1
|
Spencer AP, Vilaça A, Xavier M, Santos R, Ionescu A, Lázaro M, Leiro V, Perlson E, Guimarães SC, Maoz BM, Pêgo AP. Engineered Chitosan-Derived Nanocarrier for Efficient siRNA Delivery to Peripheral and Central Neurons. Adv Healthc Mater 2025; 14:e2500107. [PMID: 40364633 DOI: 10.1002/adhm.202500107] [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: 01/07/2025] [Revised: 04/17/2025] [Indexed: 05/15/2025]
Abstract
Gene therapy using small interfering RNA (siRNA) holds promise for treating neurological disorders by silencing specific genes, like the phosphatase and tensin homolog (PTEN) gene, which restricts axonal growth. Effective siRNA delivery to neurons, however, poses challenges due to premature nucleic acid degradation and unspecific delivery. Chitosan-based delivery systems, noted for their biocompatibility, face limitations such as low transfection efficiency and lack of neurotropism. Building on the previous successes with neuron-targeted DNA delivery using chitosan, a novel approach for siRNA delivery aimed at PTEN downregulation is proposed. This involves using thiolated trimethyl chitosan (TMCSH)-based siRNA nanoparticles functionalized with the neurotropic C-terminal fragment of the tetanus neurotoxin heavy chain (HC) for efficient delivery to peripheral and central neurons. These polyplexes demonstrate suitable physicochemical properties, biocompatibility, and no adverse effects on neuronal electrophysiology. Diverse neuronal models, including 3D ex vivo cultures and microfluidics, confirm the polyplexes' efficiency and neurospecificity. HC-functionalization significantly enhances neuronal binding, and live cell imaging reveals fivefold faster retrograde transport along axons. Furthermore, siRNA delivery targeting PTEN promoted axonal outgrowth in embryonic cortical neurons. In conclusion, the proposed polyplexes represent a promising platform for neuronal siRNA delivery, offering potential for clinical translation and therapeutic applications.
Collapse
Affiliation(s)
- Ana P Spencer
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Adriana Vilaça
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - Miguel Xavier
- International Iberian Nanotechnology Laboratory (INL), Braga, Portugal
| | - Rafael Santos
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ariel Ionescu
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - María Lázaro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Victoria Leiro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Eran Perlson
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sofia C Guimarães
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ben M Maoz
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana P Pêgo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
2
|
Subbiahanadar Chelladurai K, Selvan Christyraj JD, Rajagopalan K, Vadivelu K, Chandrasekar M, Das P, Kalimuthu K, Balamurugan N, Subramanian V, Selvan Christyraj JRS. Ex vivo functional whole organ in biomedical research: a review. J Artif Organs 2025; 28:131-145. [PMID: 39592544 DOI: 10.1007/s10047-024-01478-4] [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: 02/07/2024] [Accepted: 07/29/2024] [Indexed: 11/28/2024]
Abstract
Model systems are critical in biomedical and preclinical research. Animal and in vitro models serve an important role in our current understanding of human physiology, disease pathophysiology, and therapy development. However, if the system is between cell culture and animal models, it may be able to overcome the knowledge gap that exists in the current system. Studies employing ex vivo organs as models have not been thoroughly investigated. Though the integration of other organs and systems has an impact on many biological mechanisms and disorders, it can add a new dimension to modeling and aid in the identification of new possible therapeutic targets. Here, we have discussed why the ex vivo organ model is desirable and the importance of the inclusion of organs from diverse species, described its historical aspects, studied organs as models in scientific research, and its ex vivo stability. We also discussed, how an ex vivo organ model might help researchers better understand organ physiology, as well as organ-specific diseases and therapeutic targets. We emphasized how this ex vivo organ dynamics will be more competent than existing models, as well as what tissues or organs would have potentially viable longevity for ex vivo modeling including human tissues, organs, and/or at least biopsies and its possible advantage in clinical medicine including organ transplantation procedure and precision medicine.
Collapse
Affiliation(s)
- Karthikeyan Subbiahanadar Chelladurai
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Jackson Durairaj Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India.
| | - Kamarajan Rajagopalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Kayalvizhi Vadivelu
- Department of Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Meikandan Chandrasekar
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Puja Das
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Kalishwaralal Kalimuthu
- Rajiv Gandhi Centre for Biotechnology, Department of Biotechnology, Thiruvananthapuram, Kerala, India
| | - Nivedha Balamurugan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Vijayalakshmi Subramanian
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India
| | - Johnson Retnaraj Samuel Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science & Technology, Chennai, Tamil Nadu, India.
| |
Collapse
|
3
|
Rodrigues Salles G, Granato AEC, Viero FT, Pacheco-Soares C, Ferreira ST, Porcionatto M, Ulrich H. Self-assembly and 3D Bioprinting of Neurospheres and Evaluation of Caffeine and Photobiomodulation Effects in an Alzheimer's Disease In Vitro Model. Stem Cell Rev Rep 2025; 21:988-1000. [PMID: 40198478 DOI: 10.1007/s12015-025-10850-7] [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] [Accepted: 02/03/2025] [Indexed: 04/10/2025]
Abstract
Several in vitro models of Alzheimer's disease (AD) rely on 2D cell culture, and, more recently, 3D cultures represented by free-floating neurospheres have been used as models for the disease. The advantage of 3D over 2D cell culture is that cell-extracellular matrix and cell-cell interactions can be assessed, better representing the molecular and cellular hallmarks of the disease. In the current study, we developed two complementary 3D neurosphere models using SH-SY5Y human neuroblastoma cells to investigate AD pathology and evaluate potential therapies. First, self-assembled neurospheres were exposed to hydrogen peroxide (H₂O₂) and amyloid-beta oligomers (AβOs), inducing AD-like features such as increased production of reactive oxygen species (ROS), amyloid aggregation, and apoptosis. Treatment with caffeine or photobiomodulation (PBM) using LED irradiation significantly reduced Aβ1-42 accumulation, ROS generation, and decreased apoptosis markers. Second, 3D bioprinting of SH-SY5Y cells resulted in neurospheres with enhanced cellular organization and differentiation. These findings emphasize the advantages of 3D models for studying neurodegeneration and evaluating therapeutic strategies, bridging the gap between traditional 2D cultures and complex in vitro systems.
Collapse
Affiliation(s)
- Geisa Rodrigues Salles
- Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, SP, Brazil
- Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, SP, Brazil
| | - Alessandro E C Granato
- Department of Dental Materials and Prosthodontics, Institute of Science and Technology of São José dos Campos, São Paulo State University (UNESP), São José dos Campos, SP, Brazil
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, SP, Brazil
| | - Fernanda Tibolla Viero
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, SP, Brazil
| | - Cristina Pacheco-Soares
- Institute of Research and Development, University of Vale do Paraíba, São José dos Campos, SP, Brazil
| | - Sérgio T Ferreira
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- D'Or Institute for Research and Education, Rio de Janeiro, RJ, Brazil
| | - Marimelia Porcionatto
- Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, SP, Brazil
- National Institute of Science and Technology in Modeling Human Complex Diseases with 3D Platforms (INCT Model3D), São Paulo, SP, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, 05508-900, SP, Brazil.
| |
Collapse
|
4
|
Wang T, Li MY, Pei Z, Chen QX, Cheng QS, Li Z. Down-regulation of platelet-derived growth factor receptor β in pericytes increases blood-brain barrier permeability and significantly enhances α-synuclein in a Parkinson's Disease 3D cell model in vitro under hyperglycemic condition. Tissue Cell 2025; 93:102751. [PMID: 39847894 DOI: 10.1016/j.tice.2025.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/24/2024] [Accepted: 01/15/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Parkinson's Disease (PD) often presents with a compromised blood-brain barrier (BBB), which hyperglycemia may exacerbate. Pericytes, a key cell for BBB integrity, are potential therapeutic targets for neurodegenerative disorders. Few studies have developed 3D PD cell models incorporating neurovascular units (NVU) through the co-culture of human endothelial, pericytes, astrocytes, and SH-SY5Y cells to evaluate BBB impairment and the role of pericytes under hyperglycemic condition. METHOD A 3D PD like cell model was developed using 6-OHDA-affected SH-SY5Y cells, combined with endothelial cells, pericytes, and astrocytes through the Real Architecture for Tissue (RAFT) 3D co-culture system. PD incorporating reduced (30 % and 89 %) PDGFRβ NVU (RPN) with or without hyperglycemic model (HM) were also established. BBB permeability to sodium fluorescein was assessed, and BBB impairment was evaluated using BBB-associated proteins (ZO-1, CD54, CD144), cell-specific proteins (CD31, GFAP, PDGFRβ, CD13), tyrosine hydroxylase (TH), α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9). RESULTS PD 3D cell models incorporating RPN with or without hyperglycemia were successfully established in vitro. Graduately increased BBB impairment was observed in PD, PD with RPN, and PD with RPN combined with HM, indicated by decreased BBB-associated and cell-specific proteins, reduced TH, and increased α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9) compared to the NVU model. CONCLUSION Reduced pericyte PDGFRβ could increase BBB permeability, accelerate PD progression, and exacerbate under hyperglycemic condition.
Collapse
Affiliation(s)
- Ting Wang
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| | - Meng-Yan Li
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road II, Guangzhou 510080, PR China.
| | - Qiu-Xia Chen
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China; Guangdong Medical University, Zhanjiang, Guangdong 524023, PR China.
| | - Qiu-Sheng Cheng
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| | - Ze Li
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong 510282, PR China.
| |
Collapse
|
5
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
6
|
Mateus JC, Melo P, Aroso M, Charlot B, Aguiar P. Influence of asymmetric microchannels in the structure and function of engineered neuronal circuits. Biofabrication 2025; 17:025022. [PMID: 39908667 DOI: 10.1088/1758-5090/adb2e5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/05/2025] [Indexed: 02/07/2025]
Abstract
Understanding the intricate structure-function relationships of neuronal circuits is crucial for unraveling how the brain achieves efficient information transfer. In specific brain regions, like the hippocampus, neurons are organized in layers and form unidirectional connectivity, which is thought to help ensure controlled signal flow and information processing. In recent years, researchers have tried emulating these structural principles by providing cultured neurons with asymmetric environmental cues, namely microfluidics' microchannels, which promote directed axonal growth. Even though a few reports have claimed to achieve unidirectional connectivity ofin vitroneuronal circuits, given the lack of functional characterization, it remains unknown if this structural connectivity correlates with functional connectivity. We have replicated and tested the performance of asymmetric microchannel designs previously reported in the literature to be successful in promoting directed axonal growth, as well as other custom variations. A new variation of 'Arrowhead', termed 'Rams', was the best-performing motif with a ∼76% probability per microchannel of allowing strictly unidirectional connections at 14 din vitro. Importantly, we assessed the functional implications of these different asymmetric microchannel designs. For this purpose, we combined custom microfluidics with microelectrode array technology to record the electrophysiological activity of two segregated populations of hippocampal neurons ('Source' and 'Target'). This functional characterization revealed that up to ∼94% of the spiking activity recorded along microchannels with the 'Rams' motif propagates towards the 'Target' population. Moreover, our results indicate that these engineered circuits also tended to exhibit network-level synchronizations with defined directionality. Overall, this functional characterization of the structure-function relationships promoted by asymmetric microchannels has the potential to provide insights into how neuronal circuits use specific network architectures for effective computations. Moreover, the here-developed devices and approaches may be used in a wide range of applications, such as disease modeling or preclinical drug screening.
Collapse
Affiliation(s)
- J C Mateus
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - P Melo
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FEUP-Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - M Aroso
- INEB-Instituto Nacional de Engenharia Biomédica, Porto, Portugal
| | - B Charlot
- Institut d'Electronique et des Systèmes, CNRS, University Montpellier, Montpellier, France
| | - P Aguiar
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- FEUP-Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| |
Collapse
|
7
|
Kala S, Strutz AG, Katt ME. The Rise of Pluripotent Stem Cell-Derived Glia Models of Neuroinflammation. Neurol Int 2025; 17:6. [PMID: 39852770 PMCID: PMC11767680 DOI: 10.3390/neurolint17010006] [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: 11/20/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation is a blanket term that describes the body's complex inflammatory response in the central nervous system (CNS). It encompasses a phenotype shift to a proinflammatory state, the release of cytokines, the recruitment of peripheral immune cells, and a wide variety of other processes. Neuroinflammation has been implicated in nearly every major CNS disease ranging from Alzheimer's disease to brain cancer. Understanding and modeling neuroinflammation is critical for the identification of novel therapeutic targets in the treatment of CNS diseases. Unfortunately, the translation of findings from non-human models has left much to be desired. This review systematically discusses the role of human pluripotent stem cell (hPSC)-derived glia and supporting cells within the CNS, including astrocytes, microglia, oligodendrocyte precursor cells, pericytes, and endothelial cells, to describe the state of the field and hope for future discoveries. hPSC-derived cells offer an expanded potential to study the pathobiology of neuroinflammation and immunomodulatory cascades that impact disease progression. While much progress has been made in the development of models, there is much left to explore in the application of these models to understand the complex inflammatory response in the CNS.
Collapse
Affiliation(s)
- Srishti Kala
- Cancer Cell Biology Graduate Education Program, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Andrew G. Strutz
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Moriah E. Katt
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| |
Collapse
|
8
|
Aili D, Herland A. Engineered Hydrogels for 3D Cell Culture and Bioprinting of Human Induced Pluripotent Stem Cell-Derived Neuroepithelial Stem Cells. Methods Mol Biol 2025; 2924:223-233. [PMID: 40307646 DOI: 10.1007/978-1-0716-4530-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
This protocol outlines the synthesis and use of engineered hyaluronan-based hydrogels for 3D cell culture and bioprinting of human induced pluripotent stem cell (hiPSC)-derived neuroepithelial stem cells (lt-NES). Key steps include hydrogel formation using bioorthogonal chemistries, cell encapsulation, and 3D bioprinting with a Cellink BioX printer, enabling the creation of complex tissue models. The protocol ensures high cell viability and supports differentiation, essential for neuroscience research and drug development.
Collapse
Affiliation(s)
- Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.
| | - Anna Herland
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
- Division of Nanobiotechnology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology at Science for Life Laboratory, Solna, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
9
|
Zhao Y, Zhang C, Liang C, Lv X, Zhou D, Deng Y, Zhang R. On Multicell-Interaction Chip: In Situ Observing the Interactions between the Astrocytes with Lysosomal Dysfunction and BBB Cells. Anal Chem 2024; 96:20057-20064. [PMID: 39642021 DOI: 10.1021/acs.analchem.4c04893] [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: 12/08/2024]
Abstract
Lysosomes in astrocytes play vital roles in toxic protein degradation in the brain. Lysosomal dysfunction can lead to abnormal protein deposits, which further induce damage to neurons and the blood-brain barrier (BBB), and thereby affect the interaction between the nervous and vascular systems. Therefore, investigating the interactions between astrocytes with lysosomal dysfunction and BBB cells is of significant importance. However, the lack of effective in vitro models hinders the study of this complex system. Herein, an 8-well arrayed microfence multicell interculture chip (AMMIC) with a hydrophilically optimized surface is introduced for investigating the interactions between astrocytes and BBB cells. Then, a novel lysosome-targeted photosensitizer, IVQ-2Br, is synthesized for inducing controllable oxidative stress damage in the lysosomes of astrocytes. By the combination of the 8-well AMMIC and IVQ-2Br, a model for studying the interactions between astrocytes with lysosomal dysfunction and BBB cells has been constructed. Particularly, severe secondary injuries to BBB cells brought about by oxidative stress, including alterations in cell morphology and activity as well as notable DNA damage, are in situ observed on the 8-well AMMIC. The mediators involved in this oxidative stress injury-mediated intercellular communication are validated to be reactive oxygen species (ROS) and exosomes. This work not only presents an in vitro modeling method for studying cell-cell interactions but also demonstrates the potential of in vitro models constructed through the integration of complex microfluidic chip techniques and photosensitizers for advancing biomedical research.
Collapse
Affiliation(s)
- Yimeng Zhao
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Chen Zhang
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Chaohui Liang
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Xuefei Lv
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Di Zhou
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yulin Deng
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Ruoyao Zhang
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
10
|
Di Lisa D, Andolfi A, Masi G, Uras G, Ferrari PF, Martinoia S, Pastorino L. Impact of perfusion on neuronal development in human derived neuronal networks. APL Bioeng 2024; 8:046102. [PMID: 39364213 PMCID: PMC11446581 DOI: 10.1063/5.0221911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
Advanced in vitro models of the brain have evolved in recent years from traditional two-dimensional (2D) ones, based on rodent derived cells, to three-dimensional (3D) ones, based on human neurons derived from induced pluripotent stem cells. To address the dynamic changes of the tissue microenvironment, bioreactors are used to control the in vitro microenvironment for viability, repeatability, and standardization. However, in neuronal tissue engineering, bioreactors have primarily been used for cell expansion purposes, while microfluidic systems have mainly been employed for culturing organoids. In this study, we explored the use of a commercial perfusion bioreactor to control the culture microenvironment of neuronal cells in both 2D and 3D cultures. Namely, neurons differentiated from human induced pluripotent stem cells (iNeurons) were cultured in 2D under different constant flow rates for 72 h. The impact of different flow rates on early-stage neuronal development and synaptogenesis was assessed by morphometric characterization and synaptic analysis. Based on these results, two involving variable flow rates were developed and applied again in 2D culture. The most effective protocol, in terms of positive impact on neuronal development, was then used for a preliminary study on the application of dynamic culturing conditions to neuronal cells in 3D. To this purpose, both iNeurons, co-cultured with astrocytes, and the human neuroblastoma cells SH-SY5Y were embedded into a hydrogel and maintained under perfusion for up to 28 days. A qualitative evaluation by immunocytochemistry and confocal microscopy was carried out to assess cell morphology and the formation of a 3D neuronal network.
Collapse
Affiliation(s)
| | - Andrea Andolfi
- DIBRIS, Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Via Opera Pia 13, 16145 Genoa, Italy
| | - Giacomo Masi
- DIBRIS, Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Via Opera Pia 13, 16145 Genoa, Italy
| | | | | | | | | |
Collapse
|
11
|
Nagai H, Saito M, Iwata H. Direct conversion of urine-derived cells into functional motor neuron-like cells by defined transcription factors. Sci Rep 2024; 14:27011. [PMID: 39505927 PMCID: PMC11541886 DOI: 10.1038/s41598-024-73759-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 11/08/2024] Open
Abstract
Direct cell-type conversion of somatic cells into cell types of interest has garnered great attention because it circumvents rejuvenation and preserves the hallmarks of cellular aging (unlike induced pluripotent stem cells [iPSCs]) and is more suitable for modeling diseases with strong age-related and epigenetic contributions. Fibroblasts are commonly used for direct conversion; however, obtaining these cells requires highly invasive skin biopsies. Urine-derived cells (UDCs) are an alternative cell source and can be obtained via noninvasive procedures. Herein, induced motor neuron-like cells (iMNs) were generated from UDCs by transducing transcription factors involved in motor neuron (MN) differentiation. iMNs exhibited neuronal morphology, upregulation of pan-neuron and MN markers, and MN functionality, including spontaneous calcium oscillation and bungarotoxin-positive neuromuscular junction formation, when co-cultured with myotubes. Altogether, the findings of this study indicated that UDCs can be converted to functional MNs. This technology may allow us to understand disease pathogenesis and progression and discover biomarkers and drugs for MN-related diseases at the population level.
Collapse
Affiliation(s)
- Hiroaki Nagai
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Kanagawa, Japan.
| | - Masayo Saito
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Kanagawa, Japan
| | - Hidehisa Iwata
- Neuroscience Drug Discovery Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, 251-8555, Kanagawa, Japan.
| |
Collapse
|
12
|
Chea M, Bouvier S, Gris JC. The hemostatic system in chronic brain diseases: A new challenging frontier? Thromb Res 2024; 243:109154. [PMID: 39305718 DOI: 10.1016/j.thromres.2024.109154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
Neurological diseases (ND), including neurodegenerative diseases (NDD) and psychiatric disorders (PD), present a significant public health challenge, ranking third in Europe for disability and premature death, following cardiovascular diseases and cancers. In 2017, approximately 540 million cases of ND were reported among Europe's 925 million people, with strokes, dementia, and headaches being most prevalent. Nowadays, more and more evidence highlight the hemostasis critical role in cerebral homeostasis and vascular events. Indeed, hemostasis, thrombosis, and brain abnormalities contributing to ND form a complex and poorly understood equilibrium. Alterations in vascular biology, particularly involving the blood-brain barrier, are implicated in ND, especially dementia, and PD. While the roles of key coagulation players such as thrombin and fibrinogen are established, the roles of other hemostasis components are less clear. Moreover, the involvement of these elements in psychiatric disease pathogenesis is virtually unstudied, except in specific pathological models such as antiphospholipid syndrome. Advanced imaging techniques, primarily functional magnetic resonance imaging and its derivatives like diffusion tensor imaging, have been developed to study brain areas affected by ND and to improve our understanding of the pathophysiology of these diseases. This literature review aims to clarify the current understanding of the connections between hemostasis, thrombosis, and neurological diseases, as well as explore potential future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mathias Chea
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France.
| | - Sylvie Bouvier
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France
| | - Jean-Christophe Gris
- Department of Hematology, Nîmes University Hospital, Place du Professeur Robert Debré, Nîmes, France; Desbrest Institute of Epidemiology and Public Health, Univ Montpellier, INSERM, University of Montpellier, Montpellier, France; Faculty of Pharmaceutical and Biological Sciences, University of Montpellier, Montpellier, France; I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
13
|
Ghiasvand K, Amirfazli M, Moghimi P, Safari F, Takhshid MA. The role of neuron-like cell lines and primary neuron cell models in unraveling the complexity of neurodegenerative diseases: a comprehensive review. Mol Biol Rep 2024; 51:1024. [PMID: 39340590 DOI: 10.1007/s11033-024-09964-x] [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: 05/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs) are characterized by the progressive loss of neurons. As to developing effective therapeutic interventions, it is crucial to understand the underlying mechanisms of NDs. Cellular models have become invaluable tools for studying the complex pathogenesis of NDs, offering insights into disease mechanisms, determining potential therapeutic targets, and aiding in drug discovery. This review provides a comprehensive overview of various cellular models used in ND research, focusing on Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Cell lines, such as SH-SY5Y and PC12 cells, have emerged as valuable tools due to their ease of use, reproducibility, and scalability. Additionally, co-culture models, involving the growth of distinct cell types like neurons and astrocytes together, are highlighted for simulating brain interactions and microenvironment. While cell lines cannot fully replicate the complexity of the human brain, they provide a scalable method for examining important aspects of neurodegenerative diseases. Advancements in cell line technologies, including the incorporation of patient-specific genetic variants and improved co-culture models, hold promise for enhancing our understanding and expediting the development of effective treatments. Integrating multiple cellular models and advanced technologies offers the potential for significant progress in unraveling the intricacies of these debilitating diseases and improving patient outcomes.
Collapse
Affiliation(s)
- Kianoush Ghiasvand
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Amirfazli
- School of biological sciences, Illinois State University, Normal, United States of America
| | - Parvaneh Moghimi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
14
|
Shan X, Zhang A, Rezzonico MG, Tsai MC, Sanchez-Priego C, Zhang Y, Chen MB, Choi M, Andrade López JM, Phu L, Cramer AL, Zhang Q, Pattison JM, Rose CM, Hoogenraad CC, Jeong CG. Fully defined NGN2 neuron protocol reveals diverse signatures of neuronal maturation. CELL REPORTS METHODS 2024; 4:100858. [PMID: 39255791 PMCID: PMC11440061 DOI: 10.1016/j.crmeth.2024.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 04/26/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
NGN2-driven induced pluripotent stem cell (iPSC)-to-neuron conversion is a popular method for human neurological disease modeling. In this study, we present a standardized approach for generating neurons utilizing clonal, targeted-engineered iPSC lines with defined reagents. We demonstrate consistent production of excitatory neurons at scale and long-term maintenance for at least 150 days. Temporal omics, electrophysiological, and morphological profiling indicate continued maturation to postnatal-like neurons. Quantitative characterizations through transcriptomic, imaging, and functional assays reveal coordinated actions of multiple pathways that drive neuronal maturation. We also show the expression of disease-related genes in these neurons to demonstrate the relevance of our protocol for modeling neurological disorders. Finally, we demonstrate efficient generation of NGN2-integrated iPSC lines. These workflows, profiling data, and functional characterizations enable the development of reproducible human in vitro models of neurological disorders.
Collapse
Affiliation(s)
- Xiwei Shan
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ai Zhang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Mitchell G Rezzonico
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ming-Chi Tsai
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Yingjie Zhang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Michelle B Chen
- Department of Cellular and Tissue Genomics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Meena Choi
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Lilian Phu
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Amber L Cramer
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Qiao Zhang
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Jillian M Pattison
- Advanced Cell Engineering, Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Christopher M Rose
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Claire G Jeong
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
15
|
Pramotton FM, Spitz S, Kamm RD. Challenges and Future Perspectives in Modeling Neurodegenerative Diseases Using Organ-on-a-Chip Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403892. [PMID: 38922799 PMCID: PMC11348103 DOI: 10.1002/advs.202403892] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/01/2024] [Indexed: 06/28/2024]
Abstract
Neurodegenerative diseases (NDDs) affect more than 50 million people worldwide, posing a significant global health challenge as well as a high socioeconomic burden. With aging constituting one of the main risk factors for some NDDs such as Alzheimer's disease (AD) and Parkinson's disease (PD), this societal toll is expected to rise considering the predicted increase in the aging population as well as the limited progress in the development of effective therapeutics. To address the high failure rates in clinical trials, legislative changes permitting the use of alternatives to traditional pre-clinical in vivo models are implemented. In this regard, microphysiological systems (MPS) such as organ-on-a-chip (OoC) platforms constitute a promising tool, due to their ability to mimic complex and human-specific tissue niches in vitro. This review summarizes the current progress in modeling NDDs using OoC technology and discusses five critical aspects still insufficiently addressed in OoC models to date. Taking these aspects into consideration in the future MPS will advance the modeling of NDDs in vitro and increase their translational value in the clinical setting.
Collapse
Affiliation(s)
- Francesca Michela Pramotton
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah Spitz
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
16
|
Rotaru-Zăvăleanu AD, Dinescu VC, Aldea M, Gresita A. Hydrogel-Based Therapies for Ischemic and Hemorrhagic Stroke: A Comprehensive Review. Gels 2024; 10:476. [PMID: 39057499 PMCID: PMC11276304 DOI: 10.3390/gels10070476] [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: 06/07/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Stroke remains the second leading cause of death and a major cause of disability worldwide, significantly impacting individuals, families, and healthcare systems. This neurological emergency can be triggered by ischemic events, including small vessel arteriolosclerosis, cardioembolism, and large artery atherothromboembolism, as well as hemorrhagic incidents resulting from macrovascular lesions, venous sinus thrombosis, or vascular malformations, leading to significant neuronal damage. The resultant motor impairment, cognitive dysfunction, and emotional disturbances underscore the urgent need for effective therapeutic interventions. Recent advancements in biomaterials, particularly hydrogels, offer promising new avenues for stroke management. Hydrogels, composed of three-dimensional networks of hydrophilic polymers, are notable for their ability to absorb and retain substantial amounts of water. Commonly used polymers in hydrogel formulations include natural polymers like alginate, chitosan, and collagen, as well as synthetic polymers such as polyethylene glycol (PEG), polyvinyl alcohol (PVA), and polyacrylamide. Their customizable characteristics-such as their porosity, swelling behavior, mechanical strength, and degradation rates-make hydrogels ideal for biomedical applications, including drug delivery, cell delivery, tissue engineering, and the controlled release of therapeutic agents. This review comprehensively explores hydrogel-based approaches to both ischemic and hemorrhagic stroke therapy, elucidating the mechanisms by which hydrogels provide neuroprotection. It covers their application in drug delivery systems, their role in reducing inflammation and secondary injury, and their potential to support neurogenesis and angiogenesis. It also discusses current advancements in hydrogel technology and the significant challenges in translating these innovations from research into clinical practice. Additionally, it emphasizes the limited number of clinical trials utilizing hydrogel therapies for stroke and addresses the associated limitations and constraints, underscoring the need for further research in this field.
Collapse
Affiliation(s)
- Alexandra-Daniela Rotaru-Zăvăleanu
- Department of Epidemiology, University of Medicine and Pharmacy of Craiova, 2-4 Petru Rares Str., 200349 Craiova, Romania;
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Venera Cristina Dinescu
- Department of Health Promotion and Occupational Medicine, University of Medicine and Pharmacy of Craiova, 2–4 Petru Rares Str., 200349 Craiova, Romania
| | - Madalina Aldea
- Psychiatry Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Andrei Gresita
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 115680, USA
| |
Collapse
|
17
|
Rogal J, Zamproni LN, Nikolakopoulou P, Ygberg S, Wedell A, Wredenberg A, Herland A. Human In Vitro Models of Neuroenergetics and Neurometabolic Disturbances: Current Advances and Clinical Perspectives. Stem Cells Transl Med 2024; 13:505-514. [PMID: 38588471 PMCID: PMC11165162 DOI: 10.1093/stcltm/szae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024] Open
Abstract
Neurological conditions conquer the world; they are the leading cause of disability and the second leading cause of death worldwide, and they appear all around the world in every age group, gender, nationality, and socioeconomic class. Despite the growing evidence of an immense impact of perturbations in neuroenergetics on overall brain function, only little is known about the underlying mechanisms. Especially human insights are sparse, owing to a shortage of physiologically relevant model systems. With this perspective, we aim to explore the key steps and considerations involved in developing an advanced human in vitro model for studying neuroenergetics. We discuss biological and technological strategies to meet the requirements of a predictive model, aiming at providing a guide and inspiration for future in vitro models of neuroenergetics.
Collapse
Affiliation(s)
- Julia Rogal
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Division of Nanobiotechnology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology at Science for Life Laboratory, 17165 Solna, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, 17177 Stockholm, Sweden
| | - Laura Nicoleti Zamproni
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04039-032, Brazil
| | - Polyxeni Nikolakopoulou
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, 17177 Stockholm, Sweden
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Sofia Ygberg
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
- Neuropediatric Unit, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Anna Wedell
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institute, 17177 Stockholm, Sweden
| | - Anna Wredenberg
- Centre for Inherited Metabolic Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177 Stockholm, Sweden
| | - Anna Herland
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
- Division of Nanobiotechnology, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology at Science for Life Laboratory, 17165 Solna, Sweden
- Center for the Advancement of Integrated Medical and Engineering Sciences (AIMES), Karolinska Institute and KTH Royal Institute of Technology, 17177 Stockholm, Sweden
| |
Collapse
|
18
|
Li X, Wang M, Davis TP, Zhang L, Qiao R. Advancing Tissue Culture with Light-Driven 3D-Printed Microfluidic Devices. BIOSENSORS 2024; 14:301. [PMID: 38920605 PMCID: PMC11201418 DOI: 10.3390/bios14060301] [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: 04/29/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024]
Abstract
Three-dimensional (3D) printing presents a compelling alternative for fabricating microfluidic devices, circumventing certain limitations associated with traditional soft lithography methods. Microfluidics play a crucial role in the biomedical sciences, particularly in the creation of tissue spheroids and pharmaceutical research. Among the various 3D printing techniques, light-driven methods such as stereolithography (SLA), digital light processing (DLP), and photopolymer inkjet printing have gained prominence in microfluidics due to their rapid prototyping capabilities, high-resolution printing, and low processing temperatures. This review offers a comprehensive overview of light-driven 3D printing techniques used in the fabrication of advanced microfluidic devices. It explores biomedical applications for 3D-printed microfluidics and provides insights into their potential impact and functionality within the biomedical field. We further summarize three light-driven 3D printing strategies for producing biomedical microfluidic systems: direct construction of microfluidic devices for cell culture, PDMS-based microfluidic devices for tissue engineering, and a modular SLA-printed microfluidic chip to co-culture and monitor cells.
Collapse
Affiliation(s)
| | | | | | - Liwen Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Ruirui Qiao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
19
|
Shariff S, Kantawala B, Xochitun Gopar Franco W, Dejene Ayele N, Munyangaju I, Esam Alzain F, Nazir A, Wojtara M, Uwishema O. Tailoring epilepsy treatment: personalized micro-physiological systems illuminate individual drug responses. Ann Med Surg (Lond) 2024; 86:3557-3567. [PMID: 38846814 PMCID: PMC11152789 DOI: 10.1097/ms9.0000000000002078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/09/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Approximately 50 million people worldwide have epilepsy, with many not achieving seizure freedom. Organ-on-chip technology, which mimics organ-level physiology, could revolutionize drug development for epilepsy by replacing animal models in preclinical studies. The authors' goal is to determine if customized micro-physiological systems can lead to tailored drug treatments for epileptic patients. Materials and methods A comprehensive literature search was conducted utilizing various databases, including PubMed, Ebscohost, Medline, and the National Library of Medicine, using a predetermined search strategy. The authors focused on articles that addressed the role of personalized micro-physiological systems in individual drug responses and articles that discussed different types of epilepsy, diagnosis, and current treatment options. Additionally, articles that explored the components and design considerations of micro-physiological systems were reviewed to identify challenges and opportunities in drug development for challenging epilepsy cases. Results The micro-physiological system offers a more accurate and cost-effective alternative to traditional models for assessing drug effects, toxicities, and disease mechanisms. Nevertheless, designing patient-specific models presents critical considerations, including the integration of analytical biosensors and patient-derived cells, while addressing regulatory, material, and biological complexities. Material selection, standardization, integration of vascular systems, cost efficiency, real-time monitoring, and ethical considerations are also crucial to the successful use of this technology in drug development. Conclusion The future of organ-on-chip technology holds great promise, with the potential to integrate artificial intelligence and machine learning for personalized treatment of epileptic patients.
Collapse
Affiliation(s)
- Sanobar Shariff
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - Burhan Kantawala
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - William Xochitun Gopar Franco
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- University of Guadalajara, Guadalajara, Mexico
| | - Nitsuh Dejene Ayele
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Internal Medicine, Faculty of Medicine, Wolkite University, Wolkite, Ethiopia
| | - Isabelle Munyangaju
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Medicine and General Surgery, Sudan University Of Science and Technology, Khartoum, Sudan
| | - Fatima Esam Alzain
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Medicine and General Surgery, Sudan University Of Science and Technology, Khartoum, Sudan
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Madga Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Clinton Global Initiative University, New York, NY
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
20
|
Rahav N, Marrero D, Soffer A, Glickman E, Beldjilali‐Labro M, Yaffe Y, Tadmor K, Leichtmann‐Bardoogo Y, Ashery U, Maoz BM. Multi-Sensor Origami Platform: A Customizable System for Obtaining Spatiotemporally Precise Functional Readouts in 3D Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305555. [PMID: 38634605 PMCID: PMC11200086 DOI: 10.1002/advs.202305555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/14/2024] [Indexed: 04/19/2024]
Abstract
Bioprinting technology offers unprecedented opportunities to construct in vitro tissue models that recapitulate the 3D morphology and functionality of native tissue. Yet, it remains difficult to obtain adequate functional readouts from such models. In particular, it is challenging to position sensors in desired locations within pre-fabricated 3D bioprinted structures. At the same time, bioprinting tissue directly onto a sensing device is not feasible due to interference with the printer head. As such, a multi-sensing platform inspired by origami that overcomes these challenges by "folding" around a separately fabricated 3D tissue structure is proposed, allowing for the insertion of electrodes into precise locations, which are custom-defined using computer-aided-design software. The multi-sensing origami platform (MSOP) can be connected to a commercial multi-electrode array (MEA) system for data-acquisition and processing. To demonstrate the platform, how integrated 3D MEA electrodes can record neuronal electrical activity in a 3D model of a neurovascular unit is shown. The MSOP also enables a microvascular endothelial network to be cultured separately and integrated with the 3D tissue structure. Accordingly, how impedance-based sensors in the platform can measure endothelial barrier function is shown. It is further demonstrated the device's versatility by using it to measure neuronal activity in brain organoids.
Collapse
Affiliation(s)
- Noam Rahav
- School of Neurobiology, Biochemistry and BiophysicsThe George S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv69978Israel
| | - Denise Marrero
- Instituto de Microelectrónica de Barcelona (IMB‐CNM, CSIC)Campus UABBellaterraBarcelona08193Spain
- Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y NanomedicinaMadrid50018Spain
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Adi Soffer
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | - Emma Glickman
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
| | | | - Yakey Yaffe
- Sagol Center for Regenerative MedicineTel Aviv UniversityTel Aviv69978Israel
| | - Keshet Tadmor
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | | | - Uri Ashery
- School of Neurobiology, Biochemistry and BiophysicsThe George S. Wise Faculty of Life SciencesTel Aviv UniversityTel Aviv69978Israel
- Sagol Center for Regenerative MedicineTel Aviv UniversityTel Aviv69978Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
| | - Ben M. Maoz
- Department of Biomedical EngineeringTel Aviv UniversityTel Aviv69978Israel
- Sagol Center for Regenerative MedicineTel Aviv UniversityTel Aviv69978Israel
- Sagol School of NeuroscienceTel Aviv UniversityTel Aviv69978Israel
- The Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv69978Israel
| |
Collapse
|
21
|
Boylin K, Aquino GV, Purdon M, Abedi K, Kasendra M, Barrile R. Basic models to advanced systems: harnessing the power of organoids-based microphysiological models of the human brain. Biofabrication 2024; 16:032007. [PMID: 38749420 DOI: 10.1088/1758-5090/ad4c08] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Understanding the complexities of the human brain's function in health and disease is a formidable challenge in neuroscience. While traditional models like animals offer valuable insights, they often fall short in accurately mirroring human biology and drug responses. Moreover, recent legislation has underscored the need for more predictive models that more accurately represent human physiology. To address this requirement, human-derived cell cultures have emerged as a crucial alternative for biomedical research. However, traditional static cell culture models lack the dynamic tissue microenvironment that governs human tissue function. Advancedin vitrosystems, such as organoids and microphysiological systems (MPSs), bridge this gap by offering more accurate representations of human biology. Organoids, which are three-dimensional miniaturized organ-like structures derived from stem cells, exhibit physiological responses akin to native tissues, but lack essential tissue-specific components such as functional vascular structures and immune cells. Recent endeavors have focused on incorporating endothelial cells and immune cells into organoids to enhance vascularization, maturation, and disease modeling. MPS, including organ-on-chip technologies, integrate diverse cell types and vascularization under dynamic culture conditions, revolutionizing brain research by bridging the gap betweenin vitroandin vivomodels. In this review, we delve into the evolution of MPS, with a particular focus on highlighting the significance of vascularization in enhancing the viability, functionality, and disease modeling potential of organoids. By examining the interplay of vasculature and neuronal cells within organoids, we can uncover novel therapeutic targets and gain valuable insights into disease mechanisms, offering the promise of significant advancements in neuroscience and improved patient outcomes.
Collapse
Affiliation(s)
- Katherine Boylin
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Grace V Aquino
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Michael Purdon
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Kimia Abedi
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Magdalena Kasendra
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Riccardo Barrile
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
22
|
Asimakidou E, Tan JKS, Zeng J, Lo CH. Blood-Brain Barrier-Targeting Nanoparticles: Biomaterial Properties and Biomedical Applications in Translational Neuroscience. Pharmaceuticals (Basel) 2024; 17:612. [PMID: 38794182 PMCID: PMC11123901 DOI: 10.3390/ph17050612] [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: 03/26/2024] [Revised: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Overcoming the blood-brain barrier (BBB) remains a significant hurdle in effective drug delivery to the brain. While the BBB serves as a crucial protective barrier, it poses challenges in delivering therapeutic agents to their intended targets within the brain parenchyma. To enhance drug delivery for the treatment of neurological diseases, several delivery technologies to circumvent the BBB have been developed in the last few years. Among them, nanoparticles (NPs) are one of the most versatile and promising tools. Here, we summarize the characteristics of NPs that facilitate BBB penetration, including their size, shape, chemical composition, surface charge, and importantly, their conjugation with various biological or synthetic molecules such as glucose, transferrin, insulin, polyethylene glycol, peptides, and aptamers. Additionally, we discuss the coating of NPs with surfactants. A comprehensive overview of the common in vitro and in vivo models of the BBB for NP penetration studies is also provided. The discussion extends to discussing BBB impairment under pathological conditions and leveraging BBB alterations under pathological conditions to enhance drug delivery. Emphasizing the need for future studies to uncover the inherent therapeutic properties of NPs, the review advocates for their role beyond delivery systems and calls for efforts translating NPs to the clinic as therapeutics. Overall, NPs stand out as a highly promising therapeutic strategy for precise BBB targeting and drug delivery in neurological disorders.
Collapse
Affiliation(s)
- Evridiki Asimakidou
- Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK;
| | - Justin Kok Soon Tan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore;
- The N.1 Institute for Health, National University of Singapore, Singapore 117456, Singapore
| | - Jialiu Zeng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Chih Hung Lo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| |
Collapse
|
23
|
Cattane N, Di Benedetto MG, D'Aprile I, Riva MA, Cattaneo A. Dissecting the Long-Term Effect of Stress Early in Life on FKBP5: The Role of miR-20b-5p and miR-29c-3p. Biomolecules 2024; 14:371. [PMID: 38540789 PMCID: PMC10967956 DOI: 10.3390/biom14030371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 04/04/2024] Open
Abstract
Exposure to early-life stress (ELS) has been related to an increased susceptibility to psychiatric disorders later in life. Although the molecular mechanisms underlying this association are still under investigation, glucocorticoid signaling has been proposed to be a key mediator. Here, we used two preclinical models, the prenatal stress (PNS) animal model and an in vitro model of hippocampal progenitor cells, to assess the long-term effect of ELS on FKBP5, NR3C1, NR3C2, and FoxO1, four stress-responsive genes involved in the effects of glucocorticoids. In the hippocampus of male PNS rats sacrificed at different time points during neurodevelopment (PND 21, 40, 62), we found a statistically significant up-regulation of FKBP5 at PND 40 and PND 62 and a significant increase in FoxO1 at PND 62. Interestingly, all four genes were significantly up-regulated in differentiated cells treated with cortisol during cell proliferation. As FKBP5 was consistently modulated by PNS at adolescence (PND 40) and adulthood (PND 62) and by cortisol treatment after cell differentiation, we measured a panel of miRNAs targeting FKBP5 in the same samples where FKBP5 expression levels were available. Interestingly, both miR-20b-5p and miR-29c-3p were significantly reduced in PNS-exposed animals (both at PND40 and 62) and also in the in vitro model after cortisol exposure. Our results highlight the key role of miR-20b-5p and miR-29c-3p in sustaining the long-term effects of ELS on the stress response system, representing a mechanistic link possibly contributing to the enhanced stress-related vulnerability to mental disorders.
Collapse
Affiliation(s)
- Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Maria Grazia Di Benedetto
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Ilari D'Aprile
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Marco Andrea Riva
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
| |
Collapse
|
24
|
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
|
25
|
Salmina AB, Alexandrova OP, Averchuk AS, Korsakova SA, Saridis MR, Illarioshkin SN, Yurchenko SO. Current progress and challenges in the development of brain tissue models: How to grow up the changeable brain in vitro? J Tissue Eng 2024; 15:20417314241235527. [PMID: 38516227 PMCID: PMC10956167 DOI: 10.1177/20417314241235527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
In vitro modeling of brain tissue is a promising but not yet resolved problem in modern neurobiology and neuropharmacology. Complexity of the brain structure and diversity of cell-to-cell communication in (patho)physiological conditions make this task almost unachievable. However, establishment of novel in vitro brain models would ultimately lead to better understanding of development-associated or experience-driven brain plasticity, designing efficient approaches to restore aberrant brain functioning. The main goal of this review is to summarize the available data on methodological approaches that are currently in use, and to identify the most prospective trends in development of neurovascular unit, blood-brain barrier, blood-cerebrospinal fluid barrier, and neurogenic niche in vitro models. The manuscript focuses on the regulation of adult neurogenesis, cerebral microcirculation and fluids dynamics that should be reproduced in the in vitro 4D models to mimic brain development and its alterations in brain pathology. We discuss approaches that are critical for studying brain plasticity, deciphering the individual person-specific trajectory of brain development and aging, and testing new drug candidates in the in vitro models.
Collapse
Affiliation(s)
- Alla B Salmina
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Olga P Alexandrova
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | - Anton S Averchuk
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
- Bauman Moscow State Technical University, Moscow, Russia
| | | | | | | | | |
Collapse
|
26
|
Lisa DD, Muzzi L, Lagazzo A, Andolfi A, Martinoia S, Pastorino L. Long-term in vitroculture of 3D brain tissue model based on chitosan thermogel. Biofabrication 2023; 16:015011. [PMID: 37922538 DOI: 10.1088/1758-5090/ad0979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 11/03/2023] [Indexed: 11/07/2023]
Abstract
Methods for studying brain function and disease heavily rely onin vivoanimal models,ex-vivotissue slices, and 2D cell culture platforms. These methods all have limitations that significantly impact the clinical translatability of results. Consequently, models able to better recapitulate some aspects ofin vivohuman brain are needed as additional preclinical tools. In this context, 3D hydrogel-basedin vitromodels of the brain are considered promising tools. To create a 3D brain-on-a-chip model, a hydrogel capable of sustaining neuronal maturation over extended culture periods is required. Among biopolymeric hydrogels, chitosan-β-glycerophosphate (CHITO-β-GP) thermogels have demonstrated their versatility and applicability in the biomedical field over the years. In this study, we investigated the ability of this thermogel to encapsulate neuronal cells and support the functional maturation of a 3D neuronal network in long-term cultures. To the best of our knowledge, we demonstrated for the first time that CHITO-β-GP thermogel possesses optimal characteristics for promoting neuronal growth and the development of an electrophysiologically functional neuronal network derived from both primary rat neurons and neurons differentiated from human induced pluripotent stem cells (h-iPSCs) co-cultured with astrocytes. Specifically, two different formulations were firstly characterized by rheological, mechanical and injectability tests. Primary nervous cells and neurons differentiated from h-iPSCs were embedded into the two thermogel formulations. The 3D cultures were then deeply characterized by immunocytochemistry, confocal microscopy, and electrophysiological recordings, employing both 2D and 3D micro-electrode arrays. The thermogels supported the long-term culture of neuronal networks for up to 100 d. In conclusion, CHITO-β-GP thermogels exhibit excellent mechanical properties, stability over time under culture conditions, and bioactivity toward nervous cells. Therefore, they are excellent candidates as artificial extracellular matrices in brain-on-a-chip models, with applications in neurodegenerative disease modeling, drug screening, and neurotoxicity evaluation.
Collapse
Affiliation(s)
- Donatella Di Lisa
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Lorenzo Muzzi
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Alberto Lagazzo
- Department of Civil, Chemical and Environmental Engineering, University of Genoa, via Montallegro 1, Genoa, Italy
| | - Andrea Andolfi
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Sergio Martinoia
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| | - Laura Pastorino
- Department of Informatics, Bioengineering, Robotics and System Engineering, University of Genoa, Via all 'Opera Pia 13, 16145 Genoa, Italy
| |
Collapse
|
27
|
Sade O, Boneberg R, Weiss Y, Beldjilali-Labro M, Leichtmann-Bardoogo Y, Talpir I, Gottfried I, Ashery U, Rauti R, Maoz BM. Super-Resolution-Chip: an in-vitro platform that enables super-resolution microscopy of co-cultures and 3D systems. BIOMEDICAL OPTICS EXPRESS 2023; 14:5223-5237. [PMID: 37854575 PMCID: PMC10581794 DOI: 10.1364/boe.498038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/06/2023] [Accepted: 08/12/2023] [Indexed: 10/20/2023]
Abstract
The development of organs-on-a-chip platforms has revolutionized in-vitro cellular culture by allowing cells to be grown in an environment that better mimics human physiology. However, there is still a challenge in integrating those platforms with advanced imaging technology. This is extremely important when we want to study molecular changes and subcellular processes on the level of a single molecule using super-resolution microscopy (SRM), which has a resolution beyond the diffraction limit of light. Currently, existing platforms that include SRM have certain limitations, either as they only support 2D monocultures, without flow or as they demand a lot of production and handling. In this study, we developed a Super-Res-Chip platform, consisting of a 3D-printed chip and a porous membrane, that could be used to co-culture cells in close proximity either in 2D or in 3D while allowing SRM on both sides of the membrane. To demonstrate the functionality of the device, we co-cultured in endothelial and epithelial cells and used direct stochastic optical reconstruction microscopy (dSTORM) to investigate how glioblastoma cells affect the expression of the gap-junction protein Connexin43 in endothelial cells grown in 2D and in 3D. Cluster analysis of Connexin43 distribution revealed no difference in the number of clusters, their size, or radii, but did identify differences in their density. Furthermore, the spatial resolution was high also when the cells were imaged through the membrane (20-30 nm for x-y) and 10-20 nm when imaged directly both for 2D and 3D conditions. Overall, this chip allows to characterize of complex cellular processes on a molecular scale in an easy manner and improved the capacity for imaging in a single molecule resolution complex cellular organization.
Collapse
Affiliation(s)
- Ofir Sade
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ronja Boneberg
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yifat Weiss
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | | | | | - Itay Talpir
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Irit Gottfried
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Rossana Rauti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, 61029, Italy
| | - Ben M Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| |
Collapse
|
28
|
Reed-McBain CA, Turaga RV, Zima SRT, Abizanda Campo S, Riendeau J, Contreras Guzman E, Juang TD, Juang DS, Hampton DW, Skala MC, Ayuso JM. Microfluidic device with reconfigurable spatial temporal gradients reveals plastic astrocyte response to stroke and reperfusion. LAB ON A CHIP 2023; 23:3945-3960. [PMID: 37448230 DOI: 10.1039/d3lc00276d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/15/2023]
Abstract
As a leading cause of mortality and morbidity, stroke constitutes a significant global health burden. Ischemic stroke accounts for 80% of cases and occurs due to an arterial thrombus, which impedes cerebral blood flow and rapidly leads to cell death. As the most abundant cell type within the central nervous system, astrocytes play a critical role within the injured brain. We developed a novel microphysiological platform that permits the induction of spatiotemporally controlled nutrient gradients, allowing us to study astrocytic response during and after transient nutrient deprivation. Within 24 h of inducing starvation in the platform, nutrient deprivation led to multiple changes in astrocyte response, from metabolic perturbations to gene expression changes, and cell viability. Furthermore, we observed that nutrient restoration did not reverse the functional changes in astrocyte metabolism, which mirrors reperfusion injury observed in vivo. We also identified alterations in numerous glucose metabolism-associated genes, many of which remained upregulated or downregulated even after restoration of the nutrient supply. Together, these findings suggest that astrocyte activation during and after nutrient starvation induces plastic changes that may underpin persistent stroke-induced functional impairment. Overall, our innovative device presents interesting potential to be used in the development of new therapies to improve tissue repair and even cognitive recovery after stroke.
Collapse
Affiliation(s)
- Catherine A Reed-McBain
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, 1 S Park Street, Madison, WI, 53715, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
- UW Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA
- Centre for Clinical Brain Sciences, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Rithvik V Turaga
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, 1 S Park Street, Madison, WI, 53715, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
- UW Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA
| | - Seth R T Zima
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, 1 S Park Street, Madison, WI, 53715, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
- UW Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA
| | - Sara Abizanda Campo
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, 1 S Park Street, Madison, WI, 53715, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
- UW Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA
| | - Jeremiah Riendeau
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, 53715, USA
| | | | - Terry D Juang
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
| | - Duane S Juang
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
| | - David W Hampton
- Centre for Clinical Brain Sciences, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Euan MacDonald Centre for Motor Neurone Disease, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Melissa C Skala
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
- UW Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA
- Morgridge Institute for Research, 330 N Orchard Street, Madison, WI, 53715, USA
| | - Jose M Ayuso
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin, 1 S Park Street, Madison, WI, 53715, USA.
- Department of Biomedical Engineering, College of Engineering, University of Wisconsin, 1550 Engineering Dr, Madison, WI, 53705, USA
- UW Carbone Cancer Center, 600 Highland Avenue, Madison, WI 53792, USA
| |
Collapse
|
29
|
Schlotterose L, Beldjilali-Labro M, Hagel M, Yadid M, Flaxer C, Flaxer E, Barnea AR, Hattermann K, Shohami E, Leichtmann-Bardoogo Y, Maoz BM. Inducing Mechanical Stimuli to Tissues Grown on a Magnetic Gel Allows Deconvoluting the Forces Leading to Traumatic Brain Injury. Neurotrauma Rep 2023; 4:560-572. [PMID: 37636339 PMCID: PMC10457614 DOI: 10.1089/neur.2023.0026] [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] [Indexed: 08/29/2023] Open
Abstract
Traumatic brain injury (TBI), which is characterized by damage to the brain resulting from a sudden traumatic event, is a major cause of death and disability worldwide. It has short- and long-term effects, including neuroinflammation, cognitive deficits, and depression. TBI consists of multiple steps that may sometimes have opposing effects or mechanisms, making it challenging to investigate and translate new knowledge into effective therapies. In order to better understand and address the underlying mechanisms of TBI, we have developed an in vitro platform that allows dynamic simulation of TBI conditions by applying external magnetic forces to induce acceleration and deceleration injury, which is often observed in human TBI. Endothelial and neuron-like cells were successfully grown on magnetic gels and applied to the platform. Both cell types showed an instant response to the TBI model, but the endothelial cells were able to recover quickly-in contrast to the neuron-like cells. In conclusion, the presented in vitro model mimics the mechanical processes of acceleration/deceleration injury involved in TBI and will be a valuable resource for further research on brain injury.
Collapse
Affiliation(s)
- Luise Schlotterose
- Institute of Anatomy, Kiel University, Kiel, Germany
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Mario Hagel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Moran Yadid
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Carina Flaxer
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Eli Flaxer
- AFEKA–Tel-Aviv Academic College of Engineering, Tel-Aviv, Israel
| | - A. Ronny Barnea
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Esther Shohami
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
30
|
Brandl S, Reindl M. Blood-Brain Barrier Breakdown in Neuroinflammation: Current In Vitro Models. Int J Mol Sci 2023; 24:12699. [PMID: 37628879 PMCID: PMC10454051 DOI: 10.3390/ijms241612699] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The blood-brain barrier, which is formed by tightly interconnected microvascular endothelial cells, separates the brain from the peripheral circulation. Together with other central nervous system-resident cell types, including pericytes and astrocytes, the blood-brain barrier forms the neurovascular unit. Upon neuroinflammation, this barrier becomes leaky, allowing molecules and cells to enter the brain and to potentially harm the tissue of the central nervous system. Despite the significance of animal models in research, they may not always adequately reflect human pathophysiology. Therefore, human models are needed. This review will provide an overview of the blood-brain barrier in terms of both health and disease. It will describe all key elements of the in vitro models and will explore how different compositions can be utilized to effectively model a variety of neuroinflammatory conditions. Furthermore, it will explore the existing types of models that are used in basic research to study the respective pathologies thus far.
Collapse
Affiliation(s)
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
31
|
Buchmann S, Enrico A, Holzreuter MA, Reid M, Zeglio E, Niklaus F, Stemme G, Herland A. Probabilistic cell seeding and non-autofluorescent 3D-printed structures as scalable approach for multi-level co-culture modeling. Mater Today Bio 2023; 21:100706. [PMID: 37435551 PMCID: PMC10331311 DOI: 10.1016/j.mtbio.2023.100706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 07/13/2023] Open
Abstract
To model complex biological tissue in vitro, a specific layout for the position and numbers of each cell type is necessary. Establishing such a layout requires manual cell placement in three dimensions (3D) with micrometric precision, which is complicated and time-consuming. Moreover, 3D printed materials used in compartmentalized microfluidic models are opaque or autofluorescent, hindering parallel optical readout and forcing serial characterization methods, such as patch-clamp probing. To address these limitations, we introduce a multi-level co-culture model realized using a parallel cell seeding strategy of human neurons and astrocytes on 3D structures printed with a commercially available non-autofluorescent resin at micrometer resolution. Using a two-step strategy based on probabilistic cell seeding, we demonstrate a human neuronal monoculture that forms networks on the 3D printed structure and can establish cell-projection contacts with an astrocytic-neuronal co-culture seeded on the glass substrate. The transparent and non-autofluorescent printed platform allows fluorescence-based immunocytochemistry and calcium imaging. This approach provides facile multi-level compartmentalization of different cell types and routes for pre-designed cell projection contacts, instrumental in studying complex tissue, such as the human brain.
Collapse
Affiliation(s)
- Sebastian Buchmann
- Division of Nanobiotechnology, KTH Royal Institute of Technology, Tomtebodavägen 23a, 171 65, Solna, Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Alessandro Enrico
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
- Synthetic Physiology lab, Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100, Pavia, Italy
| | - Muriel Alexandra Holzreuter
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
| | - Michael Reid
- Department of Fiber and Polymer Technology, Wallenberg Wood Science Centre, KTH Royal Institute of Technology, Teknikringen 56-58, 100 44, Stockholm, Sweden
| | - Erica Zeglio
- Division of Nanobiotechnology, KTH Royal Institute of Technology, Tomtebodavägen 23a, 171 65, Solna, Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| | - Frank Niklaus
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
| | - Göran Stemme
- Division of Micro and Nanosystems, KTH Royal Institute of Technology, Malvinas väg 10, 100 44, Stockholm, Sweden
| | - Anna Herland
- Division of Nanobiotechnology, KTH Royal Institute of Technology, Tomtebodavägen 23a, 171 65, Solna, Sweden
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences, Department of Neuroscience, Karolinska Institute, 17177, Stockholm, Sweden
| |
Collapse
|
32
|
Rauti R, Navok S, Biran D, Tadmor K, Leichtmann-Bardoogo Y, Ron EZ, Maoz BM. Insight on Bacterial Newborn Meningitis Using a Neurovascular-Unit-on-a-Chip. Microbiol Spectr 2023; 11:e0123323. [PMID: 37222614 PMCID: PMC10269748 DOI: 10.1128/spectrum.01233-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Understanding the pathogenesis of bacterial infections is critical for combatting them. For some infections, animal models are inadequate and functional genomic studies are not possible. One example is bacterial meningitis, a life-threatening infection with high mortality and morbidity. Here, we used the newly developed, physiologically relevant, organ-on-a-chip platform integrating the endothelium with neurons, closely mimicking in vivo conditions. Using high-magnification microscopy, permeability measurements, electrophysiological recordings, and immunofluorescence staining, we studied the dynamic by which the pathogens cross the blood-brain barrier and damage the neurons. Our work opens up possibilities for performing large-scale screens with bacterial mutant libraries for identifying the virulence genes involved in meningitis and determining the role of these genes, including various capsule types, in the infection process. These data are essential for understanding and therapy of bacterial meningitis. Moreover, our system offers possibilities for the study of additional infections-bacterial, fungal, and viral. IMPORTANCE The interactions of newborn meningitis (NBM) with the neurovascular unit are very complex and are hard to study. This work presents a new platform to study NBM in a system that enables monitoring of multicellular interactions and identifies processes that were not observed before.
Collapse
Affiliation(s)
- Rossana Rauti
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sharon Navok
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Dvora Biran
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Keshet Tadmor
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Eliora Z. Ron
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
33
|
Fabbri R, Cacopardo L, Ahluwalia A, Magliaro C. Advanced 3D Models of Human Brain Tissue Using Neural Cell Lines: State-of-the-Art and Future Prospects. Cells 2023; 12:1181. [PMID: 37190089 PMCID: PMC10136913 DOI: 10.3390/cells12081181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Human-relevant three-dimensional (3D) models of cerebral tissue can be invaluable tools to boost our understanding of the cellular mechanisms underlying brain pathophysiology. Nowadays, the accessibility, isolation and harvesting of human neural cells represents a bottleneck for obtaining reproducible and accurate models and gaining insights in the fields of oncology, neurodegenerative diseases and toxicology. In this scenario, given their low cost, ease of culture and reproducibility, neural cell lines constitute a key tool for developing usable and reliable models of the human brain. Here, we review the most recent advances in 3D constructs laden with neural cell lines, highlighting their advantages and limitations and their possible future applications.
Collapse
Affiliation(s)
- Rachele Fabbri
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
| | - Ludovica Cacopardo
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of 3R Principles in Teaching and Research (Centro 3R), Italy
| | - Arti Ahluwalia
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of 3R Principles in Teaching and Research (Centro 3R), Italy
| | - Chiara Magliaro
- Research Center “E. Piaggio”, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
- Department of Information Engineering (DII), University of Pisa, Via G. Caruso 16, 56122 Pisa, Italy
- Interuniversity Center for the Promotion of 3R Principles in Teaching and Research (Centro 3R), Italy
| |
Collapse
|
34
|
Neuronal Cultures: Exploring Biophysics, Complex Systems, and Medicine in a Dish. BIOPHYSICA 2023. [DOI: 10.3390/biophysica3010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Neuronal cultures are one of the most important experimental models in modern interdisciplinary neuroscience, allowing to investigate in a control environment the emergence of complex behavior from an ensemble of interconnected neurons. Here, I review the research that we have conducted at the neurophysics laboratory at the University of Barcelona over the last 15 years, describing first the neuronal cultures that we prepare and the associated tools to acquire and analyze data, to next delve into the different research projects in which we actively participated to progress in the understanding of open questions, extend neuroscience research on new paradigms, and advance the treatment of neurological disorders. I finish the review by discussing the drawbacks and limitations of neuronal cultures, particularly in the context of brain-like models and biomedicine.
Collapse
|
35
|
Prasad T, Iyer S, Chatterjee S, Kumar M. In vivo models to study neurogenesis and associated neurodevelopmental disorders-Microcephaly and autism spectrum disorder. WIREs Mech Dis 2023:e1603. [PMID: 36754084 DOI: 10.1002/wsbm.1603] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/14/2022] [Accepted: 01/24/2023] [Indexed: 02/10/2023]
Abstract
The genesis and functioning of the central nervous system are one of the most intricate and intriguing aspects of embryogenesis. The big lacuna in the field of human CNS development is the lack of accessibility of the human brain for direct observation during embryonic and fetal development. Thus, it is imperative to establish alternative animal models to gain deep mechanistic insights into neurodevelopment, establishment of neural circuitry, and its function. Neurodevelopmental events such as neural specification, differentiation, and generation of neuronal and non-neuronal cell types have been comprehensively studied using a variety of animal models and in vitro model systems derived from human cells. The experimentations on animal models have revealed novel, mechanistic insights into neurogenesis, formation of neural networks, and function. The models, thus serve as indispensable tools to understand the molecular basis of neurodevelopmental disorders (NDDs) arising from aberrations during embryonic development. Here, we review the spectrum of in vivo models such as fruitfly, zebrafish, frog, mice, and nonhuman primates to study neurogenesis and NDDs like microcephaly and Autism Spectrum Disorder. We also discuss nonconventional models such as ascidians and the recent technological advances in the field to study neurogenesis, disease mechanisms, and pathophysiology of human NDDs. This article is categorized under: Cancer > Stem Cells and Development Congenital Diseases > Stem Cells and Development Neurological Diseases > Stem Cells and Development Congenital Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Tuhina Prasad
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sharada Iyer
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sayoni Chatterjee
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Megha Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
36
|
Gazerani P. Human Brain Organoids in Migraine Research: Pathogenesis and Drug Development. Int J Mol Sci 2023; 24:3113. [PMID: 36834522 PMCID: PMC9961184 DOI: 10.3390/ijms24043113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023] Open
Abstract
Human organoids are small, self-organized, three-dimensional (3D) tissue cultures that have started to revolutionize medical science in terms of understanding disease, testing pharmacologically active compounds, and offering novel ways to treat disease. Organoids of the liver, kidney, intestine, lung, and brain have been developed in recent years. Human brain organoids are used for understanding pathogenesis and investigating therapeutic options for neurodevelopmental, neuropsychiatric, neurodegenerative, and neurological disorders. Theoretically, several brain disorders can be modeled with the aid of human brain organoids, and hence the potential exists for understanding migraine pathogenesis and its treatment with the aid of brain organoids. Migraine is considered a brain disorder with neurological and non-neurological abnormalities and symptoms. Both genetic and environmental factors play essential roles in migraine pathogenesis and its clinical manifestations. Several types of migraines are classified, for example, migraines with and without aura, and human brain organoids can be developed from patients with these types of migraines to study genetic factors (e.g., channelopathy in calcium channels) and environmental stressors (e.g., chemical and mechanical). In these models, drug candidates for therapeutic purposes can also be tested. Here, the potential and limitations of human brain organoids for studying migraine pathogenesis and its treatment are communicated to generate motivation and stimulate curiosity for further research. This must, however, be considered alongside the complexity of the concept of brain organoids and the neuroethical aspects of the topic. Interested researchers are invited to join the network for protocol development and testing the hypothesis presented here.
Collapse
Affiliation(s)
- Parisa Gazerani
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, 0130 Oslo, Norway; or
- Centre for Intelligent Musculoskeletal Health (CIM), Faculty of Health Sciences, Oslo Metropolitan University, 0130 Oslo, Norway
- Department of Health Science and Technology, Faculty of Medicine, Aalborg University, 9220 Aalborg East, Denmark
| |
Collapse
|
37
|
Li Z, Jiang Z, Lu L, Liu Y. Microfluidic Manipulation for Biomedical Applications in the Central and Peripheral Nervous Systems. Pharmaceutics 2023; 15:pharmaceutics15010210. [PMID: 36678839 PMCID: PMC9862045 DOI: 10.3390/pharmaceutics15010210] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Physical injuries and neurodegenerative diseases often lead to irreversible damage to the organizational structure of the central nervous system (CNS) and peripheral nervous system (PNS), culminating in physiological malfunctions. Investigating these complex and diverse biological processes at the macro and micro levels will help to identify the cellular and molecular mechanisms associated with nerve degeneration and regeneration, thereby providing new options for the development of new therapeutic strategies for the functional recovery of the nervous system. Due to their distinct advantages, modern microfluidic platforms have significant potential for high-throughput cell and organoid cultures in vitro, the synthesis of a variety of tissue engineering scaffolds and drug carriers, and observing the delivery of drugs at the desired speed to the desired location in real time. In this review, we first introduce the types of nerve damage and the repair mechanisms of the CNS and PNS; then, we summarize the development of microfluidic platforms and their application in drug carriers. We also describe a variety of damage models, tissue engineering scaffolds, and drug carriers for nerve injury repair based on the application of microfluidic platforms. Finally, we discuss remaining challenges and future perspectives with regard to the promotion of nerve injury repair based on engineered microfluidic platform technology.
Collapse
|
38
|
Schlotterose L, Beldjilali-Labro M, Schneider G, Vardi O, Hattermann K, Even U, Shohami E, Haustein HD, Leichtmann-Bardoogo Y, Maoz BM. Traumatic Brain Injury in a Well: A Modular Three-Dimensional Printed Tool for Inducing Traumatic Brain Injury In vitro. Neurotrauma Rep 2023; 4:255-266. [PMID: 37095852 PMCID: PMC10122253 DOI: 10.1089/neur.2022.0072] [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] [Indexed: 04/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a major health problem that affects millions of persons worldwide every year among all age groups, mainly young children, and elderly persons. It is the leading cause of death for children under the age of 16 and is highly correlated with a variety of neuronal disorders, such as epilepsy, and neurodegenerative disease, such as Alzheimer's disease or amyotrophic lateral sclerosis. Over the past few decades, our comprehension of the molecular pathway of TBI has improved, yet despite being a major public health issue, there is currently no U.S. Food and Drug Administration-approved treatment for TBI, and a gap remains between these advances and their application to the clinical treatment of TBI. One of the major hurdles for pushing TBI research forward is the accessibility of TBI models and tools. Most of the TBI models require costume-made, complex, and expensive equipment, which often requires special knowledge to operate. In this study, we present a modular, three-dimensional printed TBI induction device, which induces, by the pulse of a pressure shock, a TBI-like injury on any standard cell-culture tool. Moreover, we demonstrate that our device can be used on multiple systems and cell types and can induce repetitive TBIs, which is very common in clinical TBI. Further, we demonstrate that our platform can recapitulate the hallmarks of TBI, which include cell death, decrease in neuronal functionality, axonal swelling (for neurons), and increase permeability (for endothelium). In addition, in view of the continued discussion on the need, benefits, and ethics of the use of animals in scientific research, this in vitro, high-throughput platform will make TBI research more accessible to other labs that prefer to avoid the use of animals yet are interested in this field. We believe that this will enable us to push the field forward and facilitate/accelerate the availability of novel treatments.
Collapse
Affiliation(s)
- Luise Schlotterose
- Institute of Anatomy, Kiel University, Kiel, Germany
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Gaya Schneider
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ofir Vardi
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Uzi Even
- School of Chemistry, Tel Aviv University, Tel Aviv, Israel
| | - Esther Shohami
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Herman D. Haustein
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | | | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Address correspondence to: Ben M. Maoz, PhD, Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
39
|
Human mini-blood-brain barrier models for biomedical neuroscience research: a review. Biomater Res 2022; 26:82. [PMID: 36527159 PMCID: PMC9756735 DOI: 10.1186/s40824-022-00332-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
The human blood-brain barrier (BBB) is a unique multicellular structure that is in critical demand for fundamental neuroscience studies and therapeutic evaluation. Despite substantial achievements in creating in vitro human BBB platforms, challenges in generating specifics of physiopathological relevance are viewed as impediments to the establishment of in vitro models. In this review, we provide insight into the development and deployment of in vitro BBB models that allow investigation of the physiology and pathology of neurological therapeutic avenues. First, we highlight the critical components, including cell sources, biomaterial glue collections, and engineering techniques to reconstruct a miniaturized human BBB. Second, we describe recent breakthroughs in human mini-BBBs for investigating biological mechanisms in neurology. Finally, we discuss the application of human mini-BBBs to medical approaches. This review provides strategies for understanding neurological diseases, a validation model for drug discovery, and a potential approach for generating personalized medicine.
Collapse
|
40
|
Current Advances in 3D Dynamic Cell Culture Systems. Gels 2022; 8:gels8120829. [PMID: 36547353 PMCID: PMC9778081 DOI: 10.3390/gels8120829] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The traditional two-dimensional (2D) cell culture methods have a long history of mimicking in vivo cell growth. However, these methods cannot fully represent physiological conditions, which lack two major indexes of the in vivo environment; one is a three-dimensional 3D cell environment, and the other is mechanical stimulation; therefore, they are incapable of replicating the essential cellular communications between cell to cell, cell to the extracellular matrix, and cellular responses to dynamic mechanical stimulation in a physiological condition of body movement and blood flow. To solve these problems and challenges, 3D cell carriers have been gradually developed to provide a 3D matrix-like structure for cell attachment, proliferation, differentiation, and communication in static and dynamic culture conditions. 3D cell carriers in dynamic culture systems could primarily provide different mechanical stimulations which further mimic the real in vivo microenvironment. In this review, the current advances in 3D dynamic cell culture approaches have been introduced, with their advantages and disadvantages being discussed in comparison to traditional 2D cell culture in static conditions.
Collapse
|
41
|
Montalà-Flaquer M, López-León CF, Tornero D, Houben AM, Fardet T, Monceau P, Bottani S, Soriano J. Rich dynamics and functional organization on topographically designed neuronal networks in vitro. iScience 2022; 25:105680. [PMID: 36567712 PMCID: PMC9768383 DOI: 10.1016/j.isci.2022.105680] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/05/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Neuronal cultures are a prominent experimental tool to understand complex functional organization in neuronal assemblies. However, neurons grown on flat surfaces exhibit a strongly coherent bursting behavior with limited functionality. To approach the functional richness of naturally formed neuronal circuits, here we studied neuronal networks grown on polydimethylsiloxane (PDMS) topographical patterns shaped as either parallel tracks or square valleys. We followed the evolution of spontaneous activity in these cultures along 20 days in vitro using fluorescence calcium imaging. The networks were characterized by rich spatiotemporal activity patterns that comprised from small regions of the culture to its whole extent. Effective connectivity analysis revealed the emergence of spatially compact functional modules that were associated with both the underpinned topographical features and predominant spatiotemporal activity fronts. Our results show the capacity of spatial constraints to mold activity and functional organization, bringing new opportunities to comprehend the structure-function relationship in living neuronal circuits.
Collapse
Affiliation(s)
- Marc Montalà-Flaquer
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain,Universitat de Barcelona Institute of Complex Systems (UBICS), E-08028 Barcelona, Spain
| | - Clara F. López-León
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain,Universitat de Barcelona Institute of Complex Systems (UBICS), E-08028 Barcelona, Spain
| | - Daniel Tornero
- Laboratory of Neural Stem Cells and Brain Damage, Institute of Neurosciences, University of Barcelona, E-08036 Barcelona, Spain
| | - Akke Mats Houben
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain,Universitat de Barcelona Institute of Complex Systems (UBICS), E-08028 Barcelona, Spain
| | - Tanguy Fardet
- Laboratoire Matière et Systèmes Complexes, Université de Paris, UMR 7057 CNRS, Paris, France,University of Tübingen, Tübingen, Germany,Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Pascal Monceau
- Laboratoire Matière et Systèmes Complexes, Université de Paris, UMR 7057 CNRS, Paris, France
| | - Samuel Bottani
- Laboratoire Matière et Systèmes Complexes, Université de Paris, UMR 7057 CNRS, Paris, France
| | - Jordi Soriano
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, E-08028 Barcelona, Spain,Universitat de Barcelona Institute of Complex Systems (UBICS), E-08028 Barcelona, Spain,Corresponding author
| |
Collapse
|
42
|
Castiglione H, Vigneron PA, Baquerre C, Yates F, Rontard J, Honegger T. Human Brain Organoids-on-Chip: Advances, Challenges, and Perspectives for Preclinical Applications. Pharmaceutics 2022; 14:2301. [PMID: 36365119 PMCID: PMC9699341 DOI: 10.3390/pharmaceutics14112301] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 09/26/2023] Open
Abstract
There is an urgent need for predictive in vitro models to improve disease modeling and drug target identification and validation, especially for neurological disorders. Cerebral organoids, as alternative methods to in vivo studies, appear now as powerful tools to decipher complex biological processes thanks to their ability to recapitulate many features of the human brain. Combining these innovative models with microfluidic technologies, referred to as brain organoids-on-chips, allows us to model the microenvironment of several neuronal cell types in 3D. Thus, this platform opens new avenues to create a relevant in vitro approach for preclinical applications in neuroscience. The transfer to the pharmaceutical industry in drug discovery stages and the adoption of this approach by the scientific community requires the proposition of innovative microphysiological systems allowing the generation of reproducible cerebral organoids of high quality in terms of structural and functional maturation, and compatibility with automation processes and high-throughput screening. In this review, we will focus on the promising advantages of cerebral organoids for disease modeling and how their combination with microfluidic systems can enhance the reproducibility and quality of these in vitro models. Then, we will finish by explaining why brain organoids-on-chips could be considered promising platforms for pharmacological applications.
Collapse
Affiliation(s)
- Héloïse Castiglione
- NETRI, 69007 Lyon, France
- Sup’Biotech/CEA-IBFJ-SEPIA, Bâtiment 60, 18 Route du Panorama, 94260 Fontenay-aux-Roses, France
| | - Pierre-Antoine Vigneron
- Sup’Biotech/CEA-IBFJ-SEPIA, Bâtiment 60, 18 Route du Panorama, 94260 Fontenay-aux-Roses, France
- Sup’Biotech, Ecole D’ingénieurs, 66 Rue Guy Môquet, 94800 Villejuif, France
| | | | - Frank Yates
- Sup’Biotech/CEA-IBFJ-SEPIA, Bâtiment 60, 18 Route du Panorama, 94260 Fontenay-aux-Roses, France
- Sup’Biotech, Ecole D’ingénieurs, 66 Rue Guy Môquet, 94800 Villejuif, France
| | | | | |
Collapse
|
43
|
Räsänen N, Harju V, Joki T, Narkilahti S. Practical guide for preparation, computational reconstruction and analysis of 3D human neuronal networks in control and ischaemic conditions. Development 2022; 149:276215. [PMID: 35929583 PMCID: PMC9440753 DOI: 10.1242/dev.200012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 06/23/2022] [Indexed: 11/20/2022]
Abstract
To obtain commensurate numerical data of neuronal network morphology in vitro, network analysis needs to follow consistent guidelines. Important factors in successful analysis are sample uniformity, suitability of the analysis method for extracting relevant data and the use of established metrics. However, for the analysis of 3D neuronal cultures, there is little coherence in the analysis methods and metrics used in different studies. Here, we present a framework for the analysis of neuronal networks in 3D. First, we selected a hydrogel that supported the growth of human pluripotent stem cell-derived cortical neurons. Second, we tested and compared two software programs for tracing multi-neuron images in three dimensions and optimized a workflow for neuronal analysis using software that was considered highly suitable for this purpose. Third, as a proof of concept, we exposed 3D neuronal networks to oxygen-glucose deprivation- and ionomycin-induced damage and showed morphological differences between the damaged networks and control samples utilizing the proposed analysis workflow. With the optimized workflow, we present a protocol for preparing, challenging, imaging and analysing 3D human neuronal cultures. Summary: An optimized protocol is presented that allows morphological, quantifiable differences between the damaged and control human neuronal networks to be detected in three-dimensional cultures.
Collapse
Affiliation(s)
- Noora Räsänen
- Tampere University, 33100, Tampere Faculty of Medicine and Health Technology , , Finland
| | - Venla Harju
- Tampere University, 33100, Tampere Faculty of Medicine and Health Technology , , Finland
| | - Tiina Joki
- Tampere University, 33100, Tampere Faculty of Medicine and Health Technology , , Finland
| | - Susanna Narkilahti
- Tampere University, 33100, Tampere Faculty of Medicine and Health Technology , , Finland
| |
Collapse
|
44
|
Habibey R, Striebel J, Schmieder F, Czarske J, Busskamp V. Long-term morphological and functional dynamics of human stem cell-derived neuronal networks on high-density micro-electrode arrays. Front Neurosci 2022; 16:951964. [PMID: 36267241 PMCID: PMC9578684 DOI: 10.3389/fnins.2022.951964] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Comprehensive electrophysiological characterizations of human induced pluripotent stem cell (hiPSC)-derived neuronal networks are essential to determine to what extent these in vitro models recapitulate the functional features of in vivo neuronal circuits. High-density micro-electrode arrays (HD-MEAs) offer non-invasive recording with the best spatial and temporal resolution possible to date. For 3 months, we tracked the morphology and activity features of developing networks derived from a transgenic hiPSC line in which neurogenesis is inducible by neurogenic transcription factor overexpression. Our morphological data revealed large-scale structural changes from homogeneously distributed neurons in the first month to the formation of neuronal clusters over time. This led to a constant shift in position of neuronal cells and clusters on HD-MEAs and corresponding changes in spatial distribution of the network activity maps. Network activity appeared as scarce action potentials (APs), evolved as local bursts with longer duration and changed to network-wide synchronized bursts with higher frequencies but shorter duration over time, resembling the emerging burst features found in the developing human brain. Instantaneous firing rate data indicated that the fraction of fast spiking neurons (150–600 Hz) increases sharply after 63 days post induction (dpi). Inhibition of glutamatergic synapses erased burst features from network activity profiles and confirmed the presence of mature excitatory neurotransmission. The application of GABAergic receptor antagonists profoundly changed the bursting profile of the network at 120 dpi. This indicated a GABAergic switch from excitatory to inhibitory neurotransmission during circuit development and maturation. Our results suggested that an emerging GABAergic system at older culture ages is involved in regulating spontaneous network bursts. In conclusion, our data showed that long-term and continuous microscopy and electrophysiology readouts are crucial for a meaningful characterization of morphological and functional maturation in stem cell-derived human networks. Most importantly, assessing the level and duration of functional maturation is key to subject these human neuronal circuits on HD-MEAs for basic and biomedical applications.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Johannes Striebel
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
| | - Felix Schmieder
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany
| | - Jürgen Czarske
- Laboratory of Measurement and Sensor System Technique, Faculty of Electrical and Computer Engineering, TU Dresden, Dresden, Germany
- Competence Center for Biomedical Computational Laser Systems (BIOLAS), TU Dresden, Dresden, Germany
- Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany
- School of Science, Institute of Applied Physics, TU Dresden, Dresden, Germany
| | - Volker Busskamp
- Department of Ophthalmology, Universitäts-Augenklinik Bonn, University of Bonn, Bonn, Germany
- *Correspondence: Volker Busskamp,
| |
Collapse
|
45
|
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
|
46
|
Abend A, Steele C, Schmidt S, Frank R, Jahnke HG, Zink M. Neuronal and glial cell co-culture organization and impedance spectroscopy on nanocolumnar TiN films for lab-on-a-chip devices. Biomater Sci 2022; 10:5719-5730. [PMID: 36039696 DOI: 10.1039/d2bm01066f] [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: 11/21/2022]
Abstract
Lab-on-a-chip devices, such as multielectrode arrays (MEAs), offer great advantages to study function and behavior of biological cells, such as neurons, outside the complex tissue structure. Nevertheless, in vitro systems can only succeed if they represent realistic conditions such as cell organization as similarly found in tissues. In our study, we employ a co-culture system of neuron-like (SH-SY5Y) and glial-like (U-87 MG) cells with various neuron-glial ratios to model different brain regions with different cellular compositions in vitro. We find that cell behavior in terms of cellular organization, as well as proliferation, depends on neuron-glial cell ratio, as well as the underlying substrate material. In fact, nanocolumnar titanium nitride (TiN nano), which exhibits improved electric properties for neural recording on MEA, shows improved biocompatible features compared to indium tin oxide (ITO). Moreover, electrochemical impedance spectroscopy experiments allow us to monitor cellular processes label-free in real-time over several days with multielectrode arrays. Additionally, electrochemical impedance experiments reveal superiority of TiN with nanocolumnar surface modification in comparison with ITO. TiN nano exhibits enhanced relative cell signals and improved signal-to-noise ratio, especially for smaller electrode sizes, which makes nanocolumnar TiN a promising candidate for research on neural recording and stimulation.
Collapse
Affiliation(s)
- Alice Abend
- Research Group Biotechnology and Biomedicine, Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany.
| | - Chelsie Steele
- Research Group Biotechnology and Biomedicine, Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany.
| | - Sabine Schmidt
- Centre for Biotechnology and Biomedicine, Molecular Biological-Biochemical Processing Technology, Leipzig University, 04103 Leipzig, Germany
| | - Ronny Frank
- Centre for Biotechnology and Biomedicine, Molecular Biological-Biochemical Processing Technology, Leipzig University, 04103 Leipzig, Germany
| | - Heinz-Georg Jahnke
- Centre for Biotechnology and Biomedicine, Molecular Biological-Biochemical Processing Technology, Leipzig University, 04103 Leipzig, Germany
| | - Mareike Zink
- Research Group Biotechnology and Biomedicine, Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany.
| |
Collapse
|
47
|
Iafrate L, Benedetti MC, Donsante S, Rosa A, Corsi A, Oreffo ROC, Riminucci M, Ruocco G, Scognamiglio C, Cidonio G. Modelling skeletal pain harnessing tissue engineering. IN VITRO MODELS 2022; 1:289-307. [PMID: 36567849 PMCID: PMC9766883 DOI: 10.1007/s44164-022-00028-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/27/2022]
Abstract
Bone pain typically occurs immediately following skeletal damage with mechanical distortion or rupture of nociceptive fibres. The pain mechanism is also associated with chronic pain conditions where the healing process is impaired. Any load impacting on the area of the fractured bone will stimulate the nociceptive response, necessitating rapid clinical intervention to relieve pain associated with the bone damage and appropriate mitigation of any processes involved with the loss of bone mass, muscle, and mobility and to prevent death. The following review has examined the mechanisms of pain associated with trauma or cancer-related skeletal damage focusing on new approaches for the development of innovative therapeutic interventions. In particular, the review highlights tissue engineering approaches that offer considerable promise in the application of functional biomimetic fabrication of bone and nerve tissues. The strategic combination of bone and nerve tissue engineered models provides significant potential to develop a new class of in vitro platforms, capable of replacing in vivo models and testing the safety and efficacy of novel drug treatments aimed at the resolution of bone-associated pain. To date, the field of bone pain research has centred on animal models, with a paucity of data correlating to the human physiological response. This review explores the evident gap in pain drug development research and suggests a step change in approach to harness tissue engineering technologies to recapitulate the complex pathophysiological environment of the damaged bone tissue enabling evaluation of the associated pain-mimicking mechanism with significant therapeutic potential therein for improved patient quality of life. Graphical abstract Rationale underlying novel drug testing platform development. Pain detected by the central nervous system and following bone fracture cannot be treated or exclusively alleviated using standardised methods. The pain mechanism and specificity/efficacy of pain reduction drugs remain poorly understood. In vivo and ex vivo models are not yet able to recapitulate the various pain events associated with skeletal damage. In vitro models are currently limited by their inability to fully mimic the complex physiological mechanisms at play between nervous and skeletal tissue and any disruption in pathological states. Robust innovative tissue engineering models are needed to better understand pain events and to investigate therapeutic regimes.
Collapse
Affiliation(s)
- Lucia Iafrate
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Maria Cristina Benedetti
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandro Rosa
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Richard O. C. Oreffo
- Bone and Joint Research Group, Stem Cells and Regeneration, Institute of Developmental Sciences, Centre for Human Development, University of Southampton, Southampton, UK
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Chiara Scognamiglio
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
| | - Gianluca Cidonio
- Center for Life Nano- & Neuro-Science (CLN2S), Istituto Italiano di Tecnologia, Rome, Italy
- Bone and Joint Research Group, Stem Cells and Regeneration, Institute of Developmental Sciences, Centre for Human Development, University of Southampton, Southampton, UK
| |
Collapse
|
48
|
Ameenudeen S, Kashif M, Banerjee S, Srinivasan H, Pandurangan AK, Waseem M. Cellular and Molecular Machinery of Neuropathic Pain: an Emerging Insight. CURRENT PHARMACOLOGY REPORTS 2022; 8:227-235. [PMID: 35646513 PMCID: PMC9125010 DOI: 10.1007/s40495-022-00294-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/17/2022] [Indexed: 12/04/2022]
Abstract
Purpose of Review Neuropathic pain (NP) has been ubiquitously characterized by lesion and its linked somatosensory system either the central nervous system (CNS) or peripheral nervous system (PNS) This PNS episode is the most prevalent site of NP origin and is found to be associated with afferent nerve fibers carrying pain signals from injured/trauma site to the CNS including the brain. Several kinds of pharmacotherapeutic drugs shuch as analgesics, anti-convulsants, and anti-depressants are being employed for the its possible interventions. The NP has been a great interest to follow different pathophysiological mechanisms which are often considered to correlate with the metabolic pathways and its mediated disease. There is paucity of knowledge to make such mechanism via NP. Recent Finding Most notably, recent pandemic outbreak of COVID-19 has also been reported in chronic pain mediated diabetes, inflammatory disorders, and cancers. There is an increasing incidence of NP and its complex mechanism has now led to identify the possible investigations of responsible genes and proteins via bioinformatics tools. The analysis might be more instrumental as collecting the genes from pain genetic database, analyzing the variants through differential gene expression (DEG) and constructing the protein–protein interaction (PPI) networks and thereby determining their upregulating and downregulating pathways. Summary This review sheds a bright light towards several mechanisms at both cellular and molecular level, correlation of NP-mediated disease mechanism and possible cell surface biomarkers (receptors), and identified genes could be more promising for their pharmacological targets.
Collapse
Affiliation(s)
- Shabnam Ameenudeen
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Mohd. Kashif
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Subhamoy Banerjee
- Department of Basic Science and Humanities, Institute of Engineering and Management, Sector V, Salt Lake, Kolkata, 700091 India
| | - Hemalatha Srinivasan
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Ashok Kumar Pandurangan
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| | - Mohammad Waseem
- School of Life Sciences, BS Abdur Rahman Crescent Institute of Science and Technology, GST Road, Vandalur, Chennai, 600048 Tamil Nadu India
| |
Collapse
|
49
|
Enhancement of Neuroglial Extracellular Matrix Formation and Physiological Activity of Dopaminergic Neural Cocultures by Macromolecular Crowding. Cells 2022; 11:cells11142131. [PMID: 35883574 PMCID: PMC9317039 DOI: 10.3390/cells11142131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/06/2023] Open
Abstract
The neuroglial extracellular matrix (ECM) provides critical support and physiological cues for the proper growth, differentiation, and function of neuronal cells in the brain. However, in most in vitro settings that study neural physiology, cells are grown as monolayers on stiff surfaces that maximize adhesion and proliferation, and, therefore, they lack the physiological cues that ECM in native neuronal tissues provides. Macromolecular crowding (MMC) is a biophysical phenomenon based on the principle of excluded volume that can be harnessed to induce native ECM deposition by cells in culture. Here, we show that MMC using two species of Ficoll with vitamin C supplementation significantly boosts deposition of relevant brain ECM by cultured human astrocytes. Dopaminergic neurons cocultured on this astrocyte–ECM bed prepared under MMC treatment showed longer and denser neuronal extensions, a higher number of pre ad post synaptic contacts, and increased physiological activity, as evidenced by higher frequency calcium oscillation, compared to standard coculture conditions. When the pharmacological activity of various compounds was tested on MMC-treated cocultures, their responses were enhanced, and for apomorphine, a D2-receptor agonist, it was inverted in comparison to control cell culture conditions, thus emulating responses observed in in vivo settings. These results indicate that macromolecular crowding can harness the ECM-building potential of human astrocytes in vitro forming an ultra-flat 3D microenvironment that makes neural cultures more physiological and pharmacological relevant.
Collapse
|
50
|
Van Breedam E, Ponsaerts P. Promising Strategies for the Development of Advanced In Vitro Models with High Predictive Power in Ischaemic Stroke Research. Int J Mol Sci 2022; 23:ijms23137140. [PMID: 35806146 PMCID: PMC9266337 DOI: 10.3390/ijms23137140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Although stroke is one of the world’s leading causes of death and disability, and more than a thousand candidate neuroprotective drugs have been proposed based on extensive in vitro and animal-based research, an effective neuroprotective/restorative therapy for ischaemic stroke patients is still missing. In particular, the high attrition rate of neuroprotective compounds in clinical studies should make us question the ability of in vitro models currently used for ischaemic stroke research to recapitulate human ischaemic responses with sufficient fidelity. The ischaemic stroke field would greatly benefit from the implementation of more complex in vitro models with improved physiological relevance, next to traditional in vitro and in vivo models in preclinical studies, to more accurately predict clinical outcomes. In this review, we discuss current in vitro models used in ischaemic stroke research and describe the main factors determining the predictive value of in vitro models for modelling human ischaemic stroke. In light of this, human-based 3D models consisting of multiple cell types, either with or without the use of microfluidics technology, may better recapitulate human ischaemic responses and possess the potential to bridge the translational gap between animal-based in vitro and in vivo models, and human patients in clinical trials.
Collapse
|