1
|
Wang P, Liu J, Zhang M, Yang J, Lian P, Cheng X, Qin J. Radiation Exposure Induced Blood-Brain Barrier Injury via Mitochondria-Mediated Sterile Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e02356. [PMID: 40433769 DOI: 10.1002/advs.202502356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Radiation-induced brain injury (RIBI) is caused by exposure to high doses of ionizing radiation and characterized by severe cognitive dysfunction and brain necrosis. However, the pathogenesis of RIBI is not fully understood, and no effective intervention is available. This work describes a blood-brain barrier (BBB) microphysiological system (MPS), that allowed to explore the responses of BBB and distinct brain cells to radiation exposure. Following acute exposure to radiation of X-ray or γ-ray, characteristic RIBI-associated pathological responses are observed, including BBB compromise, DNA breaks, inhibited cell proliferation, cell hypertrophy, and proinflammatory cytokine release. Among the distinctive types of cells, brain endothelial cells show the highest radiosensitivity as compared to other cells in the MPS. Intriguingly, X-ray and γ-ray radiation consistently induce prominent sterile inflammation responses, especially type I interferon response, in the BBB MPS. These responses are mediated by radiation-induced mitochondrial DNA release and subsequent activation of cGAS-STING signaling pathway. Furthermore, it is found abrocitinib (JAK1 inhibitor) and idebenone (mitochondrial protectant) can attenuate radiation-induced inflammation and ameliorate injuries in the BBB MPS. These findings reveal the involvement of mitochondria-mediated sterile inflammation in RIBI pathogenesis, identifying mitochondria as a potential target for new radioprotective measures.
Collapse
Affiliation(s)
- Peng Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jiayue Liu
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Min Zhang
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
| | - Juan Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, P. R. China
| | - Peihan Lian
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Xiu Cheng
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
| | - Jianhua Qin
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, P. R. China
- Disvision of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, P. R. China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100000, China
| |
Collapse
|
2
|
Loret C, Scherrer C, Rovini A, Lesage E, Richard L, Danigo A, Sturtz F, Favreau F, Faye PA, Lia AS. Addressing myelination disorders: Novel strategies using human 3D peripheral nerve model. Brain Res Bull 2025; 222:111252. [PMID: 39938756 DOI: 10.1016/j.brainresbull.2025.111252] [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: 08/28/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Peripheral myelination disorders encompass a variety of disorders that affect myelin sheaths in the peripheral nervous system. The Charcot-Marie-Tooth disease (CMT), the most common inherited peripheral neuropathy, is one of the most prevalent among them. CMT stems from a wide range of genetic causes that disrupt the nerve conduction, leading to progressive muscle weakness and atrophy, sensory loss, and motor function impairment. Historically, the study of these disorders has relied heavily on animal studies, owing to the challenges in accessing human cells. However, the advent of human induced pluripotent stem cell (hiPSC)-derived neuronal cells has addressed these limitations in the realm of peripheral myelination disorders. Despite this, obtaining myelin in these models remains an expensive, time-consuming, and material-intensive process. This study presents a novel, cost-effective method utilizing hiPSC-derived Schwann cells and motor neurons in a three-dimensional culture system. Our method successfully enabled the acquisition of myelin in a control clone within just four weeks, as confirmed by electron microscopy. Furthermore, the utility of these approaches was validated by studying CMT4C, also named AR-CMTde-SH3TC2, the most common recessive demyelinating form of CMT. This revealed defects in Schwann cell support to motor neuron neurite outgrowth and impaired myelination in disease-specific hiPSC-derived lines. This approach offers valuable insights into the pathogenesis of peripheral myelination disorders and provides a platform for testing potential therapeutic strategies.
Collapse
Affiliation(s)
- Camille Loret
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France.
| | - Camille Scherrer
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France
| | - Amandine Rovini
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France
| | - Esther Lesage
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France
| | - Laurence Richard
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Service de Neurologie, Limoges F-87000, France
| | - Aurore Danigo
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Service de Neurologie, Limoges F-87000, France
| | - Franck Sturtz
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France
| | - Frédéric Favreau
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France.
| | - Pierre-Antoine Faye
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France
| | - Anne-Sophie Lia
- University of Limoges, NeurIT UR 20218, GEIST Institute, Limoges F-87000, France; CHU Limoges, Department of Biochemistry and Molecular Genetics, Limoges F-87000, France; CHU Limoges, Department of Bioinformatics, Limoges F-87000, France
| |
Collapse
|
3
|
Patel A, Williams M, Hawkins K, Gallo L, Grillo M, Akanda N, Guo X, Lambert S, Hickman JJ. Establishment of a Serum-Free Human iPSC-Derived Model of Peripheral Myelination. ACS Biomater Sci Eng 2024; 10:7132-7143. [PMID: 39437333 PMCID: PMC11558563 DOI: 10.1021/acsbiomaterials.4c01431] [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: 08/02/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Myelination and the formation of nodes of Ranvier are essential for the rapid conduction of nerve impulses along axons in the peripheral nervous system (PNS). While many animal-based and serum-containing models of peripheral myelination have been developed, these have limited ability when it comes to studying genetic disorders affecting peripheral myelination. We report a fully induced pluripotent stem cell (iPSC)-derived human model of peripheral myelination using Schwann cells (SCs) and motoneurons, cultured in a serum-free medium on patterned and nonpatterned surfaces. Results demonstrated iPSC-derived SC-expressed early growth response protein 2 (Egr2), a key transcription factor for myelination, and after ∼30 days in coculture, hallmark features of myelination, including myelin segment and node of Ranvier formation, were observed. Myelin segments were stained for the myelin basic protein, which surrounded neurofilament-stained motoneuron axons. Clusters of voltage-gated sodium channels flanked by paranodal protein contactin-associated protein 1, indicating node of Ranvier formation, were also observed. High-resolution confocal microscopy allowed for 3D reconstruction and measurement of myelin g-ratios of myelin segments, with an average g-ratio of 0.67, consistent with reported values in the literature, indicating mature myelin segment formation. This iPSC-based model of peripheral myelination provides a platform to investigate numerous PNS diseases, including Charcot-Marie Tooth disorder, Guillian-Barre syndrome, chronic inflammatory demyelinating polyneuropathy, and antimyelin-associated glycoprotein peripheral neuropathy, with the potential for greater translatability to humans for improving the applicability for drug-screening programs.
Collapse
Affiliation(s)
- Aakash Patel
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Marnie Williams
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Kenneth Hawkins
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Leandro Gallo
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Marcella Grillo
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Nesar Akanda
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Xiufang Guo
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
| | - Stephen Lambert
- College
of Medicine, University of Central Florida, 6850 Lake Nona Blvd, Orlando, Florida 32827, United States
| | - James J. Hickman
- NanoScience
Technology Center, University of Central
Florida, 12424 Research Parkway, Suite 400, Orlando, Florida 32826, United States
- Hesperos
Inc., 12501 Research
Parkway, Suite 100, Orlando, Florida 32826, United States
| |
Collapse
|
4
|
Zhang X, Gao X, Zhang X, Yao X, Kang X. Revolutionizing Intervertebral Disc Regeneration: Advances and Future Directions in Three-Dimensional Bioprinting of Hydrogel Scaffolds. Int J Nanomedicine 2024; 19:10661-10684. [PMID: 39464675 PMCID: PMC11505483 DOI: 10.2147/ijn.s469302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/10/2024] [Indexed: 10/29/2024] Open
Abstract
Hydrogels are multifunctional platforms. Through reasonable structure and function design, they use material engineering to adjust their physical and chemical properties, such as pore size, microstructure, degradability, stimulus-response characteristics, etc. and have a variety of biomedical applications. Hydrogel three-dimensional (3D) printing has emerged as a promising technique for the precise deposition of cell-laden biomaterials, enabling the fabrication of intricate 3D structures such as artificial vertebrae and intervertebral discs (IVDs). Despite being in the early stages, 3D printing techniques have shown great potential in the field of regenerative medicine for the fabrication of various transplantable tissues within the human body. Currently, the utilization of engineered hydrogels as carriers or scaffolds for treating intervertebral disc degeneration (IVDD) presents numerous challenges. However, it remains an indispensable multifunctional manufacturing technology that is imperative in addressing the escalating issue of IVDD. Moreover, it holds the potential to serve as a micron-scale platform for a diverse range of applications. This review primarily concentrates on emerging treatment strategies for IVDD, providing an in-depth analysis of their merits and drawbacks, as well as the challenges that need to be addressed. Furthermore, it extensively explores the biological properties of hydrogels and various nanoscale biomaterial inks, compares different prevalent manufacturing processes utilized in 3D printing, and thoroughly examines the potential clinical applications and prospects of integrating 3D printing technology with hydrogels.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xidan Gao
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xuefang Zhang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xin Yao
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xin Kang
- Department of Sports Medicine, Honghui Hospital, Xi’an Jiao Tong University, Xi’An, Shaanxi, P.R. China
| |
Collapse
|
5
|
Jung S, Cheong S, Lee Y, Lee J, Lee J, Kwon MS, Oh YS, Kim T, Ha S, Kim SJ, Jo DH, Ko J, Jeon NL. Integrating Vascular Phenotypic and Proteomic Analysis in an Open Microfluidic Platform. ACS NANO 2024; 18:24909-24928. [PMID: 39208278 PMCID: PMC11394367 DOI: 10.1021/acsnano.4c05537] [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: 04/26/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
This research introduces a vascular phenotypic and proteomic analysis (VPT) platform designed to perform high-throughput experiments on vascular development. The VPT platform utilizes an open-channel configuration that facilitates angiogenesis by precise alignment of endothelial cells, allowing for a 3D morphological examination and protein analysis. We study the effects of antiangiogenic agents─bevacizumab, ramucirumab, cabozantinib, regorafenib, wortmannin, chloroquine, and paclitaxel─on cytoskeletal integrity and angiogenic sprouting, observing an approximately 50% reduction in sprouting at higher drug concentrations. Precise LC-MS/MS analyses reveal global protein expression changes in response to four of these drugs, providing insights into the signaling pathways related to the cell cycle, cytoskeleton, cellular senescence, and angiogenesis. Our findings emphasize the intricate relationship between cytoskeletal alterations and angiogenic responses, underlining the significance of integrating morphological and proteomic data for a comprehensive understanding of angiogenesis. The VPT platform not only advances our understanding of drug impacts on vascular biology but also offers a versatile tool for analyzing proteome and morphological features across various models beyond blood vessels.
Collapse
Affiliation(s)
- Sangmin Jung
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Sunghun Cheong
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Yoonho Lee
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Jungseub Lee
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Jihye Lee
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
| | - Min-Seok Kwon
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
- Department
of Public Health Science, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Sun Oh
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Target
Link Therapeutics, Inc., Seoul 04545, Republic
of Korea
| | - Taewan Kim
- Department
of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sungjae Ha
- ProvaLabs,
Inc., Seoul 08826, Republic of Korea
| | - Sung Jae Kim
- Department
of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
- SOFT
Foundry, Seoul National University, Seoul 08826, Republic of Korea
- Inter-university
Semiconductor Research Center, Seoul National
University, Seoul 08826, Republic
of Korea
| | - Dong Hyun Jo
- Department
of Anatomy and Cell Biology, Seoul National
University College of Medicine, Seoul 03080, Republic of Korea
| | - Jihoon Ko
- Department
of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic
of Korea
| | - Noo Li Jeon
- Department
of Mechanical Engineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Interdisciplinary
Program in Bioengineering, Seoul National
University, Seoul 08826, Republic
of Korea
- Institute
of Advanced Machines and Design, Seoul National
University, Seoul 08826, Republic
of Korea
- Qureator, Inc., San
Diego, California 92121, United States
| |
Collapse
|
6
|
De Vitis E, Stanzione A, Romano A, Quattrini A, Gigli G, Moroni L, Gervaso F, Polini A. The Evolution of Technology-Driven In Vitro Models for Neurodegenerative Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304989. [PMID: 38366798 DOI: 10.1002/advs.202304989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/15/2024] [Indexed: 02/18/2024]
Abstract
The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
Collapse
Affiliation(s)
- Eleonora De Vitis
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Antonella Stanzione
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Angelo Quattrini
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Giuseppe Gigli
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Lorenzo Moroni
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, Netherlands
| | - Francesca Gervaso
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
7
|
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
|
8
|
Lachance GP, Gauvreau D, Boisselier É, Boukadoum M, Miled A. Breaking Barriers: Exploring Neurotransmitters through In Vivo vs. In Vitro Rivalry. SENSORS (BASEL, SWITZERLAND) 2024; 24:647. [PMID: 38276338 PMCID: PMC11154401 DOI: 10.3390/s24020647] [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: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
Neurotransmitter analysis plays a pivotal role in diagnosing and managing neurodegenerative diseases, often characterized by disturbances in neurotransmitter systems. However, prevailing methods for quantifying neurotransmitters involve invasive procedures or require bulky imaging equipment, therefore restricting accessibility and posing potential risks to patients. The innovation of compact, in vivo instruments for neurotransmission analysis holds the potential to reshape disease management. This innovation can facilitate non-invasive and uninterrupted monitoring of neurotransmitter levels and their activity. Recent strides in microfabrication have led to the emergence of diminutive instruments that also find applicability in in vitro investigations. By harnessing the synergistic potential of microfluidics, micro-optics, and microelectronics, this nascent realm of research holds substantial promise. This review offers an overarching view of the current neurotransmitter sensing techniques, the advances towards in vitro microsensors tailored for monitoring neurotransmission, and the state-of-the-art fabrication techniques that can be used to fabricate those microsensors.
Collapse
Affiliation(s)
| | - Dominic Gauvreau
- Department Electrical Engineering, Université Laval, Québec, QC G1V 0A6, Canada; (G.P.L.); (D.G.)
| | - Élodie Boisselier
- Department Ophthalmology and Otolaryngology—Head and Neck Surgery, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Mounir Boukadoum
- Department Computer Science, Université du Québec à Montréal, Montréal, QC H2L 2C4, Canada;
| | - Amine Miled
- Department Electrical Engineering, Université Laval, Québec, QC G1V 0A6, Canada; (G.P.L.); (D.G.)
| |
Collapse
|
9
|
Xiao N, Xiong S, Zhou Z, Zhong M, Bai H, Li Q, Tang Y, Xie J. Recent progress in biomaterials-driven ferroptosis for cancer therapy. Biomater Sci 2024; 12:288-307. [PMID: 38189655 DOI: 10.1039/d3bm01832f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Ferroptosis, first suggested in 2012, is a type of non-apoptotic programmed cell death caused by the buildup of lipid peroxidation and marked by an overabundance of oxidized poly unsaturated fatty acids. During the last decade, researchers have uncovered the formation of ferroptosis and created multiple drugs aimed at it, but due to poor selectivity and pharmacokinetics, clinical application has been hindered. In recent years, biomedical discoveries and developments in nanotechnology have spurred the investigation of ferroptosis nanomaterials, providing new opportunities for the ferroptosis driven tumours treatment. Additionally, hydrogels have been widely studied in ferroptosis because of their unique 3D structure and excellent controllability. By using these biomaterials, it is possible to achieve controlled release and targeted delivery of drugs, thus increasing the potency of the drugs and minimizing adverse effects. Therefore, summarizing the biomedical nanomaterials, including hydrogels, used in ferroptosis for cancer therapy is a must. This article provides an overview of ferroptosis, detailing its properties and underlying mechanisms. It also categorizes and reviews the use of various nanomaterials in ferroptosis, along with relevant explanations and illustrations. In addition, we discuss the opportunities and challenges facing the application of nanomaterials in ferroptosis. Finally, the development prospects of this field are prospected. This review is intended to provide a foundation for the development and application of biomedical nanomaterials in ferroptosis.
Collapse
Affiliation(s)
- Nianting Xiao
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Su Xiong
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Ziwei Zhou
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Min Zhong
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Huayang Bai
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Qiyu Li
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Yaqin Tang
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| | - Jing Xie
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
10
|
Buentello DC, Garcia-Corral M, Trujillo-de Santiago G, Alvarez MM. Neuron(s)-on-a-Chip: A Review of the Design and Use of Microfluidic Systems for Neural Tissue Culture. IEEE Rev Biomed Eng 2024; 17:243-263. [PMID: 36301779 DOI: 10.1109/rbme.2022.3217486] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neuron-on-chip (NoC) systems-microfluidic devices in which neurons are cultured-have become a promising alternative to replace or minimize the use of animal models and have greatly facilitated in vitro research. Here, we review and discuss current developments in neuron-on-chip platforms, with a particular emphasis on existing biological models, culturing techniques, biomaterials, and topologies. We also discuss how the architecture, flow, and gradients affect neuronal growth, differentiation, and development. Finally, we discuss some of the most recent applications of NoCs in fundamental research (i.e., studies on the effects of electrical, mechanical/topological, or chemical stimuli) and in disease modeling.
Collapse
|
11
|
Choinière W, Petit È, Monfette V, Pelletier S, Godbout-Lavoie C, Lauzon MA. Dynamic three-dimensional coculture model: The future of tissue engineering applied to the peripheral nervous system. J Tissue Eng 2024; 15:20417314241265916. [PMID: 39139455 PMCID: PMC11320398 DOI: 10.1177/20417314241265916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/18/2024] [Indexed: 08/15/2024] Open
Abstract
Traumatic injuries to the peripheral nervous system (PNI) can lead to severe consequences such as paralysis. Unfortunately, current treatments rarely allow for satisfactory functional recovery. The high healthcare costs associated with PNS injuries, worker disability, and low patient satisfaction press for alternative solutions that surpass current standards. For the treatment of injuries with a deficit of less than 30 mm to bridge, the use of synthetic nerve conduits (NGC) is favored. However, to develop such promising therapeutic strategies, in vitro models that more faithfully mimic nerve physiology are needed. The absence of a clinically scaled model with essential elements such as a three-dimension environment and dynamic coculture has hindered progress in this field. The presented research focuses on the development of an in vitro coculture model of the peripheral nervous system (PNS) involving the use of functional biomaterial which microstructure replicates nerve topography. Initially, the behavior of neuron-derived cell lines (N) and Schwann cells (SC) in contact with a short section of biomaterial (5 mm) was studied. Subsequent investigations, using fluorescent markers and survival assays, demonstrated the synergistic effects of coculture. These optimized parameters were then applied to longer biomaterials (30 mm), equivalent to clinically used NGC. The results obtained demonstrated the possibility of maintaining an extended coculture of SC and N over a 7-day period on a clinically scaled biomaterial, observing some functionality. In the long term, the knowledge gained from this work will contribute to a better understanding of the PNS regeneration process and promote the development of future therapeutic approaches while reducing reliance on animal experimentation. This model can be used for drug screening and adapted for personalized medicine trials. Ultimately, this work fills a critical gap in current research, providing a transformative approach to study and advance treatments for PNS injuries.
Collapse
Affiliation(s)
- William Choinière
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Ève Petit
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Vincent Monfette
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Samuel Pelletier
- Department of Electrical and Informatics Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Catherine Godbout-Lavoie
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marc-Antoine Lauzon
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, QC, Canada
- Research Center on Aging, CIUSS de l’ESTRIE-CHUS, Sherbrooke, QC, Canada
- The Quebec Network for Research on Protein Function, Engineering, and Applications, Montréal, QC, Canada
| |
Collapse
|
12
|
Wang Z, Zhang Y, Li Z, Wang H, Li N, Deng Y. Microfluidic Brain-on-a-Chip: From Key Technology to System Integration and Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304427. [PMID: 37653590 DOI: 10.1002/smll.202304427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Indexed: 09/02/2023]
Abstract
As an ideal in vitro model, brain-on-chip (BoC) is an important tool to comprehensively elucidate brain characteristics. However, the in vitro model for the definition scope of BoC has not been universally recognized. In this review, BoC is divided into brain cells-on-a- chip, brain slices-on-a-chip, and brain organoids-on-a-chip according to the type of culture on the chip. Although these three microfluidic BoCs are constructed in different ways, they all use microfluidic chips as carrier tools. This method can better meet the needs of maintaining high culture activity on a chip for a long time. Moreover, BoC has successfully integrated cell biology, the biological material platform technology of microenvironment on a chip, manufacturing technology, online detection technology on a chip, and so on, enabling the chip to present structural diversity and high compatibility to meet different experimental needs and expand the scope of applications. Here, the relevant core technologies, challenges, and future development trends of BoC are summarized.
Collapse
Affiliation(s)
- Zhaohe Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yongqian Zhang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhe Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Hao Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Nuomin Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| | - Yulin Deng
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
13
|
Liu Y, Yao X, Fan C, Zhang G, Luo X, Qian Y. Microfabrication and lab-on-a-chip devices promote in vitromodeling of neural interfaces for neuroscience researches and preclinical applications. Biofabrication 2023; 16:012002. [PMID: 37832555 DOI: 10.1088/1758-5090/ad032a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 10/13/2023] [Indexed: 10/15/2023]
Abstract
Neural tissues react to injuries through the orchestration of cellular reprogramming, generating specialized cells and activating gene expression that helps with tissue remodeling and homeostasis. Simplified biomimetic models are encouraged to amplify the physiological and morphological changes during neural regeneration at cellular and molecular levels. Recent years have witnessed growing interest in lab-on-a-chip technologies for the fabrication of neural interfaces. Neural system-on-a-chip devices are promisingin vitromicrophysiological platforms that replicate the key structural and functional characteristics of neural tissues. Microfluidics and microelectrode arrays are two fundamental techniques that are leveraged to address the need for microfabricated neural devices. In this review, we explore the innovative fabrication, mechano-physiological parameters, spatiotemporal control of neural cell cultures and chip-based neurogenesis. Although the high variability in different constructs, and the restriction in experimental and analytical access limit the real-life applications of microphysiological models, neural system-on-a-chip devices have gained considerable translatability for modeling neuropathies, drug screening and personalized therapy.
Collapse
Affiliation(s)
- Yang Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xiangyun Yao
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| | - Guifeng Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Xi Luo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, People's Republic of China
| | - Yun Qian
- Department of Orthopedics, Shanghai Sixth People's Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, People's Republic of China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai 200233, People's Republic of China
| |
Collapse
|
14
|
Mutschler C, Fazal SV, Schumacher N, Loreto A, Coleman MP, Arthur-Farraj P. Schwann cells are axo-protective after injury irrespective of myelination status in mouse Schwann cell-neuron cocultures. J Cell Sci 2023; 136:jcs261557. [PMID: 37642648 PMCID: PMC10546878 DOI: 10.1242/jcs.261557] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023] Open
Abstract
Myelinating Schwann cell (SC)-dorsal root ganglion (DRG) neuron cocultures are an important technique for understanding cell-cell signalling and interactions during peripheral nervous system (PNS) myelination, injury, and regeneration. Although methods using rat SCs and neurons or mouse DRG explants are commonplace, there are no established protocols for compartmentalised myelinating cocultures with dissociated mouse cells. There consequently is a need for a coculture protocol that allows separate genetic manipulation of mouse SCs or neurons, or use of cells from different transgenic animals to complement in vivo mouse experiments. However, inducing myelination of dissociated mouse SCs in culture is challenging. Here, we describe a new method to coculture dissociated mouse SCs and DRG neurons in microfluidic chambers and induce robust myelination. Cocultures can be axotomised to study injury and used for drug treatments, and cells can be lentivirally transduced for live imaging. We used this model to investigate axon degeneration after traumatic axotomy and find that SCs, irrespective of myelination status, are axo-protective. At later timepoints after injury, live imaging of cocultures shows that SCs break up, ingest and clear axonal debris.
Collapse
Affiliation(s)
- Clara Mutschler
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Shaline V. Fazal
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Nathalie Schumacher
- Laboratory of Nervous System Disorders and Therapies, GIGA Neurosciences, University of Liège, 4000 Liège, Belgium
| | - Andrea Loreto
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Michael P. Coleman
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0PY, UK
| |
Collapse
|
15
|
Lee D, Yang K, Xie J. Advances in Nerve Injury Models on a Chip. Adv Biol (Weinh) 2023; 7:e2200227. [PMID: 36709421 DOI: 10.1002/adbi.202200227] [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: 08/17/2022] [Revised: 12/19/2022] [Indexed: 01/30/2023]
Abstract
Regeneration and functional recovery of the damaged nerve are challenging due to the need for effective therapeutic drugs, biomaterials, and approaches. The poor outcome of the treatment of nerve injury stems from the incomplete understanding of axonal biology and interactions between neurons and the surrounding environment, such as glial cells and extracellular matrix. Microfluidic devices, in combination with various injury techniques, have been applied to test biological hypotheses in nerve injury and nerve regeneration. The microfluidic devices provide multiple advantages over the in vitro cell culture on a petri dish and in vivo animal models because a specific part of the neuronal environment can be manipulated using physical and chemical interventions. In addition, single-cell behavior and interactions between neurons and glial cells can be visualized and quantified on microfluidic platforms. In this article, current in vitro nerve injury models on a chip that mimics in vivo axonal injuries and the regeneration process of axons are summarized. The microfluidic-based nerve injury models could enhance the understanding of the physiological and pathophysiological mechanisms of nerve tissues and simultaneously serve as powerful drug and biomaterial screening platforms.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kai Yang
- Department of Surgery-Plastic Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
16
|
Yu Y, Jin B, Chen J, Lou C, Guo J, Yang C, Zhao Y. Nerve-on-a-Chip Derived Biomimicking Microfibers for Peripheral Nerve Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2207536. [PMID: 37119478 PMCID: PMC10369236 DOI: 10.1002/advs.202207536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/28/2023] [Indexed: 06/19/2023]
Abstract
Fibrous scaffolds have shown their advantages in tissue engineering, such as peripheral nerve regeneration, while most of the existing fiber-shaped scaffolds are with simple structures, and the in vitro performance for nerve regeneration lacks systematic analysis. Here, novel nerve-on-a-chip derived biomimicking microfibers for peripheral nerve regeneration are presented. The microfibers with controllable core-shell structures and functionalities are generated through capillary microfluidic devices. By integrating these microfibers into a multitrack-architectured chip, and coculturing them with nerve cells as well as gradient bioactive elements, the nerve-on-a-chip with the capabilities of systematically assessing the performances of nerve fiber formation in the hollow microfibers at in vitro level is constructed. Based on a rat sciatic nerve injury model, the rapid promotion ability is demonstrated of optimized microfibers in nerve regeneration and function recovery in vivo, which implies the credibility of the nerve-on-a-chip on biomimicking microfibers evaluation for peripheral nerve regeneration. Thus, it is convinced that the organ-on-a-chip will undoubtedly open up a new chapter in evaluating biological scaffolds for in vivo tissue engineering.
Collapse
Affiliation(s)
- Yunru Yu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| | - Binghui Jin
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jinghao Chen
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Chenghao Lou
- Oujiang Laboratory, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Jiahui Guo
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Chaoyu Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325000, P. R. China
| |
Collapse
|
17
|
Aubry G, Lee HJ, Lu H. Advances in Microfluidics: Technical Innovations and Applications in Diagnostics and Therapeutics. Anal Chem 2023; 95:444-467. [PMID: 36625114 DOI: 10.1021/acs.analchem.2c04562] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Guillaume Aubry
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Hyun Jee Lee
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Hang Lu
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
18
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
19
|
Gharib G, Bütün İ, Muganlı Z, Kozalak G, Namlı İ, Sarraf SS, Ahmadi VE, Toyran E, van Wijnen AJ, Koşar A. Biomedical Applications of Microfluidic Devices: A Review. BIOSENSORS 2022; 12:1023. [PMID: 36421141 PMCID: PMC9688231 DOI: 10.3390/bios12111023] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/30/2022] [Accepted: 11/08/2022] [Indexed: 05/26/2023]
Abstract
Both passive and active microfluidic chips are used in many biomedical and chemical applications to support fluid mixing, particle manipulations, and signal detection. Passive microfluidic devices are geometry-dependent, and their uses are rather limited. Active microfluidic devices include sensors or detectors that transduce chemical, biological, and physical changes into electrical or optical signals. Also, they are transduction devices that detect biological and chemical changes in biomedical applications, and they are highly versatile microfluidic tools for disease diagnosis and organ modeling. This review provides a comprehensive overview of the significant advances that have been made in the development of microfluidics devices. We will discuss the function of microfluidic devices as micromixers or as sorters of cells and substances (e.g., microfiltration, flow or displacement, and trapping). Microfluidic devices are fabricated using a range of techniques, including molding, etching, three-dimensional printing, and nanofabrication. Their broad utility lies in the detection of diagnostic biomarkers and organ-on-chip approaches that permit disease modeling in cancer, as well as uses in neurological, cardiovascular, hepatic, and pulmonary diseases. Biosensor applications allow for point-of-care testing, using assays based on enzymes, nanozymes, antibodies, or nucleic acids (DNA or RNA). An anticipated development in the field includes the optimization of techniques for the fabrication of microfluidic devices using biocompatible materials. These developments will increase biomedical versatility, reduce diagnostic costs, and accelerate diagnosis time of microfluidics technology.
Collapse
Affiliation(s)
- Ghazaleh Gharib
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology Research and Application Centre (SUNUM), Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - İsmail Bütün
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | - Zülâl Muganlı
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | - Gül Kozalak
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - İlayda Namlı
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | | | | | - Erçil Toyran
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
| | - Andre J. van Wijnen
- Department of Biochemistry, University of Vermont, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | - Ali Koşar
- Faculty of Engineering and Natural Science, Sabanci University, Istanbul 34956, Turkey
- Sabanci University Nanotechnology Research and Application Centre (SUNUM), Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
20
|
Wang W, Gao S, Wang Y, Li Y, Yue W, Niu H, Yin F, Guo Y, Shen G. Advances in Emerging Photonic Memristive and Memristive-Like Devices. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105577. [PMID: 35945187 PMCID: PMC9534950 DOI: 10.1002/advs.202105577] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/06/2022] [Indexed: 05/19/2023]
Abstract
Possessing the merits of high efficiency, low consumption, and versatility, emerging photonic memristive and memristive-like devices exhibit an attractive future in constructing novel neuromorphic computing and miniaturized bionic electronic system. Recently, the potential of various emerging materials and structures for photonic memristive and memristive-like devices has attracted tremendous research efforts, generating various novel theories, mechanisms, and applications. Limited by the ambiguity of the mechanism and the reliability of the material, the development and commercialization of such devices are still rare and in their infancy. Therefore, a detailed and systematic review of photonic memristive and memristive-like devices is needed to further promote its development. In this review, the resistive switching mechanisms of photonic memristive and memristive-like devices are first elaborated. Then, a systematic investigation of the active materials, which induce a pivotal influence in the overall performance of photonic memristive and memristive-like devices, is highlighted and evaluated in various indicators. Finally, the recent advanced applications are summarized and discussed. In a word, it is believed that this review provides an extensive impact on many fields of photonic memristive and memristive-like devices, and lay a foundation for academic research and commercial applications.
Collapse
Affiliation(s)
- Wenxiao Wang
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Song Gao
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Yaqi Wang
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Yang Li
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Wenjing Yue
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Hongsen Niu
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Feifei Yin
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Yunjian Guo
- School of Information Science and EngineeringShandong Provincial Key Laboratory of Network Based Intelligent ComputingUniversity of JinanJinan250022China
| | - Guozhen Shen
- School of Integrated Circuits and ElectronicsBeijing Institute of TechnologyBeijing100081China
| |
Collapse
|
21
|
Nguyen T, Ho L, Moinuddin SM, Sarkar T, Saha D, Ahsan F. Multicellular Cell Seeding on a Chip: New Design and Optimization towards Commercialization. BIOSENSORS 2022; 12:bios12080587. [PMID: 36004984 PMCID: PMC9405756 DOI: 10.3390/bios12080587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/13/2022] [Accepted: 07/29/2022] [Indexed: 05/09/2023]
Abstract
This paper shows both experimental and in-depth theoretical studies (including simulations and analytical solutions) on a microfluidic platform to optimize its design and use for 3D multicellular co-culture applications, e.g., creating a tissue-on-chip model for investigating diseases such as pulmonary arterial hypertension (PAH). A tissue microfluidic chip usually has more than two channels to seed cells and supply media. These channels are often separated by barriers made of micro-posts. The optimization for the structures of these micro-posts and their spacing distances is not considered previously, especially for the aspects of rapid and cost-efficient fabrication toward scaling up and commercialization. Our experimental and theoretical (COMSOL simulations and analytical solutions) results showed the followings: (i) The cell seeding was performed successfully for this platform when the pressure drops across the two posts were significantly larger than those across the channel width. The circular posts can be used in the position of hexagonal or other shapes. (ii) In this work, circular posts are fabricated and used for the first time. They offer an excellent barrier effect, i.e., prevent the liquid and gel from migrating from one channel to another. (iii) As for rapid and cost-efficient production, our computer-aided manufacturing (CAM) simulation confirms that circular-post fabrication is much easier and more rapid than hexagonal posts when utilizing micro-machining techniques, e.g., micro-milling for creating the master mold, i.e., the shim for polymer injection molding. The findings open up a possibility for rapid, cost-efficient, large-scale fabrication of the tissue chips using micro-milling instead of expensive clean-room (soft) lithography techniques, hence enhancing the production of biochips via thermoplastic polymer injection molding and realizing commercialization.
Collapse
Affiliation(s)
- Trieu Nguyen
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA; (T.N.); (L.H.); (S.M.M.); (T.S.)
- East Bay Institute for Research & Education (EBIRE), Mather, CA 95655, USA;
| | - Linh Ho
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA; (T.N.); (L.H.); (S.M.M.); (T.S.)
| | - Sakib M. Moinuddin
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA; (T.N.); (L.H.); (S.M.M.); (T.S.)
- East Bay Institute for Research & Education (EBIRE), Mather, CA 95655, USA;
| | - Tanoy Sarkar
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA; (T.N.); (L.H.); (S.M.M.); (T.S.)
| | - Dipongkor Saha
- East Bay Institute for Research & Education (EBIRE), Mather, CA 95655, USA;
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, California Northstate University, Elk Grove, CA 95757, USA; (T.N.); (L.H.); (S.M.M.); (T.S.)
- East Bay Institute for Research & Education (EBIRE), Mather, CA 95655, USA;
- MedLuidics, Elk Grove, CA 95757, USA
- Correspondence:
| |
Collapse
|
22
|
Ko J, Park D, Lee S, Gumuscu B, Jeon NL. Engineering Organ-on-a-Chip to Accelerate Translational Research. MICROMACHINES 2022; 13:1200. [PMID: 36014122 PMCID: PMC9412404 DOI: 10.3390/mi13081200] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 02/01/2023]
Abstract
We guide the use of organ-on-chip technology in tissue engineering applications. Organ-on-chip technology is a form of microengineered cell culture platform that elaborates the in-vivo like organ or tissue microenvironments. The organ-on-chip platform consists of microfluidic channels, cell culture chambers, and stimulus sources that emulate the in-vivo microenvironment. These platforms are typically engraved into an oxygen-permeable transparent material. Fabrication of these materials requires the use of microfabrication strategies, including soft lithography, 3D printing, and injection molding. Here we provide an overview of what is an organ-on-chip platform, where it can be used, what it is composed of, how it can be fabricated, and how it can be operated. In connection with this topic, we also introduce an overview of the recent applications, where different organs are modeled on the microscale using this technology.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Korea;
| | - Dohyun Park
- Bio-MAX Institute, Seoul National University, Seoul 08826, Korea;
| | - Somin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Korea;
| | - Burcu Gumuscu
- Biosensors and Devices Laboratory, Biomedical Engineering Department, Institute for Complex Molecular Systems, Eindhoven Artificial Intelligence Systems Institute, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands;
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Korea;
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Korea;
- Institute of Advanced Machines and Design, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
23
|
Seo Y, Bang S, Son J, Kim D, Jeong Y, Kim P, Yang J, Eom JH, Choi N, Kim HN. Brain physiome: A concept bridging in vitro 3D brain models and in silico models for predicting drug toxicity in the brain. Bioact Mater 2022; 13:135-148. [PMID: 35224297 PMCID: PMC8843968 DOI: 10.1016/j.bioactmat.2021.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, adverse reactions to pharmaceuticals have been evaluated using 2D in vitro models and animal models. However, with increasing computational power, and as the key drivers of cellular behavior have been identified, in silico models have emerged. These models are time-efficient and cost-effective, but the prediction of adverse reactions to unknown drugs using these models requires relevant experimental input. Accordingly, the physiome concept has emerged to bridge experimental datasets with in silico models. The brain physiome describes the systemic interactions of its components, which are organized into a multilevel hierarchy. Because of the limitations in obtaining experimental data corresponding to each physiome component from 2D in vitro models and animal models, 3D in vitro brain models, including brain organoids and brain-on-a-chip, have been developed. In this review, we present the concept of the brain physiome and its hierarchical organization, including cell- and tissue-level organizations. We also summarize recently developed 3D in vitro brain models and link them with the elements of the brain physiome as a guideline for dataset collection. The connection between in vitro 3D brain models and in silico modeling will lead to the establishment of cost-effective and time-efficient in silico models for the prediction of the safety of unknown drugs.
Collapse
Affiliation(s)
- Yoojin Seo
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongtae Son
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongsup Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yong Jeong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Pilnam Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihun Yang
- Next&Bio Inc., Seoul, 02841, Republic of Korea
| | - Joon-Ho Eom
- Medical Device Research Division, National Institute of Food and Drug Safety Evaluation, Cheongju, 28159, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
24
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 439] [Impact Index Per Article: 109.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
25
|
Malheiro A, Harichandan A, Bernardi J, Seijas-Gamardo A, Konings GF, Volders PGA, Romano A, Mota C, Wieringa P, Moroni L. 3D culture platform of human iPSCs-derived nociceptors for peripheral nerve modelling and tissue innervation. Biofabrication 2021; 14. [PMID: 34736244 DOI: 10.1088/1758-5090/ac36bf] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/04/2021] [Indexed: 11/11/2022]
Abstract
Functional humanized in vitro nerve models are coveted as an alternative to animal models due to their ease of access, lower cost, clinical relevance and no need for recurrent animal sacrifice. To this end, we developed a sensory nerve model using induced pluripotent stem cells (iPSCs)-derived nociceptors that are electrically active and exhibit a functional response to noxious stimuli. The differentiated neurons were co-cultured with primary Schwann cells on an aligned microfibrous scaffold to produce biomimetic peripheral nerve tissue. Compared to glass coverslips, our scaffold enhances tissue development and stabilization. Using this model, we demonstrate that myelin damage can be induced from hyperglycemia exposure (glucose at 45 mM) and mitigated by epalrestat (1µM) supplementation. Through fibrin embedding of the platform, we were able to create 3D anisotropic myelinated tissue, reaching over 6.5 mm in length. Finally, as a proof-of-concept, we incorporated pancreatic pseudoislets and endometrial organoids into our nerve platform, to demonstrate the potential in generating nociceptor innervation models. In summary, we propose here an improved tool for neurobiology research with potential applications in pathology modelling, drug screening and target tissue innervation.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration department, Maastricht University, Universiteitssingel 40, Maastricht, 6200 MD, NETHERLANDS
| | - Abhishek Harichandan
- Complex Tissue Regeneration department, Maastricht University, Universiteitssingel, 40, Maastricht, 6200 MD, NETHERLANDS
| | - Joyce Bernardi
- Department of Cardiology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Adrián Seijas-Gamardo
- Complex Tissue Regeneration department, Maastricht University, Universiteitssingel 40, Maastricht, 6200 MD, NETHERLANDS
| | - Gonda F Konings
- Department of Gynaecology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Paul G A Volders
- Department of Cardiology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Andrea Romano
- Department of Gynaecology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Carlos Mota
- Department of Complex Tissue Regeneration (CTR), Maastricht University, Universiteitssingel, 40, office 3.541A, Maastricht, 6229 ER, NETHERLANDS
| | - Paul Wieringa
- Complex Tissue Regeneration, Maastricht University MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229ER, NETHERLANDS
| | - Lorenzo Moroni
- Complex Tissue Regeneration, Maastricht University, Universiteitsingel, 40, Maastricht, 6200 MD, NETHERLANDS
| |
Collapse
|
26
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
27
|
Marx U, Accastelli E, David R, Erfurth H, Koenig L, Lauster R, Ramme AP, Reinke P, Volk HD, Winter A, Dehne EM. An Individual Patient's "Body" on Chips-How Organismoid Theory Can Translate Into Your Personal Precision Therapy Approach. Front Med (Lausanne) 2021; 8:728866. [PMID: 34589503 PMCID: PMC8473633 DOI: 10.3389/fmed.2021.728866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/16/2021] [Indexed: 11/24/2022] Open
Abstract
The first concepts for reproducing human systemic organismal biology in vitro were developed over 12 years ago. Such concepts, then called human- or body-on-a-chip, claimed that microphysiological systems would become the relevant technology platform emulating the physiology and morphology of human organisms at the smallest biologically acceptable scale in vitro and, therefore, would enable the selection of personalized therapies for any patient at unprecedented precision. Meanwhile, the first human organoids-stem cell-derived complex three-dimensional organ models that expand and self-organize in vitro-have proven that in vitro self-assembly of minute premature human organ-like structures is feasible, once the respective stimuli of ontogenesis are provided to human stem cells. Such premature organoids can precisely reflect a number of distinct physiological and pathophysiological features of their respective counterparts in the human body. We now develop the human-on-a-chip concepts of the past into an organismoid theory. We describe the current concept and principles to create a series of organismoids-minute, mindless and emotion-free physiological in vitro equivalents of an individual's mature human body-by an artificially short process of morphogenetic self-assembly mimicking an individual's ontogenesis from egg cell to sexually mature organism. Subsequently, we provide the concept and principles to maintain such an individual's set of organismoids at a self-sustained functional healthy homeostasis over very long time frames in vitro. Principles how to perturb a subset of healthy organismoids by means of the natural or artificial induction of diseases are enrolled to emulate an individual's disease process. Finally, we discuss using such series of healthy and perturbed organismoids in predictively selecting, scheduling and dosing an individual patient's personalized therapy or medicine precisely. The potential impact of the organismoid theory on our healthcare system generally and the rapid adoption of disruptive personalized T-cell therapies particularly is highlighted.
Collapse
Affiliation(s)
- Uwe Marx
- Department of Medical Biotechnology, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
- TissUse GmbH, Berlin, Germany
| | | | - Rhiannon David
- Functional and Mechanistic Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Roland Lauster
- Department of Medical Biotechnology, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | | | - Petra Reinke
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
- BIH-Center for Regenerative Therapies, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- BIH-Center for Regenerative Therapies, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | |
Collapse
|
28
|
Kajtez J, Nilsson F, Fiorenzano A, Parmar M, Emnéus J. 3D biomaterial models of human brain disease. Neurochem Int 2021; 147:105043. [PMID: 33887378 DOI: 10.1016/j.neuint.2021.105043] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 01/25/2023]
Abstract
Inherent limitations of the traditional approaches to study brain function and disease, such as rodent models and 2D cell culture platforms, have led to the development of 3D in vitro cell culture systems. These systems, products of multidisciplinary efforts encompassing stem cell biology, materials engineering, and biofabrication, have quickly shown great potential to mimic biochemical composition, structural properties, and cellular morphology and diversity found in the native brain tissue. Crucial to these developments have been the advancements in stem cell technology and cell reprogramming protocols that allow reproducible generation of human subtype-specific neurons and glia in laboratory conditions. At the same time, biomaterials have been designed to provide cells in 3D with a microenvironment that mimics functional and structural aspects of the native extracellular matrix with increasing fidelity. In this article, we review the use of biomaterials in 3D in vitro models of neurological disorders with focus on hydrogel technology and with biochemical composition and physical properties of the in vivo environment as reference.
Collapse
Affiliation(s)
- Janko Kajtez
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden.
| | - Fredrik Nilsson
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden
| | - Alessandro Fiorenzano
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden
| | - Malin Parmar
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden
| | - Jenny Emnéus
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
29
|
Kim S, Ko J, Lee SR, Park D, Park S, Jeon NL. Anchor-IMPACT: A standardized microfluidic platform for high-throughput antiangiogenic drug screening. Biotechnol Bioeng 2021; 118:2524-2535. [PMID: 33764506 DOI: 10.1002/bit.27765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/16/2021] [Accepted: 03/23/2021] [Indexed: 01/22/2023]
Abstract
In vitro models are becoming more advanced to truly present physiological systems while enabling high-throughput screening and analysis. Organ-on-a-chip devices provide remarkable results through the reconstruction of a three-dimensional (3D) cellular microenvironment although they need to be further developed in terms of multiple liquid patterning principle, material properties, and scalability. Here we present a 3D anchor-based microfluidic injection-molded plastic array culture platform (Anchor-IMPACT) that enables selective, space-intensive patterning of hydrogels using anchor-island for high-throughput angiogenesis evaluation model. Anchor-IMPACT consists of a central channel and an anchor-island, integrating the array into an abbreviated 96-well plate format with a standard microscope slide size. The anchor-island enables selective 3D cell patterning without channel-to-channel contact or any hydrogel injection port using an anchor structure unlike conventional culture compartment. The hydrogel was patterned into defined regions by spontaneous capillary flow under hydrophilic conditions. We configured multiple cell patterning structures to investigate the angiogenic potency of colorectal cancer cells in Anchor-IMPACT and the morphological properties of the angiogenesis induced by the paracrine effect were evaluated. In addition, the efficacy of anticancer drugs against angiogenic sprouts was verified by following dose-dependent responses. Our results indicate that Anchor-IMPACT offers not only a model of high-throughput experimentation but also an advanced 3D cell culture platform and can significantly improve current in vitro models while providing the basis for developing predictive preclinical models for biopharmaceutical applications.
Collapse
Affiliation(s)
- Suryong Kim
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Jihoon Ko
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Seung-Ryeol Lee
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Seunghyuk Park
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul, Republic of Korea.,Institute of Advanced Machines and Design, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|