1
|
Ondriš J, Schwamborn JC, Olcese U. Functional recovery through the plastic adaptation of organoid grafts in cortical tissue. Cell Mol Life Sci 2025; 82:227. [PMID: 40490613 DOI: 10.1007/s00018-025-05767-w] [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: 02/02/2025] [Revised: 05/01/2025] [Accepted: 05/24/2025] [Indexed: 06/11/2025]
Abstract
The lack of effective therapeutic options for patients suffering from neurological impairments related to acquired brain damage requires novel translational strategies, among which transplantation of neural tissue is receiving strong attention. One of the most recent developments involves the implantation of brain organoid models, derived from embryonic or induced pluripotent stem cells, into damaged rodent cortices. While this approach is gaining popularity, the extent of graft integration within the host tissue remains poorly understood. This review aims to examine whether xenotransplanting organoids into cortical tissue induces functional recovery and plastic adaptation to the damaged implantation sites. Physiological indications of grafted organoid plasticity and integration into the host included viability, corticogenesis, vascularisation, growth, and the development of area-specific morphological identities. The functional integration into host neural circuitry has been probed by tracing of axonal projection growth according to implantation sites, but also through observations of spontaneous, stimulus evoked, and selectively tuned activity of grafted neurons. Finally, some studies also investigated whether the engraftment procedure facilitated behavioural recovery in tasks requiring motor, memory, or reward-seeking functions. Overall, organoid grafts show signs of progressive anatomical, functional, and behaviourally-relevant integration within the damaged host cortices. Yet, further investigation is necessary before this transplantation approach can be actually translated into a robust method to achieve functional restoration in patients suffering from brain damage.
Collapse
Affiliation(s)
- Juraj Ondriš
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, Amsterdam, 1098 XH, The Netherlands
| | | | - Umberto Olcese
- Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, Amsterdam, 1098 XH, The Netherlands.
| |
Collapse
|
2
|
Ajongbolo AO, Langhans SA. Brain Organoids and Assembloids-From Disease Modeling to Drug Discovery. Cells 2025; 14:842. [PMID: 40498018 PMCID: PMC12155111 DOI: 10.3390/cells14110842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2025] [Revised: 06/02/2025] [Accepted: 06/02/2025] [Indexed: 06/19/2025] Open
Abstract
Brain organoids are self-organized, three-dimensional (3D) aggregates derived from human embryonic stem cells, induced pluripotent stem cells, or primary organs with cell types and cellular architectures resembling those of the developing human brain. Recent studies have shown the use of region-specific brain organoids for modeling various diseases ranging from neurodevelopmental and neurodegenerative diseases to different brain cancers, which have numerous applications in fundamental research and the development of new drugs, personalized treatment, and regenerative medicine. Consequently, the use of brain organoids in drug discovery is complex and challenging and still an emerging area in this field. This review article summarizes the primary stem cells used in brain organoid generation, region-specific brain organoids, and the functional assays used in their characterization. In addition, we discuss the use of brain organoids in modeling neurodevelopmental and neurodegenerative diseases and pediatric brain cancers, as well as the application of organoids, assembloids, and tumoroids in cancer neuroscience. We further explore the recent advances in using brain organoids in high-throughput screening to improve their use for drug discovery.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology, Nemours Children’s Health, Wilmington, DE 19803, USA;
- Biological Sciences Graduate Program, University of Delaware, Newark, DE 19716, USA
| | - Sigrid A. Langhans
- Division of Neurology, Nemours Children’s Health, Wilmington, DE 19803, USA;
| |
Collapse
|
3
|
Huang R, Gao F, Yu L, Chen H, Zhu R. Generation of Neural Organoids and Their Application in Disease Modeling and Regenerative Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e01198. [PMID: 40411400 DOI: 10.1002/advs.202501198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/17/2025] [Indexed: 05/26/2025]
Abstract
The complexity and precision of the human nervous system have posed significant challenges for researchers seeking suitable models to elucidate refractory neural disorders. Traditional approaches, including monolayer cell cultures and animal models, often fail to replicate the intricacies of human neural tissue. The advent of organoid technology derived from stem cells has addressed many of these limitations, providing highly representative platforms for studying the structure and function of the human embryonic brain and spinal cord. Researchers have induced neural organoids with regional characteristics by mimicking morphogen gradients in neural development. Recent advancements have demonstrated the utility of neural organoids in disease modeling, offering insights into the pathophysiology of various neural disorders, as well as in the field of neural regeneration. Developmental defects in neural organoids due to the lack of microglia or vascular systems are addressed. In addition to induction methods, microfluidics is used to simulate the dynamic physiological environment; bio-manufacturing technologies are employed to regulate physical signaling and shape the structure of complex organs. These technologies further expand the construction strategies and application scope of neural organoids. With the emergence of new material paradigms and advances in AI, new possibilities in the realm of neural organoids are witnessed.
Collapse
Affiliation(s)
- Ruiqi Huang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200065, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200065, China
| | - Feng Gao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200065, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200065, China
| | - Liqun Yu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200065, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200065, China
| | - Haokun Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200065, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200065, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, 200065, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200065, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200065, China
| |
Collapse
|
4
|
Zhao Y, Wang T, Liu J, Wang Z, Lu Y. Emerging brain organoids: 3D models to decipher, identify and revolutionize brain. Bioact Mater 2025; 47:378-402. [PMID: 40026825 PMCID: PMC11869974 DOI: 10.1016/j.bioactmat.2025.01.025] [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: 10/23/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Brain organoids are an emerging in vitro 3D brain model that is integrated from pluripotent stem cells. This model mimics the human brain's developmental process and disease-related phenotypes to a certain extent while advancing the development of human brain-based biological intelligence. However, many limitations of brain organoid culture (e.g., lacking a functional vascular system, etc.) prevent in vitro-cultured organoids from truly replicating the human brain in terms of cell type and structure. To improve brain organoids' scalability, efficiency, and stability, this paper discusses important contributions of material biology and microprocessing technology in solving the related limitations of brain organoids and applying the latest imaging technology to make real-time imaging of brain organoids possible. In addition, the related applications of brain organoids, especially the development of organoid intelligence combined with artificial intelligence, are analyzed, which will help accelerate the rational design and guidance of brain organoids.
Collapse
Affiliation(s)
- Yuli Zhao
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Ting Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Jiajun Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze Wang
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
5
|
Tzekaki E, Bekiari C, Pantazaki A, Tsantarliotou M, Tsolaki M, Lavrentiadou SN. A new protocol for the development of organoids based on molecular mechanisms in the developing newborn rat brain: Prospective applications in the study of Alzheimer's disease. J Neurosci Methods 2025; 417:110404. [PMID: 39978482 DOI: 10.1016/j.jneumeth.2025.110404] [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: 11/16/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Brain organoids have emerged as powerful models for studying brain development and neurological disorders COMPARISON WITH EXISTING METHODS: Current models rely on stem cell isolation and differentiation using different growth factors. Thus, their composition varies according to the protocol followed. NEW METHOD We developed a simple protocol to generate organoids from newborn rat whole brain. It is a one-step procedure that yields organoids of consistent composition. The whole brains from 3-day old pups were digested enzymatically. All isolated cells were seeded in culture plates using a basement membrane extract (BME) matrix as a scaffold and cultured in the presence of the appropriate medium. RESULTS Hematoxylin-eosin staining of 28-day-old cultured domes revealed their structural integrity, while immunohistochemistry confirmed the presence of neurons, astrocytes, microglia, and progenitor stem cells in the structures. To assess whether these organoids can serve as a model to study brain physiopathology, and in particular neurodegenerative diseases such as Alzheimer's disease (AD), we determined how these organoids respond upon their exposure to lipopolysaccharides (LPS), a potent neuroinflammatory factor. LPS-induced amyloid precursor protein (APP), tau protein and glial fibrillary acidic protein (GFAP) expression. Moreover, the intracellular levels of IL-1β and the extracellular levels of amyloid-beta (Aβ) were also elevated. CONCLUSIONS Therefore, this simple protocol results in the generation of functional brain organoids with a consistent structure, that requires no use of varying factors that may affect the structure and function of the produced organoids, thus providing a valuable tool for the study of the physiopathology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Eleni Tzekaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Chryssa Bekiari
- Laboratory of Anatomy and Histology School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| | - Anastasia Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Maria Tsantarliotou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| | - Magda Tsolaki
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Sophia N Lavrentiadou
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece; Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| |
Collapse
|
6
|
Verma I, Seshagiri PB. Current Applications of Human Pluripotent Stem Cells in Neuroscience Research and Cell Transplantation Therapy for Neurological Disorders. Stem Cell Rev Rep 2025; 21:964-987. [PMID: 40186708 DOI: 10.1007/s12015-025-10851-6] [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] [Accepted: 02/05/2025] [Indexed: 04/07/2025]
Abstract
Many neurological diseases involving tissue damage cannot be treated with drug-based approaches, and the inaccessibility of human brain samples further hampers the study of these diseases. Human pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), provide an excellent model for studying neural development and function. PSCs can be differentiated into various neural cell types, providing a renewal source of functional human brain cells. Therefore, PSC-derived neural cells are increasingly used for multiple applications, including neurodevelopmental and neurotoxicological studies, neurological disease modeling, drug screening, and regenerative medicine. In addition, the neural cells generated from patient iPSCs can be used to study patient-specific disease signatures and progression. With the recent advances in genome editing technologies, it is possible to remove the disease-related mutations in the patient iPSCs to generate corrected iPSCs. The corrected iPSCs can differentiate into neural cells with normal physiological functions, which can be used for autologous transplantation. This review highlights the current progress in using PSCs to understand the fundamental principles of human neurodevelopment and dissect the molecular mechanisms of neurological diseases. This knowledge can be applied to develop better drugs and explore cell therapy options. We also discuss the basic requirements for developing cell transplantation therapies for neurological disorders and the current status of the ongoing clinical trials.
Collapse
Affiliation(s)
- Isha Verma
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India.
- Department of Neurology, University of Michigan, Ann Arbor, 48109, USA.
| | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
7
|
Sun Y, Ikeuchi Y, Guo F, Hyun I, Ming GL, Fu J. Bioengineering innovations for neural organoids with enhanced fidelity and function. Cell Stem Cell 2025; 32:689-709. [PMID: 40315834 PMCID: PMC12052258 DOI: 10.1016/j.stem.2025.03.014] [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: 08/05/2024] [Revised: 02/19/2025] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Neural organoids have been utilized to recapitulate different aspects of the developing nervous system. While hailed as promising experimental tools for studying human neural development and neuropathology, current neural organoids do not fully recapitulate the anatomy or microcircuitry-level functionality of the developing brain, spinal cord, or peripheral nervous system. In this review, we discuss emerging bioengineering approaches that control morphogen signals and biophysical microenvironments, which have improved the efficiency, fidelity, and utility of neural organoids. Furthermore, advancements in bioengineered tools have facilitated more sophisticated analyses of neural organoid functions and applications, including improved neural-bioelectronic interfaces and organoid-based information processing. Emerging bioethical issues associated with advanced neural organoids are also discussed. Future opportunities of neural organoid research lie in enhancing their fidelity, maturity, and complexity and expanding their applications in a scalable manner.
Collapse
Affiliation(s)
- Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA.
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-8654, Japan
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47408, USA
| | - Insoo Hyun
- Center for Life Sciences and Public Learning, Museum of Science, Boston, MA 02114, USA; Center for Bioethics, Harvard Medical School, Boston, MA 02115, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Hattori Y. Microglial colonization routes and their impacts on cellular diversity. Neurosci Res 2025:S0168-0102(25)00078-1. [PMID: 40288616 DOI: 10.1016/j.neures.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Microglia are the resident immune cells of the central nervous system. Unlike other glial cells-such as astrocytes and oligodendrocytes-which originate from neural stem cells alongside neurons, microglia derive from erythromyeloid progenitors that emerge in the yolk sac during early embryonic development. Once they reach the brain, microglia expand their population through proliferation during development. A growing body of research has revealed that microglia play diverse roles throughout life, both in physiological and pathological contexts. With recent advancements in single-cell transcriptomics, it has become increasingly evident that microglia exhibit substantial heterogeneity in their gene expression patterns. While various functions and subtypes of microglia are being uncovered, the mechanisms underlying their diversity remain largely unknown. Two key hypotheses may explain how microglial diversity arises. One possibility is that their diversity is influenced by the different colonization routes they take before settling in the brain. Alternatively, microglia may acquire distinct properties in response to their local environment. This review explores both possibilities, with a particular focus on the first hypothesis, drawing on recent findings that highlight the multiple routes microglia utilize to colonize the brain. It discusses how these processes contribute to the establishment of microglial diversity during brain development.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
9
|
Ene J, Liu C, Syed F, Sun L, Berry D, Durairaj P, Liu ZL, Zeng C, Jung S, Li Y. Biomanufacturing and lipidomics analysis of extracellular vesicles secreted by human blood vessel organoids in a vertical wheel bioreactor. Stem Cell Res Ther 2025; 16:207. [PMID: 40275401 PMCID: PMC12023677 DOI: 10.1186/s13287-025-04317-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from human organoids are phospholipid bilayer-bound nanoparticles that carry therapeutic cargo. However, the low yield of EVs remains a critical bottleneck for clinical translation. Vertical-Wheel bioreactors (VWBRs), with unique design features, facilitate the scalable production of EVs secreted by human blood vessel organoids (BVOs) under controlled shear stress, using aggregate- and microcarrier-based culture systems. METHODS Human induced pluripotent stem cell-derived BVOs cultured as aggregates or on Synthemax II microcarriers within VWBRs (40 and 80 rpm) were compared to static controls. The organoids were characterized by metabolite profiling, flow cytometry, and gene expression of EV biogenesis markers. EVs were characterized by nanoparticle tracking analysis, electron microscopy, and Western blotting. Lipidomics provided insights into EV lipid composition, while functional assays assessed the impact of EVs in a D-galactose-induced senescence model. RESULTS VWBR cultures showed more aerobic metabolism and higher expression of EV biogenesis genes compared to the static control. EVs from different conditions were comparable in size, but the yields were significantly higher for microcarrier and dynamic cultures than static aggregates. Lipidomic profiling revealed minimal variation (< 0.36%) in total lipid content; however, distinct differences were identified in lipid chain lengths and saturation levels, affecting key pathways such as sphingolipid and neurotrophin signaling. Human BVO EVs demonstrated the abilities of reducing oxidative stress and increasing cell proliferation in vitro. CONCLUSIONS Human BVOs differentiated in VWBRs (in particular 40 rpm) produce 2-3 fold higher yield of EVs (per mL) than static control. The bio manufactured EVs from VWBRs have exosomal characteristics and therapeutic cargo, showing functional properties in in vitro assays. This innovative approach establishes VWBRs as a scalable platform for producing functional EVs with defined lipid profiles and therapeutic potential, paving the way for future in vivo studies.
Collapse
Affiliation(s)
- Justice Ene
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
| | - Chang Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
| | - Falak Syed
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, 32306, USA
| | - Danyale Berry
- Department of Industrial and Manufacture Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
- High Performance Materials Institute, Florida State University, Tallahassee, FL, 32310, USA
| | - Pradeepraj Durairaj
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
| | - Zixiang Leonardo Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
| | - Changchun Zeng
- Department of Industrial and Manufacture Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA
- High Performance Materials Institute, Florida State University, Tallahassee, FL, 32310, USA
| | | | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, 32310, USA.
| |
Collapse
|
10
|
Li Y, Sun XY, Zeng PM, Luo ZG. Neural Responses to Hypoxic Injury in a Vascularized Cerebral Organoid Model. Neurosci Bull 2025:10.1007/s12264-025-01396-2. [PMID: 40261528 DOI: 10.1007/s12264-025-01396-2] [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: 11/26/2024] [Accepted: 01/21/2025] [Indexed: 04/24/2025] Open
Abstract
Hypoxic injury (HI) in the prenatal period often causes neonatal neurological disabilities. Due to the difficulty in obtaining clinical samples, the molecular and cellular mechanisms remain unclear. Here we use vascularized cerebral organoids to investigate the hypoxic injury phenotype and explore the intercellular interactions between vascular and neural tissues under hypoxic conditions. Our results indicate that fused vascularized cerebral organoids exhibit broader hypoxic responses and larger decreases in panels of neural development-related genes when exposed to low oxygen levels compared to single cerebral organoids. Interestingly, vessels also exhibit neural protective effects on T-box brain protein 2+ intermediate progenitors (IPs), which are markedly lost in HI cerebral organoids. Furthermore, we identify the role of bone morphogenic protein signaling in protecting IPs. Thus, this study has established an in vitro organoid system that can be used to study the contribution of vessels to brain injury under hypoxic conditions and provides a strategy for the identification of intervention targets.
Collapse
Affiliation(s)
- Yang Li
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, 201210, China
| | - Xin-Yao Sun
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, 201210, China
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Peng-Ming Zeng
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, 201210, China
| | - Zhen-Ge Luo
- School of Life Science and Technology & State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
11
|
Papetti AV, Jin M, Ma Z, Stillitano AC, Jiang P. Chimeric brain models: Unlocking insights into human neural development, aging, diseases, and cell therapies. Neuron 2025:S0896-6273(25)00256-9. [PMID: 40300597 DOI: 10.1016/j.neuron.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Human-rodent chimeric brain models serve as a unique platform for investigating the pathophysiology of human cells within a living brain environment. These models are established by transplanting human tissue- or human pluripotent stem cell (hPSC)-derived macroglial, microglial, or neuronal lineage cells, as well as cerebral organoids, into the brains of host animals. This approach has opened new avenues for exploring human brain development, disease mechanisms, and regenerative processes. Here, we highlight recent advancements in using chimeric models to study human neural development, aging, and disease. Additionally, we explore the potential applications of these models for studying human glial cell-replacement therapies, studying in vivo human glial-to-neuron reprogramming, and harnessing single-cell omics and advanced functional assays to uncover detailed insights into human neurobiology. Finally, we discuss strategies to enhance the precision and translational relevance of these models, expanding their impact in stem cell and neuroscience research.
Collapse
Affiliation(s)
- Ava V Papetti
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alessandro C Stillitano
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA.
| |
Collapse
|
12
|
Gaweda-Walerych K, Aragona V, Lodato S, Sitek EJ, Narożańska E, Buratti E. Progranulin deficiency in the brain: the interplay between neuronal and non-neuronal cells. Transl Neurodegener 2025; 14:18. [PMID: 40234992 PMCID: PMC12001433 DOI: 10.1186/s40035-025-00475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 04/17/2025] Open
Abstract
Heterozygous mutations in GRN gene lead to insufficient levels of the progranulin (PGRN) protein, resulting in frontotemporal dementia (FTD) with TAR DNA-binding protein 43 (TDP-43) inclusions, classified pathologically as frontotemporal lobar degeneration (FTLD-TDP). Homozygous GRN mutations are exceedingly rare and cause neuronal ceroid lipofuscinosis 11, a lysosomal storage disease with onset in young adulthood, or an FTD syndrome with late-onset manifestations. In this review, we highlight the broad spectrum of clinical phenotypes associated with PGRN deficiency, including primary progressive aphasia and behavioral variant of frontotemporal dementia. We explore these phenotypes alongside relevant rodent and in vitro human models, ranging from the induced pluripotent stem cell-derived neural progenitors, neurons, microglia, and astrocytes to genetically engineered heterotypic organoids containing both neurons and astrocytes. We summarize advantages and limitations of these models in recapitulating the main FTLD-GRN hallmarks, highlighting the role of non-cell-autonomous mechanisms in the formation of TDP-43 pathology, neuroinflammation, and neurodegeneration. Data obtained from patients' brain tissues and biofluids, in parallel with single-cell transcriptomics, demonstrate the complexity of interactions among the highly heterogeneous cellular clusters present in the brain, including neurons, astrocytes, microglia, oligodendroglia, endothelial cells, and pericytes. Emerging evidence has revealed that PGRN deficiency is associated with cell cluster-specific, often conserved, genetic and molecular phenotypes in the central nervous system. In this review, we focus on how these distinct cellular populations and their dysfunctional crosstalk contribute to neurodegeneration and neuroinflammation in FTD-GRN. Specifically, we characterize the phenotypes of lipid droplet-accumulating microglia and alterations of myelin lipid content resulting from lysosomal dysfunction caused by PGRN deficiency. Additionally, we consider how the deregulation of glia-neuron communication affects the exchange of organelles such as mitochondria, and the removal of excess toxic products such as protein aggregates, in PGRN-related neurodegeneration.
Collapse
Affiliation(s)
- Katarzyna Gaweda-Walerych
- Department of Neurogenetics and Functional Genomics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106, Warsaw, Poland.
| | - Vanessa Aragona
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Levi Montalicini 4, Pieve Emanuele, 20072, Milan, Italy
- Neurodevelopment Biology Lab, IRCCS Humanitas Research Hospital, via Manzoni, 56, Rozzano, 20089, Milan, Italy
| | - Emilia J Sitek
- Division of Neurological and Psychiatric Nursing, Laboratory of Clinical Neuropsychology, Neurolinguistics, and Neuropsychotherapy, Faculty of Health Sciences, Medical University of Gdansk, 80-210, Gdansk, Poland.
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland.
| | - Ewa Narożańska
- Neurology Department, St. Adalbert Hospital, Copernicus PL, 80-462, Gdansk, Poland
| | - Emanuele Buratti
- Molecular Pathology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), AREA Science Park, 34149, Trieste, Italy
| |
Collapse
|
13
|
Zhou L, Huang J, Li C, Gu Q, Li G, Li ZA, Xu J, Zhou J, Tuan RS. Organoids and organs-on-chips: Recent advances, applications in drug development, and regulatory challenges. MED 2025; 6:100667. [PMID: 40220744 DOI: 10.1016/j.medj.2025.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/30/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025]
Abstract
Organoids and organs-on-chips (OoCs) are rapidly evolving technologies for creating miniature human tissue models. They can mimic complex physiological functions and pathological conditions, offering more realistic platforms for disease modeling, drug screening, precision medicine, and regenerative therapies. The passing of the FDA Modernization Act 2.0 has reduced animal testing requirements for drug trials, marking a significant milestone in using advanced in vitro models such as organoids and OoCs for therapeutic discovery. Apart from technical and ethical challenges, regulatory issues persist in ensuring the reliability, scientificity, and applicability of these models in drug development. This perspective explores the concept, advancements, pros and cons, and applications of organoids and OoCs, particularly in drug research and development. It also examines global regulatory agencies' policies and actions on using these models in drug evaluation, aiming to guide industry standard setting and advance regulatory science.
Collapse
Affiliation(s)
- Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingjing Huang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Cun Li
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qi Gu
- Key Laboratory of Organ Regeneration and Reconstruction, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gang Li
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital of the Southern Medical University, Guangzhou, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jie Zhou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Rocky S Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
14
|
Sullivan R, Ahrens Q, Mills-Huffnagle SL, Elcheva IA, Hicks SD. A human iPSC-derived midbrain neural stem cell model of prenatal opioid exposure and withdrawal: A proof of concept study. PLoS One 2025; 20:e0319418. [PMID: 40168407 PMCID: PMC11960892 DOI: 10.1371/journal.pone.0319418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/30/2025] [Indexed: 04/03/2025] Open
Abstract
A growing body of clinical literature has described neurodevelopmental delays in infants with chronic prenatal opioid exposure and withdrawal. Despite this, the mechanism of how opioids impact the developing brain remains unknown. Here, we developed an in vitro model of prenatal morphine exposure and withdrawal using healthy human induced pluripotent stem cell (iPSC)-derived midbrain neural progenitors in monolayer. To optimize our model, we identified that a longer neural induction and regional patterning period increases expression of canonical opioid receptors mu and kappa in midbrain neural progenitors compared to a shorter protocol (OPRM1, two-tailed t-test, p = 0.004; OPRK1, p = 0.0003). Next, we showed that the midbrain neural progenitors derived from a longer iPSC neural induction also have scant toll-like receptor 4 (TLR4) expression, a key player in neonatal opioid withdrawal syndrome pathophysiology. During morphine withdrawal, differentiating neural progenitors experience cyclic adenosine monophosphate overshoot compared to cell exposed to vehicle (p = 0.0496) and morphine exposure conditions (p, = 0.0136, 1-way ANOVA). Finally, we showed that morphine exposure and withdrawal alters proportions of differentiated progenitor cell fates (2-way ANOVA, F = 16.05, p < 0.0001). Chronic morphine exposure increased proportions of nestin positive progenitors (p = 0.0094), and decreased proportions of neuronal nuclear antigen positive neurons (NEUN) (p = 0.0047) compared to those exposed to vehicle. Morphine withdrawal decreased proportions of glial fibrillary acidic protein positive cells of astrocytic lineage (p = 0.044), and increased proportions of NEUN-positive neurons (p < 0.0001) compared to those exposed to morphine only. Applications of this paradigm include mechanistic studies underscoring neural progenitor cell fate commitments in early neurodevelopment during morphine exposure and withdrawal.
Collapse
MESH Headings
- Humans
- Neural Stem Cells/metabolism
- Neural Stem Cells/drug effects
- Neural Stem Cells/cytology
- Mesencephalon/cytology
- Mesencephalon/drug effects
- Mesencephalon/metabolism
- Mesencephalon/pathology
- Induced Pluripotent Stem Cells/cytology
- Induced Pluripotent Stem Cells/metabolism
- Induced Pluripotent Stem Cells/drug effects
- Morphine/adverse effects
- Morphine/pharmacology
- Female
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/pharmacology
- Cell Differentiation/drug effects
- Pregnancy
- Substance Withdrawal Syndrome/pathology
- Substance Withdrawal Syndrome/metabolism
- Prenatal Exposure Delayed Effects
- Toll-Like Receptor 4/metabolism
- Proof of Concept Study
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/genetics
Collapse
Affiliation(s)
- Rhea Sullivan
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Quinn Ahrens
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Sara L. Mills-Huffnagle
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Irina A. Elcheva
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Steven D. Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
15
|
Jeong Y, Son S, Park J, Kim CY, Kim J. Antidepressant aripiprazole induces adverse effects on neural development during cortex organoid generation. Reprod Toxicol 2025; 133:108862. [PMID: 39971107 DOI: 10.1016/j.reprotox.2025.108862] [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: 11/03/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
A significant number of women experience anxiety and depressive symptoms during pregnancy, leading to the prescription of antidepressants, including aripiprazole. However, although a few animal studies have reported its developmental toxicity, there is a lack of research on the potential risks aripiprazole may pose to the fetus, particularly regarding neural development, as well as an absence of appropriate models to verify these effects. Therefore, this study investigates the impact of aripiprazole on neural development using cortex organoids, which can effectively model human brain development and function while overcoming interspecies differences. Cortex organoids were generated and exposed to aripiprazole at concentrations of 0.3-9 µM over 4 weeks. We assessed morphological changes, cell viability, gene expression, immunofluorescence staining, and electrophysiological function. The results revealed that aripiprazole led to significant reductions in organoid size and increased cell death, particularly at higher concentrations. Immunofluorescence analysis showed abnormalities in the expression patterns of neural stem cells and neuronal markers. Additionally, real-time PCR demonstrated decreased expression of genes related to neural stem cells, neural differentiation and migration, maturation, synaptogenesis, and gliogenesis, along with increased apoptosis-related gene expression. Electrophysiological recordings indicated impaired neural activity, evidenced by reduced mean firing rates. Our study is the first to demonstrate that aripiprazole induces adverse effects on neural development across functional, molecular, and morphological aspects. The findings will aid in a better understanding of the risks associated with antidepressant use during pregnancy in terms of neural development and suggest that cortex organoids are a valuable model for evaluating potential neurodevelopmental toxicants.
Collapse
Affiliation(s)
- Youngin Jeong
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Suil Son
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiyun Park
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Jin Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea.
| |
Collapse
|
16
|
Huang Z, He C, Wang G, Zhu M, Tong X, Feng Y, Zhang C, Dong S, Harim Y, Liu HK, Zhou W, Lan F, Feng W. Mutation of CHD7 impairs the output of neuroepithelium transition that is reversed by the inhibition of EZH2. Mol Psychiatry 2025:10.1038/s41380-025-02990-6. [PMID: 40164694 DOI: 10.1038/s41380-025-02990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 03/07/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Haploinsufficiency of CHD7 (Chromo-Helicase-DNA binding protein 7) causes a severe congenital disease CHARGE syndrome. Brain anomaly such as microcephaly and olfactory bulb agenesis seen in CHARGE patients have not been mimicked in previous animal models. Here, we uncover an indispensable function of CHD7 in the neuroepithelium (NE) but not in the neural stem cells (NSCs) after NE transition. Loss of Chd7 in mouse NE resulted in CHARGE-like brain anomalies due to reduced proliferation and differentiation of neural stem and progenitor cells, which were recapitulated in CHD7 KO human forebrain organoids. Mechanistically, we find that CHD7 activates neural transcription factors by removing the repressive histone mark H3K27me3 and promoting chromatin accessibility. Importantly, neurodevelopmental defects caused by CHD7 loss in human brain organoids and mice were ameliorated by the inhibition of H3K27me3 methyltransferase EZH2. Altogether, by implementing appropriate experimental models, we uncover the pathogenesis of CHD7-associated neurodevelopmental diseases, and identify a potential therapeutic opportunity for CHARGE syndrome.
Collapse
Affiliation(s)
- Zhuxi Huang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenxi He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Guangfu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ming Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiaoyu Tong
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Yi Feng
- State Key Laboratory of Medical Neurobiology, Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Chenyang Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shuhua Dong
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yassin Harim
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Hai-Kun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, 69120, Germany
| | - Wenhao Zhou
- Division of Neonatology and Center for Newborn Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fei Lan
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Weijun Feng
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Kim S, Woo Y, Um D, Chun I, Noh SJ, Ji HA, Jung N, Goo BS, Yoo JY, Mun DJ, Nghi TD, Nhung TTM, Han SH, Lee SB, Lee W, Yun J, So KH, Kim DK, Jang H, Suh Y, Rah JC, Baek ST, Yoon KJ, Kim MS, Kim TK, Park SK. Perturbed cell fate decision by schizophrenia-associated AS3MT d2d3 isoform during corticogenesis. SCIENCE ADVANCES 2025; 11:eadp8271. [PMID: 40153497 PMCID: PMC11952104 DOI: 10.1126/sciadv.adp8271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 02/24/2025] [Indexed: 03/30/2025]
Abstract
The neurodevelopmental theory of schizophrenia emphasizes early brain development in its etiology. Genome-wide association studies have linked schizophrenia to genetic variations of AS3MT (arsenite methyltransferase) gene, particularly the increased expression of AS3MTd2d3 isoform. To investigate the biological basis of this association with schizophrenia pathophysiology, we established a transgenic mouse model (AS3MTd2d3-Tg) ectopically expressing AS3MTd2d3 at the cortical neural stem cells. AS3MTd2d3-Tg mice exhibited enlarged ventricles and deficits in sensorimotor gating and sociability. Single-cell and single-nucleus RNA sequencing analyses of AS3MTd2d3-Tg brains revealed cell fate imbalances and altered excitatory neuron composition. AS3MTd2d3 localized to centrosome, disrupting mitotic spindle orientation and differentiation in developing neocortex and organoids, in part through NPM1 (Nucleophosmin 1). The structural analysis identified that hydrophobic residues exposed in AS3MTd2d3 are critical for its pathogenic function. Therefore, our findings may help to explain the early pathological features of schizophrenia.
Collapse
Affiliation(s)
- Seunghyun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Youngsik Woo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dahun Um
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Inseop Chun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Su-Jin Noh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Hyeon Ah Ji
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Namyoung Jung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Bon Seong Goo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jin Yeong Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dong Jin Mun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Tran Diem Nghi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Truong Thi My Nhung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Seung Hyeon Han
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Su Been Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Wonhyeok Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jonghyeok Yun
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Ki Hurn So
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Dae-Kyum Kim
- Division of Thoracic and Upper Gastrointestinal Surgery, Department of Surgery, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3G 1A4, Canada
- Cancer Research Program, Research Institute of McGill University Health Centre, Montreal, Quebec H4A 3J1, Canada
| | - Hyunsoo Jang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejon 34141, Republic of Korea
| | - Yeongjun Suh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jong-Cheol Rah
- Korea Brain Research Institute, Daegu 41062, Republic of Korea
| | - Seung Tae Baek
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejon 34141, Republic of Korea
| | - Min-Sung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03772, Republic of Korea
| |
Collapse
|
18
|
Lin Z, Wang W, Liu R, Li Q, Lee J, Hirschler C, Liu J. Cyborg organoids integrated with stretchable nanoelectronics can be functionally mapped during development. Nat Protoc 2025:10.1038/s41596-025-01147-7. [PMID: 40140634 DOI: 10.1038/s41596-025-01147-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/31/2024] [Indexed: 03/28/2025]
Abstract
Organoids are in vitro miniaturized cellular models of organs that offer opportunities for studying organ development, disease mechanisms and drug screening. Understanding the complex processes governing organoid development and function requires methods suitable for the continuous, long-term monitoring of cell activities (for example, electrophysiological and mechanical activity) at single-cell resolution throughout the entire three-dimensional (3D) structure. Cyborg organoid technology addresses this need by seamlessly integrating stretchable mesh nanoelectronics with tissue-like properties, such as tissue-level flexibility, subcellular feature size and mesh-like networks, into 3D organoids through a 2D-to-3D tissue reconfiguration process during organogenesis. This approach enables longitudinal, tissue-wide, single-cell functional mapping, thereby overcoming the limitations of existing techniques including recording duration, spatial coverage, and the ability to maintain stable contact with the tissue during organoid development. This protocol describes the fabrication and characterization of stretchable mesh nanoelectronics, their electrical performance, their integration with organoids and the acquisition of long-term functional organoid activity requiring multimodal data analysis techniques. Cyborg organoid technology represents a transformative tool for investigating organoid development and function, with potential for improving in vitro disease models, drug screening and personalized medicine. The procedure is suitable for users within a multidisciplinary team with expertise in stem cell biology, tissue engineering, nanoelectronics fabrication, electrophysiology and data science.
Collapse
Affiliation(s)
- Zuwan Lin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wenbo Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Qiang Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jaeyong Lee
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Charles Hirschler
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA.
| |
Collapse
|
19
|
Ahmed AA, Alegret N, Almeida B, Alvarez-Puebla R, Andrews AM, Ballerini L, Barrios-Capuchino JJ, Becker C, Blick RH, Bonakdar S, Chakraborty I, Chen X, Cheon J, Chilla G, Coelho Conceicao AL, Delehanty J, Dulle M, Efros AL, Epple M, Fedyk M, Feliu N, Feng M, Fernández-Chacón R, Fernandez-Cuesta I, Fertig N, Förster S, Garrido JA, George M, Guse AH, Hampp N, Harberts J, Han J, Heekeren HR, Hofmann UG, Holzapfel M, Hosseinkazemi H, Huang Y, Huber P, Hyeon T, Ingebrandt S, Ienca M, Iske A, Kang Y, Kasieczka G, Kim DH, Kostarelos K, Lee JH, Lin KW, Liu S, Liu X, Liu Y, Lohr C, Mailänder V, Maffongelli L, Megahed S, Mews A, Mutas M, Nack L, Nakatsuka N, Oertner TG, Offenhäusser A, Oheim M, Otange B, Otto F, Patrono E, Peng B, Picchiotti A, Pierini F, Pötter-Nerger M, Pozzi M, Pralle A, Prato M, Qi B, Ramos-Cabrer P, Genger UR, Ritter N, Rittner M, Roy S, Santoro F, Schuck NW, Schulz F, Şeker E, Skiba M, Sosniok M, Stephan H, Wang R, Wang T, Wegner KD, Weiss PS, Xu M, Yang C, Zargarian SS, Zeng Y, Zhou Y, Zhu D, Zierold R, Parak WJ. Interfacing with the Brain: How Nanotechnology Can Contribute. ACS NANO 2025; 19:10630-10717. [PMID: 40063703 PMCID: PMC11948619 DOI: 10.1021/acsnano.4c10525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 03/26/2025]
Abstract
Interfacing artificial devices with the human brain is the central goal of neurotechnology. Yet, our imaginations are often limited by currently available paradigms and technologies. Suggestions for brain-machine interfaces have changed over time, along with the available technology. Mechanical levers and cable winches were used to move parts of the brain during the mechanical age. Sophisticated electronic wiring and remote control have arisen during the electronic age, ultimately leading to plug-and-play computer interfaces. Nonetheless, our brains are so complex that these visions, until recently, largely remained unreachable dreams. The general problem, thus far, is that most of our technology is mechanically and/or electrically engineered, whereas the brain is a living, dynamic entity. As a result, these worlds are difficult to interface with one another. Nanotechnology, which encompasses engineered solid-state objects and integrated circuits, excels at small length scales of single to a few hundred nanometers and, thus, matches the sizes of biomolecules, biomolecular assemblies, and parts of cells. Consequently, we envision nanomaterials and nanotools as opportunities to interface with the brain in alternative ways. Here, we review the existing literature on the use of nanotechnology in brain-machine interfaces and look forward in discussing perspectives and limitations based on the authors' expertise across a range of complementary disciplines─from neuroscience, engineering, physics, and chemistry to biology and medicine, computer science and mathematics, and social science and jurisprudence. We focus on nanotechnology but also include information from related fields when useful and complementary.
Collapse
Affiliation(s)
- Abdullah
A. A. Ahmed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Department
of Physics, Faculty of Applied Science, Thamar University, Dhamar 87246, Yemen
| | - Nuria Alegret
- Biogipuzkoa
HRI, Paseo Dr. Begiristain
s/n, 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bethany Almeida
- Department
of Chemical and Biomolecular Engineering, Clarkson University, Potsdam, New York 13699, United States
| | - Ramón Alvarez-Puebla
- Universitat
Rovira i Virgili, 43007 Tarragona, Spain
- ICREA, 08010 Barcelona, Spain
| | - Anne M. Andrews
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- Neuroscience
Interdepartmental Program, University of
California, Los Angeles, Los Angeles, California 90095, United States
- Department
of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience
& Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Laura Ballerini
- Neuroscience
Area, International School for Advanced
Studies (SISSA/ISAS), Trieste 34136, Italy
| | | | - Charline Becker
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Robert H. Blick
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Shahin Bonakdar
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- National
Cell Bank Department, Pasteur Institute
of Iran, P.O. Box 1316943551, Tehran, Iran
| | - Indranath Chakraborty
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Nano Science and Technology, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Xiaodong Chen
- Innovative
Center for Flexible Devices (iFLEX), Max Planck − NTU Joint
Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jinwoo Cheon
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
- Department
of Chemistry, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Gerwin Chilla
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - James Delehanty
- U.S. Naval
Research Laboratory, Washington, D.C. 20375, United States
| | - Martin Dulle
- JCNS-1, Forschungszentrum
Jülich, 52428 Jülich, Germany
| | | | - Matthias Epple
- Inorganic
Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, 45117 Essen, Germany
| | - Mark Fedyk
- Center
for Neuroengineering and Medicine, UC Davis, Sacramento, California 95817, United States
| | - Neus Feliu
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Miao Feng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Rafael Fernández-Chacón
- Instituto
de Biomedicina de Sevilla (IBiS), Hospital
Universitario Virgen del Rocío/Consejo Superior de Investigaciones
Científicas/Universidad de Sevilla, 41013 Seville, Spain
- Departamento
de Fisiología Médica y Biofísica, Facultad de
Medicina, Universidad de Sevilla, CIBERNED,
ISCIII, 41013 Seville, Spain
| | | | - Niels Fertig
- Nanion
Technologies GmbH, 80339 München, Germany
| | | | - Jose A. Garrido
- ICREA, 08010 Barcelona, Spain
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
| | | | - Andreas H. Guse
- The Calcium
Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Norbert Hampp
- Fachbereich
Chemie, Universität Marburg, 35032 Marburg, Germany
| | - Jann Harberts
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Drug Delivery,
Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne
Centre for Nanofabrication, Victorian Node
of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
| | - Jili Han
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Hauke R. Heekeren
- Executive
University Board, Universität Hamburg, 20148 Hamburg Germany
| | - Ulrich G. Hofmann
- Section
for Neuroelectronic Systems, Department for Neurosurgery, University Medical Center Freiburg, 79108 Freiburg, Germany
- Faculty
of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Malte Holzapfel
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | | | - Yalan Huang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Patrick Huber
- Institute
for Materials and X-ray Physics, Hamburg
University of Technology, 21073 Hamburg, Germany
- Center
for X-ray and Nano Science CXNS, Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Taeghwan Hyeon
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Sven Ingebrandt
- Institute
of Materials in Electrical Engineering 1, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcello Ienca
- Institute
for Ethics and History of Medicine, School of Medicine and Health, Technische Universität München (TUM), 81675 München, Germany
| | - Armin Iske
- Fachbereich
Mathematik, Universität Hamburg, 20146 Hamburg, Germany
| | - Yanan Kang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Dae-Hyeong Kim
- Center
for Nanoparticle Research, Institute for Basic Science (IBS), Seoul 08826, Republic of Korea
- School
of Chemical and Biological Engineering, and Institute of Chemical
Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kostas Kostarelos
- Catalan
Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, 08193 Bellaterra, Spain
- Centre
for Nanotechnology in Medicine, Faculty of Biology, Medicine &
Health and The National Graphene Institute, University of Manchester, Manchester M13 9PL, United
Kingdom
| | - Jae-Hyun Lee
- Institute
for Basic Science Center for Nanomedicine, Seodaemun-gu, Seoul 03722, Korea
- Advanced
Science Institute, Yonsei University, Seodaemun-gu, Seoul 03722, Korea
| | - Kai-Wei Lin
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sijin Liu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Xin Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yang Liu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Christian Lohr
- Fachbereich
Biologie, Universität Hamburg, 20146 Hamburg, Germany
| | - Volker Mailänder
- Department
of Dermatology, Center for Translational Nanomedicine, Universitätsmedizin der Johannes-Gutenberg,
Universität Mainz, 55131 Mainz, Germany
- Max Planck
Institute for Polymer Research, Ackermannweg 10, 55129 Mainz, Germany
| | - Laura Maffongelli
- Institute
of Medical Psychology, University of Lübeck, 23562 Lübeck, Germany
| | - Saad Megahed
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Physics
Department, Faculty of Science, Al-Azhar
University, 4434104 Cairo, Egypt
| | - Alf Mews
- Fachbereich
Chemie, Universität Hamburg, 20146 Hamburg, Germany
| | - Marina Mutas
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Leroy Nack
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Nako Nakatsuka
- Laboratory
of Chemical Nanotechnology (CHEMINA), Neuro-X
Institute, École Polytechnique Fédérale de Lausanne
(EPFL), Geneva CH-1202, Switzerland
| | - Thomas G. Oertner
- Institute
for Synaptic Neuroscience, University Medical
Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Andreas Offenhäusser
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Martin Oheim
- Université
Paris Cité, CNRS, Saints Pères
Paris Institute for the Neurosciences, 75006 Paris, France
| | - Ben Otange
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Ferdinand Otto
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Enrico Patrono
- Institute
of Physiology, Czech Academy of Sciences, Prague 12000, Czech Republic
| | - Bo Peng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | | - Filippo Pierini
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Pötter-Nerger
- Head and
Neurocenter, Department of Neurology, University
Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maria Pozzi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Arnd Pralle
- University
at Buffalo, Department of Physics, Buffalo, New York 14260, United States
| | - Maurizio Prato
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Bing Qi
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- School
of Life Sciences, Southern University of
Science and Technology, Shenzhen, 518055, China
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque Research and Technology
Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Basque
Foundation for Science, Ikerbasque, 48013 Bilbao, Spain
| | - Ute Resch Genger
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Norbert Ritter
- Executive
Faculty Board, Faculty for Mathematics, Informatics and Natural Sciences, Universität Hamburg, 20345 Hamburg, Germany
| | - Marten Rittner
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Sathi Roy
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
- Department
of Mechanical Engineering, Indian Institute
of Technology Kharagpur, Kharagpur 721302, India
| | - Francesca Santoro
- Institute
of Biological Information Processing - Bioelectronics, Forschungszentrum Jülich, 52425 Jülich, Germany
- Faculty
of Electrical Engineering and Information Technology, RWTH Aachen, 52074 Aachen, Germany
| | - Nicolas W. Schuck
- Institute
of Psychology, Universität Hamburg, 20146 Hamburg, Germany
- Max Planck
Research Group NeuroCode, Max Planck Institute
for Human Development, 14195 Berlin, Germany
- Max Planck
UCL Centre for Computational Psychiatry and Ageing Research, 14195 Berlin, Germany
| | - Florian Schulz
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Erkin Şeker
- University
of California, Davis, Davis, California 95616, United States
| | - Marvin Skiba
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Martin Sosniok
- Zentrum
für Angewandte Nanotechnologie CAN, Fraunhofer-Institut für Angewandte Polymerforschung IAP, 20146 Hamburg, Germany
| | - Holger Stephan
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical
Cancer Research, 01328 Dresden, Germany
| | - Ruixia Wang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- Deutsches
Elektronen-Synchrotron DESY, 22607 Hamburg, Germany
| | - Ting Wang
- State Key
Laboratory of Organic Electronics and Information Displays & Jiangsu
Key Laboratory for Biosensors, Institute of Advanced Materials (IAM),
Jiangsu National Synergetic Innovation Center for Advanced Materials
(SICAM), Nanjing University of Posts and
Telecommunications, Nanjing 210023, China
| | - K. David Wegner
- Division
Biophotonics, Federal Institute for Materials Research and Testing
(BAM), 12489 Berlin, Germany
| | - Paul S. Weiss
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, Los
Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Bioengineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Department
of Materials Science and Engineering, University
of California, Los Angeles, Los
Angeles, California 90095, United States
| | - Ming Xu
- State Key
Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- University
of the Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxi Yang
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Seyed Shahrooz Zargarian
- Department
of Biosystems and Soft Matter, Institute
of Fundamental Technological Research, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Yuan Zeng
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Yaofeng Zhou
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | - Dingcheng Zhu
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
- College
of Material, Chemistry and Chemical Engineering, Key Laboratory of
Organosilicon Chemistry and Material Technology, Ministry of Education,
Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou 311121, China
| | - Robert Zierold
- Fachbereich
Physik, Universität Hamburg, 22761 Hamburg, Germany
| | | |
Collapse
|
20
|
Zheng H, Feng Y, Tang J, Yu F, Wang Z, Xu J, Hai C, Jiang M, Cheng Y, Shao Z, Ma N, Lobie PE, Ma S. Astrocyte-secreted cues promote neural maturation and augment activity in human forebrain organoids. Nat Commun 2025; 16:2845. [PMID: 40122897 PMCID: PMC11930946 DOI: 10.1038/s41467-025-58295-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Brain organoids have been proposed as suitable human brain model candidates for a variety of applications. However, the lack of appropriate maturation limits the transferability of such functional tools. Here, we present a method to facilitate neuronal maturation by integrating astrocyte-secreted factors into hPSC-derived 2D and 3D neural culture systems. We demonstrate that protein- and nutrient-enriched astrocyte-conditioned medium (ACM) accelerates neuronal differentiation with enlarged neuronal layer and the overproduction of deep-layer cortical neurons. We captured the elevated changes in the functional activity of neuronal networks within ACM-treated organoids using comprehensive electrophysiological recordings. Furthermore, astrocyte-secreted cues can induce lipid droplet accumulation in neural cultures, offering protective effects in neural differentiation to withstand cellular stress. Together, these data indicate the potential of astrocyte secretions to promote neural maturation.
Collapse
Affiliation(s)
- Honghui Zheng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Yilin Feng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Jiyuan Tang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Feifei Yu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China
| | - Jiani Xu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Cheng Hai
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Mingyue Jiang
- Guangzhou National Laboratory, Guangzhou, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Yifan Cheng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
| | - Zhicheng Shao
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institute of Pediatrics, National Children's Medical Center, Children's Hospital, Fudan University, Shanghai, China
| | - Ning Ma
- School of Basic Medical Sciences, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Peter E Lobie
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen, China.
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China.
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education Institutes, Tsinghua Shenzhen International Graduate School, Shenzhen, China.
| |
Collapse
|
21
|
Jin Y, Guo Y, Li Q, Wu L, Ge Y, Zhao J. Non-Invasive and Long-Term Electrophysiological Monitoring Sensors for Cerebral Organoids Differentiation. BIOSENSORS 2025; 15:173. [PMID: 40136970 PMCID: PMC11940203 DOI: 10.3390/bios15030173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
Cerebral organoids derived from human induced pluripotent stem cells (iPSCs) have emerged as powerful in vitro models for studying human brain development and neurological disorders. Understanding the electrophysiological properties of these organoids is crucial for evaluating their functional maturity and potential applications. However, the differentiation and maturation of stem cells into cerebral organoids is a long, slow, and error-prone process. Hence, it is vitally crucial to establish a non-invasive method of monitoring the process over a long period of time. In this study, a planar microelectrode array (MEA) with platinum (Pt) black electroplating is designed to monitor the electrophysiological activities and pharmacological responses of cerebral organoids using an external neural signal acquisition system interfaced with the MEA. The planar MEA with Pt black electroplating has a significantly reduced electrode impedance and exhibits a robust capability for the real-time detection of spontaneous neural activities, including extracellular spikes and local field potentials. Distinct electrophysiological signal strengths in cerebral organoids were observed at early and late developmental stages. Further pharmacological stimulations showed that 30 mM KCl would induce a marked increase in spike rate, indicating an enhancement of neuronal depolarization and an elevation of network excitability. This robust response to KCl stimulation in mature networks serves as a reliable indicator of neural maturity in cerebral organoids and underscores the platform's potential for drug screening applications. This work highlights the integration of MEA technology with cerebral organoids, offering a powerful platform for real-time electrophysiological monitoring. It provides new insights into the functional maturation of neural networks and establishes a reliable system for drug screening and disease modeling, facilitating future research into human brain physiology and pathology.
Collapse
Affiliation(s)
- Yan Jin
- College of Sciences, Shanghai Institute of Technology, Shanghai 201418, China; (Y.J.); (Y.G.)
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Yixun Guo
- College of Sciences, Shanghai Institute of Technology, Shanghai 201418, China; (Y.J.); (Y.G.)
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Qiushi Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Lei Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Yuqing Ge
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Frontier Innovation Research Institute, Shanghai 201108, China
| |
Collapse
|
22
|
Mattingly Z, Chetty S. Untangling the Molecular Mechanisms Contributing to Autism Spectrum Disorder Using Stem Cells. Autism Res 2025; 18:476-485. [PMID: 39989339 DOI: 10.1002/aur.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neuro developmental condition characterized by significant genetic and phenotypic variability, making diagnosis and treatment challenging. The heterogeneity of ASD-associated genetic variants and the absence of clear causal factors in many cases complicate personalized care. Traditional models, such as postmortem brain tissue and animal studies, have provided valuable insights but are limited in capturing the dynamic processes and human-specific aspects of ASD pathology. Recent advances in human induced pluripotent stem cell (iPSC) technology have transformed ASD research by enabling the generation of patient-derived neural cells in both two-dimensional cultures and three-dimensional brain organoid models. These models retain the donor's genetic background, allowing researchers to investigate disease-specific cellular and molecular mechanisms while identifying potential therapeutic targets tailored to individual patients. This commentary highlights how stem cell-based approaches are advancing our understanding of ASD and paving the way for more personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zoe Mattingly
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sundari Chetty
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Kim E, Jeong E, Hong YM, Jeong I, Kim J, Kwon YW, Park YG, Lee J, Choi S, Kim JY, Lee JH, Cho SW, Park JU. Magnetically reshapable 3D multi-electrode arrays of liquid metals for electrophysiological analysis of brain organoids. Nat Commun 2025; 16:2011. [PMID: 40016200 PMCID: PMC11868496 DOI: 10.1038/s41467-024-55752-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 12/23/2024] [Indexed: 03/01/2025] Open
Abstract
To comprehend the volumetric neural connectivity of a brain organoid, it is crucial to monitor the spatiotemporal electrophysiological signals within the organoid, known as intra-organoid signals. However, previous methods risked damaging the three-dimensional (3D) cytoarchitecture of organoids, either through sectioning or inserting rigid needle-like electrodes. Also, the limited numbers of electrodes in fixed positions with non-adjustable electrode shapes were insufficient for examining the complex neural activity throughout the organoid. Herein, we present a magnetically reshapable 3D multi-electrode array (MEA) using direct printing of liquid metals for electrophysiological analysis of brain organoids. The adaptable distribution and the softness of these printed electrodes facilitate the spatiotemporal recording of intra-organoid signals. Furthermore, the unique capability to reshape these soft electrodes within the organoid using magnetic fields allows a single electrode in the MEA to record from multiple points, effectively increasing the recording site density without the need for additional electrodes.
Collapse
Affiliation(s)
- Enji Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yeon-Mi Hong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Junghoon Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yong Won Kwon
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Young-Geun Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Jiin Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suah Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ju-Young Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea.
| | - Seung-Woo Cho
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea.
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea.
- Department of Neurosurgery, Yonsei University College of Medicine, Yonsei, Republic of Korea.
- Yonsei-KIST Convergence Research Institute, Seoul, 03722, Republic of Korea.
| |
Collapse
|
24
|
Noh S, Park Y, Kim B, Mun JY. Structural Analysis of Cerebral Organoids Using Confocal Microscopy and Transmission/Scanning Electron Microscopy. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2025; 31:ozae119. [PMID: 39999189 DOI: 10.1093/mam/ozae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 11/10/2024] [Indexed: 02/27/2025]
Abstract
Cerebral organoid cultures from human-induced pluripotent stem cells are widely used to study complex human brain development; however, there is still limited ultrastructural information regarding the development. In this study, we examined the structural details of cerebral organoids using various microscopy techniques. Two protocols were chosen as representative methods for the development of brain organoids: the classic whole-cerebral organoid (Whole-CO) culture technique, and the air-liquid interface-cerebral organoid (ALI-CO) culture technique. Immunostained confocal laser scanning microscopy (CLSM) revealed the formation of the CTIP2- and TBR1-positive cortical deep layer on days 90 and 150, depending on the developmental progress of both methods. Furthermore, the presence of astrocytes and oligodendrocytes was verified through immunostained CLSM utilizing two-dimensional and three-dimensional reconstruction images after a 150-day period. Transmission electron microscopy analysis revealed nanometer-resolution details of the cellular organelles and neuron-specific structures including synapses and myelin. Large-area scanning electron microscopy confirmed the well-developed neuronal connectivity from each culture method on day 150. Using those microscopy techniques, we clearly showed significant details within two representative culture protocols, the Whole-CO and ALI-CO culture methods. These multi-level images provide ultrastructural insight into the features of cerebral organoids depending on the developmental stage.
Collapse
Affiliation(s)
- Seulgi Noh
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Yurim Park
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Beomsue Kim
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| | - Ji Young Mun
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| |
Collapse
|
25
|
Yang J, Jiang Y, Li M, Wu K, Wei S, Zhao Y, Shen J, Du F, Chen Y, Deng S, Xiao Z, Yuan W, Wu X. Organoid, organ-on-a-chip and traditional Chinese medicine. Chin Med 2025; 20:22. [PMID: 39940016 PMCID: PMC11823035 DOI: 10.1186/s13020-025-01071-8] [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: 07/24/2024] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
In the past few years, the emergence of organoids and organ-on-a-chip (OOAC) technologies, which are complementary to animal models and two-dimensional cell culture methods and can better simulate the internal environment of the human body, provides a new platform for traditional Chinese medicine (TCM) studies. Organoids and OOAC techniques have been increasingly applied in the fields of drug screening, drug assessment and development, personalized therapies, and developmental biology, and there have been some application cases in the TCM studies. In this review, we summarized the current status of using organoid and OOAC technologies in TCM research and provide key insights for future study. It is believed that organoid and OOAC technologies will play more and more important roles in research and make greater contributions to the innovative development of TCM.
Collapse
Affiliation(s)
- Jiayue Yang
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yu Jiang
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Mingxing Li
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Ke Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shulin Wei
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yueshui Zhao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Jing Shen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Fukuan Du
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Yu Chen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Shuai Deng
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Zhangang Xiao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
- Gulin County Hospital of Traditional Chinese Medicine, Luzhou, 646500, China
- School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, 621000, Sichuan, China
| | - Wen Yuan
- School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, 621000, Sichuan, China.
| | - Xu Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Departments of Paediatrics & Paediatric Care, Luzhou People's Hospital, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
26
|
Narasipura SD, Zayas JP, Ash MK, Reyes AF, Shull T, Gambut S, Szczerkowski JLA, McKee C, Schneider JR, Lorenzo-Redondo R, Al-Harthi L, Mamede JI. Inflammatory responses revealed through HIV infection of microglia-containing cerebral organoids. J Neuroinflammation 2025; 22:36. [PMID: 39930449 PMCID: PMC11808982 DOI: 10.1186/s12974-025-03353-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
Cerebral organoids (COs) are valuable tools for studying the intricate interplay between glial cells and neurons in brain development and disease, including HIV-associated neuroinflammation. We developed a novel approach to generate microglia containing COs (CO-iMs) by co-culturing hematopoietic progenitors and inducing pluripotent stem cells. This approach allowed for the differentiation of microglia within the organoids concomitantly with the neuronal progenitors. Compared with conventional COs, CO-iMs were more efficient at generating CD45+/CD11b+/Iba-1+ microglia and presented a physiologically relevant proportion of microglia (~ 7%). CO-iMs presented substantially increased expression of microglial homeostatic and sensome markers as well as markers for the complement cascade. CO-iMs are susceptible to HIV infection, resulting in a significant increase in several pro-inflammatory cytokines/chemokines, which are abrogated by the addition of antiretrovirals. Thus, CO-iM is a robust model for deciphering neuropathogenesis, neuroinflammation, and viral infections of brain cells in a 3D culture system.
Collapse
Affiliation(s)
- Srinivas D Narasipura
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Janet P Zayas
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Michelle K Ash
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Anjelica F Reyes
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Tanner Shull
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Stephanie Gambut
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - James L A Szczerkowski
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Charia McKee
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Jeffrey R Schneider
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lena Al-Harthi
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA.
| | - João I Mamede
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
27
|
Siwakoti U, Jones SA, Kumbhare D, Cui XT, Castagnola E. Recent Progress in Flexible Microelectrode Arrays for Combined Electrophysiological and Electrochemical Sensing. BIOSENSORS 2025; 15:100. [PMID: 39997002 PMCID: PMC11853293 DOI: 10.3390/bios15020100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
Understanding brain function requires advanced neural probes to monitor electrical and chemical signaling across multiple timescales and brain regions. Microelectrode arrays (MEAs) are widely used to record neurophysiological activity across various depths and brain regions, providing single-unit resolution for extended periods. Recent advancements in flexible MEAs, built on micrometer-thick polymer substrates, have improved integration with brain tissue by mimicking the brain's soft nature, reducing mechanical trauma and inflammation. These flexible, subcellular-scale MEAs can record stable neural signals for months, making them ideal for long-term studies. In addition to electrical recording, MEAs have been functionalized for electrochemical neurotransmitter detection. Electroactive neurotransmitters, such as dopamine, serotonin, and adenosine, can be directly measured via electrochemical methods, particularly on carbon-based surfaces. For non-electroactive neurotransmitters like acetylcholine, glutamate, and γ-aminobutyric acid, alternative strategies, such as enzyme immobilization and aptamer-based recognition, are employed to generate electrochemical signals. This review highlights recent developments in flexible MEA fabrication and functionalization to achieve both electrochemical and electrophysiological recordings, minimizing sensor fowling and brain damage when implanted long-term. It covers multi-time scale neurotransmitter detection, development of conducting polymer and nanomaterial composite coatings to enhance sensitivity, incorporation of enzyme and aptamer-based recognition methods, and the integration of carbon electrodes on flexible MEAs. Finally, it summarizes strategies to acquire electrochemical and electrophysiological measurements from the same device.
Collapse
Affiliation(s)
- Umisha Siwakoti
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (U.S.); (S.A.J.)
| | - Steven A. Jones
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (U.S.); (S.A.J.)
| | - Deepak Kumbhare
- Department of Neurosurgery, Louisiana State University Health Sciences, Shreveport, LA 71103, USA;
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburg, Pittsburgh, PA 15260, USA;
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Elisa Castagnola
- Department of Biomedical Engineering, Louisiana Tech University, Ruston, LA 71272, USA; (U.S.); (S.A.J.)
- Department of Bioengineering, University of Pittsburg, Pittsburgh, PA 15260, USA;
- Institute for Micromanufacturing, Louisiana Tech University, Ruston, LA 71272, USA
| |
Collapse
|
28
|
Glass MR, Matoba N, Beltran AA, Patel NK, Farah TM, Eswar K, Bhargava S, Huang K, Curtin I, Ahmed S, Srivastava M, Drake E, Davis LT, Yeturi M, Sun K, Love MI, Simon JM, St John T, Marrus N, Pandey J, Estes A, Dager S, Schultz RT, Botteron K, Evans A, Kim SH, Styner M, McKinstry RC, Collins DL, Volk H, Benke K, Zwaigenbaum L, Hazlett H, Beltran AS, Girault JB, Shen MD, Piven J, Stein JL. Early cell cycle genes in cortical organoid progenitors predict interindividual variability in infant brain growth trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637106. [PMID: 39974968 PMCID: PMC11839139 DOI: 10.1101/2025.02.07.637106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell (iPSC) derived cortical organoids (hCOs) model neurogenesis on an individual's genetic background. The degree to which hCO phenotypes recapitulate the brain growth of the participants from which they were derived is not well established. We generated up to 3 iPSC clones from each of 18 participants in the Infant Brain Imaging Study, who have undergone longitudinal brain imaging during infancy. We identified consistent hCO morphology and cortical cell types across clones from the same participant. hCO cross-sectional area and production of cortical hem cells were associated with in vivo cortical growth rates. Cell cycle associated genes expression in early progenitors at the crux of fate decision trajectories were correlated with cortical growth rate from 6-12 months of age, and were enriched in microcephaly and neurodevelopmental disorder genes. Our data suggest the hCOs capture inter-individual variation in cortical cell types influencing infant cortical surface area expansion.
Collapse
Affiliation(s)
- Madison R Glass
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Alvaro A Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Tala M Farah
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karthik Eswar
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Shivam Bhargava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karen Huang
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Ian Curtin
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Sara Ahmed
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Mary Srivastava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Emma Drake
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Liam T Davis
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Meghana Yeturi
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Kexin Sun
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jeremy M Simon
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Present address: Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Present address: Department of Biostatistics, Harvard T.H. Chan School of Public Health, MA, USA
| | - Tanya St John
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Natasha Marrus
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Juhi Pandey
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Annette Estes
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Stephen Dager
- Department of Radiology, University of Washington; Seattle, WA, USA
- Department of Bioengineering, University of Washington; Seattle, WA, USA
| | - Robert T Schultz
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Kelly Botteron
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Alan Evans
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Psychiatry, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Sun Hyung Kim
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Martin Styner
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Computer Sciences, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Robert C McKinstry
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - D Louis Collins
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Heather Volk
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Kelly Benke
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Lonnie Zwaigenbaum
- Department of Developmental Pediatrics, University of Alberta; Edmonton, AB, CA
| | - Heather Hazlett
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Adriana S Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jessica B Girault
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Mark D Shen
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
- Lead contact
| |
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 2025; 60:101036. [PMID: 39567282 PMCID: PMC11825288 DOI: 10.1016/j.dmpk.2024.101036] [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] [Indexed: 11/22/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding its 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 SUD using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUD 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, USA
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA.
| |
Collapse
|
30
|
Ajongbolo AO, Langhans SA. YAP/TAZ-associated cell signaling - at the crossroads of cancer and neurodevelopmental disorders. Front Cell Dev Biol 2025; 13:1522705. [PMID: 39936032 PMCID: PMC11810912 DOI: 10.3389/fcell.2025.1522705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
YAP/TAZ (Yes-associated protein/paralog transcriptional co-activator with PDZ-binding domain) are transcriptional cofactors that are the key and major downstream effectors of the Hippo signaling pathway. Both are known to play a crucial role in defining cellular outcomes, including cell differentiation, cell proliferation, and apoptosis. Aside from the canonical Hippo signaling cascade with the key components MST1/2 (mammalian STE20-like kinase 1/2), SAV1 (Salvador homologue 1), MOB1A/B (Mps one binder kinase activator 1A/B) and LATS1/2 (large tumor suppressor kinase 1/2) upstream of YAP/TAZ, YAP/TAZ activation is also influenced by numerous other signaling pathways. Such non-canonical regulation of YAP/TAZ includes well-known growth factor signaling pathways such as the epidermal growth factor receptor (EGFR)/ErbB family, Notch, and Wnt signaling as well as cell-cell adhesion, cell-matrix interactions and mechanical cues from a cell's microenvironment. This puts YAP/TAZ at the center of a complex signaling network capable of regulating developmental processes and tissue regeneration. On the other hand, dysregulation of YAP/TAZ signaling has been implicated in numerous diseases including various cancers and neurodevelopmental disorders. Indeed, in recent years, parallels between cancer development and neurodevelopmental disorders have become apparent with YAP/TAZ signaling being one of these pathways. This review discusses the role of YAP/TAZ in brain development, cancer and neurodevelopmental disorders with a special focus on the interconnection in the role of YAP/TAZ in these different conditions.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
- Biological Sciences Graduate Program, University of Delaware, Newark, DE, United States
| | - Sigrid A. Langhans
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
| |
Collapse
|
31
|
Park S, Min CH, Choi E, Choi JS, Park K, Han S, Choi W, Jang HJ, Cho KO, Kim M. Long-term tracking of neural and oligodendroglial development in large-scale human cerebral organoids by noninvasive volumetric imaging. Sci Rep 2025; 15:2536. [PMID: 39833280 PMCID: PMC11747076 DOI: 10.1038/s41598-025-85455-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Human cerebral organoids serve as a quintessential model for deciphering the complexities of brain development in a three-dimensional milieu. However, imaging these organoids, particularly when they exceed several millimeters in size, has been curtailed by the technical impediments such as phototoxicity, slow imaging speeds, and inadequate resolution and imaging depth. Addressing these pivotal challenges, our study has pioneered a high-speed scanning microscope, synergistically coupled with advanced computational image processing. This ensemble has empowered us to monitor the intricate dynamics of neuron and oligodendrocyte development within cerebral organoids across a trajectory of approximately two months. Line-shaped illumination mitigates photodamage and, alongside refined spatial gating, maximizes signal collection through integrating with computational processing. The integration of deconvolution and compressive sensing has improved image contrast by 6-fold, elucidating fine features of the neurites. Thus, noninvasive imaging enabled us to perform long-term tracking of neural and oligodendroglial development in the large-scale human cerebral organoid. Furthermore, our sophisticated volumetric segmentation algorithm has yielded a robust four-dimensional quantitative analysis, encapsulating both neuronal and oligodendroglial maturation. Collectively, these advances mark a significant advancement in the field of neurodevelopment, providing a powerful tool for in-depth study of complex brain organoid systems.
Collapse
Affiliation(s)
- Sangjun Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Cheol Hong Min
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Eunjin Choi
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jeong-Sun Choi
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Kyungjin Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedical Engineering, UNIST, Ulsan, 44919, Korea
| | - Seokyoung Han
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Mechanical Engineering, University of Louisville, Louisville, KY, 40208, USA
| | - Wonjun Choi
- Park Systems Corp, Suwon, 16229, Gyeonggi-do, Korea
| | - Hyun-Jong Jang
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, Seoul, 06591, Korea
| | - Kyung-Ok Cho
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Catholic Neuroscience Institute, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, Seoul, 06591, Korea.
| | - Moonseok Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
32
|
Winden KD, Gisser I, Sahin M. Using cortical organoids to understand the pathogenesis of malformations of cortical development. Front Neurosci 2025; 18:1522652. [PMID: 39881808 PMCID: PMC11774837 DOI: 10.3389/fnins.2024.1522652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025] Open
Abstract
Malformations of cortical development encompass a broad range of disorders associated with abnormalities in corticogenesis. Widespread abnormalities in neuronal formation or migration can lead to small head size or microcephaly with disorganized placement of cell types. Specific, localized malformations are termed focal cortical dysplasias (FCD). Neurodevelopmental disorders are common in all types of malformations of cortical development with the most prominent being refractory epilepsy, behavioral disorders such as autism spectrum disorder (ASD), and learning disorders. Several genetic pathways have been associated with these disorders from control of cell cycle and cytoskeletal dynamics in global malformations to variants in growth factor signaling pathways, especially those interacting with the mechanistic target of rapamycin (mTOR), in FCDs. Despite advances in understanding these disorders, the underlying developmental pathways that lead to lesion formation and mechanisms through which defects in cortical development cause specific neurological symptoms often remains unclear. One limitation is the difficulty in modeling these disorders, as animal models frequently do not faithfully mirror the human phenotype. To circumvent this obstacle, many investigators have turned to three-dimensional human stem cell models of the brain, known as organoids, because they recapitulate early neurodevelopmental processes. High throughput analysis of these organoids presents a promising opportunity to model pathophysiological processes across the breadth of malformations of cortical development. In this review, we highlight advances in understanding the pathophysiology of brain malformations using organoid models.
Collapse
Affiliation(s)
| | | | - Mustafa Sahin
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
33
|
Yan H, Abdulla A, Wang A, Ding S, Zhang M, Zhang Y, Zhuang TY, Wu L, Wang Y, Ren R, Jiang L, Ding X. Time-Lapse Acquisition of Both Freely Secreted Proteome and Exosome Encapsulated Proteome in Live Organoids' Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406509. [PMID: 39573935 PMCID: PMC11727246 DOI: 10.1002/advs.202406509] [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: 06/12/2024] [Revised: 10/24/2024] [Indexed: 01/14/2025]
Abstract
Proteomic communications in neighboring microenvironments during early organ development is a dynamic process that continuously reshapes human embryonic stem cells (hESCs) developmental fate. Such dynamic proteomic alteration in the microenvironment consists of both freely secreted proteome and exosome-encapsulated proteome. Simultaneous monitoring of the time-lapse shift of both proteomes with live organoids remains technically challenging. Here, a continuous organoid secretion/encapsulation proteome tandem LC-MS/MS (COSEP-LCM) is introduced, which permits time-lapse monitoring of proteomic alterations both in free secretion form and in exosome encapsulated form at live organoids' microenvironment. Continuous growth of human cerebral organoids (COs) and free-secretion/exosome-encapsulation proteomics acquisition with COSEP-LCM for 60 days is demonstrated. SERPINF1, F5, and EFNB1 are initially enriched inside exosomes as encapsulated excretion and then gradually enriched outside exosomes as freely secreted excretion, while C3 is initially enriched outside exosomes as freely secreted excretion and gradually enriched inside exosomes as encapsulated excretion. Such dynamic excretion pattern paradigm shift may imply critical developmental strategy evolution during early human cerebral development. COSEP-LCM offers a platform technique for continuous inside/outside exosome proteomics co-analysis in live organoids' microenvironment.
Collapse
Affiliation(s)
- Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Aiting Wang
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Shuyu Ding
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Manlin Zhang
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Yizhi Zhang
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Tsz Yui Zhuang
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Leqi Wu
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Yan Wang
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Rongrong Ren
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalShanghai Jiaotong University School of MedicineShanghai200092P. R. China
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care UnitXinhua HospitalSchool of Medicine and School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030P. R. China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineShanghai Jiao Tong UniversityShanghai200030P. R. China
| |
Collapse
|
34
|
Jin M, Ma Z, Zhang H, Papetti AV, Dang R, Stillitano AC, Zou L, Goldman SA, Jiang P. Human-Mouse Chimeric Brain Models to Study Human Glial-Neuronal and Macroglial-Microglial Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601990. [PMID: 39005270 PMCID: PMC11244967 DOI: 10.1101/2024.07.03.601990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Human-mouse chimeric brain models, generated by transplanting human induced pluripotent stem cell (hiPSC)-derived neural cells, are valuable for studying the development and function of human neural cells in vivo. Understanding glial-glial and glial-neuronal interactions is essential for unraveling the complexities of brain function and developing treatments for neurological disorders. To explore these interactions between human neural cells in vivo, we co-engrafted hiPSC-derived neural progenitor cells together with primitive macrophage progenitors into the neonatal mouse brain. This approach creates human-mouse chimeric brains containing human microglia, macroglia (astroglia and oligodendroglia), and neurons. Using super-resolution imaging and 3D reconstruction techniques, we examine the dynamics between human neurons and glia, and observe human microglia pruning synapses of human neurons, and often engulfing neurons themselves. Single-cell RNA sequencing analysis of the chimeric brain uncovers a close recapitulation of the human glial progenitor cell population, along with a dynamic stage in astroglial development that mirrors the processes found in the human brain. Furthermore, cell-cell communication analysis highlights significant neuronal-glial and macroglial-microglial interactions, especially the interaction between adhesion molecules neurexins and neuroligins between neurons and astroglia, emphasizing their key role in synaptogenesis. We also observed interactions between microglia and astroglia mediated by SPP1, crucial for promoting microglia growth and astrogliosis, and the PTN-MK pathways, instrumental in homeostatic maintenance and development in macroglial progenitors. This innovative co-transplantation model opens up new avenues for exploring the complex pathophysiological mechanisms underlying human neurological diseases. It holds particular promise for studying disorders where glial-neuronal interactions and non-cell-autonomous effects play crucial roles.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Haiwei Zhang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Ava V. Papetti
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Rui Dang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Lisa Zou
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
35
|
Zhao X, Sun Q, Shou Y, Chen W, Wang M, Qu W, Huang X, Li Y, Wang C, Gu Y, Ji C, Shu Q, Li X. A human forebrain organoid model reveals the essential function of GTF2IRD1-TTR-ERK axis for the neurodevelopmental deficits of Williams syndrome. eLife 2024; 13:RP98081. [PMID: 39671308 PMCID: PMC11643624 DOI: 10.7554/elife.98081] [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] [Indexed: 12/15/2024] Open
Abstract
Williams syndrome (WS; OMIM#194050) is a rare disorder, which is caused by the microdeletion of one copy of 25-27 genes, and WS patients display diverse neuronal deficits. Although remarkable progresses have been achieved, the mechanisms for these distinct deficits are still largely unknown. Here, we have shown that neural progenitor cells (NPCs) in WS forebrain organoids display abnormal proliferation and differentiation capabilities, and synapse formation. Genes with altered expression are related to neuronal development and neurogenesis. Single cell RNA-seq (scRNA-seq) data analysis revealed 13 clusters in healthy control and WS organoids. WS organoids show an aberrant generation of excitatory neurons. Mechanistically, the expression of transthyretin (TTR) are remarkably decreased in WS forebrain organoids. We have found that GTF2IRD1 encoded by one WS associated gene GTF2IRD1 binds to TTR promoter regions and regulates the expression of TTR. In addition, exogenous TTR can activate ERK signaling and rescue neurogenic deficits of WS forebrain organoids. Gtf2ird1-deficient mice display similar neurodevelopmental deficits as observed in WS organoids. Collectively, our study reveals critical function of GTF2IRD1 in regulating neurodevelopment of WS forebrain organoids and mice through regulating TTR-ERK pathway.
Collapse
Affiliation(s)
- Xingsen Zhao
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
- The Institute of Translational Medicine, School of Medicine, Zhejiang UniversityHangzhouChina
- Binjiang Institute of Zhejiang UniversityHangzhouChina
| | - Qihang Sun
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
- The Institute of Translational Medicine, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Yikai Shou
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Weijun Chen
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Mengxuan Wang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
- The Institute of Translational Medicine, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Wenzheng Qu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Xiaoli Huang
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Ying Li
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Chao Wang
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Yan Gu
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Chai Ji
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Qiang Shu
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Xuekun Li
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang UniversityHangzhouChina
- The Institute of Translational Medicine, School of Medicine, Zhejiang UniversityHangzhouChina
- Binjiang Institute of Zhejiang UniversityHangzhouChina
| |
Collapse
|
36
|
Shukla S, Schwartz JL, Walsh C, Wong WM, Patel V, Hsieh YP, Onwuasoanya C, Chen S, Offenhäusser A, Cauwenberghs G, Santoro F, Muotri AR, Yeo GW, Chalasani SH, Jahed Z. Supra- and sub-threshold intracellular-like recording of 2D and 3D neuronal networks using nanopillar electrode arrays. MICROSYSTEMS & NANOENGINEERING 2024; 10:184. [PMID: 39632788 PMCID: PMC11618331 DOI: 10.1038/s41378-024-00817-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024]
Abstract
The brain integrates activity across networks of interconnected neurons to generate behavioral outputs. Several physiological and imaging-based approaches have been previously used to monitor responses of individual neurons. While these techniques can identify cellular responses greater than the neuron's action potential threshold, less is known about the events that are smaller than this threshold or are localized to subcellular compartments. Here we use NEAs to obtain temporary intracellular access to neurons allowing us to record information-rich data that indicates action potentials, and sub-threshold electrical activity. We demonstrate these recordings from primary hippocampal neurons, induced pluripotent stem cell-derived (iPSC) neurons, and iPSC-derived brain organoids. Moreover, our results show that our arrays can record activity from subcellular compartments of the neuron. We suggest that these data might enable us to correlate activity changes in individual neurons with network behavior, a key goal of systems neuroscience.
Collapse
Affiliation(s)
- Shivani Shukla
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Joshua L Schwartz
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Callum Walsh
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Wen Mai Wong
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Vrund Patel
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yu-Peng Hsieh
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Chichi Onwuasoanya
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shaoming Chen
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Andreas Offenhäusser
- Institute of Biological Information Processing-Bioelectronics, IBI-3, Forschungszentrum Jülich, Jülich, 52428, Germany
| | - Gert Cauwenberghs
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Francesca Santoro
- Institute of Biological Information Processing-Bioelectronics, IBI-3, Forschungszentrum Jülich, Jülich, 52428, Germany
- Neuroelectronic Interfaces, Faculty of Electrical Engineering and IT, RWTH Aachen, Aachen, 52074, Germany
| | - Alysson R Muotri
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Center for Academic Research and Training in Anthropogeny (CARTA) and Archealization (ArchC), University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Stem Cell Education and Integrated Space Stem Cell Orbital Research (ISSCOR) Center University of California San Diego, La Jolla, CA, 92093, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Stem Cell Institute Innovation Center, University of California San Diego, La Jolla, CA, 92093, USA
- Center for RNA Technologies and Therapeutics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sreekanth H Chalasani
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Zeinab Jahed
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
37
|
Amin ND, Kelley KW, Kaganovsky K, Onesto M, Hao J, Miura Y, McQueen JP, Reis N, Narazaki G, Li T, Kulkarni S, Pavlov S, Pașca SP. Generating human neural diversity with a multiplexed morphogen screen in organoids. Cell Stem Cell 2024; 31:1831-1846.e9. [PMID: 39642864 DOI: 10.1016/j.stem.2024.10.016] [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: 07/24/2023] [Revised: 07/31/2024] [Accepted: 10/28/2024] [Indexed: 12/09/2024]
Abstract
Morphogens choreograph the generation of remarkable cellular diversity in the developing nervous system. Differentiation of stem cells in vitro often relies upon the combinatorial modulation of these signaling pathways. However, the lack of a systematic approach to understand morphogen-directed differentiation has precluded the generation of many neural cell populations, and the general principles of regional specification and maturation remain incomplete. Here, we developed an arrayed screen of 14 morphogen modulators in human neural organoids cultured for over 70 days. Deconvolution of single-cell-multiplexed RNA sequencing data revealed design principles of brain region specification. We tuned neural subtype diversity to generate a tachykinin 3 (TAC3)-expressing striatal interneuron type within assembloids. To circumvent limitations of in vitro neuronal maturation, we used a neonatal rat transplantation strategy that enabled human Purkinje neurons to develop their hallmark complex dendritic branching. This comprehensive platform yields insights into the factors influencing stem cell-derived neural diversification and maturation.
Collapse
Affiliation(s)
- Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Konstantin Kaganovsky
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Massimo Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Jin Hao
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - James P McQueen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Noah Reis
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Genta Narazaki
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Tommy Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Shravanti Kulkarni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Sergey Pavlov
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA.
| |
Collapse
|
38
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
39
|
Kelley KW, Revah O, Gore F, Kaganovsky K, Chen X, Deisseroth K, Pașca SP. Host circuit engagement of human cortical organoids transplanted in rodents. Nat Protoc 2024; 19:3542-3567. [PMID: 39075308 DOI: 10.1038/s41596-024-01029-4] [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: 11/06/2023] [Accepted: 05/22/2024] [Indexed: 07/31/2024]
Abstract
Human neural organoids represent promising models for studying neural function; however, organoids grown in vitro lack certain microenvironments and sensory inputs that are thought to be essential for maturation. The transplantation of patient-derived neural organoids into animal hosts helps overcome some of these limitations and offers an approach for neural organoid maturation and circuit integration. Here, we describe a method for transplanting human stem cell-derived cortical organoids (hCOs) into the somatosensory cortex of newborn rats. The differentiation of human induced pluripotent stem cells into hCOs occurs over 30-60 days, and the transplantation procedure itself requires ~0.5-1 hours per animal. The use of neonatal hosts provides a developmentally appropriate stage for circuit integration and allows the generation and experimental manipulation of a unit of human neural tissue within the cortex of a living animal host. After transplantation, animals can be maintained for hundreds of days, and transplanted hCO growth can be monitored by using brain magnetic resonance imaging. We describe the assessment of human neural circuit function in vivo by monitoring genetically encoded calcium responses and extracellular activity. To demonstrate human neuron-host functional integration, we also describe a procedure for engaging host neural circuits and for modulating animal behavior by using an optogenetic behavioral training paradigm. The transplanted human neurons can then undergo ex vivo characterization across modalities including dendritic morphology reconstruction, single-nucleus transcriptomics, optogenetic manipulation and electrophysiology. This approach may enable the discovery of cellular phenotypes from patient-derived cells and uncover mechanisms that contribute to human brain evolution from previously inaccessible developmental stages.
Collapse
Affiliation(s)
- Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Felicity Gore
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Konstantin Kaganovsky
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
40
|
Dave B, Tailor J. Human stem cell models to unravel brain cancer. BMC Cancer 2024; 24:1465. [PMID: 39609728 PMCID: PMC11603633 DOI: 10.1186/s12885-024-13187-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Pre-clinical animal models of human brain tumors have been invaluable tools for studying cancer pathogenesis and exploring novel treatment modalities. Such models recapitulate important aspects of the human disease such as the stem-progenitor-differentiated cell hierarchy. Although powerful, we argue that animal models are inherently limited in their ability to phenocopy certain important aspects of human brain tumor biology. We specifically highlight the inability of mouse models to generate certain forms aggressive pediatric medulloblastoma likely owing to cellular, anatomic, and genetic differences between the human and mouse brains. Additionally, we review some limitations of human brain tumor derived cell lines and outline why they are a sub-optimal system for purposes of pre-clinical modeling. Below, we present the case for human stem cell-based models of brain tumors, focusing mainly on glioblastoma and medulloblastoma. Drawing on several recently published studies, we review the exciting progress that has been made towards modeling human brain tumors using two-dimensional adherent stem cell cultures and three-dimensional organoids. We identify the important advances arrived at using these human stem cell-based models and suggest opportunities for future work in this direction. In this review article, we aim to highlight the utility and promises of human stem cell-based models of brain tumors as a complementary system to traditional transgenic animal and cell line systems.
Collapse
Affiliation(s)
- Biren Dave
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Jignesh Tailor
- Division of Pediatric Neurosurgery, Riley Hospital for Children, Indianapolis, IN, USA.
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
41
|
Nishimura H, Li Y. Human pluripotent stem cell-derived models of the hippocampus. Int J Biochem Cell Biol 2024; 177:106695. [PMID: 39557338 DOI: 10.1016/j.biocel.2024.106695] [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: 08/31/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
The hippocampus is a crucial structure of the brain, recognised for its roles in the formation of memory, and our ability to navigate the world. Despite its importance, clear understanding of how the human hippocampus develops and its contribution to disease is limited due to the inaccessible nature of the human brain. In this regard, the advent of human pluripotent stem cell (hPSC) technologies has enabled the study of human biology in an unprecedented manner, through the ability to model development and disease as both 2D monolayers and 3D organoids. In this review, we explore the existing efforts to derive the hippocampal lineage from hPSCs and evaluate the various aspects of the in vivo hippocampus that are replicated in vitro. In addition, we highlight key diseases that have been modelled using hPSC-derived cultures and offer our perspective on future directions for this emerging field.
Collapse
Affiliation(s)
- Haruka Nishimura
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Yun Li
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
42
|
Müller Y, Lengacher L, Friscourt F, Quairiaux C, Stoppini L, Magistretti PJ, Lengacher S, Finsterwald C. Epileptiform activity in brain organoids derived from patient with Glucose Transporter 1 Deficiency Syndrome. Front Neurosci 2024; 18:1498801. [PMID: 39605786 PMCID: PMC11599213 DOI: 10.3389/fnins.2024.1498801] [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: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Glucose Transporter 1-Deficiency Syndrome (GLUT1-DS) is a rare genetic disorder caused by mutations in the gene encoding for GLUT1 and characterized by impaired glucose uptake in the brain. This leads to brain hypometabolism and the development of symptoms that include epilepsy, motor dysfunctions and cognitive impairment. The development of patient-specific in vitro models is a valuable tool for understanding the pathophysiology of rare genetic disorders and testing new therapeutic interventions. Methods In this study, we generated brain organoids from induced pluripotent stem cells (iPSCs) derived either from a GLUT1-DS patient or a healthy individual. The functional organoids were analyzed for cellular composition, maturity, and electrophysiological activity using a custom-made microelectrode array (MEA) platform, which allowed for the detection of spikes, burst patterns, and epileptiform discharges. Results Immunostaining revealed a similar distribution of neurons and astrocytes in both healthy and GLUT1-DS brain organoids, though GLUT1-DS brain organoids exhibited reduced cellular density and smaller overall size. Electrophysiological recordings demonstrated functional spike profiles in both organoid types. Notably, our study demonstrates that brain organoids derived from a GLUT1-DS patient exhibit distinct epileptiform activity and heightened sensitivity to glucose deprivation, reflecting key features of the disorder. Discussion These findings validate the use of brain organoids as a model for studying GLUT1-DS and highlight their potential for testing novel therapeutic strategies aimed at improving glucose metabolism and managing epilepsy in patients.
Collapse
Affiliation(s)
| | | | - F. Friscourt
- Functional Brain Mapping Lab, Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
- Neurosurgery Clinic, Department of Clinical Neuroscience, University Hospital Geneva, Geneva, Switzerland
| | - C. Quairiaux
- Functional Brain Mapping Lab, Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - L. Stoppini
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | | | | | | |
Collapse
|
43
|
Hu N, Shi JX, Chen C, Xu HH, Chang ZH, Hu PF, Guo D, Zhang XW, Shao WW, Fan X, Zuo JC, Ming D, Li XH. Constructing organoid-brain-computer interfaces for neurofunctional repair after brain injury. Nat Commun 2024; 15:9580. [PMID: 39505863 PMCID: PMC11541701 DOI: 10.1038/s41467-024-53858-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/24/2024] [Indexed: 11/08/2024] Open
Abstract
The reconstruction of damaged neural circuits is critical for neurological repair after brain injury. Classical brain-computer interfaces (BCIs) allow direct communication between the brain and external controllers to compensate for lost functions. Importantly, there is increasing potential for generalized BCIs to input information into the brains to restore damage, but their effectiveness is limited when a large injured cavity is caused. Notably, it might be overcome by transplantation of brain organoids into the damaged region. Here, we construct innovative BCIs mediated by implantable organoids, coined as organoid-brain-computer interfaces (OBCIs). We assess the prolonged safety and feasibility of the OBCIs, and explore neuroregulatory strategies. OBCI stimulation promotes progressive differentiation of grafts and enhances structural-functional connections within organoids and the host brain, promising to repair the damaged brain via regenerating and regulating, potentially directing neurons to preselected targets and recovering functional neural networks in the future.
Collapse
Affiliation(s)
- Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Jian-Xin Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Hai-Huan Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
- Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Peng-Fei Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Di Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Wen-Wei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Xiu Fan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Jia-Chen Zuo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China
| | - Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China.
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, China.
| |
Collapse
|
44
|
Zhou G, Li R, Sheng S, Huang J, Zhou F, Wei Y, Liu H, Su J. Organoids and organoid extracellular vesicles-based disease treatment strategies. J Nanobiotechnology 2024; 22:679. [PMID: 39506799 PMCID: PMC11542470 DOI: 10.1186/s12951-024-02917-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Organoids are "mini-organs" that self-organize and differentiate from stem cells under in vitro 3D culture conditions, mimicking the spatial structure and function of tissues in vivo. Extracellular vesicles (EVs) are nanoscale phospholipid bilayer vesicles secreted by living cells, rich in bioactive molecules, with excellent biocompatibility and low immunogenicity. Compared to EVs, organoid-derived EVs (OEVs) exhibit higher yield and enhanced biological functions. Organoids possess stem cell characteristics, and OEVs are capable of delivering active substances, making both highly promising for medical applications. In this review, we provide an overview of the fundamental biological principles of organoids and OEVs, and discuss their current applications in disease treatment. We then focus on the differences between OEVs and traditional EVs. Subsequently, we present methods for the engineering modification of OEVs. Finally, we critically summarize the advantages and challenges of organoids and OEVs. In conclusion, we believe that a deeper understanding of organoids and OEVs will provide innovative solutions to complex diseases.
Collapse
Affiliation(s)
- Guangyin Zhou
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Ruiyang Li
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jingtao Huang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, China.
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
45
|
Nakamura Y, Shimada IS, Maroofian R, Falabella M, Zaki MS, Fujimoto M, Sato E, Takase H, Aoki S, Miyauchi A, Koshimizu E, Miyatake S, Arioka Y, Honda M, Higashi T, Miya F, Okubo Y, Ogawa I, Scardamaglia A, Miryounesi M, Alijanpour S, Ahmadabadi F, Herkenrath P, Dafsari HS, Velmans C, Al Balwi M, Vitobello A, Denommé-Pichon AS, Jeanne M, Civit A, Abdel-Hamid MS, Naderi H, Darvish H, Bakhtiari S, Kruer MC, Carroll CJ, Ghayoor Karimiani E, Khailany RA, Abdulqadir TA, Ozaslan M, Bauer P, Zifarelli G, Seifi T, Zamani M, Al Alam C, Alvi JR, Sultan T, Efthymiou S, Pope SAS, Haginoya K, Matsunaga T, Osaka H, Matsumoto N, Ozaki N, Ohkawa Y, Oki S, Tsunoda T, Pitceathly RDS, Taketomi Y, Houlden H, Murakami M, Kato Y, Saitoh S. Biallelic null variants in PNPLA8 cause microcephaly by reducing the number of basal radial glia. Brain 2024; 147:3949-3967. [PMID: 39082157 PMCID: PMC11531855 DOI: 10.1093/brain/awae185] [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: 05/18/2023] [Revised: 05/05/2024] [Accepted: 05/20/2024] [Indexed: 11/05/2024] Open
Abstract
Patatin-like phospholipase domain-containing lipase 8 (PNPLA8), one of the calcium-independent phospholipase A2 enzymes, is involved in various physiological processes through the maintenance of membrane phospholipids. Biallelic variants in PNPLA8 have been associated with a range of paediatric neurodegenerative disorders. However, the phenotypic spectrum, genotype-phenotype correlations and the underlying mechanisms are poorly understood. Here, we newly identified 14 individuals from 12 unrelated families with biallelic ultra-rare variants in PNPLA8 presenting with a wide phenotypic spectrum of clinical features. Analysis of the clinical features of current and previously reported individuals (25 affected individuals across 20 families) showed that PNPLA8-related neurological diseases manifest as a continuum ranging from variable developmental and/or degenerative epileptic-dyskinetic encephalopathy to childhood-onset neurodegeneration. We found that complete loss of PNPLA8 was associated with the more profound end of the spectrum, with congenital microcephaly. Using cerebral organoids generated from human induced pluripotent stem cells, we found that loss of PNPLA8 led to developmental defects by reducing the number of basal radial glial cells and upper-layer neurons. Spatial transcriptomics revealed that loss of PNPLA8 altered the fate specification of apical radial glial cells, as reflected by the enrichment of gene sets related to the cell cycle, basal radial glial cells and neural differentiation. Neural progenitor cells lacking PNPLA8 showed a reduced amount of lysophosphatidic acid, lysophosphatidylethanolamine and phosphatidic acid. The reduced number of basal radial glial cells in patient-derived cerebral organoids was rescued, in part, by the addition of lysophosphatidic acid. Our data suggest that PNPLA8 is crucial to meet phospholipid synthetic needs and to produce abundant basal radial glial cells in human brain development.
Collapse
Affiliation(s)
- Yuji Nakamura
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Issei S Shimada
- Department of Cell Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Reza Maroofian
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Masanori Fujimoto
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Emi Sato
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hiroshi Takase
- Core Laboratory, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Shiho Aoki
- Department of Pediatrics, Jichi Medical University, Tochigi 3290498, Japan
| | - Akihiko Miyauchi
- Department of Pediatrics, Jichi Medical University, Tochigi 3290498, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 2360004, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 2360004, Japan
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama 2360004, Japan
| | - Yuko Arioka
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya 4668550, Japan
| | - Mizuki Honda
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 7398526, Japan
| | - Takayoshi Higashi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan
| | - Fuyuki Miya
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, 1608582, Japan
| | - Yukimune Okubo
- Department of Pediatric Neurology, Miyagi Children's Hospital, Sendai 9893126, Japan
| | - Isamu Ogawa
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Annarita Scardamaglia
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1516745811, Iran
| | - Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1516745811, Iran
| | - Farzad Ahmadabadi
- Pediatric Neurology Department, Faculty of Medicine, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran 1546815514, Iran
| | - Peter Herkenrath
- Department of Pediatrics and Center for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne 50937, Germany
| | - Hormos Salimi Dafsari
- Department of Pediatrics and Center for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne 50937, Germany
- Max-Planck-Institute for Biology of Ageing, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Clara Velmans
- Faculty of Medicine and University Hospital Cologne, Institute of Human Genetics, University of Cologne, Cologne 50931, Germany
| | - Mohammed Al Balwi
- Department of Pathology and Laboratory Medicine, College of Medicine, KSAU-HS, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Antonio Vitobello
- Functional Unit for Diagnostic Innovation in Rare Diseases, FHU-TRANSLAD, Dijon Bourgogne University Hospital, Dijon 21000, France
- INSERM UMR1231 GAD ‘Génétique des Anomalies du Développement’, FHU-TRANSLAD, University of Burgundy, Dijon 21000, France
| | - Anne-Sophie Denommé-Pichon
- Functional Unit for Diagnostic Innovation in Rare Diseases, FHU-TRANSLAD, Dijon Bourgogne University Hospital, Dijon 21000, France
- INSERM UMR1231 GAD ‘Génétique des Anomalies du Développement’, FHU-TRANSLAD, University of Burgundy, Dijon 21000, France
| | - Médéric Jeanne
- Genetics Department, University Hospital of Tours, Tours 37044, France
- UMR 1253, iBrain, University of Tours, INSERM, Tours 37032, France
| | - Antoine Civit
- Genetics Department, University Hospital of Tours, Tours 37044, France
| | - Mohamed S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Hamed Naderi
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 4918936316, Iran
| | - Hossein Darvish
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 4918936316, Iran
| | - Somayeh Bakhtiari
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
- Departments of Child Health, Neurology, Cellular & Molecular Medicine and Program in Genetics, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Michael C Kruer
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
- Departments of Child Health, Neurology, Cellular & Molecular Medicine and Program in Genetics, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Christopher J Carroll
- Genetics Section, Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Ehsan Ghayoor Karimiani
- Genetics Section, Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Rozhgar A Khailany
- Department of Basic Science, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Talib Adil Abdulqadir
- Department of Pediatrics, College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Mehmet Ozaslan
- Department of Biology, Division of Molecular Biology and Genetics, Gaziantep University, Gaziantep 27410, Turkey
| | | | | | - Tahere Seifi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 83151-61355, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz 61556-89467, Iran
| | - Mina Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 83151-61355, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz 61556-89467, Iran
| | - Chadi Al Alam
- Pediatrics and Pediatric Neurology, American Center for Psychiatry and Neurology, Abu Dhabi 108699, UAE
| | - Javeria Raza Alvi
- Department of Pediatric Neurology, the Children’s Hospital and the University of Child Health Sciences, Lahore 54600, Pakistan
| | - Tipu Sultan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Simon A S Pope
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Kazuhiro Haginoya
- Department of Pediatric Neurology, Miyagi Children's Hospital, Sendai 9893126, Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Tochigi 3290498, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 2360004, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya 4668550, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 8128582, Japan
| | - Shinya Oki
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 8600811, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo 113-0033, Japan
- Laboratory for Medical Science Mathematics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan
| | - Yoichi Kato
- Department of Cell Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| |
Collapse
|
46
|
Teles E Silva AL, Yokota-Moreno BY, Branquinho MS, Salles GR, de Souza TC, de Carvalho RA, Batista G, Varella Branco E, Griesi-Oliveira K, Passos Bueno MR, Porcionatto MA, Herai RH, Gamarra LF, Sertié AL. Generation and characterization of cortical organoids from iPSC-derived dental pulp stem cells using traditional and innovative approaches. Neurochem Int 2024; 180:105854. [PMID: 39241808 DOI: 10.1016/j.neuint.2024.105854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cortical organoids derived from human induced pluripotent stem cells (hiPSCs) represent a powerful in vitro experimental system to investigate human brain development and disease, often inaccessible to direct experimentation. However, despite steady progress in organoid technology, several limitations remain, including high cost and variability, use of hiPSCs derived from tissues harvested invasively, unexplored three-dimensional (3D) structural features and neuronal connectivity. Here, using a cost-effective and reproducible protocol as well as conventional two-dimensional (2D) immunostaining, we show that cortical organoids generated from hiPSCs obtained by reprogramming stem cells from human exfoliated deciduous teeth (SHED) recapitulate key aspects of human corticogenesis, such as polarized organization of neural progenitor zones with the presence of outer radial glial stem cells, and differentiation of superficial- and deep-layer cortical neurons and glial cells. We also show that 3D bioprinting and magnetic resonance imaging of intact cortical organoids are alternative and complementary approaches to unravel critical features of the 3D architecture of organoids. Finally, extracellular electrical recordings in whole organoids showed functional neuronal networks. Together, our findings suggest that SHED-derived cortical organoids constitute an attractive model of human neurodevelopment, and support the notion that a combination of 2D and 3D techniques to analyze organoid structure and function may help improve this promising technology.
Collapse
Affiliation(s)
| | | | | | - Geisa Rodrigues Salles
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Ronald Almeida de Carvalho
- Pontifícia Universidade Católica Do Paraná, Escola de Medicina, Laboratório de Bioinformática e Neurogenética, Curitiba, Paraná, Brazil
| | - Gabriel Batista
- Pontifícia Universidade Católica Do Paraná, Escola de Medicina, Laboratório de Bioinformática e Neurogenética, Curitiba, Paraná, Brazil
| | - Elisa Varella Branco
- Centro de Estudos Do Genoma Humano e Células Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Maria Rita Passos Bueno
- Centro de Estudos Do Genoma Humano e Células Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Roberto Hirochi Herai
- Pontifícia Universidade Católica Do Paraná, Escola de Medicina, Laboratório de Bioinformática e Neurogenética, Curitiba, Paraná, Brazil
| | | | | |
Collapse
|
47
|
Pallavicini G, Moccia A, Iegiani G, Parolisi R, Peirent ER, Berto GE, Lorenzati M, Tshuva RY, Ferraro A, Balzac F, Turco E, Salvi SU, Myklebust HF, Wang S, Eisenberg J, Chitale M, Girgla NS, Boda E, Reiner O, Buffo A, Di Cunto F, Bielas SL. Modeling primary microcephaly with human brain organoids reveals fundamental roles of CIT kinase activity. J Clin Invest 2024; 134:e175435. [PMID: 39316437 PMCID: PMC11527453 DOI: 10.1172/jci175435] [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: 09/20/2023] [Accepted: 09/06/2024] [Indexed: 09/26/2024] Open
Abstract
Brain size and cellular heterogeneity are tightly regulated by species-specific proliferation and differentiation of multipotent neural progenitor cells (NPCs). Errors in this process are among the mechanisms of primary hereditary microcephaly (MCPH), a group of disorders characterized by reduced brain size and intellectual disability. Biallelic citron rho-interacting serine/threonine kinase (CIT) missense variants that disrupt kinase function (CITKI/KI) and frameshift loss-of-function variants (CITFS/FS) are the genetic basis for MCPH17; however, the function of CIT catalytic activity in brain development and NPC cytokinesis is unknown. Therefore, we created the CitKI/KI mouse model and found that it did not phenocopy human microcephaly, unlike biallelic CitFS/FS animals. Nevertheless, both Cit models exhibited binucleation, DNA damage, and apoptosis. To investigate human-specific mechanisms of CIT microcephaly, we generated CITKI/KI and CITFS/FS human forebrain organoids. We found that CITKI/KI and CITFS/FS organoids lost cytoarchitectural complexity, transitioning from pseudostratified to simple neuroepithelium. This change was associated with defects that disrupted the polarity of NPC cytokinesis, in addition to elevating apoptosis. Together, our results indicate that both CIT catalytic and scaffolding functions in NPC cytokinesis are critical for human corticogenesis. Species differences in corticogenesis and the dynamic 3D features of NPC mitosis underscore the utility of human forebrain organoid models for understanding human microcephaly.
Collapse
Affiliation(s)
- Gianmarco Pallavicini
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | | | - Giorgia Iegiani
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Roberta Parolisi
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Emily R. Peirent
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Gaia Elena Berto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Martina Lorenzati
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Rami Y. Tshuva
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Alessia Ferraro
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Fiorella Balzac
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Emilia Turco
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | | | | | | | - Julia Eisenberg
- Department of Human Genetics and
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | | - Enrica Boda
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Annalisa Buffo
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Ferdinando Di Cunto
- Neuroscience Institute Cavalieri Ottolenghi, Turin, Italy
- Department of Neuroscience “Rita Levi Montalcini,” University of Turin, Turin, Italy
| | - Stephanie L. Bielas
- Department of Human Genetics and
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Cui X, Li X, Zheng H, Su Y, Zhang S, Li M, Hao X, Zhang S, Hu Z, Xia Z, Shi C, Xu Y, Mao C. Human midbrain organoids: a powerful tool for advanced Parkinson's disease modeling and therapy exploration. NPJ Parkinsons Dis 2024; 10:189. [PMID: 39428415 PMCID: PMC11491477 DOI: 10.1038/s41531-024-00799-8] [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: 01/06/2023] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by the loss of dopaminergic neurons in the substantia nigra. Despite progress, the pathogenesis remains unclear. Human midbrain organoids (hMLOs) have emerged as a promising model for studying PD, drug screening, and potential treatments. This review discusses the development of hMLOs, their application in PD research, and current challenges in organoid construction, highlighting possible optimization strategies.
Collapse
Affiliation(s)
- Xin Cui
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinwei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuyu Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Neuro-Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China.
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
49
|
Tian C, Cai H, Ao Z, Gu L, Li X, Niu VC, Bondesson M, Gu M, Mackie K, Guo F. Engineering human midbrain organoid microphysiological systems to model prenatal PFOS exposure. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174478. [PMID: 38964381 PMCID: PMC11404128 DOI: 10.1016/j.scitotenv.2024.174478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Perfluorooctane sulfonate (PFOS), a class of synthetic chemicals detected in various environmental compartments, has been associated with dysfunctions of the human central nervous system (CNS). However, the underlying neurotoxicology of PFOS exposure is largely understudied due to the lack of relevant human models. Here, we report bioengineered human midbrain organoid microphysiological systems (hMO-MPSs) to recapitulate the response of a fetal human brain to multiple concurrent PFOS exposure conditions. Each hMO-MPS consists of an hMO on a fully 3D printed holder device with a perfusable organoid adhesion layer for enhancing air-liquid interface culturing. Leveraging the unique, simply-fabricated holder devices, hMO-MPSs are scalable, easy to use, and compatible with conventional well-plates, and allow easy transfer onto a multiple-electrode array (MEA) system for plug-and-play measurement of neural activity. Interestingly, the neural activity of hMO-MPSs initially increased and subsequently decreased by exposure to a concentration range of 0, 30, 100, to 300 μM of PFOS. Furthermore, PFOS exposure impaired neural development and promoted neuroinflammation in the engineered hMO-MPSs. Along with PFOS, our platform is broadly applicable for studies toxicology of various other environmental pollutants.
Collapse
Affiliation(s)
- Chunhui Tian
- 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
| | - Zheng Ao
- 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
| | - Xiang Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Vivian C Niu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States; Bloomington High School South, Bloomington, IN 47401, United States
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN 47405, United States
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, OH 45229, Cincinnati, United States; University of Cincinnati School of Medicine, OH 45229, Cincinnati, 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
|
50
|
Whye D, Norabuena EM, Srinivasan GR, Wood D, Polanco TJ, Makhortova NR, Sahin M, Buttermore ED. A Hybrid 2D-to-3D in vitro Differentiation Platform Improves Outcomes of Cerebral Cortical Organoid Generation in hiPSCs. Curr Protoc 2024; 4:e70022. [PMID: 39400999 DOI: 10.1002/cpz1.70022] [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] [Indexed: 10/15/2024]
Abstract
Three-dimensional (3D) cerebral cortical organoids are popular in vitro cellular model systems widely used to study human brain development and disease, compared to traditional stem cell-derived methods that use two-dimensional (2D) monolayer cultures. Despite the advancements made in protocol development for cerebral cortical organoid derivation over the past decade, limitations due to biological, mechanistic, and technical variables remain in generating these complex 3D cellular systems. Building from our previously established differentiation system, we have made modifications to our existing 3D cerebral cortical organoid protocol that resolve several of these technical and biological challenges when working with diverse groups of human induced pluripotent stem cell (hiPSC) lines. This improved protocol blends a 2D monolayer culture format for the specification of neural stem cells and expansion of neuroepithelial progenitor cells with a 3D system for improved self-aggregation and subsequent organoid development. Furthermore, this "hybrid" approach is amenable to both an accelerated cerebral cortical organoid protocol as well as an alternative long-term differentiation protocol. In addition to establishing a hybrid technical format, this protocol also offers phenotypic and morphological characterization of stage-specific cellular profiles using antibodies and fluorescent-based dyes for live cell imaging. © 2024 Wiley Periodicals LLC. Basic Protocol 1: hiPSC-based 2D monolayer specification into neural stem cells (NSCs) Basic Protocol 2: Serial passaging and 2D monolayer expansion of neuroepithelial progenitor cells (NPCs) Support Protocol 1: Direct cryopreservation and rapid thawing of NSCs and NPCs Basic Protocol 3: Bulk aggregation of 3D neurospheres and accelerated cerebral cortical organoid differentiation Alternate Protocol 1: Bulk aggregation of 3D neurospheres and long-term cerebral cortical organoid differentiation Support Protocol 2: High-throughput 3D neurosphere formation and 2D neurosphere migration assay Support Protocol 3: LIVE/DEAD stain cell imaging assay of 3D neurospheres Support Protocol 4: NeuroFluor NeuO live cell dye for 3D cerebral cortical organoids.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Erika M Norabuena
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Gayathri Rajaram Srinivasan
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Taryn J Polanco
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Nina R Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth D Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|