode Arrays. Cells 2021; 11:cells11010106. [PMID: 35011667 PMCID: PMC8750870 DOI: 10.3390/cells11010106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 12/26/2022] Open
Abstract
Human pluripotent stem cell (hPSC)-derived neuron cultures have emerged as models of electrical activity in the human brain. Microelectrode arrays (MEAs) measure changes in the extracellular electric potential of cell cultures or tissues and enable the recording of neuronal network activity. MEAs have been applied to both human subjects and hPSC-derived brain models. Here, we review the literature on the functional characterization of hPSC-derived two- and three-dimensional brain models with MEAs and examine their network function in physiological and pathological contexts. We also summarize MEA results from the human brain and compare them to the literature on MEA recordings of hPSC-derived brain models. MEA recordings have shown network activity in two-dimensional hPSC-derived brain models that is comparable to the human brain and revealed pathology-associated changes in disease models. Three-dimensional hPSC-derived models such as brain organoids possess a more relevant microenvironment, tissue architecture and potential for modeling the network activity with more complexity than two-dimensional models. hPSC-derived brain models recapitulate many aspects of network function in the human brain and provide valid disease models, but certain advancements in differentiation methods, bioengineering and available MEA technology are needed for these approaches to reach their full potential.
Collapse
|
41
|
Mateus JC, Lopes C, Aroso M, Costa AR, Gerós A, Meneses J, Faria P, Neto E, Lamghari M, Sousa MM, Aguiar P. Bidirectional flow of action potentials in axons drives activity dynamics in neuronal cultures. J Neural Eng 2021; 18. [PMID: 34891149 DOI: 10.1088/1741-2552/ac41db] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
Objective. Recent technological advances are revealing the complex physiology of the axon and challenging long-standing assumptions. Namely, while most action potential (AP) initiation occurs at the axon initial segment in central nervous system neurons, initiation in distal parts of the axon has been reported to occur in both physiological and pathological conditions. The functional role of these ectopic APs, if exists, is still not clear, nor its impact on network activity dynamics.Approach. Using an electrophysiology platform specifically designed for assessing axonal conduction we show here for the first time regular and effective bidirectional axonal conduction in hippocampal and dorsal root ganglia cultures. We investigate and characterize this bidirectional propagation both in physiological conditions and after distal axotomy.Main results.A significant fraction of APs are not coming from the canonical synapse-dendrite-soma signal flow, but instead from signals originating at the distal axon. Importantly, antidromic APs may carry information and can have a functional impact on the neuron, as they consistently depolarize the soma. Thus, plasticity or gene transduction mechanisms triggered by soma depolarization can also be affected by these antidromic APs. Conduction velocity is asymmetrical, with antidromic conduction being slower than orthodromic.Significance.Altogether these findings have important implications for the study of neuronal functionin vitro, reshaping our understanding on how information flows in neuronal cultures.
Collapse
Affiliation(s)
- J C Mateus
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Cdf Lopes
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - M Aroso
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - A R Costa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - A Gerós
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,FEUP-Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - J Meneses
- CDRSP-IPL-Centre for Rapid and Sustainable Product Development-Instituto Politécnico de Leiria, Marinha Grande, Portugal.,IBEB-Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - P Faria
- CDRSP-IPL-Centre for Rapid and Sustainable Product Development-Instituto Politécnico de Leiria, Marinha Grande, Portugal
| | - E Neto
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - M Lamghari
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - M M Sousa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - P Aguiar
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
42
|
Ji J, Ren X, Zorlutuna P. Cardiac Cell Patterning on Customized Microelectrode Arrays for Electrophysiological Recordings. MICROMACHINES 2021; 12:mi12111351. [PMID: 34832763 PMCID: PMC8619285 DOI: 10.3390/mi12111351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Cardiomyocytes (CMs) and fibroblast cells are two essential elements for cardiac tissue structure and function. The interactions between them can alter cardiac electrophysiology and thus contribute to cardiac diseases, such as arrhythmogenesis. One possible explanation is that fibroblasts can directly affect cardiac electrophysiology through electrical coupling with CMs. Therefore, detecting the electrical activities in the CM-fibroblast network is vital for understanding the coupling dynamics among them. Current commercialized platforms for studying cardiac electrophysiology utilize planar microelectrode arrays (MEAs) to record the extracellular field potential (FP) in real-time, but the prearranged electrode configuration highly limits the measurement capabilities at specific locations. Here, we report a custom-designed MEA device with a novel micropatterning method to construct a controlled network of neonatal rat CMs (rCMs) and fibroblast connections for monitoring the electrical activity of rCM-fibroblast co-cultures in a spatially controlled fashion. For the micropatterning of the co-culture, surface topographical features and mobile blockers were used to control the initial attachment locations of a mixture of rCMs and fibroblasts, to form separate beating rCM-fibroblast clusters while leaving empty space for fibroblast growth to connect these clusters. Once the blockers are removed, the proliferating fibroblasts connect and couple the separate beating clusters. Using this method, electrical activity of both rCMs and human-induced-pluripotent-stem-cell-derived cardiomyocytes (iCMs) was examined. The coupling dynamics were studied through the extracellular FP and impedance profile recorded from the MEA device, indicating that the fibroblast bridge provided an RC-type coupling of physically separate rCM-containing clusters and enabled synchronization of these clusters.
Collapse
|
43
|
Duc P, Vignes M, Hugon G, Sebban A, Carnac G, Malyshev E, Charlot B, Rage F. Human neuromuscular junction on micro-structured microfluidic devices implemented with a custom micro electrode array (MEA). LAB ON A CHIP 2021; 21:4223-4236. [PMID: 34559171 DOI: 10.1039/d1lc00497b] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In the neuromuscular system, signal transmission from motor neurons (MNs) to innervated muscle fibers is crucial for their synaptic function, viability, and maintenance. In order to better understand human neuromuscular junction (hNMJ) functionality, it is important to develop on-a-chip devices with human cells. To investigate this cell network, microfluidic platforms are useful to grow different cell types in isolated compartments. Such devices have already been developed to study in vitro neuronal circuitry. Here, we combined microfluidics with two techniques: soft lithography and custom microelectrodes array (MEA). Our goal was to create hNMJs on a specific pattern of electrodes to stimulate pre-synaptic axons and record post-synaptic muscle activity. Micromachining was used to create structurations to guide muscle growth above electrodes, without impairing axon propagation, therefore optimizing the effectiveness of activity recording. Electrodes were also arranged to be aligned with the microfluidic chambers in order to specifically stimulate axons that were growing between the two compartments. Isolation of the two cell types allows for the selective treatment of neurons or muscle fibers to assess NMJ functionality hallmarks. Altogether, this microfluidic/microstructured/MEA platform allowed mature and functional in vitro hNMJ modelling. We demonstrate that electrical activation of MNs can trigger recordable extracellular muscle action potentials. This study provides evidence for a physiologically relevant model to mimic a hNMJ that will in the future be a powerful tool, more sensitive than calcium imaging, to better understand and characterize NMJs and their disruption in neurodegenerative diseases.
Collapse
Affiliation(s)
- Pauline Duc
- IGMM, University of Montpellier, CNRS, Montpellier, France.
| | - Michel Vignes
- IBMM UMR5247, University of Montpellier, CNRS, Montpellier, France
| | - Gérald Hugon
- PhyMedExp, INSERM U1046, CNRS UMR9214, Université de Montpellier, Montpellier, France
| | - Audrey Sebban
- IES, CNRS University of Montpellier, Montpellier 34095, France
| | - Gilles Carnac
- PhyMedExp, INSERM U1046, CNRS UMR9214, Université de Montpellier, Montpellier, France
| | - Eugene Malyshev
- IES, CNRS University of Montpellier, Montpellier 34095, France
| | - Benoît Charlot
- IES, CNRS University of Montpellier, Montpellier 34095, France
| | - Florence Rage
- IGMM, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
44
|
Tran HT, Lucas MS, Ishikawa T, Shahmoradian SH, Padeste C. A Compartmentalized Neuronal Cell-Culture Platform Compatible With Cryo-Fixation by High-Pressure Freezing for Ultrastructural Imaging. Front Neurosci 2021; 15:726763. [PMID: 34566569 PMCID: PMC8455873 DOI: 10.3389/fnins.2021.726763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/17/2021] [Indexed: 12/29/2022] Open
Abstract
The human brain contains a wide array of billions of neurons and interconnections, which are often simplified for analysis in vitro using compartmentalized microfluidic devices for neuronal cell culturing, to better understand neuronal development and disease. However, such devices are traditionally incompatible for high-pressure freezing and high-resolution nanoscale imaging and analysis of their sub-cellular processes by methods including electron microscopy. Here we develop a novel compartmentalized neuronal co-culture platform allowing reconstruction of neuronal networks with high variable spatial control, which is uniquely compatible for high-pressure freezing. This cryo-fixation method is well-established to enable high-fidelity preservation of the reconstructed neuronal networks and their sub-cellular processes in a near-native vitreous state without requiring chemical fixatives. To direct the outgrowth of neurites originating from two distinct groups of neurons growing in the two different compartments, polymer microstructures akin to microchannels are fabricated atop of sapphire disks. Two populations of neurons expressing either enhanced green fluorescent protein (EGFP) or mCherry were grown in either compartment, facilitating the analysis of the specific interactions between the two separate groups of cells. Neuronally differentiated PC12 cells, murine hippocampal and striatal neurons were successfully used in this context. The design of this device permits direct observation of entire neuritic processes within microchannels by optical microscopy with high spatial and temporal resolution, prior to processing for high-pressure freezing and electron microscopy. Following freeze substitution, we demonstrate that it is possible to process the neuronal networks for ultrastructural imaging by electron microscopy. Several key features of the embedded neuronal networks, including mitochondria, synaptic vesicles, axonal terminals, microtubules, with well-preserved ultrastructures were observed at high resolution using focused ion beam - scanning electron microscopy (FIB-SEM) and serial sectioning - transmission electron microscopy (TEM). These results demonstrate the compatibility of the platform with optical microscopy, high-pressure freezing and electron microscopy. The platform can be extended to neuronal models of brain disease or development in future studies, enabling the investigation of subcellular processes at the nanoscale within two distinct groups of neurons in a functional neuronal pathway, as well as pharmacological testing and drug screening.
Collapse
Affiliation(s)
- Hung Tri Tran
- Laboratory of Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland
| | - Miriam S. Lucas
- Scientific Center for Optical and Electron Microscopy ScopeM, ETH Zürich, Zurich, Switzerland
| | - Takashi Ishikawa
- Laboratory of Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland
| | | | - Celestino Padeste
- Laboratory of Nanoscale Biology, Paul Scherrer Institute, Villigen, Switzerland
| |
Collapse
|
45
|
Soheilmoghaddam F, Rumble M, Cooper-White J. High-Throughput Routes to Biomaterials Discovery. Chem Rev 2021; 121:10792-10864. [PMID: 34213880 DOI: 10.1021/acs.chemrev.0c01026] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many existing clinical treatments are limited in their ability to completely restore decreased or lost tissue and organ function, an unenviable situation only further exacerbated by a globally aging population. As a result, the demand for new medical interventions has increased substantially over the past 20 years, with the burgeoning fields of gene therapy, tissue engineering, and regenerative medicine showing promise to offer solutions for full repair or replacement of damaged or aging tissues. Success in these fields, however, inherently relies on biomaterials that are engendered with the ability to provide the necessary biological cues mimicking native extracellular matrixes that support cell fate. Accelerating the development of such "directive" biomaterials requires a shift in current design practices toward those that enable rapid synthesis and characterization of polymeric materials and the coupling of these processes with techniques that enable similarly rapid quantification and optimization of the interactions between these new material systems and target cells and tissues. This manuscript reviews recent advances in combinatorial and high-throughput (HT) technologies applied to polymeric biomaterial synthesis, fabrication, and chemical, physical, and biological screening with targeted end-point applications in the fields of gene therapy, tissue engineering, and regenerative medicine. Limitations of, and future opportunities for, the further application of these research tools and methodologies are also discussed.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Madeleine Rumble
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| |
Collapse
|
46
|
Herreros P, Ballesteros-Esteban LM, Laguna MF, Leyva I, Sendiña-Nadal I, Holgado M. Neuronal circuits on a chip for biological network monitoring. Biotechnol J 2021; 16:e2000355. [PMID: 33984186 DOI: 10.1002/biot.202000355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 03/16/2021] [Accepted: 04/28/2021] [Indexed: 11/09/2022]
Abstract
Cultured neuronal networks (CNNs) are a robust model to closely investigate neuronal circuits' formation and monitor their structural properties evolution. Typically, neurons are cultured in plastic plates or, more recently, in microfluidic platforms with potentially a wide variety of neuroscience applications. As a biological protocol, cell culture integration with a microfluidic system provides benefits such as accurate control of cell seeding area, culture medium renewal, or lower exposure to contamination. The objective of this report is to present a novel neuronal network on a chip device, including a chamber, fabricated from PDMS, vinyl and glass connected to a microfluidic platform to perfuse the continuous flow of culture medium. Network growth is compared in chips and traditional Petri dishes to validate the microfluidic chip performance. The network assessment is performed by computing relevant topological measures like the number of connected neurons, the clustering coefficient, and the shortest path between any pair of neurons throughout the culture's life. The results demonstrate that neuronal circuits on a chip have a more stable network structure and lifespan than developing in conventional settings, and therefore this setup is an advantageous alternative to current culture methods. This technology could lead to challenging applications such as batch drug testing of in vitro cell culture models. From the engineering perspective, a device's advantage is the chance to develop custom designs more efficiently than other microfluidic systems.
Collapse
Affiliation(s)
- Pedro Herreros
- Group of Optics, Photonics and Biophotonics (GOFB), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Luis M Ballesteros-Esteban
- Complex Systems Group & GISC, Universidad Rey Juan Carlos, Madrid, Spain.,Group of Biological Networks, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - María Fe Laguna
- Group of Optics, Photonics and Biophotonics (GOFB), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain.,Departamento de Física Aplicada e Ingeniería de Materiales, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, Madrid, Spain
| | - Inmaculada Leyva
- Complex Systems Group & GISC, Universidad Rey Juan Carlos, Madrid, Spain.,Group of Biological Networks, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - Irene Sendiña-Nadal
- Complex Systems Group & GISC, Universidad Rey Juan Carlos, Madrid, Spain.,Group of Biological Networks, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - Miguel Holgado
- Group of Optics, Photonics and Biophotonics (GOFB), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain.,Departamento de Física Aplicada e Ingeniería de Materiales, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, Madrid, Spain
| |
Collapse
|
47
|
Pigareva Y, Gladkov A, Kolpakov V, Mukhina I, Bukatin A, Kazantsev VB, Pimashkin A. Experimental Platform to Study Spiking Pattern Propagation in Modular Networks In Vitro. Brain Sci 2021; 11:brainsci11060717. [PMID: 34071257 PMCID: PMC8229331 DOI: 10.3390/brainsci11060717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 12/31/2022] Open
Abstract
The structured organization of connectivity in neural networks is associated with highly efficient information propagation and processing in the brain, in contrast with disordered homogeneous network architectures. Using microfluidic methods, we engineered modular networks of cultures using dissociated cells with unidirectional synaptic connections formed by asymmetric microchannels. The complexity of the microchannel geometry defined the strength of the synaptic connectivity and the properties of spiking activity propagation. In this study, we developed an experimental platform to study the effects of synaptic plasticity on a network level with predefined locations of unidirectionally connected cellular assemblies using multisite extracellular electrophysiology.
Collapse
Affiliation(s)
- Yana Pigareva
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (Y.P.); (A.G.); (V.K.); (I.M.); (V.B.K.)
| | - Arseniy Gladkov
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (Y.P.); (A.G.); (V.K.); (I.M.); (V.B.K.)
- Cell Technology Department, Central Research Laboratory, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Vladimir Kolpakov
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (Y.P.); (A.G.); (V.K.); (I.M.); (V.B.K.)
| | - Irina Mukhina
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (Y.P.); (A.G.); (V.K.); (I.M.); (V.B.K.)
- Cell Technology Department, Central Research Laboratory, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | - Anton Bukatin
- The Laboratory of Renewable Energy Sources, Alferov Saint-Petersburg National Research Academic University of the Russian Academy of Sciences, 194021 Saint-Petersburg, Russia;
- The Laboratory of Bio and Chemosensor Microsystems, Institute for Analytical Instrumentation of the RAS, 198095 Saint-Petersburg, Russia
| | - Victor B. Kazantsev
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (Y.P.); (A.G.); (V.K.); (I.M.); (V.B.K.)
- Neuroscience and Cognitive Technology Laboratory, Center for Technologies in Robotics and Mechatronics Components, Innopolis University, 1 Universitetskaya Str., 420500 Innopolis, Russia
- Center for Neurotechnology and Machine Learning, Immanuel Kant Baltic Federal University, 14 Nevsky Str., 236016 Kaliningrad, Russia
| | - Alexey Pimashkin
- Neurotechnology Department, Lobachevsky State University of Nizhny Novgorod, 603950 Nizhny Novgorod, Russia; (Y.P.); (A.G.); (V.K.); (I.M.); (V.B.K.)
- Correspondence:
| |
Collapse
|
48
|
Advances in 3D neuronal microphysiological systems: towards a functional nervous system on a chip. In Vitro Cell Dev Biol Anim 2021; 57:191-206. [PMID: 33438114 PMCID: PMC7802613 DOI: 10.1007/s11626-020-00532-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022]
Abstract
Microphysiological systems (MPS) designed to study the complexities of the peripheral and central nervous systems have made marked improvements over the years and have allowed researchers to assess in two and three dimensions the functional interconnectivity of neuronal tissues. The recent generation of brain organoids has further propelled the field into the nascent recapitulation of structural, functional, and effective connectivities which are found within the native human nervous system. Herein, we will review advances in culture methodologies, focused especially on those of human tissues, which seek to bridge the gap from 2D cultures to hierarchical and defined 3D MPS with the end goal of developing a robust nervous system-on-a-chip platform. These advances have far-reaching implications within basic science, pharmaceutical development, and translational medicine disciplines.
Collapse
|
49
|
Liu W, Sun M, Han K, Hu R, Liu D, Wang J. Comprehensive Evaluation of Stable Neuronal Cell Adhesion and Culture on One-Step Modified Polydimethylsiloxane Using Functionalized Pluronic. ACS OMEGA 2020; 5:32753-32760. [PMID: 33376913 PMCID: PMC7758976 DOI: 10.1021/acsomega.0c05190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/02/2020] [Indexed: 06/12/2023]
Abstract
Polydimethylsiloxane (PDMS) is a popular and property-advantageous material for developing biomedical microsystems and advancing cell microengineering. The requirement of constructing a robust cell-adhesive PDMS interface drives the exploration of simple, straightforward, and applicable surface modification methods. Here, a comprehensive evaluation of highly stable neuronal cell adhesion and culture on the PDMS surface modified in one step using functionalized Pluronic is presented. According to multiple comparative tests, this modification is sufficiently verified to enable more significant cell adhesion and spreading in both quantity and stability, higher neuronal differentiation and viability/growth, more complete formation of the neuronal network, and stabler neuronal cell culture than the common coating tools on the PDMS substrate. The comparable and even superior cellular effects of this modification on PDMS to the standard coating of polystyrene for in vitro neurological research are demonstrated. Long-term microfluidic neuron culture with stable adhesion and high differentiation on the modified PDMS interface is accomplished, too. The achievement provides a detailed experimental demonstration of this simple and effective modification for strengthening neuronal cell culture on the PDMS substrate, which is useful for potential applications in the fields of neurobiology, neuron microengineering, and brain-on-a-chip.
Collapse
Affiliation(s)
- Wenming Liu
- Departments
of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
- Department
of Chemistry, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Meilin Sun
- Departments
of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Kai Han
- Departments
of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Rui Hu
- Departments
of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Dan Liu
- Departments
of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Jinyi Wang
- Department
of Chemistry, College of Chemistry and Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
50
|
Pelkonen A, Mzezewa R, Sukki L, Ryynänen T, Kreutzer J, Hyvärinen T, Vinogradov A, Aarnos L, Lekkala J, Kallio P, Narkilahti S. A modular brain-on-a-chip for modelling epileptic seizures with functionally connected human neuronal networks. Biosens Bioelectron 2020; 168:112553. [DOI: 10.1016/j.bios.2020.112553] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/01/2020] [Accepted: 08/23/2020] [Indexed: 12/22/2022]
|