1
|
Ryu JR, Ko K, Sun W. Polarization of organoids by bioengineered symmetry breaking. IBRO Neurosci Rep 2024; 17:22-31. [PMID: 38881849 PMCID: PMC11176950 DOI: 10.1016/j.ibneur.2024.05.002] [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: 10/27/2023] [Accepted: 05/08/2024] [Indexed: 06/18/2024] Open
Abstract
Symmetry breaking leading to axis formation and spatial patterning is crucial for achieving more accurate recapitulation of human development in organoids. While these processes can occur spontaneously by self-organizing capabilities of pluripotent stem cells, they can often result in variation in structure and composition of cell types within organoids. To address this limitation, bioengineering techniques that utilize geometric, topological and stiffness factors are increasingly employed to enhance control and consistency. Here, we review how spontaneous manners and engineering tools such as micropattern, microfluidics, biomaterials, etc. can facilitate the process of symmetry breaking leading to germ layer patterning and the formation of anteroposterior and dorsoventral axes in blastoids, gastruloids, neuruloids and neural organoids. Furthermore, brain assembloids, which are composed of multiple brain regions through fusion processes are discussed. The overview of organoid polarization in terms of patterning tools can offer valuable insights for enhancing the physiological relevance of organoid system.
Collapse
Affiliation(s)
- Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kahee Ko
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
2
|
Li M, Yuan Y, Hou Z, Hao S, Jin L, Wang B. Human brain organoid: trends, evolution, and remaining challenges. Neural Regen Res 2024; 19:2387-2399. [PMID: 38526275 PMCID: PMC11090441 DOI: 10.4103/1673-5374.390972] [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: 06/19/2023] [Revised: 09/26/2023] [Accepted: 10/28/2023] [Indexed: 03/26/2024] Open
Abstract
Advanced brain organoids provide promising platforms for deciphering the cellular and molecular processes of human neural development and diseases. Although various studies and reviews have described developments and advancements in brain organoids, few studies have comprehensively summarized and analyzed the global trends in this area of neuroscience. To identify and further facilitate the development of cerebral organoids, we utilized bibliometrics and visualization methods to analyze the global trends and evolution of brain organoids in the last 10 years. First, annual publications, countries/regions, organizations, journals, authors, co-citations, and keywords relating to brain organoids were identified. The hotspots in this field were also systematically identified. Subsequently, current applications for brain organoids in neuroscience, including human neural development, neural disorders, infectious diseases, regenerative medicine, drug discovery, and toxicity assessment studies, are comprehensively discussed. Towards that end, several considerations regarding the current challenges in brain organoid research and future strategies to advance neuroscience will be presented to further promote their application in neurological research.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhan Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zongkun Hou
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
3
|
Chooi WH, Wu Y, Ng SY. Defined hydrogels for spinal cord organoids: challenges and potential applications. Neural Regen Res 2024; 19:2329-2330. [PMID: 38526259 PMCID: PMC11090425 DOI: 10.4103/nrr.nrr-d-23-01665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/07/2023] [Accepted: 12/26/2023] [Indexed: 03/26/2024] Open
Affiliation(s)
- Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuewen Wu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
| | - Shi-Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
4
|
Arnaldi P, Casarotto E, Relucenti M, Bellese G, Gagliani MC, Crippa V, Castagnola P, Cortese K. A NSC-34 cell line-derived spheroid model: Potential and challenges for in vitro evaluation of neurodegeneration. Microsc Res Tech 2024; 87:2785-2800. [PMID: 38988205 DOI: 10.1002/jemt.24651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
Three-dimensional (3D) spheroid models aim to bridge the gap between traditional two-dimensional (2D) cultures and the complex in vivo tissue environment. These models, created by self-clustering cells to mimic a 3D environment with surrounding extracellular framework, provide a valuable research tool. The NSC-34 cell line, generated by fusing mouse spinal cord motor neurons and neuroblastoma cells, is essential for studying neurodegenerative diseases like amyotrophic lateral sclerosis (ALS), where abnormal protein accumulation, such as TAR-DNA-binding protein 43 (TDP-43), occurs in affected nerve cells. However, NSC-34 behavior in a 3D context remains underexplored, and this study represents the first attempt to create a 3D model to determine its suitability for studying pathology. We generated NSC-34 spheroids using a nonadhesive hydrogel-based template and characterized them for 6 days. Light microscopy revealed that NSC-34 cells in 3D maintained high viability, a distinct round shape, and forming stable membrane connections. Scanning electron microscopy identified multiple tunnel-like structures, while ultrastructural analysis highlighted nuclear bending and mitochondria alterations. Using inducible GFP-TDP-43-expressing NSC-34 spheroids, we explored whether 3D structure affected TDP-43 expression, localization, and aggregation. Spheroids displayed nuclear GFP-TDP-43 expression, albeit at a reduced level compared with 2D cultures and generated both TDP-35 fragments and TDP-43 aggregates. This study sheds light on the distinctive behavior of NSC-34 in 3D culture, suggesting caution in the use of the 3D model for ALS or TDP-43 pathologies. Yet, it underscores the spheroids' potential for investigating fundamental cellular mechanisms, cell adaptation in a 3D context, future bioreactor applications, and drug penetration studies. RESEARCH HIGHLIGHTS: 3D spheroid generation: NSC-34 spheroids, developed using a hydrogel-based template, showed high viability and distinct shapes for 6 days. Structural features: advanced microscopy identified tunnel-like structures and nuclear and mitochondrial changes in the spheroids. Protein dynamics: the study observed how 3D structures impact TDP-43 behavior, with altered expression but similar aggregation patterns to 2D cultures. Research implications: this study reveals the unique behavior of NSC-34 in 3D culture, suggests a careful approach to use this model for ALS or TDP-43 pathologies, and highlights its potential in cellular mechanism research and drug testing applications.
Collapse
Affiliation(s)
- Pietro Arnaldi
- Department of Experimental Medicine, Cellular Electron Microscopy Lab, University of Genoa, Genoa, Italy
| | - Elena Casarotto
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Department of Excellence 2018-2027, University of Milan, Milan, Italy
| | - Michela Relucenti
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Grazia Bellese
- Department of Experimental Medicine, Cellular Electron Microscopy Lab, University of Genoa, Genoa, Italy
| | - Maria Cristina Gagliani
- Department of Experimental Medicine, Cellular Electron Microscopy Lab, University of Genoa, Genoa, Italy
| | - Valeria Crippa
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Department of Excellence 2018-2027, University of Milan, Milan, Italy
| | | | - Katia Cortese
- Department of Experimental Medicine, Cellular Electron Microscopy Lab, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
5
|
Onesto MM, Kim JI, Pasca SP. Assembloid models of cell-cell interaction to study tissue and disease biology. Cell Stem Cell 2024:S1934-5909(24)00360-6. [PMID: 39454582 DOI: 10.1016/j.stem.2024.09.017] [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: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Neurodevelopment involves the migration, projection, and integration of various cell types across different regions of the nervous system. Assembloids are self-organizing systems formed by the integration of multiple organoids or cell types. Here, we outline the generation and application of assembloids. We illustrate how assembloids recapitulate critical neurodevelopmental steps, like migration, axon projection, and circuit formation, and how they are starting to provide biological insights into neuropsychiatric disorders. Additionally, we review how assembloids can be used to study properties emerging from cell-cell interactions within non-neural tissues. Overall, assembloid platforms represent a powerful tool for discovering human biology and developing therapeutics.
Collapse
Affiliation(s)
- Massimo M Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Sergiu P Pasca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Kim JI, Miura Y, Li MY, Revah O, Selvaraj S, Birey F, Meng X, Thete MV, Pavlov SD, Andersen J, Pașca AM, Porteus MH, Huguenard JR, Pașca SP. Human assembloids reveal the consequences of CACNA1G gene variants in the thalamocortical pathway. Neuron 2024:S0896-6273(24)00692-5. [PMID: 39419023 DOI: 10.1016/j.neuron.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Abnormalities in thalamocortical crosstalk can lead to neuropsychiatric disorders. Variants in CACNA1G, which encodes the α1G subunit of the thalamus-enriched T-type calcium channel, are associated with absence seizures, intellectual disability, and schizophrenia, but the cellular and circuit consequences of these genetic variants in humans remain unknown. Here, we developed a human assembloid model of the thalamocortical pathway to dissect the contribution of genetic variants in T-type calcium channels. We discovered that the M1531V CACNA1G variant associated with seizures led to changes in T-type currents in thalamic neurons, as well as correlated hyperactivity of thalamic and cortical neurons in assembloids. By contrast, CACNA1G loss, which has been associated with risk of schizophrenia, resulted in abnormal thalamocortical connectivity that was related to both increased spontaneous thalamic activity and aberrant axonal projections. These results illustrate the utility of multi-cellular systems for interrogating human genetic disease risk variants at both cellular and circuit level.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Min-Yin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sridhar Selvaraj
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Fikri Birey
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Xiangling Meng
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Sergey D Pavlov
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Jimena Andersen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Anca M Pașca
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Cheng JL, Cook AL, Talbot J, Perry S. How is Excitotoxicity Being Modelled in iPSC-Derived Neurons? Neurotox Res 2024; 42:43. [PMID: 39405005 PMCID: PMC11480214 DOI: 10.1007/s12640-024-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Excitotoxicity linked either to environmental causes (pesticide and cyanotoxin exposure), excitatory neurotransmitter imbalance, or to intrinsic neuronal hyperexcitability, is a pathological mechanism central to neurodegeneration in amyotrophic lateral sclerosis (ALS). Investigation of excitotoxic mechanisms using in vitro and in vivo animal models has been central to understanding ALS mechanisms of disease. In particular, advances in induced pluripotent stem cell (iPSC) technologies now provide human cell-based models that are readily amenable to environmental and network-based excitotoxic manipulations. The cell-type specific differentiation of iPSC, combined with approaches to modelling excitotoxicity that include editing of disease-associated gene variants, chemogenetics, and environmental risk-associated exposures make iPSC primed to examine gene-environment interactions and disease-associated excitotoxic mechanisms. Critical to this is knowledge of which neurotransmitter receptor subunits are expressed by iPSC-derived neuronal cultures being studied, how their activity responds to antagonists and agonists of these receptors, and how to interpret data derived from multi-parameter electrophysiological recordings. This review explores how iPSC-based studies have contributed to our understanding of ALS-linked excitotoxicity and highlights novel approaches to inducing excitotoxicity in iPSC-derived neurons to further our understanding of its pathological pathways.
Collapse
Affiliation(s)
- Jan L Cheng
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Anthony L Cook
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Jana Talbot
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia.
| |
Collapse
|
8
|
Miura Y, Kim JI, Jurjuț O, Kelley KW, Yang X, Chen X, Thete MV, Revah O, Cui B, Pachitariu M, Pașca SP. Assembloid model to study loop circuits of the human nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.617729. [PMID: 39463945 PMCID: PMC11507680 DOI: 10.1101/2024.10.13.617729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Neural circuits connecting the cerebral cortex, the basal ganglia and the thalamus are fundamental networks for sensorimotor processing and their dysfunction has been consistently implicated in neuropsychiatric disorders 1-9 . These recursive, loop circuits have been investigated in animal models and by clinical neuroimaging, however, direct functional access to developing human neurons forming these networks has been limited. Here, we use human pluripotent stem cells to reconstruct an in vitro cortico-striatal-thalamic-cortical circuit by creating a four-part loop assembloid. More specifically, we generate regionalized neural organoids that resemble the key elements of the cortico-striatal-thalamic-cortical circuit, and functionally integrate them into loop assembloids using custom 3D-printed biocompatible wells. Volumetric and mesoscale calcium imaging, as well as extracellular recordings from individual parts of these assembloids reveal the emergence of synchronized patterns of neuronal activity. In addition, a multi-step rabies retrograde tracing approach demonstrate the formation of neuronal connectivity across the network in loop assembloids. Lastly, we apply this system to study heterozygous loss of ASH1L gene associated with autism spectrum disorder and Tourette syndrome and discover aberrant synchronized activity in disease model assembloids. Taken together, this human multi-cellular platform will facilitate functional investigations of the cortico-striatal-thalamic-cortical circuit in the context of early human development and in disease conditions.
Collapse
|
9
|
Xu Y, Shen Y. The Assembly of Miniaturized Droplets toward Functional Architectures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2404366. [PMID: 39380419 DOI: 10.1002/smll.202404366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/19/2024] [Indexed: 10/10/2024]
Abstract
Recent explorations of bioengineering have generated new concepts and strategies for the processing of soft and functional materials. Droplet assembly techniques can address problems in the construction of extremely soft architectures by expanding the manufacturing capabilities using droplets containing liquid or hydrogels including weak hydrogels. This Perspective sets out to provide a brief overview of this growing field, and discusses the challenges and opportunities ahead. The study highlights the recent key advances of materials and architectures from hitherto effective droplet-assembly technologies, as well as the applications in biomedical and bioengineering fields from artificial tissues to bioreactors. It is envisaged that these assembled architectures, as nature-inspired models, will stimulate the discovery of biomaterials and miniaturized platforms for interdisciplinary research in health, biotechnology, and sustainability.
Collapse
Affiliation(s)
- Yufan Xu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Yi Shen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
10
|
Alam El Din DM, Shin J, Lysinger A, Roos MJ, Johnson EC, Shafer TJ, Hartung T, Smirnova L. Organoid intelligence for developmental neurotoxicity testing. Front Cell Neurosci 2024; 18:1480845. [PMID: 39440004 PMCID: PMC11493634 DOI: 10.3389/fncel.2024.1480845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
The increasing prevalence of neurodevelopmental disorders has highlighted the need for improved testing methods to determine developmental neurotoxicity (DNT) hazard for thousands of chemicals. This paper proposes the integration of organoid intelligence (OI); leveraging brain organoids to study neuroplasticity in vitro, into the DNT testing paradigm. OI brings a new approach to measure the impacts of xenobiotics on plasticity mechanisms - a critical biological process that is not adequately covered in current DNT in vitro assays. Finally, the integration of artificial intelligence (AI) techniques will further facilitate the analysis of complex brain organoid data to study these plasticity mechanisms.
Collapse
Affiliation(s)
- Dowlette-Mary Alam El Din
- Center for Alternatives to Animal Testing, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Jeongwon Shin
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alexandra Lysinger
- Center for Alternatives to Animal Testing, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Matthew J. Roos
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States
| | - Erik C. Johnson
- Research and Exploratory Development Department, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States
| | - Timothy J. Shafer
- Center for Computational Toxicology and Exposure, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
- Center for Alternatives to Animal Testing Europe, University of Konstanz, Konstanz, Germany
- Doerenkamp-Zbinden Chair for Evidence-based Toxicology, Baltimore, MD, United States
| | - Lena Smirnova
- Center for Alternatives to Animal Testing, Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
11
|
Pang W, Zhu J, Yang K, Zhu X, Zhou W, Jiang L, Zhuang X, Liu Y, Wei J, Lu X, Yin Y, Chen Z, Xiang Y. Generation of human region-specific brain organoids with medullary spinal trigeminal nuclei. Cell Stem Cell 2024; 31:1501-1512.e8. [PMID: 39208804 DOI: 10.1016/j.stem.2024.08.004] [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: 01/10/2024] [Revised: 06/16/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Brain organoids with nucleus-specific identities provide unique platforms for studying human brain development and diseases at a finer resolution. Despite its essential role in vital body functions, the medulla of the hindbrain has seen a lack of in vitro models, let alone models resembling specific medullary nuclei, including the crucial spinal trigeminal nucleus (SpV) that relays peripheral sensory signals to the thalamus. Here, we report a method to differentiate human pluripotent stem cells into region-specific brain organoids resembling the dorsal domain of the medullary hindbrain. Importantly, organoids specifically recapitulated the development of the SpV derived from the dorsal medulla. We also developed an organoid system to create the trigeminothalamic projections between the SpV and the thalamus by fusing these organoids, namely human medullary SpV-like organoids (hmSpVOs), with organoids representing the thalamus (hThOs). Our study provides a platform for understanding SpV development, nucleus-based circuit organization, and related disorders in the human brain.
Collapse
Affiliation(s)
- Wei Pang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinkui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaona Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yantong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoxiang Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziling Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
12
|
Leconte M, Bonne G, Bertrand AT. Recent insights in striated muscle laminopathies. Curr Opin Neurol 2024; 37:509-514. [PMID: 38989655 DOI: 10.1097/wco.0000000000001297] [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: 07/12/2024]
Abstract
PURPOSE OF REVIEW To highlight recent insights in different aspects of striated muscle laminopathies (SMLs) related to LMNA mutations. RECENT FINDINGS Clinical and genetic studies allow better patient management and diagnosis, with confirmation of ventricular tachyarrhythmias (VTA) risk prediction score to help with ICD implantation and development of models to help with classification of LMNA variants of uncertain significance. From a pathophysiology perspective, characterization of lamin interactomes in different contexts revealed new lamin A/C partners. Expression or function modulation of these partners evidenced them as potential therapeutic targets. After a positive phase 2, the first phase 3 clinical trial, testing a p38 inhibitor targeting the life-threatening cardiac disease of SML, has been recently stopped, thus highlighting the need for new therapeutic approaches together with new animal and cell models. SUMMARY Since the first LMNA mutation report in 1999, lamin A/C structure and functions have been actively explored to understand the SML pathophysiology. The latest discoveries of partners and altered pathways, highlight the importance of lamin A/C at the nuclear periphery and in the nucleoplasm. Modulation of altered pathways allowed some benefits, especially for cardiac involvement. However, additional studies are still needed to fully assess treatment efficacy and safety.
Collapse
Affiliation(s)
- Marine Leconte
- Sorbonne Université, Inserm, Institut de Myologie, Centre de recherche en Myologie, Paris, France
| | | | | |
Collapse
|
13
|
Cadena MA, Sing A, Taylor K, Jin L, Ning L, Amoli MS, Singh Y, Lanjewar SN, Tomov ML, Serpooshan V, Sloan SA. A 3D Bioprinted Cortical Organoid Platform for Modeling Human Brain Development. Adv Healthc Mater 2024; 13:e2401603. [PMID: 38815975 PMCID: PMC11518656 DOI: 10.1002/adhm.202401603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/24/2024] [Indexed: 06/01/2024]
Abstract
The ability to promote three-dimensional (3D) self-organization of induced pluripotent stem cells into complex tissue structures called organoids presents new opportunities for the field of developmental biology. Brain organoids have been used to investigate principles of neurodevelopment and neuropsychiatric disorders and serve as a drug screening and discovery platform. However, brain organoid cultures are currently limited by a lacking ability to precisely control their extracellular environment. Here, this work employs 3D bioprinting to generate a high-throughput, tunable, and reproducible scaffold for controlling organoid development and patterning. Additionally, this approach supports the coculture of organoids and vascular cells in a custom architecture containing interconnected endothelialized channels. Printing fidelity and mechanical assessments confirm that fabricated scaffolds closely match intended design features and exhibit stiffness values reflective of the developing human brain. Using organoid growth, viability, cytoarchitecture, proliferation, and transcriptomic benchmarks, this work finds that organoids cultured within the bioprinted scaffold long-term are healthy and have expected neuroectodermal differentiation. Lastly, this work confirms that the endothelial cells (ECs) in printed channel structures can migrate toward and infiltrate into the embedded organoids. This work demonstrates a tunable 3D culturing platform that can be used to create more complex and accurate models of human brain development and underlying diseases.
Collapse
Affiliation(s)
- Melissa A. Cadena
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anson Sing
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kylie Taylor
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Linqi Jin
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Liqun Ning
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Mechanical Engineering, Cleveland State University, Cleveland, OH, United States
| | - Mehdi Salar Amoli
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - Yamini Singh
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
| | - The Brain Organoid Hub
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Samantha N. Lanjewar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Martin L. Tomov
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, US
| | - Steven A. Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
14
|
Jiang Y, Zhou R, Liao F, Kong G, Zeng J, Wu Y, Li X, Wang B, Qi F, Chen S, Zhu Q, Gu L, Zheng C. Unraveling radiation-induced skeletal muscle damage: Insights from a 3D human skeletal muscle organoid model. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119792. [PMID: 38936620 DOI: 10.1016/j.bbamcr.2024.119792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/28/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND Three-dimensional (3D) organoids derived from human pluripotent stem cells (hPSCs) have revolutionized in vitro tissue modeling, offering a unique opportunity to replicate physiological tissue organization and functionality. This study investigates the impact of radiation on skeletal muscle response using an innovative in vitro human 3D skeletal muscle organoids (hSMOs) model derived from hPSCs. METHODS The hSMOs model was established through a differentiation protocol faithfully recapitulating embryonic myogenesis and maturation via paraxial mesodermal differentiation of hPSCs. Key skeletal muscle characteristics were confirmed using immunofluorescent staining and RT-qPCR. Subsequently, the hSMOs were exposed to a clinically relevant dose of 2 Gy of radiation, and their response was analyzed using immunofluorescent staining and RNA-seq. RESULTS The hSMO model faithfully recapitulated embryonic myogenesis and maturation, maintaining key skeletal muscle characteristics. Following exposure to 2 Gy of radiation, histopathological analysis revealed deficits in hSMOs expansion, differentiation, and repair response across various cell types at early (30 min) and intermediate (18 h) time points post-radiation. Immunofluorescent staining targeting γH2AX and 53BP1 demonstrated elevated levels of foci per cell, particularly in PAX7+ cells, during early and intermediate time points, with a distinct kinetic pattern showing a decrease at 72 h. RNA-seq data provided comprehensive insights into the DNA damage response within the hSMOs. CONCLUSIONS Our findings highlight deficits in expansion, differentiation, and repair response in hSMOs following radiation exposure, enhancing our understanding of radiation effects on skeletal muscle and contributing to strategies for mitigating radiation-induced damage in this context.
Collapse
Affiliation(s)
- Yifei Jiang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Runtao Zhou
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Fawei Liao
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Ganggang Kong
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jingguang Zeng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Yixun Wu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Xubo Li
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Bo Wang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Fangze Qi
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Shiju Chen
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Qintang Zhu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Liqiang Gu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China
| | - Canbin Zheng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou 510080, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou 510080, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou 510080, China.
| |
Collapse
|
15
|
Mierke CT. Bioprinting of Cells, Organoids and Organs-on-a-Chip Together with Hydrogels Improves Structural and Mechanical Cues. Cells 2024; 13:1638. [PMID: 39404401 PMCID: PMC11476109 DOI: 10.3390/cells13191638] [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: 08/20/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
The 3D bioprinting technique has made enormous progress in tissue engineering, regenerative medicine and research into diseases such as cancer. Apart from individual cells, a collection of cells, such as organoids, can be printed in combination with various hydrogels. It can be hypothesized that 3D bioprinting will even become a promising tool for mechanobiological analyses of cells, organoids and their matrix environments in highly defined and precisely structured 3D environments, in which the mechanical properties of the cell environment can be individually adjusted. Mechanical obstacles or bead markers can be integrated into bioprinted samples to analyze mechanical deformations and forces within these bioprinted constructs, such as 3D organoids, and to perform biophysical analysis in complex 3D systems, which are still not standard techniques. The review highlights the advances of 3D and 4D printing technologies in integrating mechanobiological cues so that the next step will be a detailed analysis of key future biophysical research directions in organoid generation for the development of disease model systems, tissue regeneration and drug testing from a biophysical perspective. Finally, the review highlights the combination of bioprinted hydrogels, such as pure natural or synthetic hydrogels and mixtures, with organoids, organoid-cell co-cultures, organ-on-a-chip systems and organoid-organ-on-a chip combinations and introduces the use of assembloids to determine the mutual interactions of different cell types and cell-matrix interferences in specific biological and mechanical environments.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
16
|
Li J, Wiesinger A, Fokkert L, Bakker P, de Vries DK, Tijsen AJ, Pinto YM, Verkerk AO, Christoffels VM, Boink GJJ, Devalla HD. Modeling the atrioventricular conduction axis using human pluripotent stem cell-derived cardiac assembloids. Cell Stem Cell 2024:S1934-5909(24)00294-7. [PMID: 39260368 DOI: 10.1016/j.stem.2024.08.008] [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: 12/20/2023] [Revised: 06/19/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
The atrioventricular (AV) conduction axis provides electrical continuity between the atrial and ventricular chambers. The "nodal" cardiomyocytes populating this region (AV canal in the embryo, AV node from fetal stages onward) propagate impulses slowly, ensuring sequential contraction of the chambers. Dysfunction of AV nodal tissue causes severe disturbances in rhythm and contraction, and human models that capture its salient features are limited. Here, we report an approach for the reproducible generation of AV canal cardiomyocytes (AVCMs) with in vivo-like gene expression and electrophysiological profiles. We created the so-called "assembloids" composed of atrial, AVCM, and ventricular spheroids, which effectively recapitulated unidirectional conduction and the "fast-slow-fast" activation pattern typical for the vertebrate heart. We utilized these systems to reveal intracellular calcium mishandling as the basis of LMNA-associated AV conduction block. In sum, our study introduces novel cell differentiation and tissue construction strategies to facilitate the study of complex disorders affecting heart rhythm.
Collapse
Affiliation(s)
- Jiuru Li
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Alexandra Wiesinger
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Lianne Fokkert
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Priscilla Bakker
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Dylan K de Vries
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Anke J Tijsen
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands.
| |
Collapse
|
17
|
Mazzini L, De Marchi F, Buzanska L, Follenzi A, Glover JC, Gelati M, Lombardi I, Maioli M, Mesa-Herrera F, Mitrečić D, Olgasi C, Pivoriūnas A, Sanchez-Pernaute R, Sgromo C, Zychowicz M, Vescovi A, Ferrari D. Current status and new avenues of stem cell-based preclinical and therapeutic approaches in amyotrophic lateral sclerosis. Expert Opin Biol Ther 2024; 24:933-954. [PMID: 39162129 DOI: 10.1080/14712598.2024.2392307] [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: 01/24/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
INTRODUCTION Cell therapy development represents a critical challenge in amyotrophic lateral sclerosis (ALS) research. Despite more than 20 years of basic and clinical research, no definitive safety and efficacy results of cell-based therapies for ALS have been published. AREAS COVERED This review summarizes advances using stem cells (SCs) in pre-clinical studies to promote clinical translation and in clinical trials to treat ALS. New technologies have been developed and new experimental in vitro and animal models are now available to facilitate pre-clinical research in this field and to determine the most promising approaches to pursue in patients. New clinical trial designs aimed at developing personalized SC-based treatment with biological endpoints are being defined. EXPERT OPINION Knowledge of the basic biology of ALS and on the use of SCs to study and potentially treat ALS continues to grow. However, a consensus has yet to emerge on how best to translate these results into therapeutic applications. The selection and follow-up of patients should be based on clinical, biological, and molecular criteria. Planning of SC-based clinical trials should be coordinated with patient profiling genetically and molecularly to achieve personalized treatment. Much work within basic and clinical research is still needed to successfully transition SC therapy in ALS.
Collapse
Affiliation(s)
- Letizia Mazzini
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Fabiola De Marchi
- ALS Center, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Leonora Buzanska
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Antonia Follenzi
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Novara, Italy
- Dipartimento Attività Integrate Ricerca Innovazione, Azienda Ospedaliero-Universitaria SS. Antonio e Biagio e C. Arrigo, Alessandria, Italy
| | - Joel Clinton Glover
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital; Laboratory of Neural Development and Optical Recording (NDEVOR), Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Maurizio Gelati
- Unità Produttiva per Terapie Avanzate (UPTA), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ivan Lombardi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Center for Developmental Biology and Reprogramming-CEDEBIOR, University of Sassari, Sassari, Italy
| | - Fatima Mesa-Herrera
- Reprogramming and Neural Regeneration Lab, BioBizkaia Health Research Institute, Barakaldo, Spain
| | - Dinko Mitrečić
- Laboratory for Stem Cells, Croatian Institute for Brain Research and Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Cristina Olgasi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Rosario Sanchez-Pernaute
- Reprogramming and Neural Regeneration Lab, BioBizkaia Health Research Institute, Barakaldo, Spain
- Ikerbaske, Basque Foundation for Science, Bilbao, Spain
| | - Chiara Sgromo
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, Novara, Italy
| | - Marzena Zychowicz
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Angelo Vescovi
- Unità Produttiva per Terapie Avanzate (UPTA), IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Daniela Ferrari
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| |
Collapse
|
18
|
Rizzuti M, Melzi V, Brambilla L, Quetti L, Sali L, Ottoboni L, Meneri M, Ratti A, Verde F, Ticozzi N, Comi GP, Corti S, Abati E. Shaping the Neurovascular Unit Exploiting Human Brain Organoids. Mol Neurobiol 2024; 61:6642-6657. [PMID: 38334812 PMCID: PMC11338975 DOI: 10.1007/s12035-024-03998-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Brain organoids, three-dimensional cell structures derived from pluripotent stem cells, closely mimic key aspects of the human brain in vitro, providing a powerful tool for studying neurodevelopment and disease. The neuroectodermal induction protocol employed for brain organoid generation primarily gives rise to the neural cellular component but lacks the vital vascular system, which is crucial for the brain functions by regulating differentiation, migration, and circuit formation, as well as delivering oxygen and nutrients. Many neurological diseases are caused by dysfunctions of cerebral microcirculation, making vascularization of human brain organoids an important tool for pathogenetic and translational research. Experimentally, the creation of vascularized brain organoids has primarily focused on the fusion of vascular and brain organoids, on organoid transplantation in vivo, and on the use of microfluidic devices to replicate the intricate microenvironment of the human brain in vitro. This review summarizes these efforts and highlights the importance of studying the neurovascular unit in a forward-looking perspective of leveraging their use for understanding and treating neurological disorders.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Linda Ottoboni
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
- Department Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Federico Verde
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Nicola Ticozzi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
19
|
Xu X, Gao Y, Dai J, Wang Q, Wang Z, Liang W, Zhang Q, Ma W, Liu Z, Luo H, Qiao Z, Li L, Wang Z, Chen L, Zhang Y, Xiong Z. Gastric Cancer Assembloids Derived from Patient-Derived Xenografts: A Preclinical Model for Therapeutic Drug Screening. SMALL METHODS 2024; 8:e2400204. [PMID: 38948952 DOI: 10.1002/smtd.202400204] [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: 02/07/2024] [Revised: 05/18/2024] [Indexed: 07/02/2024]
Abstract
The construction of reliable preclinical models is crucial for understanding the molecular mechanisms involved in gastric cancer and for advancing precision medicine. Currently, existing in vitro tumor models often do not accurately replicate the human gastric cancer environment and are unsuitable for high-throughput therapeutic drug screening. In this study, droplet microfluidic technology is employed to create novel gastric cancer assembloids by encapsulating patient-derived xenograft gastric cancer cells and patient stromal cells in Gelatin methacryloyl (GelMA)-Gelatin-Matrigel microgels. The usage of GelMA-Gelatin-Matrigel composite hydrogel effectively alleviated cell aggregation and sedimentation during the assembly process, allowing for the handling of large volumes of cell-laden hydrogel and the uniform generation of assembloids in a high-throughput manner. Notably, the patient-derived xenograft assembloids exhibited high consistency with primary tumors at both transcriptomic and histological levels, and can be efficiently scaled up for preclinical drug screening efforts. Furthermore, the drug screening results clearly demonstrated that the in vitro assembloid model closely mirrored in vivo drug responses. Thus, these findings suggest that gastric cancer assembloids, which effectively replicate the in vivo tumor microenvironment, show promise for enabling more precise high-throughput drug screening and predicting the clinical outcomes of various drugs.
Collapse
Affiliation(s)
- Xinxin Xu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yunhe Gao
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jianli Dai
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Qianqian Wang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zixuan Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Wenquan Liang
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qing Zhang
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Wenbo Ma
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zibo Liu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Hao Luo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Zhi Qiao
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li Li
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zijian Wang
- Medical School of Chinese PLA, Beijing, 100853, China
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lin Chen
- Senior Department of General Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Institute of New Materials and Advanced Manufacturing, Beijing Academy of Science and Technology, Beijing, 100089, China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
20
|
Grass T, Dokuzluoglu Z, Buchner F, Rosignol I, Thomas J, Caldarelli A, Dalinskaya A, Becker J, Rost F, Marass M, Wirth B, Beyer M, Bonaguro L, Rodriguez-Muela N. Isogenic patient-derived organoids reveal early neurodevelopmental defects in spinal muscular atrophy initiation. Cell Rep Med 2024; 5:101659. [PMID: 39067446 PMCID: PMC11384962 DOI: 10.1016/j.xcrm.2024.101659] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024]
Abstract
Whether neurodevelopmental defects underlie postnatal neuronal death in neurodegeneration is an intriguing hypothesis only recently explored. Here, we focus on spinal muscular atrophy (SMA), a neuromuscular disorder caused by reduced survival of motor neuron (SMN) protein levels leading to spinal motor neuron (MN) loss and muscle wasting. Using the first isogenic patient-derived induced pluripotent stem cell (iPSC) model and a spinal cord organoid (SCO) system, we show that SMA SCOs exhibit abnormal morphological development, reduced expression of early neural progenitor markers, and accelerated expression of MN progenitor and MN markers. Longitudinal single-cell RNA sequencing reveals marked defects in neural stem cell specification and fewer MNs, favoring mesodermal progenitors and muscle cells, a bias also seen in early SMA mouse embryos. Surprisingly, SMN2-to-SMN1 conversion does not fully reverse these developmental abnormalities. These suggest that early neurodevelopmental defects may underlie later MN degeneration, indicating that postnatal SMN-increasing interventions might not completely amend SMA pathology in all patients.
Collapse
Affiliation(s)
- Tobias Grass
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany.
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Felix Buchner
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Ines Rosignol
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Joshua Thomas
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Antonio Caldarelli
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Anna Dalinskaya
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Jutta Becker
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering, TUD, Dresden, Germany
| | - Michele Marass
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Center for Rare Diseases, University Hospital of Cologne, Cologne, Germany
| | - Marc Beyer
- Systems Medicine, DZNE, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE & University of Bonn and West German Genome Center, Bonn, Germany; Immunogenomics & Neurodegeneration, DZNE, Bonn, Germany
| | - Lorenzo Bonaguro
- Systems Medicine, DZNE, Bonn, Germany; Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Natalia Rodriguez-Muela
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany; Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
21
|
Kosik KS. Why brain organoids are not conscious yet. PATTERNS (NEW YORK, N.Y.) 2024; 5:101011. [PMID: 39233695 PMCID: PMC11368692 DOI: 10.1016/j.patter.2024.101011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/06/2024]
Abstract
Rapid advances in human brain organoid technologies have prompted the question of their consciousness. Although brain organoids resemble many facets of the brain, their shortcomings strongly suggest that they do not fit any of the operational definitions of consciousness. As organoids gain internal processing systems through statistical learning and closed loop algorithms, interact with the external world, and become embodied through fusion with other organ systems, questions of biosynthetic consciousness will arise.
Collapse
Affiliation(s)
- Kenneth S. Kosik
- Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
22
|
Hanlon KS, Cheng M, Ferrer RM, Ryu JR, Lee B, De La Cruz D, Patel N, Espinoza P, Santoscoy MC, Gong Y, Ng C, Nguyen DM, Nammour J, Clark SW, Heine VM, Sun W, Kozarsky K, Maguire CA. In vivo selection in non-human primates identifies AAV capsids for on-target CSF delivery to spinal cord. Mol Ther 2024; 32:2584-2603. [PMID: 38845196 PMCID: PMC11405149 DOI: 10.1016/j.ymthe.2024.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/16/2024] Open
Abstract
Systemic administration of adeno-associated virus (AAV) vectors for spinal cord gene therapy has challenges including toxicity at high doses and pre-existing immunity that reduces efficacy. Intrathecal (IT) delivery of AAV vectors into cerebral spinal fluid can avoid many issues, although distribution of the vector throughout the spinal cord is limited, and vector entry to the periphery sometimes initiates hepatotoxicity. Here we performed biopanning in non-human primates (NHPs) with an IT injected AAV9 peptide display library. We identified top candidates by sequencing inserts of AAV DNA isolated from whole tissue, nuclei, or nuclei from transgene-expressing cells. These barcoded candidates were pooled with AAV9 and compared for biodistribution and transgene expression in spinal cord and liver of IT injected NHPs. Most candidates displayed increased retention in spinal cord compared with AAV9. Greater spread from the lumbar to the thoracic and cervical regions was observed for several capsids. Furthermore, several capsids displayed decreased biodistribution to the liver compared with AAV9, providing a high on-target/low off-target biodistribution. Finally, we tested top candidates in human spinal cord organoids and found them to outperform AAV9 in efficiency of transgene expression in neurons and astrocytes. These capsids have potential to serve as leading-edge delivery vehicles for spinal cord-directed gene therapies.
Collapse
Affiliation(s)
- Killian S Hanlon
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA; University College London, London, UK
| | - Ming Cheng
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Roberto Montoro Ferrer
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, University of Amsterdam, Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan, Amsterdam, the Netherlands
| | - Jae Ryun Ryu
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Boram Lee
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | - Demitri De La Cruz
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Nikita Patel
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Paula Espinoza
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Miguel C Santoscoy
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Yi Gong
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Carrie Ng
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Diane M Nguyen
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Josette Nammour
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA
| | - Sean W Clark
- SwanBio Therapeutics, Bala Cynwyd, PA 19005, USA
| | - Vivi M Heine
- Department of Child and Adolescent Psychiatry, Emma Center for Personalized Medicine, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, De Boelelaan, Amsterdam, the Netherland
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, Seoul, Republic of Korea
| | | | - Casey A Maguire
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02115, USA; Molecular Neurogenetics Unit, Massachusetts General Hospital, Charlestown, MA 02129, USA; Harvard Medical School, Boston, MA 02116, USA.
| |
Collapse
|
23
|
Nishimura K, Osaki H, Tezuka K, Nakashima D, Numata S, Masamizu Y. Recent advances and applications of human brain models. Front Neural Circuits 2024; 18:1453958. [PMID: 39161368 PMCID: PMC11330844 DOI: 10.3389/fncir.2024.1453958] [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: 06/24/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Recent advances in human pluripotent stem cell (hPSC) technologies have prompted the emergence of new research fields and applications for human neurons and brain organoids. Brain organoids have gained attention as an in vitro model system that recapitulates the higher structure, cellular diversity and function of the brain to explore brain development, disease modeling, drug screening, and regenerative medicine. This progress has been accelerated by abundant interactions of brain organoid technology with various research fields. A cross-disciplinary approach with human brain organoid technology offers a higher-ordered advance for more accurately understanding the human brain. In this review, we summarize the status of neural induction in two- and three-dimensional culture systems from hPSCs and the modeling of neurodegenerative diseases using brain organoids. We also highlight the latest bioengineered technologies for the assembly of spatially higher-ordered neural tissues and prospects of brain organoid technology toward the understanding of the potential and abilities of the human brain.
Collapse
Affiliation(s)
- Kaneyasu Nishimura
- Laboratory of Functional Brain Circuit Construction, Graduate School of Brain Science, Doshisha University, Kyotanabe, Japan
| | | | | | | | | | | |
Collapse
|
24
|
Nebol A, Gouti M. A new era in neuromuscular junction research: current advances in self-organized and assembled in vitro models. Curr Opin Genet Dev 2024; 87:102229. [PMID: 39047588 DOI: 10.1016/j.gde.2024.102229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/27/2024]
Abstract
Understanding the development and function of the human neuromuscular system is crucial for deciphering the mechanisms of neuromuscular disorders and developing effective therapies. However, limitations of animal models necessitate the development of human-specific in vitro models to study such complex diseases effectively. Here, we discuss different approaches for in vitro neuromuscular junction (NMJ) modeling: complex self-organized models that rely on the inherent abilities of cells to form NMJs based on embryonic developmental principles and assembled models that depend on integrating different cell types for controlled NMJ formation. Finally, we discuss the advantages and limitations of these models and the need for continued advancements enhanced by bioengineering approaches to deepen our understanding of human NMJ biology and pave the way for personalized medicine.
Collapse
Affiliation(s)
- Aylin Nebol
- Max Delbrück Center, Berlin 13125, Germany. https://twitter.com/@aylinnebol
| | - Mina Gouti
- Max Delbrück Center, Berlin 13125, Germany.
| |
Collapse
|
25
|
Cassel de Camps C, Rostami S, Xu V, Li C, Lépine P, Durcan TM, Moraes C. Microfabricated dynamic brain organoid cocultures to assess the effects of surface geometry on assembloid formation. Biotechnol J 2024; 19:e2400070. [PMID: 39167558 DOI: 10.1002/biot.202400070] [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: 02/01/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024]
Abstract
Organoids have emerged as valuable tools for the study of development and disease. Assembloids are formed by integrating multiple organoid types to create more complex models. However, the process by which organoids integrate to form assembloids remains unclear and may play an important role in the resulting organoid structure. Here, a microfluidic platform is developed that allows separate culture of distinct organoid types and provides the capacity to partially control the geometry of the resulting organoid surfaces. Removal of a microfabricated barrier then allows the shaped and positioned organoids to interact and form an assembloid. When midbrain and unguided brain organoids were allowed to assemble with a defined spacing between them, axonal projections from midbrain organoids and cell migration out of unguided organoids were observed and quantitatively measured as the two types of organoids fused together. Axonal projection directions were statistically biased toward other midbrain organoids, and unguided organoid surface geometry was found to affect cell invasion. This platform provides a tool to observe cellular interactions between organoid surfaces that are spaced apart in a controlled manner, and may ultimately have value in exploring neuronal migration, axon targeting, and assembloid formation mechanisms.
Collapse
Affiliation(s)
| | - Sabra Rostami
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Vanessa Xu
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Chen Li
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
| | - Paula Lépine
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada
| | - Thomas M Durcan
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute and Hospital, McGill University, Montréal, QC, Canada
| | - Christopher Moraes
- Department of Biomedical Engineering, McGill University, Montréal, QC, Canada
- Department of Chemical Engineering, McGill University, Montréal, QC, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| |
Collapse
|
26
|
Lou Y, Ma J, Hu Y, Yao X, Liu Y, Wu M, Jia G, Chen Y, Chai R, Xia M, Li W. Integration of Functional Human Auditory Neural Circuits Based on a 3D Carbon Nanotube System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309617. [PMID: 38889308 PMCID: PMC11348147 DOI: 10.1002/advs.202309617] [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: 12/09/2023] [Revised: 04/27/2024] [Indexed: 06/20/2024]
Abstract
The physiological interactions between the peripheral and central auditory systems are crucial for auditory information transmission and perception, while reliable models for auditory neural circuits are currently lacking. To address this issue, mouse and human neural pathways are generated by utilizing a carbon nanotube nanofiber system. The super-aligned pattern of the scaffold renders the axons of the bipolar and multipolar neurons extending in a parallel direction. In addition, the electrical conductivity of the scaffold maintains the electrophysiological activity of the primary mouse auditory neurons. The mouse and human primary neurons from peripheral and central auditory units in the system are then co-cultured and showed that the two kinds of neurons form synaptic connections. Moreover, neural progenitor cells of the cochlea and auditory cortex are derived from human embryos to generate region-specific organoids and these organoids are assembled in the nanofiber-combined 3D system. Using optogenetic stimulation, calcium imaging, and electrophysiological recording, it is revealed that functional synaptic connections are formed between peripheral neurons and central neurons, as evidenced by calcium spiking and postsynaptic currents. The auditory circuit model will enable the study of the auditory neural pathway and advance the search for treatment strategies for disorders of neuronal connectivity in sensorineural hearing loss.
Collapse
Affiliation(s)
- Yiyun Lou
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Jiaoyao Ma
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yangnan Hu
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Xiaoying Yao
- Obstetrics and Gynecology HospitalFudan UniversityShanghai200011China
| | - Yaoqian Liu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Mingxuan Wu
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Gaogan Jia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
| | - Yan Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| | - Renjie Chai
- State Key Laboratory of Digital Medical EngineeringDepartment of Otolaryngology Head and Neck SurgeryZhongda HospitalSchool of Life Sciences and TechnologyAdvanced Institute for Life and HealthJiangsu Province High‐Tech Key Laboratory for Bio‐Medical ResearchSoutheast UniversityNanjing210096China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
| | - Mingyu Xia
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT HospitalState Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain ScienceFudan UniversityShanghai200031China
- Institutes of Biomedical SciencesFudan UniversityShanghai200032China
- NHC Key Laboratory of Hearing MedicineFudan UniversityShanghai200031China
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain ScienceFudan UniversityShanghai200032China
| |
Collapse
|
27
|
Gribaudo S, Robert R, van Sambeek B, Mirdass C, Lyubimova A, Bouhali K, Ferent J, Morin X, van Oudenaarden A, Nedelec S. Self-organizing models of human trunk organogenesis recapitulate spinal cord and spine co-morphogenesis. Nat Biotechnol 2024; 42:1243-1253. [PMID: 37709912 DOI: 10.1038/s41587-023-01956-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/18/2023] [Indexed: 09/16/2023]
Abstract
Integrated in vitro models of human organogenesis are needed to elucidate the multi-systemic events underlying development and disease. Here we report the generation of human trunk-like structures that model the co-morphogenesis, patterning and differentiation of the human spine and spinal cord. We identified differentiation conditions for human pluripotent stem cells favoring the formation of an embryo-like extending antero-posterior (AP) axis. Single-cell and spatial transcriptomics show that somitic and spinal cord differentiation trajectories organize along this axis and can self-assemble into a neural tube surrounded by somites upon extracellular matrix addition. Morphogenesis is coupled with AP patterning mechanisms, which results, at later stages of organogenesis, in in vivo-like arrays of neural subtypes along a neural tube surrounded by spine and muscle progenitors contacted by neuronal projections. This integrated system of trunk development indicates that in vivo-like multi-tissue co-morphogenesis and topographic organization of terminal cell types can be achieved in human organoids, opening windows for the development of more complex models of organogenesis.
Collapse
Affiliation(s)
- Simona Gribaudo
- Institut du Fer à Moulin, Paris, France
- Inserm, UMR-S 1270, Paris, France
- Sorbonne Université, Science and Engineering Faculty, Paris, France
| | - Rémi Robert
- Institut du Fer à Moulin, Paris, France
- Inserm, UMR-S 1270, Paris, France
- Sorbonne Université, Science and Engineering Faculty, Paris, France
| | - Björn van Sambeek
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Camil Mirdass
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
| | - Anna Lyubimova
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kamal Bouhali
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Julien Ferent
- Institut du Fer à Moulin, Paris, France
- Inserm, UMR-S 1270, Paris, France
- Sorbonne Université, Science and Engineering Faculty, Paris, France
| | - Xavier Morin
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
| | - Alexander van Oudenaarden
- Oncode Institute, Utrecht, The Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stéphane Nedelec
- Institut du Fer à Moulin, Paris, France.
- Inserm, UMR-S 1270, Paris, France.
- Sorbonne Université, Science and Engineering Faculty, Paris, France.
| |
Collapse
|
28
|
El It F, Faivre L, Thauvin-Robinet C, Vitobello A, Duplomb L. [The contribution of cerebral organoids to the understanding and treatment of rare genetic diseases with neurodevelopmental disorders]. Med Sci (Paris) 2024; 40:643-652. [PMID: 39303116 DOI: 10.1051/medsci/2024100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Rare genetic diseases with neurodevelopmental disorders (NDDs) encompass several heterogeneous conditions (autism spectrum disorder (ASD), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), specific learning disorder (SLD), among others). Currently, few treatments are available for these patients. The difficulty in accessing human brain samples and the discrepancies between human and animal models highlight the need for new research approaches. One promising approach is the use of the cerebral organoids. These 3D, self-organized structures, generated from induced pluripotent stem cells (iPSCs), enable the reproduction of the stages of human brain development, from the proliferation of neural stem cells to their differentiation into neurons, oligodentrocytes, and astrocytes. Cerebral organoids hold great promise in understanding brain development and in the search for treatments.
Collapse
Affiliation(s)
- Fatima El It
- UMR1231 Inserm, Génétique des anomalies du développement (GAD), université de Bourgogne Franche-Comté, Dijon, France - FHU TRANSLAD, CHU Dijon, Dijon, France
| | - Laurence Faivre
- UMR1231 Inserm, Génétique des anomalies du développement (GAD), université de Bourgogne Franche-Comté, Dijon, France - FHU TRANSLAD, CHU Dijon, Dijon, France - Centre de référence des anomalies du développement et syndromes malformatifs, CHU Dijon, Dijon, France
| | - Christel Thauvin-Robinet
- UMR1231 Inserm, Génétique des anomalies du développement (GAD), université de Bourgogne Franche-Comté, Dijon, France - FHU TRANSLAD, CHU Dijon, Dijon, France - Centre de référence des anomalies du développement et syndromes malformatifs, CHU Dijon, Dijon, France
| | - Antonio Vitobello
- UMR1231 Inserm, Génétique des anomalies du développement (GAD), université de Bourgogne Franche-Comté, Dijon, France - FHU TRANSLAD, CHU Dijon, Dijon, France - Unité fonctionnelle innovation en diagnostic génomique des maladies rares, CHU Dijon, Dijon, France
| | - Laurence Duplomb
- UMR1231 Inserm, Génétique des anomalies du développement (GAD), université de Bourgogne Franche-Comté, Dijon, France - FHU TRANSLAD, CHU Dijon, Dijon, France
| |
Collapse
|
29
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2024; 58:101031. [PMID: 39146603 DOI: 10.1016/j.dmpk.2024.101031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding their neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUDs using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUDs in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, United States
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, United States
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, United States.
| |
Collapse
|
30
|
Bock M, Hong SJ, Zhang S, Yu Y, Lee S, Shin H, Choi BH, Han I. Morphogenetic Designs, and Disease Models in Central Nervous System Organoids. Int J Mol Sci 2024; 25:7750. [PMID: 39062993 PMCID: PMC11276855 DOI: 10.3390/ijms25147750] [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: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Since the emergence of the first cerebral organoid (CO) in 2013, advancements have transformed central nervous system (CNS) research. Initial efforts focused on studying the morphogenesis of COs and creating reproducible models. Numerous methodologies have been proposed, enabling the design of the brain organoid to represent specific regions and spinal cord structures. CNS organoids now facilitate the study of a wide range of CNS diseases, from infections to tumors, which were previously difficult to investigate. We summarize the major advancements in CNS organoids, concerning morphogenetic designs and disease models. We examine the development of fabrication procedures and how these advancements have enabled the generation of region-specific brain organoids and spinal cord models. We highlight the application of these organoids in studying various CNS diseases, demonstrating the versatility and potential of organoid models in advancing our understanding of complex conditions. We discuss the current challenges in the field, including issues related to reproducibility, scalability, and the accurate recapitulation of the in vivo environment. We provide an outlook on prospective studies and future directions. This review aims to provide a comprehensive overview of the state-of-the-art CNS organoid research, highlighting key developments, current challenges, and prospects in the field.
Collapse
Affiliation(s)
- Minsung Bock
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Sung Jun Hong
- Research Competency Milestones Program, School of Medicine, CHA University, Seongnam-si 13488, Republic of Korea;
- Department of Medicine, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea
| | - Songzi Zhang
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Yerin Yu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Somin Lee
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Haeeun Shin
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Byung Hyune Choi
- Department of Biomedical Science, Inha University College of Medicine, Incheon 22212, Republic of Korea;
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
- Advanced Regenerative Medicine Research Center, CHA Future Medicine Research Institute, Seongnam-si 13488, Republic of Korea
| |
Collapse
|
31
|
Soydan HE, Doğan A. Muscle Organoid and Assembloid Systems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 38980551 DOI: 10.1007/5584_2024_816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Skeletal muscle is one of the most complex and largest tissues that perform important processes in the body, including performing voluntary movements and maintaining body temperature. Disruption of muscle homeostasis results in the development of several disorders, including diabetes and sarcopenia. To study the developmental and regenerative dynamics of skeletal muscle and the mechanism behind muscle diseases, it is important to model skeletal muscle and diseases in vitro. Since skeletal muscle has a complex structure and interaction with other tissues and cells that are required to perform their function, conventional 2D cultures are not sufficient to model the skeletal muscle with their interactions. Advances in the field of organoids and assembloids will enable the establishment of more complex and realistic tissue or disease models which cannot be fully recapitulated in conventional 2D culture systems for use in several areas, including disease research, regenerative, and tissue biology. To overcome these limitations, 3D organoid systems and assembloid systems are promising because of their success in recapitulating the complex structural organization, function, and cellular interactions of skeletal muscle.
Collapse
Affiliation(s)
- Hazar Eren Soydan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
32
|
Kalpana K, Rao C, Semrau S, Zhang B, Noggle S, Fossati V. Generating Neuroimmune Assembloids Using Human Induced Pluripotent Stem Cell (iPSC)-Derived Cortical Organoids and Microglia. Methods Mol Biol 2024. [PMID: 38976205 DOI: 10.1007/7651_2024_554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
The emergence of brain organoids has revolutionized our understanding of neurodevelopment and neurological diseases by providing an in vitro model system that recapitulates key aspects of human brain development. However, conventional organoid protocols often overlook the role of microglia, the resident immune cells of the central nervous system. Microglia dysfunction is implicated in various neurological disorders, highlighting the need for their inclusion in organoid models. Here, we present a novel method for generating neuroimmune assembloids using human-induced pluripotent stem cell (iPSC)-derived cortical organoids and microglia. Building upon our previous work generating myelinating cortical organoids, we extend our methodology to include the integration of microglia, ensuring their long-term survival and maturation within the organoids. We describe two integration methods: one involving direct addition of microglia progenitors to the organoids and an alternative approach where microglia and dissociated neuronal progenitors are aggregated together in a defined ratio. To facilitate downstream analysis, we also describe a dissociation protocol for single-cell RNA sequencing (scRNA-seq) and provide guidance on fixation, cryosectioning, and immunostaining of assembloid structures. Overall, our protocol provides a comprehensive framework for generating neuroimmune assembloids, offering researchers a valuable tool for studying the interactions between neural cell types and immune cells in the context of neurological diseases.
Collapse
Affiliation(s)
- Kriti Kalpana
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Chandrika Rao
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Stefan Semrau
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Bin Zhang
- Department of Genetics & Genomic Sciences, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Mount Sinai Center for Transformative Disease Modeling, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott Noggle
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, USA.
| |
Collapse
|
33
|
Wan S, Aregueta Robles U, Poole-Warren L, Esrafilzadeh D. Advances in 3D tissue models for neural engineering: self-assembled versus engineered tissue models. Biomater Sci 2024; 12:3522-3549. [PMID: 38829222 DOI: 10.1039/d4bm00317a] [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: 06/05/2024]
Abstract
Neural tissue engineering has emerged as a promising field that aims to create functional neural tissue for therapeutic applications, drug screening, and disease modelling. It is becoming evident in the literature that this goal requires development of three-dimensional (3D) constructs that can mimic the complex microenvironment of native neural tissue, including its biochemical, mechanical, physical, and electrical properties. These 3D models can be broadly classified as self-assembled models, which include spheroids, organoids, and assembloids, and engineered models, such as those based on decellularized or polymeric scaffolds. Self-assembled models offer advantages such as the ability to recapitulate neural development and disease processes in vitro, and the capacity to study the behaviour and interactions of different cell types in a more realistic environment. However, self-assembled constructs have limitations such as lack of standardised protocols, inability to control the cellular microenvironment, difficulty in controlling structural characteristics, reproducibility, scalability, and lengthy developmental timeframes. Integrating biomimetic materials and advanced manufacturing approaches to present cells with relevant biochemical, mechanical, physical, and electrical cues in a controlled tissue architecture requires alternate engineering approaches. Engineered scaffolds, and specifically 3D hydrogel-based constructs, have desirable properties, lower cost, higher reproducibility, long-term stability, and they can be rapidly tailored to mimic the native microenvironment and structure. This review explores 3D models in neural tissue engineering, with a particular focus on analysing the benefits and limitations of self-assembled organoids compared with hydrogel-based engineered 3D models. Moreover, this paper will focus on hydrogel based engineered models and probe their biomaterial components, tuneable properties, and fabrication techniques that allow them to mimic native neural tissue structures and environment. Finally, the current challenges and future research prospects of 3D neural models for both self-assembled and engineered models in neural tissue engineering will be discussed.
Collapse
Affiliation(s)
- Shuqian Wan
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Ulises Aregueta Robles
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
- Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
34
|
Urrestizala-Arenaza N, Cerchio S, Cavaliere F, Magliaro C. Limitations of human brain organoids to study neurodegenerative diseases: a manual to survive. Front Cell Neurosci 2024; 18:1419526. [PMID: 39049825 PMCID: PMC11267621 DOI: 10.3389/fncel.2024.1419526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
In 2013, M. Lancaster described the first protocol to obtain human brain organoids. These organoids, usually generated from human-induced pluripotent stem cells, can mimic the three-dimensional structure of the human brain. While they recapitulate the salient developmental stages of the human brain, their use to investigate the onset and mechanisms of neurodegenerative diseases still faces crucial limitations. In this review, we aim to highlight these limitations, which hinder brain organoids from becoming reliable models to study neurodegenerative diseases such as Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). Specifically, we will describe structural and biological impediments, including the lack of an aging footprint, angiogenesis, myelination, and the inclusion of functional and immunocompetent microglia—all important factors in the onset of neurodegeneration in AD, PD, and ALS. Additionally, we will discuss technical limitations for monitoring the microanatomy and electrophysiology of these organoids. In parallel, we will propose solutions to overcome the current limitations, thereby making human brain organoids a more reliable tool to model neurodegeneration.
Collapse
Affiliation(s)
- Nerea Urrestizala-Arenaza
- Achucarro Basque Center for Neuroscience, The Basque Biomodels Platform for Human Research (BBioH), Leioa, Spain
| | - Sonia Cerchio
- Centro di Ricerca “E. Piaggio” – University of Pisa, Pisa, Italy
| | - Fabio Cavaliere
- Achucarro Basque Center for Neuroscience, The Basque Biomodels Platform for Human Research (BBioH), Leioa, Spain
- Fundación Biofisica Bizkaia, Leioa, Spain
| | - Chiara Magliaro
- Centro di Ricerca “E. Piaggio” – University of Pisa, Pisa, Italy
- Department of Information Engineering, University of Pisa, Pisa, Italy
| |
Collapse
|
35
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [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: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
36
|
Raman R. Biofabrication of Living Actuators. Annu Rev Biomed Eng 2024; 26:223-245. [PMID: 38959387 DOI: 10.1146/annurev-bioeng-110122-013805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The impact of tissue engineering has extended beyond a traditional focus in medicine to the rapidly growing realm of biohybrid robotics. Leveraging living actuators as functional components in machines has been a central focus of this field, generating a range of compelling demonstrations of robots capable of muscle-powered swimming, walking, pumping, gripping, and even computation. In this review, we highlight key advances in fabricating tissue-scale cardiac and skeletal muscle actuators for a range of functional applications. We discuss areas for future growth including scalable manufacturing, integrated feedback control, and predictive modeling and also propose methods for ensuring inclusive and bioethics-focused pedagogy in this emerging discipline. We hope this review motivates the next generation of biomedical engineers to advance rational design and practical use of living machines for applications ranging from telesurgery to manufacturing to on- and off-world exploration.
Collapse
Affiliation(s)
- Ritu Raman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
37
|
Onesto MM, Amin ND, Pan C, Chen X, Reis N, Valencia AM, Hudacova Z, McQueen JP, Tessier-Lavigne M, Paşca SP. Midline Assembloids Reveal Regulators of Human Axon Guidance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600229. [PMID: 38979350 PMCID: PMC11230451 DOI: 10.1101/2024.06.26.600229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Organizers are specialized cell populations that orchestrate cell patterning and axon guidance in the developing nervous system. Although non-human models have led to fundamental discoveries about the organization of the nervous system midline by the floor plate, an experimental model of human floor plate would enable broader insights into regulation of human neurodevelopment and midline connectivity. Here, we have developed stem cell-derived organoids resembling human floor plate (hFpO) and assembled them with spinal cord organoids (hSpO) to generate midline assembloids (hMA). We demonstrate that hFpO promote Sonic hedgehog-dependent ventral patterning of human spinal progenitors and Netrin-dependent guidance of human commissural axons, paralleling non-human models. To investigate evolutionary-divergent midline regulators, we profiled the hFpO secretome and identified 27 evolutionarily divergent genes between human and mouse. Utilizing the hMA platform, we targeted these candidates in an arrayed CRISPR knockout screen and reveal that GALNT2 , a gene involved in O-linked glycosylation, impairs floor plate-mediated guidance of commissural axons in humans. This novel platform extends prior axon guidance discoveries into human-specific neurobiology with implications for mechanisms of nervous system evolution and neurodevelopmental disorders.
Collapse
|
38
|
Couturier N, Hörner SJ, Nürnberg E, Joazeiro C, Hafner M, Rudolf R. Aberrant evoked calcium signaling and nAChR cluster morphology in a SOD1 D90A hiPSC-derived neuromuscular model. Front Cell Dev Biol 2024; 12:1429759. [PMID: 38966427 PMCID: PMC11222430 DOI: 10.3389/fcell.2024.1429759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024] Open
Abstract
Familial amyotrophic lateral sclerosis (ALS) is a progressive neuromuscular disorder that is due to mutations in one of several target genes, including SOD1. So far, clinical records, rodent studies, and in vitro models have yielded arguments for either a primary motor neuron disease, or a pleiotropic pathogenesis of ALS. While mouse models lack the human origin, in vitro models using human induced pluripotent stem cells (hiPSC) have been recently developed for addressing ALS pathogenesis. In spite of improvements regarding the generation of muscle cells from hiPSC, the degree of maturation of muscle cells resulting from these protocols has remained limited. To fill these shortcomings, we here present a new protocol for an enhanced myotube differentiation from hiPSC with the option of further maturation upon coculture with hiPSC-derived motor neurons. The described model is the first to yield a combination of key myogenic maturation features that are consistent sarcomeric organization in association with complex nAChR clusters in myotubes derived from control hiPSC. In this model, myotubes derived from hiPSC carrying the SOD1 D90A mutation had reduced expression of myogenic markers, lack of sarcomeres, morphologically different nAChR clusters, and an altered nAChR-dependent Ca2+ response compared to control myotubes. Notably, trophic support provided by control hiPSC-derived motor neurons reduced nAChR cluster differences between control and SOD1 D90A myotubes. In summary, a novel hiPSC-derived neuromuscular model yields evidence for both muscle-intrinsic and nerve-dependent aspects of neuromuscular dysfunction in SOD1-based ALS.
Collapse
Affiliation(s)
- Nathalie Couturier
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Sarah Janice Hörner
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Elina Nürnberg
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Claudio Joazeiro
- Center for Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, Mannheim, Germany
| | - Rüdiger Rudolf
- CeMOS, Mannheim University of Applied Sciences, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, Mannheim, Germany
| |
Collapse
|
39
|
Jiang Y, Zhou R, Wu Y, Kong G, Zeng J, Li X, Wang B, Gu C, Liao F, Qi F, Zhu Q, Gu L, Zheng C. In vitro modeling of skeletal muscle ischemia-reperfusion injury based on sphere differentiation culture from human pluripotent stem cells. Exp Cell Res 2024; 439:114111. [PMID: 38823471 DOI: 10.1016/j.yexcr.2024.114111] [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: 02/12/2024] [Revised: 04/14/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Skeletal muscle ischemia-reperfusion (IR) injury poses significant challenges due to its local and systemic complications. Traditional studies relying on two-dimensional (2D) cell culture or animal models often fall short of faithfully replicating the human in vivo environment, thereby impeding the translational process from animal research to clinical applications. Three-dimensional (3D) constructs, such as skeletal muscle spheroids with enhanced cell-cell interactions from human pluripotent stem cells (hPSCs) offer a promising alternative by partially mimicking human physiological cellular environment in vivo processes. This study aims to establish an innovative in vitro model, human skeletal muscle spheroids based on sphere differentiation from hPSCs, to investigate human skeletal muscle developmental processes and IR mechanisms within a controlled laboratory setting. By eticulously recapitulating embryonic myogenesis through paraxial mesodermal differentiation of neuro-mesodermal progenitors, we successfully established 3D skeletal muscle spheroids that mirror the dynamic colonization observed during human skeletal muscle development. Co-culturing human skeletal muscle spheroids with spinal cord spheroids facilitated the formation of neuromuscular junctions, providing functional relevance to skeletal muscle spheroids. Furthermore, through oxygen-glucose deprivation/re-oxygenation treatment, 3D skeletal muscle spheroids provide insights into the molecular events and pathogenesis of IR injury. The findings presented in this study significantly contribute to our understanding of skeletal muscle development and offer a robust platform for in vitro studies on skeletal muscle IR injury, holding potential applications in drug testing, therapeutic development, and personalized medicine within the realm of skeletal muscle-related pathologies.
Collapse
Affiliation(s)
- Yifei Jiang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Runtao Zhou
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Yixun Wu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Ganggang Kong
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China; Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingguang Zeng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Xubo Li
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Bo Wang
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Cheng Gu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China; Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fawei Liao
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Fangze Qi
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Qintang Zhu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Liqiang Gu
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Canbin Zheng
- Department of Microsurgery, Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangdong Province Engineering Laboratory for Soft Tissue Biofabrication, Guangzhou, China; Guangdong Provincial Peripheral Nerve Tissue Engineering and Technology Research Center, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China.
| |
Collapse
|
40
|
Yan Y, Cho AN. Human Brain In Vitro Model for Pathogen Infection-Related Neurodegeneration Study. Int J Mol Sci 2024; 25:6522. [PMID: 38928228 PMCID: PMC11204318 DOI: 10.3390/ijms25126522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Recent advancements in stem cell biology and tissue engineering have revolutionized the field of neurodegeneration research by enabling the development of sophisticated in vitro human brain models. These models, including 2D monolayer cultures, 3D organoids, organ-on-chips, and bioengineered 3D tissue models, aim to recapitulate the cellular diversity, structural organization, and functional properties of the native human brain. This review highlights how these in vitro brain models have been used to investigate the effects of various pathogens, including viruses, bacteria, fungi, and parasites infection, particularly in the human brain cand their subsequent impacts on neurodegenerative diseases. Traditional studies have demonstrated the susceptibility of different 2D brain cell types to infection, elucidated the mechanisms underlying pathogen-induced neuroinflammation, and identified potential therapeutic targets. Therefore, current methodological improvement brought the technology of 3D models to overcome the challenges of 2D cells, such as the limited cellular diversity, incomplete microenvironment, and lack of morphological structures by highlighting the need for further technological advancements. This review underscored the significance of in vitro human brain cell from 2D monolayer to bioengineered 3D tissue model for elucidating the intricate dynamics for pathogen infection modeling. These in vitro human brain cell enabled researchers to unravel human specific mechanisms underlying various pathogen infections such as SARS-CoV-2 to alter blood-brain-barrier function and Toxoplasma gondii impacting neural cell morphology and its function. Ultimately, these in vitro human brain models hold promise as personalized platforms for development of drug compound, gene therapy, and vaccine. Overall, we discussed the recent progress in in vitro human brain models, their applications in studying pathogen infection-related neurodegeneration, and future directions.
Collapse
Affiliation(s)
- Yuwei Yan
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
| | - Ann-Na Cho
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
41
|
Sandoval SO, Cappuccio G, Kruth K, Osenberg S, Khalil SM, Méndez-Albelo NM, Padmanabhan K, Wang D, Niciu MJ, Bhattacharyya A, Stein JL, Sousa AMM, Waxman EA, Buttermore ED, Whye D, Sirois CL, Williams A, Maletic-Savatic M, Zhao X. Rigor and reproducibility in human brain organoid research: Where we are and where we need to go. Stem Cell Reports 2024; 19:796-816. [PMID: 38759644 PMCID: PMC11297560 DOI: 10.1016/j.stemcr.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/19/2024] Open
Abstract
Human brain organoid models have emerged as a promising tool for studying human brain development and function. These models preserve human genetics and recapitulate some aspects of human brain development, while facilitating manipulation in an in vitro setting. Despite their potential to transform biology and medicine, concerns persist about their fidelity. To fully harness their potential, it is imperative to establish reliable analytic methods, ensuring rigor and reproducibility. Here, we review current analytical platforms used to characterize human forebrain cortical organoids, highlight challenges, and propose recommendations for future studies to achieve greater precision and uniformity across laboratories.
Collapse
Affiliation(s)
- Soraya O Sandoval
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Gerarda Cappuccio
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Karina Kruth
- Department of Psychiatry, University of Iowa Health Care, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Sivan Osenberg
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Saleh M Khalil
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Natasha M Méndez-Albelo
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA; Molecular Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Krishnan Padmanabhan
- Department of Neuroscience, Center for Visual Science, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester NY 14642, USA
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Departments of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mark J Niciu
- Department of Psychiatry, University of Iowa Health Care, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Health Care, Iowa City, IA 52242, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - André M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Elisa A Waxman
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Epilepsy and NeuroDevelopmental Disorders (ENDD), The Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Elizabeth D Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA; F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, MA, USA
| | - Carissa L Sirois
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Aislinn Williams
- Department of Psychiatry, University of Iowa Health Care, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Health Care, Iowa City, IA 52242, USA.
| | - Mirjana Maletic-Savatic
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA; Center for Drug Discovery, Baylor College of Medicine, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
42
|
Tamada A, Muguruma K. Recapitulation and investigation of human brain development with neural organoids. IBRO Neurosci Rep 2024; 16:106-117. [PMID: 39007085 PMCID: PMC11240300 DOI: 10.1016/j.ibneur.2023.12.006] [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] [Indexed: 07/16/2024] Open
Abstract
Organoids are 3D cultured tissues derived from stem cells that resemble the structure of living organs. Based on the accumulated knowledge of neural development, neural organoids that recapitulate neural tissue have been created by inducing self-organized neural differentiation of stem cells. Neural organoid techniques have been applied to human pluripotent stem cells to differentiate 3D human neural tissues in culture. Various methods have been developed to generate neural tissues of different regions. Currently, neural organoid technology has several significant limitations, which are being overcome in an attempt to create neural organoids that more faithfully recapitulate the living brain. The rapidly advancing neural organoid technology enables the use of living human neural tissue as research material and contributes to our understanding of the development, structure and function of the human nervous system, and is expected to be used to overcome neurological diseases and for regenerative medicine.
Collapse
Affiliation(s)
- Atsushi Tamada
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Keiko Muguruma
- Department of iPS Cell Applied Medicine, Faculty of Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
43
|
Gregor A, Zweier C. Modelling phenotypes, variants and pathomechanisms of syndromic diseases in different systems. MED GENET-BERLIN 2024; 36:121-131. [PMID: 38854643 PMCID: PMC11154186 DOI: 10.1515/medgen-2024-2020] [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: 06/11/2024]
Abstract
In this review we describe different model organisms and systems that are commonly used to study syndromic disorders. Different use cases in modeling diseases, underlying pathomechanisms and specific effects of certain variants are elucidated. We also highlight advantages and limitations of different systems. Models discussed include budding yeast, the nematode worm, the fruit fly, the frog, zebrafish, mice and human cell-based systems.
Collapse
Affiliation(s)
- Anne Gregor
- University of BernDepartment of Human GeneticsInselspital Bern3010BernSwitzerland
| | | |
Collapse
|
44
|
Suong DNA, Imamura K, Kato Y, Inoue H. Design of neural organoids engineered by mechanical forces. IBRO Neurosci Rep 2024; 16:190-195. [PMID: 38328799 PMCID: PMC10847990 DOI: 10.1016/j.ibneur.2024.01.004] [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: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
Neural organoids consist of three-dimensional tissue derived from pluripotent stem cells that could recapitulate key features of the human brain. During the past decade, organoid technology has evolved in the field of human brain science by increasing the quality and applicability of its products. Among them, a novel approach involving the design of neural organoids engineered by mechanical forces has emerged. This review describes previous approaches for the generation of neural organoids, the engineering of neural organoids by mechanical forces, and future challenges for the application of mechanical forces in the design of neural organoids.
Collapse
Affiliation(s)
- Dang Ngoc Anh Suong
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Keiko Imamura
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical‑Risk Avoidance Based On iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE MultiMix Corporation, Saitama, Japan
| | - Haruhisa Inoue
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical‑Risk Avoidance Based On iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| |
Collapse
|
45
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [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: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
46
|
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: 0] [Impact Index Per Article: 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
|
47
|
Wu S, Hong Y, Chu C, Gan Y, Li X, Tao M, Wang D, Hu H, Zheng Z, Zhu Q, Han X, Zhu W, Xu M, Dong Y, Liu Y, Guo X. Construction of human 3D striato-nigral assembloids to recapitulate medium spiny neuronal projection defects in Huntington's disease. Proc Natl Acad Sci U S A 2024; 121:e2316176121. [PMID: 38771878 PMCID: PMC11145230 DOI: 10.1073/pnas.2316176121] [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: 09/26/2023] [Accepted: 04/22/2024] [Indexed: 05/23/2024] Open
Abstract
The striato-nigral (Str-SN) circuit is composed of medium spiny neuronal projections that are mainly sent from the striatum to the midbrain substantial nigra (SN), which is essential for regulating motor behaviors. Dysfunction of the Str-SN circuitry may cause a series of motor disabilities that are associated with neurodegenerative disorders, such as Huntington's disease (HD). Although the etiology of HD is known as abnormally expanded CAG repeats of the huntingtin gene, treatment of HD remains tremendously challenging. One possible reason is the lack of effective HD model that resembles Str-SN circuitry deficits for pharmacological studies. Here, we first differentiated striatum-like organoids from human pluripotent stem cells (hPSCs), containing functional medium spiny neurons (MSNs). We then generated 3D Str-SN assembloids by assembling striatum-like organoids with midbrain SN-like organoids. With AAV-hSYN-GFP-mediated viral tracing, extensive MSN projections from the striatum to the SN are established, which formed synaptic connection with GABAergic neurons in SN organoids and showed the optically evoked inhibitory postsynaptic currents and electronic field potentials by labeling the striatum-like organoids with optogenetic virus. Furthermore, these Str-SN assembloids exhibited enhanced calcium activity compared to that of individual striatal organoids. Importantly, we further demonstrated the reciprocal projection defects in HD iPSC-derived assembloids, which could be ameliorated by treatment of brain-derived neurotrophic factor. Taken together, these findings suggest that Str-SN assembloids could be used for identifying MSN projection defects and could be applied as potential drug test platforms for HD.
Collapse
Affiliation(s)
- Shanshan Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yuan Hong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Chu Chu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yixia Gan
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, China
| | - Xinrui Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Mengdan Tao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- School of Biological Science and Medical Engineering Southeast University, Sipailou, Nanjing210096, China
| | - Da Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Hao Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Zhilong Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing211166, China
| | - Qian Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Xiao Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Wanying Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Min Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
- School of Biological Science and Medical Engineering Southeast University, Sipailou, Nanjing210096, China
| | - Xing Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing211166, China
- Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu226001, China
| |
Collapse
|
48
|
Voitiuk K, Seiler ST, Pessoa de Melo M, Geng J, Hernandez S, Schweiger HE, Sevetson JL, Parks DF, Robbins A, Torres-Montoya S, Ehrlich D, Elliott MAT, Sharf T, Haussler D, Mostajo-Radji MA, Salama SR, Teodorescu M. A feedback-driven IoT microfluidic, electrophysiology, and imaging platform for brain organoid studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585237. [PMID: 38559212 PMCID: PMC10979982 DOI: 10.1101/2024.03.15.585237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The analysis of tissue cultures, particularly brain organoids, takes a high degree of coordination, measurement, and monitoring. We have developed an automated research platform enabling independent devices to achieve collaborative objectives for feedback-driven cell culture studies. Unified by an Internet of Things (IoT) architecture, our approach enables continuous, communicative interactions among various sensing and actuation devices, achieving precisely timed control of in vitro biological experiments. The framework integrates microfluidics, electrophysiology, and imaging devices to maintain cerebral cortex organoids and monitor their neuronal activity. The organoids are cultured in custom, 3D-printed chambers attached to commercial microelectrode arrays for electrophysiology monitoring. Periodic feeding is achieved using programmable microfluidic pumps. We developed computer vision fluid volume estimations of aspirated media, achieving high accuracy, and used feedback to rectify deviations in microfluidic perfusion during media feeding/aspiration cycles. We validated the system with a 7-day study of mouse cerebral cortex organoids, comparing manual and automated protocols. The automated experimental samples maintained robust neural activity throughout the experiment, comparable with the control samples. The automated system enabled hourly electrophysiology recordings that revealed dramatic temporal changes in neuron firing rates not observed in once-a-day recordings.
Collapse
|
49
|
Wang F, Song P, Wang J, Wang S, Liu Y, Bai L, Su J. Organoid bioinks: construction and application. Biofabrication 2024; 16:032006. [PMID: 38697093 DOI: 10.1088/1758-5090/ad467c] [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: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Organoids have emerged as crucial platforms in tissue engineering and regenerative medicine but confront challenges in faithfully mimicking native tissue structures and functions. Bioprinting technologies offer a significant advancement, especially when combined with organoid bioinks-engineered formulations designed to encapsulate both the architectural and functional elements of specific tissues. This review provides a rigorous, focused examination of the evolution and impact of organoid bioprinting. It emphasizes the role of organoid bioinks that integrate key cellular components and microenvironmental cues to more accurately replicate native tissue complexity. Furthermore, this review anticipates a transformative landscape invigorated by the integration of artificial intelligence with bioprinting techniques. Such fusion promises to refine organoid bioink formulations and optimize bioprinting parameters, thus catalyzing unprecedented advancements in regenerative medicine. In summary, this review accentuates the pivotal role and transformative potential of organoid bioinks and bioprinting in advancing regenerative therapies, deepening our understanding of organ development, and clarifying disease mechanisms.
Collapse
Affiliation(s)
- Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Jian Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Sicheng Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200444, People's Republic of China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, People's Republic of China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
| |
Collapse
|
50
|
Wu Y, Cheng J, Qi J, Hang C, Dong R, Low BC, Yu H, Jiang X. Three-dimensional liquid metal-based neuro-interfaces for human hippocampal organoids. Nat Commun 2024; 15:4047. [PMID: 38744873 PMCID: PMC11094048 DOI: 10.1038/s41467-024-48452-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 05/01/2024] [Indexed: 05/16/2024] Open
Abstract
Human hippocampal organoids (hHOs) derived from human induced pluripotent stem cells (hiPSCs) have emerged as promising models for investigating neurodegenerative disorders, such as schizophrenia and Alzheimer's disease. However, obtaining the electrical information of these free-floating organoids in a noninvasive manner remains a challenge using commercial multi-electrode arrays (MEAs). The three-dimensional (3D) MEAs developed recently acquired only a few neural signals due to limited channel numbers. Here, we report a hippocampal cyborg organoid (cyb-organoid) platform coupling a liquid metal-polymer conductor (MPC)-based mesh neuro-interface with hHOs. The mesh MPC (mMPC) integrates 128-channel multielectrode arrays distributed on a small surface area (~2*2 mm). Stretchability (up to 500%) and flexibility of the mMPC enable its attachment to hHOs. Furthermore, we show that under Wnt3a and SHH activator induction, hHOs produce HOPX+ and PAX6+ progenitors and ZBTB20+PROX1+ dentate gyrus (DG) granule neurons. The transcriptomic signatures of hHOs reveal high similarity to the developing human hippocampus. We successfully detect neural activities from hHOs via the mMPC from this cyb-organoid. Compared with traditional planar devices, our non-invasive coupling offers an adaptor for recording neural signals from 3D models.
Collapse
Affiliation(s)
- Yan Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Jinhao Cheng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jie Qi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chen Hang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Ruihua Dong
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|