1
|
Zhu S, Diao S, Liu X, Zhang Z, Liu F, Chen W, Lu X, Luo H, Cheng X, Liao Q, Li Z, Chen J. Biomaterial-based strategies: a new era in spinal cord injury treatment. Neural Regen Res 2025; 20:3476-3500. [PMID: 40095657 PMCID: PMC11974648 DOI: 10.4103/nrr.nrr-d-24-00844] [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: 07/29/2024] [Revised: 09/02/2024] [Accepted: 12/16/2024] [Indexed: 03/19/2025] Open
Abstract
Enhancing neurological recovery and improving the prognosis of spinal cord injury have gained research attention recently. Spinal cord injury is associated with a complex molecular and cellular microenvironment. This complexity has prompted researchers to elucidate the underlying pathophysiological mechanisms and changes and to identify effective treatment strategies. Traditional approaches for spinal cord injury repair include surgery, oral or intravenous medications, and administration of neurotrophic factors; however, the efficacy of these approaches remains inconclusive, and serious adverse reactions continue to be a concern. With advancements in tissue engineering and regenerative medicine, emerging strategies for spinal cord injury repair now involve nanoparticle-based nanodelivery systems, scaffolds, and functional recovery techniques that incorporate biomaterials, bioengineering, stem cell, and growth factors as well as three-dimensional bioprinting. Ideal biomaterial scaffolds should not only provide structural support for neuron migration, adhesion, proliferation, and differentiation but also mimic the mechanical properties of natural spinal cord tissue. Additionally, these scaffolds should facilitate axon growth and neurogenesis by offering adjustable topography and a range of physical and biochemical cues. The three-dimensionally interconnected porous structure and appropriate physicochemical properties enabled by three-dimensional biomimetic printing technology can maximize the potential of biomaterials used for treating spinal cord injury. Therefore, correct selection and application of scaffolds, coupled with successful clinical translation, represent promising clinical objectives to enhance the treatment efficacy for and prognosis of spinal cord injury. This review elucidates the key mechanisms underlying the occurrence of spinal cord injury and regeneration post-injury, including neuroinflammation, oxidative stress, axon regeneration, and angiogenesis. This review also briefly discusses the critical role of nanodelivery systems used for repair and regeneration of injured spinal cord, highlighting the influence of nanoparticles and the factors that affect delivery efficiency. Finally, this review highlights tissue engineering strategies and the application of biomaterial scaffolds for the treatment of spinal cord injury. It discusses various types of scaffolds, their integrations with stem cells or growth factors, and approaches for optimization of scaffold design.
Collapse
Affiliation(s)
- Shihong Zhu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Sijun Diao
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhujun Zhang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Fujun Liu
- Department of Ophthalmology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiyue Lu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huiyang Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xu Cheng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Liao
- Department of Pharmacy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhongyu Li
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Zhu H, Yao C, Xu Z, Shang G, Peng J, Xie H, Qian T, Qiu Z, Maeso L, Mao M, Liao Y, Jiang Y, Li D, Orive G, Boccaccini AR. Recent advances in 3D models of the nervous system for neural regeneration research and drug development. Acta Biomater 2025:S1742-7061(25)00421-0. [PMID: 40490242 DOI: 10.1016/j.actbio.2025.06.013] [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: 03/25/2025] [Revised: 05/12/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
The development of drugs for nervous diseases poses distinctive difficulties owing to the incomplete understanding of the physiology and complex pathogenesis of the multifaceted central (CNS) and peripheral (PNS) nervous systems. Conventional animal tests and in vitro two-dimensional (2D) cell cultures fail to reproduce the sophisticated structure of natural human tissues, hindering the new drug discovery process. The emerging three-dimensional (3D) neural tissue models, including organoids, organ-on-chips and 3D-printed neural scaffolds, can provide an improved reproduction of the critical features, structural complexity, biological functions, dynamic circulation micro-environment and cell-matrix/cell interactions of the nervous systems. This review examines state-of-the-art 3D models for neural physiology/pathology, emphasizing their drug development applications. Fundamental advantages of various in vitro 3D neural models for investigating the mechanisms of nerve regeneration and disorders in both the CNS and PNS are compared in terms of the different modeling techniques. In addition, the applications of 3D neural models in drug development are summarized covering a range of areas such as disease modeling for basic research, pharmacokinetic and pharmacodynamic testing for drug screening and drug safety evaluation. Furthermore, current challenges and future outlook of biomimetic models and the existing bottlenecks hindering their successful translation into clinical use are discussed. STATEMENT OF SIGNIFICANCE: This review highlights the groundbreaking potential of 3D neural models-organoids, organ-on-chips, and 3D-printed scaffolds-to revolutionize neurological research and drug development. Unlike conventional methods, these models replicate the intricate structure and function of human nervous systems, enabling precise study of diseases like Alzheimer's, spinal injuries, and brain tumors. By synthesizing recent advancements, the review compares techniques, their applications in drug screening and personalized medicine, and addresses challenges in model accuracy and scalability. Bridging neuroscience, engineering, and pharmacology, this work provides a roadmap for researchers to innovate therapies. Its insights are critical for accelerating drug discovery and improving treatment outcomes, making it essential for scientists and clinicians tackling neurological disorders.
Collapse
Affiliation(s)
- Hui Zhu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China.
| | - Cong Yao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zhengqi Xu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Guojin Shang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Huangfan Xie
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Tingyu Qian
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Lidia Maeso
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Mao Mao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yucheng Liao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi' an, Shaanxi, China.
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, P. R. China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an 710049, P. R. China; State Industry-Education Integration Center for Medical Innovations, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria 01007, Spain
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Material Science and Engineering, University of Erlangen-Nuremberg, 91085 Erlangen, Germany
| |
Collapse
|
3
|
Kumar D, Gupta S, Gupta V, Tanwar R, Chandel A. Engineering the Future of Regenerative Medicines in Gut Health with Stem Cell-Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10893-w. [PMID: 40380985 DOI: 10.1007/s12015-025-10893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The advent of intestinal organoids, three-dimensional structures derived from stem cells, has significantly advanced the field of biology by providing robust in vitro models that closely mimic the architecture and functionality of the human intestine. These organoids, generated from induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), or adult stem cells, possess remarkable capabilities for self-renewal, differentiation into diverse intestinal cell types, and functional recapitulation of physiological processes, including nutrient absorption, epithelial barrier integrity, and host-microbe interactions. The utility of intestinal organoids has been extensively demonstrated in disease modeling, drug screening, and personalized medicine. Notable examples include iPSC-derived organoids, which have been effectively employed to model enteric infections, and ESC-derived organoids, which have provided critical insights into fetal intestinal development. Patient-derived organoids have emerged as powerful tools for investigating personalized therapeutics and regenerative interventions for conditions such as inflammatory bowel disease (IBD), cystic fibrosis, and colorectal cancer. Preclinical studies involving transplantation of human intestinal organoids into murine models have shown promising outcomes, including functional integration, epithelial restoration, and immune system interactions. Despite these advancements, several challenges persist, particularly in achieving reproducibility, scalability, and maturation of organoids, which hinder their widespread clinical translation. Addressing these limitations requires the establishment of standardized protocols for organoid generation, culture, storage, and analysis to ensure reproducibility and comparability of findings across studies. Nevertheless, intestinal organoids hold immense promise for transforming our understanding of gastrointestinal pathophysiology, enhancing drug development pipelines, and advancing personalized medicine. By bridging the gap between preclinical research and clinical applications, these organoids represent a paradigm shift in the exploration of novel therapeutic strategies and the investigation of gut-associated diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India.
| | - Sonia Gupta
- Swami Devi Dyal Group of Professional Institute, Panchkula, India
| | - Vrinda Gupta
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Rajni Tanwar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Anchal Chandel
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
4
|
Cai H, Tian C, Chen L, Yang Y, Sun AX, McCracken K, Tchieu J, Gu M, Mackie K, Guo F. Vascular network-inspired diffusible scaffolds for engineering functional midbrain organoids. Cell Stem Cell 2025; 32:824-837.e5. [PMID: 40101722 DOI: 10.1016/j.stem.2025.02.010] [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: 08/27/2024] [Revised: 01/03/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025]
Abstract
Organoids, 3D organ-like tissue cultures derived from stem cells, show promising potential for developmental biology, drug discovery, and regenerative medicine. However, the function and phenotype of current organoids, especially neural organoids, are still limited by insufficient diffusion of oxygen, nutrients, metabolites, signaling molecules, and drugs. Herein, we present vascular network-inspired diffusible (VID) scaffolds to mimic physiological diffusion physics for generating functional organoids and phenotyping their drug response. Specifically, the VID scaffolds, 3D-printed meshed tubular channel networks, successfully engineer human midbrain organoids almost without necrosis and hypoxia in commonly used well plates. Compared with conventional organoids, these engineered organoids develop more physiologically relevant features and functions, including midbrain-specific identity, oxygen metabolism, neuronal maturation, and network activity. Moreover, these engineered organoids also better recapitulate pharmacological responses, such as neural activity changes to fentanyl exposure, compared with conventional organoids with significant diffusion limits. This platform may provide insights for organoid development and therapeutic innovation.
Collapse
Affiliation(s)
- Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Chunhui Tian
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Lei Chen
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Yang Yang
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Alfred Xuyang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioral Disorders, 8 College Road, Singapore 169857, Singapore
| | - Kyle McCracken
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Jason Tchieu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Mingxia Gu
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47405, USA.
| |
Collapse
|
5
|
Sun Y, Wang X, Zhang DY, Zhang Z, Bhattarai JP, Wang Y, Park KH, Dong W, Hung YF, Yang Q, Zhang F, Rajamani K, Mu S, Kennedy BC, Hong Y, Galanaugh J, Sambangi A, Kim SH, Wheeler G, Gonçalves T, Wang Q, Geschwind DH, Kawaguchi R, Viaene AN, Helbig I, Kessler SK, Hoke A, Wang H, Xu F, Binder ZA, Isaac Chen H, Pai ELL, Stone S, Nasrallah MP, Christian KM, Fuccillo M, Toni N, Wu Z, Cheng HJ, O'Rourke DM, Ma M, Ming GL, Song H. Brain-wide neuronal circuit connectome of human glioblastoma. Nature 2025; 641:222-231. [PMID: 39821165 DOI: 10.1038/s41586-025-08634-7] [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: 12/01/2023] [Accepted: 01/10/2025] [Indexed: 01/19/2025]
Abstract
Glioblastoma (GBM) infiltrates the brain and can be synaptically innervated by neurons, which drives tumour progression1,2. Synaptic inputs onto GBM cells identified so far are largely short range and glutamatergic3,4. The extent of GBM integration into the brain-wide neuronal circuitry remains unclear. Here we applied rabies virus-mediated and herpes simplex virus-mediated trans-monosynaptic tracing5,6 to systematically investigate circuit integration of human GBM organoids transplanted into adult mice. We found that GBM cells from multiple patients rapidly integrate into diverse local and long-range neural circuits across the brain. Beyond glutamatergic inputs, we identified various neuromodulatory inputs, including synapses between basal forebrain cholinergic neurons and GBM cells. Acute acetylcholine stimulation induces long-lasting elevation of calcium oscillations and transcriptional reprogramming of GBM cells into a more motile state via the metabotropic CHRM3 receptor. CHRM3 activation promotes GBM cell motility, whereas its downregulation suppresses GBM cell motility and prolongs mouse survival. Together, these results reveal the striking capacity for human GBM cells to rapidly and robustly integrate into anatomically diverse neuronal networks of different neurotransmitter systems. Our findings further support a model in which rapid connectivity and transient activation of upstream neurons may lead to a long-lasting increase in tumour fitness.
Collapse
Affiliation(s)
- Yusha Sun
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xin Wang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Y Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Janardhan P Bhattarai
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yingqi Wang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristen H Park
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Weifan Dong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yun-Fen Hung
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Qian Yang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keerthi Rajamani
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Shang Mu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Benjamin C Kennedy
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yan Hong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jamie Galanaugh
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Abhijeet Sambangi
- Sidney Kimmel Medical College, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Sang Hoon Kim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Garrett Wheeler
- Department of Neuroscience and Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tiago Gonçalves
- Department of Neuroscience and Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qing Wang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Angela N Viaene
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha K Kessler
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huadong Wang
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fuqiang Xu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - H Isaac Chen
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Emily Ling-Lin Pai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara Stone
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - MacLean P Nasrallah
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly M Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Fuccillo
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicolas Toni
- Center for Psychiatric Neurosciences, Lausanne University Hospital, Lausanne, Switzerland
| | - Zhuhao Wu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hwai-Jong Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Minghong Ma
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Glioblastoma Translational Center of Excellence, The Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Birtele M, Lancaster M, Quadrato G. Modelling human brain development and disease with organoids. Nat Rev Mol Cell Biol 2025; 26:389-412. [PMID: 39668188 DOI: 10.1038/s41580-024-00804-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/14/2024]
Abstract
Organoids are systems derived from pluripotent stem cells at the interface between traditional monolayer cultures and in vivo animal models. The structural and functional characteristics of organoids enable the modelling of early stages of brain development in a physiologically relevant 3D environment. Moreover, organoids constitute a tool with which to analyse how individual genetic variation contributes to the susceptibility and progression of neurodevelopmental disorders. This Roadmap article describes the features of brain organoids, focusing on the neocortex, and their advantages and limitations - in comparison with other model systems - for the study of brain development, evolution and disease. We highlight avenues for enhancing the physiological relevance of brain organoids by integrating bioengineering techniques and unbiased high-throughput analyses, and discuss future applications. As organoids advance in mimicking human brain functions, we address the ethical and societal implications of this technology.
Collapse
Affiliation(s)
- Marcella Birtele
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Madeline Lancaster
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
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
|
Diao XJ, Soto C, Wang F, Wang Y, Wu YC, Mukherjee A. The potential of brain organoids in addressing the heterogeneity of synucleinopathies. Cell Mol Life Sci 2025; 82:188. [PMID: 40293500 PMCID: PMC12037466 DOI: 10.1007/s00018-025-05686-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: 09/16/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Synucleinopathies are a group of diseases characterized by neuronal and glial accumulation of α-synuclein (aSyn) linked with different clinical presentations, including Parkinson's disease (PD), Parkinson's disease with dementia (PDD), Dementia with Lewy Bodies (DLB) and Multiple system atrophy (MSA). Interestingly, the structure of the aSyn aggregates can vary across different synucleinopathies. Currently, it is unclear how the aSyn protein can aggregate into diverse structures and affect distinct cell types and various brain regions, leading to different clinical symptoms. Recent advances in induced pluripotent stem cells (iPSCs)-based brain organoids (BOs) technology provide an unprecedented opportunity to define the etiology of synucleinopathies in human brain cells within their three-dimensional (3D) context. In this review, we will summarize current advances in investigating the mechanisms of synucleinopathies using BOs and discuss the scope of this platform to define mechanisms underlining the selective vulnerability of cell types and brain regions in synucleinopathies.
Collapse
Affiliation(s)
- Xiao-Jun Diao
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fei Wang
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Abhisek Mukherjee
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
9
|
Wu SR, Nowakowski TJ. Exploring human brain development and disease using assembloids. Neuron 2025; 113:1133-1150. [PMID: 40107269 PMCID: PMC12022838 DOI: 10.1016/j.neuron.2025.02.010] [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: 05/08/2024] [Revised: 01/10/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
How the human brain develops and what goes awry in neurological disorders represent two long-lasting questions in neuroscience. Owing to the limited access to primary human brain tissue, insights into these questions have been largely gained through animal models. However, there are fundamental differences between developing mouse and human brain, and neural organoids derived from human pluripotent stem cells (hPSCs) have recently emerged as a robust experimental system that mimics self-organizing and multicellular features of early human brain development. Controlled integration of multiple organoids into assembloids has begun to unravel principles of cell-cell interactions. Moreover, patient-derived or genetically engineered hPSCs provide opportunities to investigate phenotypic correlates of neurodevelopmental disorders and to develop therapeutic hypotheses. Here, we outline the advances in technologies that facilitate studies by using assembloids and summarize their applications in brain development and disease modeling. Lastly, we discuss the major roadblocks of the current system and potential solutions.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
10
|
Maisumu G, Willerth S, Nestor M, Waldau B, Schülke S, Nardi FV, Ahmed O, Zhou Y, Durens M, Liang B, Yakoub AM. Brain organoids: building higher-order complexity and neural circuitry models. Trends Biotechnol 2025:S0167-7799(25)00046-0. [PMID: 40221251 DOI: 10.1016/j.tibtech.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/09/2024] [Accepted: 02/07/2025] [Indexed: 04/14/2025]
Abstract
Brain organoids are 3D tissue models of the human brain that are derived from pluripotent stem cells (PSCs). They have enabled studies that were previously stymied by the inaccessibility of human brain tissue or the limitations of mouse models of some brain diseases. Despite their enormous potential, brain organoids have had significant limitations that prevented them from recapitulating the full complexity of the human brain and reduced their utility in disease studies. We describe recent progress in addressing these limitations, especially building complex organoids that recapitulate the interactions between multiple brain regions, and reconstructing in vitro the neural circuitry present in in vivo. These major advances in the human brain organoid technology will remarkably facilitate brain disease modeling and neuroscience research.
Collapse
Affiliation(s)
- Gulimiheranmu Maisumu
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA; Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - Stephanie Willerth
- Department of Biomedical Engineering, University of Victoria, Victoria, BC, Canada
| | - Michael Nestor
- National Academies of Sciences, Engineering, and Medicine, Washington, DC, USA
| | - Ben Waldau
- Department of Neurological Surgery, University of California Davis, Sacramento, CA, USA
| | - Stefan Schülke
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany; Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Francesco V Nardi
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA; Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - Osama Ahmed
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA; Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - You Zhou
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA
| | - Madel Durens
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bo Liang
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - Abraam M Yakoub
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
11
|
Liu X, Zhou Z, Zhang Y, Zhong H, Cai X, Guan R. Recent progress on the organoids: Techniques, advantages and applications. Biomed Pharmacother 2025; 185:117942. [PMID: 40043462 DOI: 10.1016/j.biopha.2025.117942] [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: 12/11/2024] [Revised: 01/30/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Organoids are a cutting-edge technology in the life sciences field, with applications in precision medicine, bionic organs, and toxicological evaluations of chemicals. Their 3D structure closely resembles that of real organs, allowing more accurate functional mimicry. The 3D organoid culture system can simulate the growth state of cells in vivo and establish a suspension culture system for organoid 3D culture by using scaffold-less or scaffold technology to avoid direct contact between cells and plastic culture vessels. Furthermore, organoids can simulate the pathophysiological state of tissues and organs in vitro. This paper primarily discusses the construction methodologies, as well as the advantages and disadvantages of 3D culture systems for both scaffold-free organoids and scaffolded organoids. This review also summarizes the application of organoid models in chemical toxicology evaluation, drug screening and functional evaluation, establishment of in vitro disease models, and research on disease occurrence and potential mechanisms. The aim is to provide a reference for the research and practical applications of organoid-related scientific fields.
Collapse
Affiliation(s)
- Xiaofeng Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Zhiyuan Zhou
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yao Zhang
- Zhejiang Provincial Key Lab for Chem and Bio Processing Technology of Farm Produces, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiulei Cai
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Moganshan Institute ZJUT, Kangqian District, Deqing 313200, China.
| |
Collapse
|
12
|
Rao C, Semrau S, Fossati V. Decoding microglial functions in Alzheimer's disease: insights from human models. Trends Immunol 2025; 46:310-323. [PMID: 40113535 PMCID: PMC11993344 DOI: 10.1016/j.it.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Microglia, key orchestrators of the brain's immune responses, play a pivotal role in the progression of Alzheimer's disease (AD). Emerging human models, including stem cell-derived microglia and cerebral organoids, are transforming our understanding of microglial contributions to AD pathology. In this review, we highlight how these models have uncovered human-specific microglial responses to amyloid plaques and their regulation of neuroinflammation, which are not recapitulated in animal models. We also illustrate how advanced human models that better mimic brain physiology and AD pathology are providing unprecedented insights into the multifaceted roles of microglia. These innovative approaches, combined with sophisticated technologies for cell editing and analysis, are shaping AD research and opening new avenues for therapeutic interventions targeting microglia.
Collapse
Affiliation(s)
- Chandrika Rao
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Stefan Semrau
- The New York Stem Cell Foundation Research Institute, New York, NY, USA
| | - Valentina Fossati
- The New York Stem Cell Foundation Research Institute, New York, NY, USA.
| |
Collapse
|
13
|
Ouaidat S, Bellapianta A, Ammer-Pickhardt F, Taghipour T, Bolz M, Salti A. Exploring organoid and assembloid technologies: a focus on retina and brain. Expert Rev Mol Med 2025; 27:e14. [PMID: 40145178 PMCID: PMC12011387 DOI: 10.1017/erm.2025.9] [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: 11/13/2024] [Revised: 02/21/2025] [Accepted: 03/21/2025] [Indexed: 03/28/2025]
Abstract
BACKGROUND The recent emergence of three-dimensional organoids and their utilization as in vitro disease models confirmed the complexities behind organ-specific functions and unravelled the importance of establishing suitable human models for various applications. Also, in light of persistent challenges associated with their use, researchers have been striving to establish more advanced structures (i.e. assembloids) that can help address the limitations presented in the current organoids. METHODS In this review, we discuss the distinct organoid types that are available to date, with a special focus on retinal and brain organoids, and highlight their importance in disease modelling. RESULTS We refer to published research to explore the extent to which retinal and brain organoids can serve as potential alternatives to organ/cell transplants and direct our attention to the topic of photostimulation in retinal organoids. Additionally, we discuss the advantages of incorporating microfluidics and organ-on-a-chip devices for boosting retinal organoid performance. The challenges of organoids leading to the subsequent development of assembloid fusion models are also presented. CONCLUSION In conclusion, organoid technology has laid the foundation for generating upgraded models that not only better replicate in vivo systems but also allow for a deeper comprehension of disease pathophysiology.
Collapse
Affiliation(s)
- Sara Ouaidat
- Research Group Cellular and Molecular Ophthalmology, University Clinic for Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Alessandro Bellapianta
- Research Group Cellular and Molecular Ophthalmology, University Clinic for Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Franziska Ammer-Pickhardt
- Research Group Cellular and Molecular Ophthalmology, University Clinic for Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
- Department of Biosciences & Medical Biology, Paris-Lodron-University of Salzburg (PLUS), Salzburg, Austria
| | - Tara Taghipour
- Research Group Cellular and Molecular Ophthalmology, University Clinic for Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Matthias Bolz
- Research Group Cellular and Molecular Ophthalmology, University Clinic for Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Ahmad Salti
- Research Group Cellular and Molecular Ophthalmology, University Clinic for Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| |
Collapse
|
14
|
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
|
15
|
Duenki T, Ikeuchi Y. Insulative Compression of Neuronal Tissues on Microelectrode Arrays by Perfluorodecalin Enhances Electrophysiological Measurements. Adv Healthc Mater 2025; 14:e2403771. [PMID: 39757474 PMCID: PMC11874680 DOI: 10.1002/adhm.202403771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Indexed: 01/07/2025]
Abstract
Microelectrode array (MEA) techniques provide a powerful method for exploration of neural network dynamics. A critical challenge is to interface 3D neural tissues including neural organoids with the flat MEAs surface, as it is essential to place neurons near to the electrodes for recording weak extracellular signals of neurons. To enhance performance of MEAs, most research have focused on improving their surface treatment, while little attention has been given to improve the tissue-MEA interactions from the medium side. Here, a strategy is introduced to augment MEA measurements by overlaying perfluorodecalin (PFD), a biocompatible fluorinated solvent, over neural tissues. Laying PFD over cerebral organoids insulates and compresses the tissues on MEA, which significantly enhances electrophysiological recordings. Even subtle signals such as the propagation of action potentials in bundled axons of motor nerve organoids can be detected with the technique. Moreover, PFD stabilizes tissues in acute recordings and its transparency allows optogenetic manipulations. This research highlights the potential of PFD as a tool for refining electrophysiological measurements of in vitro neuronal cultures. This can open new avenues to leverage precision of neuroscientific investigations and expanding the toolkit for in vitro studies of neural function and connectivity.
Collapse
Affiliation(s)
- Tomoya Duenki
- Institute of Industrial ScienceThe University of TokyoMeguroTokyo153‐8505Japan
- Institute for AI and BeyondThe University of TokyoBunkyoTokyo113‐8655Japan
- Department of Chemistry and BiotechnologyThe University of TokyoBunkyoTokyo113‐8655Japan
- LIMMSCNRS‐Institute of Industrial ScienceThe University of TokyoIRL 2820MeguroTokyo153‐8505Japan
| | - Yoshiho Ikeuchi
- Institute of Industrial ScienceThe University of TokyoMeguroTokyo153‐8505Japan
- Institute for AI and BeyondThe University of TokyoBunkyoTokyo113‐8655Japan
- Department of Chemistry and BiotechnologyThe University of TokyoBunkyoTokyo113‐8655Japan
- LIMMSCNRS‐Institute of Industrial ScienceThe University of TokyoIRL 2820MeguroTokyo153‐8505Japan
| |
Collapse
|
16
|
Gu B, Ma Q, Li J, Xu W, Xie Y, Lu P, Yu K, Huo Z, Li X, Peng J, Jiang Y, Li D, He J. Multi-material Electrohydrodynamic Printing of Bioelectronics with Sub-Microscale 3D Gold Pillars for In Vitro Extra- and Intra-Cellular Electrophysiological Recordings. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407969. [PMID: 39792774 PMCID: PMC11884540 DOI: 10.1002/advs.202407969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/16/2024] [Indexed: 01/12/2025]
Abstract
Micro/nanoscale 3D bioelectrodes gain increasing interest for electrophysiological recording of electroactive cells. Although 3D printing has shown promise to flexibly fabricate 3D bioelectronics compared with conventional microfabrication, relatively-low resolution limits the printed bioelectrode for high-quality signal monitoring. Here, a novel multi-material electrohydrodynamic printing (EHDP) strategy is proposed to fabricate bioelectronics with sub-microscale 3D gold pillars for in vitro electrophysiological recordings. EHDP is employed to fabricate conductive circuits for signal transmission, which are passivated by polyimide via extrusion-based printing. Laser-assisted EHDP is developed to produce 3D gold pillars featuring a diameter of 0.64 ± 0.04 µm. The 3D gold pillars demonstrate stable conductivity under the cell-culture environment. Living cells can conformally grow onto these sub-microscale 3D pillars with a height below 5 µm, which facilitates the highly-sensitive recording of extracellular signals with amplitudes <15 µV. The 3D pillars can apply electroporation currents to reversibly open the cellular membrane for intracellular recording, facilitating the measurement of subtle cellular electrophysiological activities. As a proof-of-concept demonstration, fully-printed chips with multiple culturing chambers and sensing bioelectronics are fabricated for zone-specific electrophysiological recording in drug testing. The proposed multi-material EHDP strategy enables rapid prototyping of organ-on-a-chip systems with 3D bioelectronics for high-quality electrophysiological recordings.
Collapse
Affiliation(s)
- Bingsong Gu
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Qihang Ma
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Jiaxin Li
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Wangkai Xu
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Yuke Xie
- Laboratory of Neurological Diseases and Brain FunctionThe Affiliated Hospital of Southwest Medical UniversityLuzhou64600P. R. China
| | - Peng Lu
- Laboratory of Neurological Diseases and Brain FunctionThe Affiliated Hospital of Southwest Medical UniversityLuzhou64600P. R. China
| | - Kun Yu
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Ziyao Huo
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Xiao Li
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Jianhua Peng
- Laboratory of Neurological Diseases and Brain FunctionThe Affiliated Hospital of Southwest Medical UniversityLuzhou64600P. R. China
| | - Yong Jiang
- Laboratory of Neurological Diseases and Brain FunctionThe Affiliated Hospital of Southwest Medical UniversityLuzhou64600P. R. China
| | - Dichen Li
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems EngineeringXi'an Jiaotong UniversityXi'an710049P. R. China
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical DevicesXi'an Jiaotong UniversityXi'an710049P. R. China
- State Industry‐Education Integration Center for Medical InnovationsXi'an Jiaotong UniversityXi'an710049P. R. China
| |
Collapse
|
17
|
Gu J, Liu F, Li L, Mao J. Advances and Challenges in Modeling Autosomal Dominant Polycystic Kidney Disease: A Focus on Kidney Organoids. Biomedicines 2025; 13:523. [PMID: 40002937 PMCID: PMC11852630 DOI: 10.3390/biomedicines13020523] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a prevalent hereditary disorder characterized by distinct phenotypic variability that has posed challenges for advancing in-depth research. Recent advancements in kidney organoid construction technologies have enabled researchers to simulate kidney development and create simplified in vitro experimental environments, allowing for more direct observation of how genetic mutations drive pathological phenotypes and disrupt physiological functions. Emerging technologies, such as microfluidic bioreactor culture systems and single-cell transcriptomics, have further supported the development of complex ADPKD organoids, offering robust models for exploring disease mechanisms and facilitating drug discovery. Nevertheless, significant challenges remain in constructing more accurate ADPKD disease models. This review will summarize recent advances in ADPKD organoid construction, focusing on the limitations of the current techniques and the critical issues that need to be addressed for future breakthroughs. New and Noteworthy: This review presents recent advancements in ADPKD organoid construction, particularly iPSC-derived models, offering new insights into disease mechanisms and drug discovery. It focuses on challenges such as limited vascularization and maturity, proposing potential solutions through emerging technologies. The ongoing optimization of ADPKD organoid models is expected to enhance understanding of the disease and drive breakthroughs in disease mechanisms and targeted therapy development.
Collapse
Affiliation(s)
| | | | | | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310058, China; (J.G.); (F.L.); (L.L.)
| |
Collapse
|
18
|
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
|
19
|
Dony L, Krontira AC, Kaspar L, Ahmad R, Demirel IS, Grochowicz M, Schäfer T, Begum F, Sportelli V, Raimundo C, Koedel M, Labeur M, Cappello S, Theis FJ, Cruceanu C, Binder EB. Chronic exposure to glucocorticoids amplifies inhibitory neuron cell fate during human neurodevelopment in organoids. SCIENCE ADVANCES 2025; 11:eadn8631. [PMID: 39951527 PMCID: PMC11827642 DOI: 10.1126/sciadv.adn8631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Disruptions in the tightly regulated process of human brain development have been linked to increased risk for brain and mental illnesses. While the genetic contribution to these diseases is well established, important environmental factors have been less studied at molecular and cellular levels. Here, we used single-cell and cell type-specific techniques to investigate the effect of glucocorticoid (GC) exposure, a mediator of antenatal environmental risk, on gene regulation and lineage specification in unguided human neural organoids. We characterized the transcriptional response to chronic GC exposure during neural differentiation and studied the underlying gene regulatory networks by integrating single-cell transcriptomics with chromatin accessibility data. We found lasting cell type-specific changes that included autism risk genes and several transcription factors associated with neurodevelopment. Chronic GC exposure influenced lineage specification primarily by priming the inhibitory neuron lineage through transcription factors like PBX3. We provide evidence for convergence of genetic and environmental risk factors through a common mechanism of altering lineage specification.
Collapse
Affiliation(s)
- Leander Dony
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
| | - Anthi C. Krontira
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Lea Kaspar
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Ruhel Ahmad
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Ilknur Safak Demirel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Tim Schäfer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Fatema Begum
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Vincenza Sportelli
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
| | - Catarina Raimundo
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Maik Koedel
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Marta Labeur
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Silvia Cappello
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- Physiological Genomics, Biomedical Center (BMC), LMU Munich Faculty of Medicine, 82152 Planegg-Martinsried, Germany
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, 85748 Garching bei München, Germany
| | - Cristiana Cruceanu
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elisabeth B. Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
20
|
Kalla J, Pfneissl J, Mair T, Tran L, Egger G. A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cell Oncol (Dordr) 2025; 48:1-26. [PMID: 38806997 PMCID: PMC11850459 DOI: 10.1007/s13402-024-00960-8] [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] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.
Collapse
Affiliation(s)
- Jessica Kalla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
21
|
Sun D, Zhang K, Zheng F, Yang G, Yang M, Xu Y, Qin Y, Lin M, Li Y, Tan J, Li Q, Qu X, Li G, Bian L, Zhu C. Matrix Viscoelasticity Controls Differentiation of Human Blood Vessel Organoids into Arterioles and Promotes Neovascularization in Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2410802. [PMID: 39686788 DOI: 10.1002/adma.202410802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Stem cell-derived blood vessel organoids are embedded in extracellular matrices to stimulate vessel sprouting. Although vascular organoids in 3D collagen I-Matrigel gels are currently available, they are primarily capillaries composed of endothelial cells (ECs), pericytes, and mesenchymal stem-like cells, which necessitate mature arteriole differentiation for neovascularization. In this context, the hypothesis that matrix viscoelasticity regulates vascular development is investigated in 3D cultures by encapsulating blood vessel organoids within viscoelastic gelatin/β-CD assembly dynamic hydrogels or methacryloyl gelatin non-dynamic hydrogels. The vascular organoids within the dynamic hydrogel demonstrate enhanced angiogenesis and differentiation into arterioles containing smooth muscle cells. The dynamic hydrogel mechanical microenvironment promotes vascular patterning and arteriolar differentiation by elevating notch receptor 3 signaling in mesenchymal stem cells and downregulating platelet-derived growth factor B expression in ECs. Transplantation of vascular organoids in vivo, along with the dynamic hydrogel, leads to the reassembly of arterioles and restoration of cardiac function in infarcted hearts. These findings indicate that the viscoelastic properties of the matrix play a crucial role in controlling the vascular organization and differentiation processes, suggesting an exciting potential for its application in regenerative medicine.
Collapse
Affiliation(s)
- Dayu Sun
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Feiyang Zheng
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Guanyuan Yang
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Mingcan Yang
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Youqian Xu
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Yinhua Qin
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Mingxin Lin
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yanzhao Li
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Ju Tan
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Qiyu Li
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Xiaohang Qu
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Gang Li
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chuhong Zhu
- Department of Anatomy, Engineering Research Center of the Ministry of Education for Tissue and Organ Regeneration and Manufacturing, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Third Military Medical University, Chongqing, 400038, P. R. China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, P. R. China
| |
Collapse
|
22
|
Dawoody Nejad L, Pioro EP. Modeling ALS with Patient-Derived iPSCs: Recent Advances and Future Potentials. Brain Sci 2025; 15:134. [PMID: 40002468 PMCID: PMC11852857 DOI: 10.3390/brainsci15020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a terminal complex neurodegenerative disease, with 10-15% of cases being familial and the majority being sporadic with no known cause. There are no animal models for the 85-90% of sporadic ALS cases. More creative, sophisticated models of ALS disease are required to unravel the mysteries of this complicated disease. While ALS patients urgently require new medications and treatments, suitable preclinical in vitro models for drug screening are lacking. Therefore, human-derived induced pluripotent stem cell (hiPSC) technology offers the opportunity to model diverse and unreachable cell types in a culture dish. In this review, we focus on recent hiPSC-derived ALS neuronal and non-neuronal models to examine the research progress of current ALS 2D monocultures, co-cultures, and more complex 3D-model organoids. Despite the challenges inherent to hiPSC-based models, their application to preclinical drug studies is enormous.
Collapse
Affiliation(s)
| | - Erik P. Pioro
- Djavad Mowafaghian Centre for Brain Health, Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| |
Collapse
|
23
|
Schickel E, Bender T, Kaysan L, Hufgard S, Mayer M, Grosshans DR, Thielemann C, Schroeder IS. Human cerebral organoids model tumor infiltration and migration supported by astrocytes in an autologous setting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635456. [PMID: 39974912 PMCID: PMC11838324 DOI: 10.1101/2025.01.29.635456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Efforts to achieve precise and efficient tumor targeting of highly malignant brain tumors are constrained by the dearth of appropriate models to study the effects and potential side effects of radiation, chemotherapy, and immunotherapy on the most complex human organ, the brain. We established a cerebral organoid model of brain tumorigenesis in an autologous setting by overexpressing c-MYC as one of the most common oncogenes in brain tumors. GFP + /c-MYC high cells were isolated from tumor organoids and used in two different culture approaches: assembloids comprising of a normal cerebral organoid with a GFP + /c-MYC high tumor sphere and co-culture of cerebral organoid slices at air-liquid interface with GFP + /c-MYC high cells. GFP + /c-MYC high cells used in both approaches exhibited tumor-like properties, including overexpression of the c-MYC oncogene, high proliferative and invasive potential, and an immature phenotype as evidenced by increased expression of Ki-67, VIM, and CD133. Organoids and organoid slices served as suitable scaffolds for infiltrating tumor-like cells. Using our highly reproducible and powerful model system that allows long-term culture, we demonstrated that the migratory and infiltrative potential of tumor-like cells is shaped by the environment in which glia cells provide support to tumor-like cells.
Collapse
|
24
|
Pagliaro A, Artegiani B, Hendriks D. Emerging approaches to enhance human brain organoid physiology. Trends Cell Biol 2025:S0962-8924(24)00254-X. [PMID: 39826996 DOI: 10.1016/j.tcb.2024.12.001] [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: 08/08/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Brain organoids are important 3D models for studying human brain development, disease, and evolution. To overcome some of the existing limitations that affect organoid quality, reproducibility, characteristics, and in vivo resemblance, current efforts are directed to improve their physiological relevance by exploring different, yet interconnected, routes. In this review, these approaches and their latest developments are discussed, including stem cell optimization, refining morphogen administration strategies, altering the extracellular matrix (ECM) niche, and manipulating tissue architecture to mimic in vivo brain morphogenesis. Additionally, strategies to increase cell diversity and enhance organoid maturation, such as establishing co-cultures, assembloids, and organoid in vivo xenotransplantation, are reviewed. We explore how these various factors can be tuned and intermingled and speculate on future avenues towards even more physiologically-advanced brain organoids.
Collapse
Affiliation(s)
- Anna Pagliaro
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
25
|
Sun J, Ahmed I, Brown J, Khosrotehrani K, Shafiee A. The empowering influence of air-liquid interface culture on skin organoid hair follicle development. BURNS & TRAUMA 2025; 13:tkae070. [PMID: 39822647 PMCID: PMC11736897 DOI: 10.1093/burnst/tkae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 01/19/2025]
Abstract
Background Rodent models have been widely used to investigate skin development, but do not account for significant differences in composition compared to human skin. On the other hand, two-dimensional and three-dimensional engineered skin models still lack the complex features of human skin such as appendages and pigmentation. Recently, hair follicle containing skin organoids (SKOs) with a stratified epidermis, and dermis layer have been generated as floating spheres from human-induced pluripotent stem cells (hiPSCs). Methods The current study aims to investigate the generation of hiPSCs-derived SKOs using an air-liquid interface (ALI) model on transwell membranes (T-SKOs) and compares their development with conventional floating culture in low-attachment plates (F-SKOs). Results Mature SKOs containing an epidermis, dermis, and appendages are created in both T-SKO and F-SKO conditions. It was found that the hair follicles are smaller and shorter in the F-SKO compared with T-SKOs. Additionally, the ALI conditions contribute to enhanced hair follicle numbers than conventional floating culture. Conclusions Together, this study demonstrates the significant influence of transwell culture on the morphogenesis of hair follicles within SKOs and highlights the potential for refinement of skin model engineering for advancing dermatology and skin research.
Collapse
Affiliation(s)
- Jane Sun
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
| | - Imaan Ahmed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
| | - Jason Brown
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
| | - Kiarash Khosrotehrani
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
| | - Abbas Shafiee
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4102Australia
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Queensland Health, Brisbane, QLD, 4029Australia
| |
Collapse
|
26
|
Chen X, Sun G, Feng L, Tian E, Shi Y. Human iPSC-derived microglial cells protect neurons from neurodegeneration in long-term cultured adhesion brain organoids. Commun Biol 2025; 8:30. [PMID: 39789340 PMCID: PMC11718079 DOI: 10.1038/s42003-024-07401-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Brain organoid models have greatly facilitated our understanding of human brain development and disease. However, key brain cell types, such as microglia, are lacking in most brain organoid models. Because microglia have been shown to play important roles in brain development and pathologies, attempts have been made to add microglia to brain organoids through co-culture. However, only short-term microglia-organoid co-cultures can be established, and it remains challenging to have long-lasting survival of microglia in organoids to mimic long-term residency of microglia in the brain. In this study, we developed an adhesion brain organoid (ABO) platform that allows prolonged culture of brain organoids (greater than a year). Moreover, the long-term (LT)-ABO system contains abundant astrocytes and can support prolonged survival and ramification of microglia. Furthermore, we showed that microglia in the LT-ABO could protect neurons from neurodegeneration by increasing synaptic density and reducing p-Tau level and cell death in the LT-ABO. Therefore, the microglia-containing LT-ABO platform generated in this study provides a promising human cellular model for studying neuron-glia and glia-glia interactions in brain development and the pathogenesis of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Xianwei Chen
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Guoqiang Sun
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Lizhao Feng
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - E Tian
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| |
Collapse
|
27
|
van der Molen T, Spaeth A, Chini M, Hernandez S, Kaurala GA, Schweiger HE, Duncan C, McKenna S, Geng J, Lim M, Bartram J, Dendukuri A, Zhang Z, Gonzalez-Ferrer J, Bhaskaran-Nair K, Blauvelt LJ, Harder CR, Petzold LR, Alam El Din DM, Laird J, Schenke M, Smirnova L, Colquitt BM, Mostajo-Radji MA, Hansma PK, Teodorescu M, Hierlemann A, Hengen KB, Hanganu-Opatz IL, Kosik KS, Sharf T. Protosequences in brain organoids model intrinsic brain states Authors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.29.573646. [PMID: 38234832 PMCID: PMC10793448 DOI: 10.1101/2023.12.29.573646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Neuronal firing sequences are thought to be the basic building blocks of neural coding and information broadcasting within the brain. However, when sequences emerge during neurodevelopment remains unknown. We demonstrate that structured firing sequences are present in spontaneous activity of human and murine brain organoids and ex vivo neonatal brain slices from the murine somatosensory cortex. We observed a balance between temporally rigid and flexible firing patterns that are emergent phenomena in human and murine brain organoids and early postnatal murine somatosensory cortex, but not in primary dissociated cortical cultures. Our findings suggest that temporal sequences do not arise in an experience-dependent manner, but are rather constrained by an innate preconfigured architecture established during neurogenesis. These findings highlight the potential for brain organoids to further explore how exogenous inputs can be used to refine neuronal circuits and enable new studies into the genetic mechanisms that govern assembly of functional circuitry during early human brain development.
Collapse
Affiliation(s)
- Tjitse van der Molen
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Alex Spaeth
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mattia Chini
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Sebastian Hernandez
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gregory A. Kaurala
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Cole Duncan
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sawyer McKenna
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jinghui Geng
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Max Lim
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Julian Bartram
- Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland
| | - Aditya Dendukuri
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Zongren Zhang
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106
| | - Jesus Gonzalez-Ferrer
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Kiran Bhaskaran-Nair
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Lon J. Blauvelt
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
| | - Cole R.K. Harder
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
| | - Linda R. Petzold
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Dowlette-Mary Alam El Din
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jason Laird
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Maren Schenke
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bradley M. Colquitt
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA 95064, USA
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - Paul K. Hansma
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Physics, University of California Santa Barbara, Santa Barbara, CA 93106
| | - Mircea Teodorescu
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zürich, Klingelbergstrasse 48, 4056 Basel, Switzerland
| | - Keith B. Hengen
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Ileana L. Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Kenneth S. Kosik
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Tal Sharf
- UC Santa Cruz Genomics Institute, University of California Santa Cruz, Santa Cruz, CA 95060, USA
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
- Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
28
|
Yin Y, Zhou W, Zhu J, Chen Z, Jiang L, Zhuang X, Chen J, Wei J, Lu X, Liu Y, Pang W, Zhang Q, Cao Y, Li Z, Zhu Y, Xiang Y. Generation of self-organized neuromusculoskeletal tri-tissue organoids from human pluripotent stem cells. Cell Stem Cell 2025; 32:157-171.e8. [PMID: 39657678 DOI: 10.1016/j.stem.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/26/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024]
Abstract
The human body function requires crosstalk between different tissues. An essential crosstalk is in the neuromusculoskeletal (NMS) axis involving neural, muscular, and skeletal tissues, which is challenging to model using human cells. Here, we describe the generation of three-dimensional, NMS tri-tissue organoids (hNMSOs) from human pluripotent stem cells through a co-development strategy. Staining, single-nucleus RNA sequencing, and spatial transcriptome profiling revealed the co-emergence and self-organization of neural, muscular, and skeletal lineages within individual organoids, and the neural domains of hNMSOs obtained a ventral-specific identity and produced motor neurons innervating skeletal muscles. The neural, muscular, and skeletal regions of hNMSOs exhibited maturation and established functional connections during development. Notably, structural, functional, and transcriptomic analyses revealed that skeletal support in hNMSOs benefited human muscular development. Modeling with hNMSOs also unveiled the neuromuscular alterations following pathological skeletal degeneration. Together, our study provides an accessible experimental model for future studies of human NMS crosstalk and abnormality.
Collapse
Affiliation(s)
- Yao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinkui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziling Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jia Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoxiang Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yantong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Pang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qinzhi Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yajing Cao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhuoya Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuyan Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
29
|
Taurin S, Alzahrani R, Aloraibi S, Ashi L, Alharmi R, Hassani N. Patient-derived tumor organoids: A preclinical platform for personalized cancer therapy. Transl Oncol 2025; 51:102226. [PMID: 39622151 DOI: 10.1016/j.tranon.2024.102226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/11/2024] Open
Abstract
Patient-derived tumor organoids (PDTOs) represent a significant advancement in cancer research and personalized medicine. These organoids, derived from various cancer types, have shown the ability to retain the genetic and molecular characteristics of the original tumors, allowing for the detailed study of tumor biology and drug responses on an individual basis. The success rates of establishing PDTOs vary widely and are influenced by factors such as cancer type, tissue quality, and media composition. Furthermore, the dynamic nature of organoid cultures may also lead to unique molecular characteristics that deviate from the original tumors, affecting their interpretation in clinical settings without the implementation of rigorous validation and establishment of standardized protocols. Recent studies have supported the correlation between PDTOs and the corresponding patient response. Although these studies involved a small number of patients, they promoted the integration of PDTOs in observational and interventional clinical trials to advance translational cancer therapies.
Collapse
Affiliation(s)
- Sebastien Taurin
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain.
| | - Reem Alzahrani
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Sahar Aloraibi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Layal Ashi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Rawan Alharmi
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| | - Noora Hassani
- Department of Molecular Medicine, College of Medicine and Health Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
30
|
Carnicer‐Lombarte A, Malliaras GG, Barone DG. The Future of Biohybrid Regenerative Bioelectronics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2408308. [PMID: 39564751 PMCID: PMC11756040 DOI: 10.1002/adma.202408308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/13/2024] [Indexed: 11/21/2024]
Abstract
Biohybrid regenerative bioelectronics are an emerging technology combining implantable devices with cell transplantation. Once implanted, biohybrid regenerative devices integrate with host tissue. The combination of transplant and device provides an avenue to both replace damaged or dysfunctional tissue, and monitor or control its function with high precision. While early challenges in the fusion of the biological and technological components limited development of biohybrid regenerative technologies, progress in the field has resulted in a rapidly increasing number of applications. In this perspective the great potential of this emerging technology for the delivery of therapy is discussed, including both recent research progress and potential new directions. Then the technology barriers are discussed that will need to be addressed to unlock the full potential of biohybrid regenerative devices.
Collapse
Affiliation(s)
| | - George G. Malliaras
- Department of EngineeringElectrical Engineering DivisionUniversity of CambridgeCambridgeCB3 0FAUK
| | - Damiano G. Barone
- Department of EngineeringElectrical Engineering DivisionUniversity of CambridgeCambridgeCB3 0FAUK
- Department of Neurosurgery, Houston MethodistHouston77030USA
- Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0QQUK
| |
Collapse
|
31
|
Abdulla A, Yan H, Chen S, Wu L, Chen XS, Zhang Y, Zhang M, Zhuang TY, Ahmad KZ, Lin J, Ding X, Jiang L. A multichannel microfluidic device for revealing the neurotoxic effects of Bisphenol S on cerebral organoids under low-dose constant exposure. Biosens Bioelectron 2025; 267:116754. [PMID: 39332252 DOI: 10.1016/j.bios.2024.116754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/20/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024]
Abstract
Bisphenol S is a widely used plasticizer in manufacturing daily supplies, while little was known about its adverse effect on human health, especially on fetal brain development. Due to the complexity and subtlety of the brain, it remains challenging to reveal the hazardous effects of environmental pollution on human fetal brain development. Taking advantage of stem cell application, cerebral organoids generated from stem cells are becoming powerful tools for understanding brain development and drug toxicity testing models. Here, we developed a microfluidic chip for cerebral organoid culturing to reveal the neurotoxicity of low-dose constant BPS exposure on cerebral organoids. The organoids in our microfluidic system could be continuously cultured for 34 days and expressed all the essential properties of the cerebral organoids. Exposure to BPS was initiated from day 20 for concessive two weeks. The neurotoxic effects were evaluated by immunofluorescence staining and proteomics, and verified by quantitative real-time PCR. Our results indicated BPS exposure would inhibit neuron differentiation, hinder the Wnt signaling pathway, and cause alteration of signaling molecule expressions in brain regionalization. Even exposure to a low dose of BPS constantly might cause neurotoxicity during fetal brain development. Altogether, the multichannel microfluidic chip offers a general platform technique to reveal the effects of different hazardous chemicals on cerebral organoids.
Collapse
Affiliation(s)
- Aynur Abdulla
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Haoni Yan
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Shujin Chen
- Ministry of Education, Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Leqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xu-Sen Chen
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yizhi Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Manlin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Tsz Yui Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Khan Zara Ahmad
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institute, Sweden
| | - Jinjin Lin
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China; School of Psychology, Shaanxi Normal University, Xi' an 710062, Shaanxi Province, China
| | - Xianting Ding
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China; State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200092, China.
| |
Collapse
|
32
|
Stone ML, Lee HH, Levine EM. Agarose hydrogel-mediated electroporation method for retinal tissue cultured at the air-liquid interface. iScience 2024; 27:111299. [PMID: 39628577 PMCID: PMC11612790 DOI: 10.1016/j.isci.2024.111299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/29/2024] [Accepted: 10/29/2024] [Indexed: 12/06/2024] Open
Abstract
It is advantageous to culture the ex vivo retina and other tissues at the air-liquid interface to allow for more efficient gas exchange. However, gene delivery to these cultures can be challenging. Electroporation is a fast and robust method of gene delivery, but typically requires submergence in liquid buffer for electrical current flow. We have developed a submergence-free electroporation technique that incorporates an agarose hydrogel disk between the positive electrode and retina. Inner retinal neurons and Müller glia are transfected with increased propensity toward Müller glia transfection after extended time in culture. We also observed an increase in BrdU incorporation in Müller glia following electrical stimulation, and variation in detection of transfected cells from expression vectors with different promoters. This method advances our ability to use ex vivo retinal tissue for genetic studies and should be adaptable for other tissues cultured at an air-liquid interface.
Collapse
Affiliation(s)
- Megan L. Stone
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville TN 37232, USA
| | - Hannah H. Lee
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville TN 37232, USA
| | - Edward M. Levine
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville TN 37232, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville TN 37232, USA
| |
Collapse
|
33
|
Voitiuk K, Seiler ST, Pessoa de Melo M, Geng J, van der Molen T, Hernandez S, Schweiger HE, Sevetson JL, Parks DF, Robbins A, Torres-Montoya S, Ehrlich D, Elliott MAT, Sharf T, Haussler D, Mostajo-Radji MA, Salama SR, Teodorescu M. A feedback-driven brain organoid platform enables automated maintenance and high-resolution neural activity monitoring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585237. [PMID: 38559212 PMCID: PMC10979982 DOI: 10.1101/2024.03.15.585237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The analysis of tissue cultures, particularly brain organoids, requires a sophisticated integration and coordination of multiple technologies for monitoring and measuring. We have developed an automated research platform enabling independent devices to achieve collaborative objectives for feedback-driven cell culture studies. Our approach enables continuous, communicative, non-invasive interactions within an Internet of Things (IoT) architecture among various sensing and actuation devices, achieving precisely timed control of in vitro biological experiments. The framework integrates microfluidics, electrophysiology, and imaging devices to maintain cerebral cortex organoids while measuring their neuronal activity. The organoids are cultured in custom, 3D-printed chambers affixed to commercial microelectrode arrays. Periodic feeding is achieved using programmable microfluidic pumps. We developed a computer vision fluid volume estimator used as feedback to rectify deviations in microfluidic perfusion during media feeding/aspiration cycles. We validated the system with a set of 7-day studies of mouse cerebral cortex organoids, comparing manual and automated protocols. The automated protocols were validated in maintaining robust neural activity throughout the experiment. The automated system enabled hourly electrophysiology recordings for the 7-day studies. Median neural unit firing rates increased for every sample and dynamic patterns of organoid firing rates were revealed by high-frequency recordings. Surprisingly, feeding did not affect firing rate. Furthermore, performing media exchange during a recording showed no acute effects on firing rate, enabling the use of this automated platform for reagent screening studies.
Collapse
Affiliation(s)
- Kateryna Voitiuk
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
| | - Spencer T. Seiler
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mirella Pessoa de Melo
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of
California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jinghui Geng
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of
California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tjitse van der Molen
- Neuroscience Research Institute, University of California Santa
Barbara, Santa Barbara, CA 93106, USA
- Department of Molecular, Cellular and Developmental Biology,
University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Sebastian Hernandez
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of
California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Hunter E. Schweiger
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Molecular, Cell, and Developmental Biology,
University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jess L. Sevetson
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Molecular, Cell, and Developmental Biology,
University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - David F. Parks
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
| | - Ash Robbins
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of
California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sebastian Torres-Montoya
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of
California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Drew Ehrlich
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Computational Media, University of California Santa
Cruz, Santa Cruz, CA 95064, USA
| | - Matthew A. T. Elliott
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tal Sharf
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
| | - David Haussler
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mohammed A. Mostajo-Radji
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Molecular, Cell, and Developmental Biology,
University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sofie R. Salama
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California
Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Molecular, Cell, and Developmental Biology,
University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Mircea Teodorescu
- Genomics Institute, University of California Santa Cruz, Santa
Cruz, CA 95064, USA
- Department of Electrical and Computer Engineering, University of
California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
34
|
Ji Y, Sun Y. Advancements in Organoid Culture Technologies: Current Trends and Innovations. Stem Cells Dev 2024; 33:631-644. [PMID: 39509169 DOI: 10.1089/scd.2024.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Organoids have emerged as valuable tools in investigating disease mechanisms, drug efficacy, and personalized medicine due to their capacity to recapitulate crucial aspects of tissue physiology, including cell-cell interactions, heterogeneity, microenvironmental cues, and drug responses. Despite their broad applicability across various research domains, conventional organoid culture methods are plagued by several limitations that hinder research progress. These limitations include the inability to faithfully recreate tissue microenvironments, immune contexts, and vascular systems. Fortunately, ongoing advancements in organoid culture techniques are addressing these shortcomings. In this review, we provide a comprehensive overview of current mainstream organoid culture protocols. By evaluating these protocols, researchers can identify the most suitable experimental methods, thereby optimizing resource allocation and experimental outcomes.
Collapse
Affiliation(s)
- Yanwei Ji
- College of Life Sciences, Jilin Agricultural University, Changchun City, People's Republic of China
| | - Yang Sun
- College of Life Sciences, Jilin Agricultural University, Changchun City, People's Republic of China
| |
Collapse
|
35
|
Yang X, Forró C, Li TL, Miura Y, Zaluska TJ, Tsai CT, Kanton S, McQueen JP, Chen X, Mollo V, Santoro F, Pașca SP, Cui B. Kirigami electronics for long-term electrophysiological recording of human neural organoids and assembloids. Nat Biotechnol 2024; 42:1836-1843. [PMID: 38253880 PMCID: PMC11260907 DOI: 10.1038/s41587-023-02081-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Abstract
Realizing the full potential of organoids and assembloids to model neural development and disease will require improved methods for long-term, minimally invasive recording of electrical activity. Current technologies, such as patch clamp, penetrating microelectrodes, planar electrode arrays and substrate-attached flexible electrodes, do not allow chronic recording of organoids in suspension, which is necessary to preserve architecture. Inspired by kirigami art, we developed flexible electronics that transition from a two-dimensional to a three-dimensional basket-like configuration with either spiral or honeycomb patterns to accommodate the long-term culture of organoids in suspension. Here we show that this platform, named kirigami electronics (KiriE), integrates with and enables chronic recording of cortical organoids for up to 120 days while preserving their morphology, cytoarchitecture and cell composition. We demonstrate integration of KiriE with optogenetic and pharmacological manipulation and modeling phenotypes related to a genetic disease. Moreover, KiriE can capture corticostriatal connectivity in assembloids following optogenetic stimulation. Thus, KiriE will enable investigation of disease and activity patterns underlying nervous system assembly.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Csaba Forró
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
- Institute for Biological I nformation Processing-Bioelectronics, IBI-3, Forschungszentrum Jülich, Jülich, Germany
| | - Thomas L Li
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | | | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Sabina Kanton
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - James P McQueen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Valentina Mollo
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
- Institute for Biological I nformation Processing-Bioelectronics, IBI-3, Forschungszentrum Jülich, Jülich, Germany
- Neuroelectronic Interfaces, RWTH Aachen, Aachen, Germany
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA.
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
36
|
Gunapala KM, Gadban A, Noreen F, Schär P, Benvenisty N, Taylor V. Ascorbic Acid Ameliorates Molecular and Developmental Defects in Human-Induced Pluripotent Stem Cell and Cerebral Organoid Models of Fragile X Syndrome. Int J Mol Sci 2024; 25:12718. [PMID: 39684429 DOI: 10.3390/ijms252312718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Fragile X Syndrome (FX) is the most common form of inherited cognitive impairment and falls under the broader category of Autism Spectrum Disorders (ASD). FX is caused by a CGG trinucleotide repeat expansion in the non-coding region of the X-linked Fragile X Messenger Ribonucleoprotein 1 (FMR1) gene, leading to its hypermethylation and epigenetic silencing. Animal models of FX rely on the deletion of the Fmr1 gene, which fails to replicate the epigenetic silencing mechanism of the FMR1 gene observed in human patients. Human stem cells carrying FX repeat expansions have provided a better understanding of the basis of epigenetic silencing of FMR1. Previous studies have found that 5-Azacytidine (5Azac) can reverse this methylation; however, 5Azac can be toxic, which may limit its therapeutic potential. Here, we show that the dietary factor Ascorbic Acid (AsA) can reduce DNA methylation in the FMR1 locus and lead to an increase in FMR1 gene expression in FX iPSCs and cerebral organoids. In addition, AsA treatment rescued neuronal gene expression and morphological defects observed in FX iPSC-derived cerebral organoids. Hence, we demonstrate that the dietary co-factor AsA can partially revert the molecular and morphological defects seen in human FX models in vitro. Our findings have implications for the development of novel therapies for FX in the future.
Collapse
Affiliation(s)
- Keith M Gunapala
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Aseel Gadban
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Faiza Noreen
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 4031 Basel, Switzerland
| | - Primo Schär
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
37
|
Sit TPH, Feord RC, Dunn AWE, Chabros J, Oluigbo D, Smith HH, Burn L, Chang E, Boschi A, Yuan Y, Gibbons GM, Khayat-Khoei M, De Angelis F, Hemberg E, Hemberg M, Lancaster MA, Lakatos A, Eglen SJ, Paulsen O, Mierau SB. MEA-NAP: A flexible network analysis pipeline for neuronal 2D and 3D organoid multielectrode recordings. CELL REPORTS METHODS 2024; 4:100901. [PMID: 39520988 PMCID: PMC11706071 DOI: 10.1016/j.crmeth.2024.100901] [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: 05/20/2024] [Revised: 08/01/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Microelectrode array (MEA) recordings are commonly used to compare firing and burst rates in neuronal cultures. MEA recordings can also reveal microscale functional connectivity, topology, and network dynamics-patterns seen in brain networks across spatial scales. Network topology is frequently characterized in neuroimaging with graph theoretical metrics. However, few computational tools exist for analyzing microscale functional brain networks from MEA recordings. Here, we present a MATLAB MEA network analysis pipeline (MEA-NAP) for raw voltage time series acquired from single- or multi-well MEAs. Applications to 3D human cerebral organoids or 2D human-derived or murine cultures reveal differences in network development, including topology, node cartography, and dimensionality. MEA-NAP incorporates multi-unit template-based spike detection, probabilistic thresholding for determining significant functional connections, and normalization techniques for comparing networks. MEA-NAP can identify network-level effects of pharmacologic perturbation and/or disease-causing mutations and thus can provide a translational platform for revealing mechanistic insights and screening new therapeutic approaches. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Timothy P H Sit
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK; Queen Square Institute of Neurology, University College London, WC1N 3BG London, UK
| | - Rachael C Feord
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - Alexander W E Dunn
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - Jeremi Chabros
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - David Oluigbo
- Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Hugo H Smith
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - Lance Burn
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - Elise Chang
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - Alessio Boschi
- Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA; Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Yin Yuan
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - George M Gibbons
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK; John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, CB2 0PY Cambridge, UK
| | - Mahsa Khayat-Khoei
- Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA
| | | | - Erik Hemberg
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Martin Hemberg
- Gene Lay Institute for Immunology and Inflammation, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Madeline A Lancaster
- MRC Laboratory for Molecular Biology, University of Cambridge, CB2 0QH Cambridge, UK
| | - Andras Lakatos
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, CB2 0PY Cambridge, UK; Cambridge University Hospitals, Cambridge Biomedical Campus, CB2 0QQ Cambridge, UK
| | - Stephen J Eglen
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, CB3 0WA Cambridge, UK
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK
| | - Susanna B Mierau
- Department of Physiology, Development and Neuroscience, University of Cambridge, CB2 3DY Cambridge, UK; Department of Neurology, Brigham & Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Huang R, Zhu Y, Chen H, Yu L, Liu Z, Liu Y, Wang Z, He X, Yang L, Xu X, Bai Y, Chen B, Zhu R. Progress in spinal cord organoid research: advancing understanding of neural development, disease modelling, and regenerative medicine. BIOMATERIALS TRANSLATIONAL 2024; 5:355-371. [PMID: 39872925 PMCID: PMC11764192 DOI: 10.12336/biomatertransl.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 01/30/2025]
Abstract
Stem cell-derived spinal cord organoids (SCOs) have revolutionised the study of spinal cord development and disease mechanisms, offering a three-dimensional model that recapitulates the complexity of native tissue. This review synthesises recent advancements in SCO technology, highlighting their role in modelling spinal cord morphogenesis and their application in neurodegenerative disease research. We discuss the methodological breakthroughs in inducing regional specification and cellular diversity within SCOs, which have enhanced their predictive ability for drug screening and their relevance in mimicking pathological conditions such as neurodegenerative diseases and neuromuscular disorders. Despite these strides, challenges in achieving vascularisation and mature neuronal integration persist. The future of SCOs lies in addressing these limitations, potentially leading to transformative impactions in regenerative medicine and therapeutic development.
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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yanjing 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Zhibo Liu
- 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, China
| | - Yuchen Liu
- 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, China
| | - Zhaojie Wang
- 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xiaolie He
- 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Li Yang
- 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xu Xu
- 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yuxin Bai
- 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Bairu 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, 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, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| |
Collapse
|
39
|
Li XH, Guo D, Chen LQ, Chang ZH, Shi JX, Hu N, Chen C, Zhang XW, Bao SQ, Chen MM, Ming D. Low-intensity ultrasound ameliorates brain organoid integration and rescues microcephaly deficits. Brain 2024; 147:3817-3833. [PMID: 38739753 DOI: 10.1093/brain/awae150] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Human brain organoids represent a remarkable platform for modelling neurological disorders and a promising brain repair approach. However, the effects of physical stimulation on their development and integration remain unclear. Here, we report that low-intensity ultrasound significantly increases neural progenitor cell proliferation and neuronal maturation in cortical organoids. Histological assays and single-cell gene expression analyses revealed that low-intensity ultrasound improves the neural development in cortical organoids. Following organoid grafts transplantation into the injured somatosensory cortices of adult mice, longitudinal electrophysiological recordings and histological assays revealed that ultrasound-treated organoid grafts undergo advanced maturation. They also exhibit enhanced pain-related gamma-band activity and more disseminated projections into the host brain than the untreated groups. Finally, low-intensity ultrasound ameliorates neuropathological deficits in a microcephaly brain organoid model. Hence, low-intensity ultrasound stimulation advances the development and integration of brain organoids, providing a strategy for treating neurodevelopmental disorders and repairing cortical damage.
Collapse
Affiliation(s)
- Xiao-Hong Li
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Di Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Li-Qun Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Zhe-Han Chang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Jian-Xin Shi
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Nan Hu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Chong Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Xiao-Wang Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Shuang-Qing Bao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Meng-Meng Chen
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
40
|
Wu Y, Li X, Liu H, Yang X, Li R, Zhao H, Shang Z. Organoids in the oral and maxillofacial region: present and future. Int J Oral Sci 2024; 16:61. [PMID: 39482304 PMCID: PMC11528035 DOI: 10.1038/s41368-024-00324-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/18/2024] [Accepted: 09/12/2024] [Indexed: 11/03/2024] Open
Abstract
The oral and maxillofacial region comprises a variety of organs made up of multiple soft and hard tissue, which are anatomically vulnerable to the pathogenic factors of trauma, inflammation, and cancer. The studies of this intricate entity have been long-termly challenged by a lack of versatile preclinical models. Recently, the advancements in the organoid industry have provided novel strategies to break through this dilemma. Here, we summarize the existing biological and engineering approaches that were employed to generate oral and maxillofacial organoids. Then, we detail the use of modified co-culture methods, such as cell cluster co-inoculation and air-liquid interface culture technology to reconstitute the vascular network and immune microenvironment in assembled organoids. We further retrospect the existing oral and maxillofacial assembled organoids and their potential to recapitulate the homeostasis in parental tissues such as tooth, salivary gland, and mucosa. Finally, we discuss how the next-generation organoids may benefit to regenerative and precision medicine for treatment of oral-maxillofacial illness.
Collapse
Affiliation(s)
- Yufei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanzhe Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Rui Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hui Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School of Stomatology-Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
41
|
He Z, Dony L, Fleck JS, Szałata A, Li KX, Slišković I, Lin HC, Santel M, Atamian A, Quadrato G, Sun J, Pașca SP, Camp JG, Theis FJ, Treutlein B. An integrated transcriptomic cell atlas of human neural organoids. Nature 2024; 635:690-698. [PMID: 39567792 PMCID: PMC11578878 DOI: 10.1038/s41586-024-08172-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 10/08/2024] [Indexed: 11/22/2024]
Abstract
Human neural organoids, generated from pluripotent stem cells in vitro, are useful tools to study human brain development, evolution and disease. However, it is unclear which parts of the human brain are covered by existing protocols, and it has been difficult to quantitatively assess organoid variation and fidelity. Here we integrate 36 single-cell transcriptomic datasets spanning 26 protocols into one integrated human neural organoid cell atlas totalling more than 1.7 million cells1-26. Mapping to developing human brain references27-30 shows primary cell types and states that have been generated in vitro, and estimates transcriptomic similarity between primary and organoid counterparts across protocols. We provide a programmatic interface to browse the atlas and query new datasets, and showcase the power of the atlas to annotate organoid cell types and evaluate new organoid protocols. Finally, we show that the atlas can be used as a diverse control cohort to annotate and compare organoid models of neural disease, identifying genes and pathways that may underlie pathological mechanisms with the neural models. The human neural organoid cell atlas will be useful to assess organoid fidelity, characterize perturbed and diseased states and facilitate protocol development.
Collapse
Affiliation(s)
- Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| | - Leander Dony
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jonas Simon Fleck
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Artur Szałata
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- School of Computation, Information, and Technology, Technical University of Munich, Munich, Germany
| | - Katelyn X Li
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Irena Slišković
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Hsiu-Chuan Lin
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Malgorzata Santel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Alexander Atamian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jieran Sun
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - J Gray Camp
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Fabian J Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- School of Computation, Information, and Technology, Technical University of Munich, Munich, Germany.
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
42
|
Assendorp N, Fossati M, Libé-Philippot B, Christopoulou E, Depp M, Rapone R, Dingli F, Loew D, Vanderhaeghen P, Charrier C. CTNND2 moderates the pace of synaptic maturation and links human evolution to synaptic neoteny. Cell Rep 2024; 43:114797. [PMID: 39352808 DOI: 10.1016/j.celrep.2024.114797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/01/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024] Open
Abstract
Human-specific genes are potential drivers of brain evolution. Among them, SRGAP2C has contributed to the emergence of features characterizing human cortical synapses, including their extended period of maturation. SRGAP2C inhibits its ancestral copy, the postsynaptic protein SRGAP2A, but the synaptic molecular pathways differentially regulated in humans by SRGAP2 proteins remain largely unknown. Here, we identify CTNND2, a protein implicated in severe intellectual disability (ID) in Cri-du-Chat syndrome, as a major partner of SRGAP2. We demonstrate that CTNND2 slows synaptic maturation and promotes neuronal integrity. During postnatal development, CTNND2 moderates neuronal excitation and excitability. In adults, it supports synapse maintenance. While CTNND2 deficiency is deleterious and results in synaptic loss of SYNGAP1, another major ID-associated protein, the human-specific protein SRGAP2C, enhances CTNND2 synaptic accumulation in human neurons. Our findings suggest that CTNND2 regulation by SRGAP2C contributes to synaptic neoteny in humans and link human-specific and ID genes at the synapse.
Collapse
Affiliation(s)
- Nora Assendorp
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Matteo Fossati
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Eirini Christopoulou
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Marine Depp
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Roberta Rapone
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France
| | - Florent Dingli
- Institut Curie, PSL Research University, CurieCore Tech Mass Spectrometry Proteomics, 75005 Paris, France
| | - Damarys Loew
- Institut Curie, PSL Research University, CurieCore Tech Mass Spectrometry Proteomics, 75005 Paris, France
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KUL, 3000 Leuven, Belgium
| | - Cécile Charrier
- Institut de Biologie de l'ENS (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Research University, 75005 Paris, France.
| |
Collapse
|
43
|
Martinelli E, Akouissi O, Liebi L, Furfaro I, Maulà D, Savoia N, Remy A, Nikles L, Roux A, Stoppini L, Lacour SP. The e-Flower: A hydrogel-actuated 3D MEA for brain spheroid electrophysiology. SCIENCE ADVANCES 2024; 10:eadp8054. [PMID: 39413178 PMCID: PMC11482305 DOI: 10.1126/sciadv.adp8054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
Traditional microelectrode arrays (MEAs) are limited to measuring electrophysiological activity in two dimensions, failing to capture the complexity of three-dimensional (3D) tissues such as neural organoids and spheroids. Here, we introduce a flower-shaped MEA (e-Flower) that can envelop submillimeter brain spheroids following actuation by the sole addition of the cell culture medium. Inspired by soft microgrippers, its actuation mechanism leverages the swelling properties of a polyacrylic acid hydrogel grafted to a polyimide substrate hosting the electrical interconnects. Compatible with standard electrophysiology recording systems, the e-Flower does not require additional equipment or solvents and is ready to use with preformed 3D tissues. We designed an e-Flower achieving a curvature as low as 300 micrometers within minutes, a value tunable by the choice of reswelling media and hydrogel cross-linker concentration. Furthermore, we demonstrate the ability of the e-Flower to detect spontaneous neural activity across the spheroid surface, demonstrating its potential for comprehensive neural signal recording.
Collapse
Affiliation(s)
- Eleonora Martinelli
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Outman Akouissi
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
- Bertarelli Foundation Chair in Translational NeuroEngineering, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Luca Liebi
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Ivan Furfaro
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Desirée Maulà
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Nathan Savoia
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Antoine Remy
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Laetitia Nikles
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Adrien Roux
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Luc Stoppini
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| | - Stéphanie P. Lacour
- Laboratory for Soft Bioelectronic Interfaces, Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| |
Collapse
|
44
|
Lancaster MA. Unraveling mechanisms of human brain evolution. Cell 2024; 187:5838-5857. [PMID: 39423803 PMCID: PMC7617105 DOI: 10.1016/j.cell.2024.08.052] [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: 02/15/2024] [Revised: 06/19/2024] [Accepted: 08/28/2024] [Indexed: 10/21/2024]
Abstract
Evolutionary changes in human brain structure and function have enabled our specialized cognitive abilities. How these changes have come about genetically and functionally has remained an open question. However, new methods are providing a wealth of information about the genetic, epigenetic, and transcriptomic differences that set the human brain apart. Combined with in vitro models that allow access to developing brain tissue and the cells of our closest living relatives, the puzzle pieces are now coming together to yield a much more complete picture of what is actually unique about the human brain. The challenge now will be linking these observations and making the jump from correlation to causation. However, elegant genetic manipulations are now possible and, when combined with model systems such as organoids, will uncover a mechanistic understanding of how evolutionary changes at the genetic level have led to key differences in development and function that enable human cognition.
Collapse
Affiliation(s)
- Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge, UK; Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
45
|
Lefèvre C, Cook GM, Dinan AM, Torii S, Stewart H, Gibbons G, Nicholson AS, Echavarría-Consuegra L, Meredith LW, Lulla V, McGovern N, Kenyon JC, Goodfellow I, Deane JE, Graham SC, Lakatos A, Lambrechts L, Brierley I, Irigoyen N. Zika viruses encode 5' upstream open reading frames affecting infection of human brain cells. Nat Commun 2024; 15:8822. [PMID: 39394194 PMCID: PMC11470053 DOI: 10.1038/s41467-024-53085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
Zika virus (ZIKV), an emerging mosquito-borne flavivirus, is associated with congenital neurological complications. Here, we investigate potential pathological correlates of virus gene expression in representative ZIKV strains through RNA sequencing and ribosome profiling. In addition to the single long polyprotein found in all flaviviruses, we identify the translation of unrecognised upstream open reading frames (uORFs) in the genomic 5' region. In Asian/American strains, ribosomes translate uORF1 and uORF2, whereas in African strains, the two uORFs are fused into one (African uORF). We use reverse genetics to examine the impact on ZIKV fitness of different uORFs mutant viruses. We find that expression of the African uORF and the Asian/American uORF1 modulates virus growth and tropism in human cortical neurons and cerebral organoids, suggesting a potential role in neurotropism. Although the uORFs are expressed in mosquito cells, we do not see a measurable effect on transmission by the mosquito vector in vivo. The discovery of ZIKV uORFs sheds new light on the infection of the human brain cells by this virus and raises the question of their existence in other neurotropic flaviviruses.
Collapse
Affiliation(s)
- Charlotte Lefèvre
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Georgia M Cook
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Adam M Dinan
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Shiho Torii
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Hazel Stewart
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - George Gibbons
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Alex S Nicholson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | - Luke W Meredith
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Valeria Lulla
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Naomi McGovern
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Julia C Kenyon
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Janet E Deane
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Stephen C Graham
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - András Lakatos
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, Cambridge, UK
| | - Louis Lambrechts
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Ian Brierley
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
46
|
Pang W, Zhu J, Yang K, Zhu X, Zhou W, Jiang L, Zhuang X, Liu Y, Wei J, Lu X, Yin Y, Chen Z, Xiang Y. Generation of human region-specific brain organoids with medullary spinal trigeminal nuclei. Cell Stem Cell 2024; 31:1501-1512.e8. [PMID: 39208804 DOI: 10.1016/j.stem.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/16/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Brain organoids with nucleus-specific identities provide unique platforms for studying human brain development and diseases at a finer resolution. Despite its essential role in vital body functions, the medulla of the hindbrain has seen a lack of in vitro models, let alone models resembling specific medullary nuclei, including the crucial spinal trigeminal nucleus (SpV) that relays peripheral sensory signals to the thalamus. Here, we report a method to differentiate human pluripotent stem cells into region-specific brain organoids resembling the dorsal domain of the medullary hindbrain. Importantly, organoids specifically recapitulated the development of the SpV derived from the dorsal medulla. We also developed an organoid system to create the trigeminothalamic projections between the SpV and the thalamus by fusing these organoids, namely human medullary SpV-like organoids (hmSpVOs), with organoids representing the thalamus (hThOs). Our study provides a platform for understanding SpV development, nucleus-based circuit organization, and related disorders in the human brain.
Collapse
Affiliation(s)
- Wei Pang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinkui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaona Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yantong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoxiang Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziling Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
47
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [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: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|
48
|
Gu L, Cai H, Chen L, Gu M, Tchieu J, Guo F. Functional Neural Networks in Human Brain Organoids. BME FRONTIERS 2024; 5:0065. [PMID: 39314749 PMCID: PMC11418062 DOI: 10.34133/bmef.0065] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/12/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Human brain organoids are 3-dimensional brain-like tissues derived from human pluripotent stem cells and hold promising potential for modeling neurological, psychiatric, and developmental disorders. While the molecular and cellular aspects of human brain organoids have been intensively studied, their functional properties such as organoid neural networks (ONNs) are largely understudied. Here, we summarize recent research advances in understanding, characterization, and application of functional ONNs in human brain organoids. We first discuss the formation of ONNs and follow up with characterization strategies including microelectrode array (MEA) technology and calcium imaging. Moreover, we highlight recent studies utilizing ONNs to investigate neurological diseases such as Rett syndrome and Alzheimer's disease. Finally, we provide our perspectives on the future challenges and opportunities for using ONNs in basic research and translational applications.
Collapse
Affiliation(s)
- Longjun Gu
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Lei Chen
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Jason Tchieu
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Pulmonary Biology, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- University of Cincinnati School of Medicine, Cincinnati, OH 45229, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering,
Indiana University Bloomington, Bloomington, IN 47405, USA
| |
Collapse
|
49
|
Petersilie L, Kafitz KW, Neu LA, Heiduschka S, Le S, Prigione A, Rose CR. Protocol for the generation of cultured cortical brain organoid slices. STAR Protoc 2024; 5:103212. [PMID: 39128007 PMCID: PMC11369419 DOI: 10.1016/j.xpro.2024.103212] [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: 05/06/2024] [Revised: 05/23/2024] [Accepted: 06/27/2024] [Indexed: 08/13/2024] Open
Abstract
Three-dimensional brain organoids from human pluripotent stem cells are a powerful tool for studying human neural networks. Here, we present a protocol for generating cortical brain organoid slices (cBOS) derived from regionalized cortical organoids and grown at the air-liquid interphase. We provide steps for slicing organoids and maintaining them in long-term culture. We then detail approaches for quality control including the evaluation of cell death and cellular identity. Finally, we describe procedures for the expression of a genetically encoded nanosensor for ATP. For complete details on the use and execution of this protocol, please refer to Petersilie et al.1.
Collapse
Affiliation(s)
- Laura Petersilie
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany.
| | - Karl W Kafitz
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Louis A Neu
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Sonja Heiduschka
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Stephanie Le
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany.
| |
Collapse
|
50
|
van der Geest AT, Jakobs CE, Ljubikj T, Huffels CFM, Cañizares Luna M, Vieira de Sá R, Adolfs Y, de Wit M, Rutten DH, Kaal M, Zwartkruis MM, Carcolé M, Groen EJN, Hol EM, Basak O, Isaacs AM, Westeneng HJ, van den Berg LH, Veldink JH, Schlegel DK, Pasterkamp RJ. Molecular pathology, developmental changes and synaptic dysfunction in (pre-) symptomatic human C9ORF72-ALS/FTD cerebral organoids. Acta Neuropathol Commun 2024; 12:152. [PMID: 39289761 PMCID: PMC11409520 DOI: 10.1186/s40478-024-01857-1] [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: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
A hexanucleotide repeat expansion (HRE) in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Human brain imaging and experimental studies indicate early changes in brain structure and connectivity in C9-ALS/FTD, even before symptom onset. Because these early disease phenotypes remain incompletely understood, we generated iPSC-derived cerebral organoid models from C9-ALS/FTD patients, presymptomatic C9ORF72-HRE (C9-HRE) carriers, and controls. Our work revealed the presence of all three C9-HRE-related molecular pathologies and developmental stage-dependent size phenotypes in cerebral organoids from C9-ALS/FTD patients. In addition, single-cell RNA sequencing identified changes in cell type abundance and distribution in C9-ALS/FTD organoids, including a reduction in the number of deep layer cortical neurons and the distribution of neural progenitors. Further, molecular and cellular analyses and patch-clamp electrophysiology detected various changes in synapse structure and function. Intriguingly, organoids from all presymptomatic C9-HRE carriers displayed C9-HRE molecular pathology, whereas the extent to which more downstream cellular defects, as found in C9-ALS/FTD models, were detected varied for the different presymptomatic C9-HRE cases. Together, these results unveil early changes in 3D human brain tissue organization and synaptic connectivity in C9-ALS/FTD that likely constitute initial pathologies crucial for understanding disease onset and the design of therapeutic strategies.
Collapse
Affiliation(s)
- Astrid T van der Geest
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Channa E Jakobs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marina de Wit
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Daan H Rutten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marthe Kaal
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maria M Zwartkruis
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mireia Carcolé
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Ewout J N Groen
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Adrian M Isaacs
- UK Dementia Research Institute at UCL and Dept. of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|