1
|
de Carvalho MT, Henriques-Pereira M, Monteiro MV, Lamghari M, Mano JF, Gaspar VM. Innervating 3D in vitro models: bioengineering and design blueprints. Trends Biotechnol 2025:S0167-7799(25)00178-7. [PMID: 40517115 DOI: 10.1016/j.tibtech.2025.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 05/02/2025] [Accepted: 05/15/2025] [Indexed: 06/16/2025]
Abstract
Innervation plays a key role in tissue homeostasis, disease, and repair throughout our lifetimes. Strategies for emulating innervation and its bioinstructive influence on surrounding cells are thus highly desirable to upgrade the organotypic features of human in vitro models. We delve into the latest strategies for generating innervated 3D models that mimic native innervation patterns, and highlight recent advances in bioengineering living platforms for emulating the effect of innervation during tissue regeneration or for recapitulating tumor-nerve interplay. We present an overview of the key design blueprints for generating innervated tissue models, highlight the challenges to be overcome, and discuss the biomedical breakthroughs that can branch from such in vitro platforms.
Collapse
Affiliation(s)
- Mariana-Tomás de Carvalho
- Department of Chemistry, Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Cellularis Biomodels, Parque de Ciência e Inovação (PCI) Creative Science Park, Aveiro Region, Via do Conhecimento, 3830-352 Ílhavo, Portugal
| | - Margarida Henriques-Pereira
- Department of Chemistry, Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Cellularis Biomodels, Parque de Ciência e Inovação (PCI) Creative Science Park, Aveiro Region, Via do Conhecimento, 3830-352 Ílhavo, Portugal
| | - Maria V Monteiro
- Department of Chemistry, Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Cellularis Biomodels, Parque de Ciência e Inovação (PCI) Creative Science Park, Aveiro Region, Via do Conhecimento, 3830-352 Ílhavo, Portugal
| | - Meriem Lamghari
- Instituto de Inovação e Investigação em Saúde (i3S), Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - João F Mano
- Department of Chemistry, Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Cellularis Biomodels, Parque de Ciência e Inovação (PCI) Creative Science Park, Aveiro Region, Via do Conhecimento, 3830-352 Ílhavo, Portugal.
| | - Vítor M Gaspar
- Department of Chemistry, Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; Cellularis Biomodels, Parque de Ciência e Inovação (PCI) Creative Science Park, Aveiro Region, Via do Conhecimento, 3830-352 Ílhavo, Portugal.
| |
Collapse
|
2
|
Wu J, Chen X, Zhang J, Wettschurack K, Robinson M, Li W, Zhao Y, Yoo YE, Deming BA, Abeyaratna AD, Que Z, Du D, Tegtmeyer M, Yuan C, Skarnes WC, Rochet JC, Wu LJ, Yang Y. Human microglia in brain assembloids display region-specific diversity and respond to hyperexcitable neurons carrying SCN2A mutation: Microglial diversity and response in assembloids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.06.04.657874. [PMID: 40501840 PMCID: PMC12157492 DOI: 10.1101/2025.06.04.657874] [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: 06/22/2025]
Abstract
Microglia critically shape neuronal circuit development and function, yet their region-specific properties and roles in distinct circuits of the human brain remain poorly understood. In this study, we generated region-specific brain organoids (cortical, striatal, and midbrain), each integrated with human microglia, to fill this critical gap. Single-cell RNA sequencing uncovered six distinct microglial subtypes exhibiting unique regional signatures, including a subtype highly enriched for the GABAB receptor gene within striatal organoids. To investigate the contributions of microglia to neural circuitry, we created microglia-incorporated midbrain-striatal assembloids, modeling a core circuit node for many neuropsychiatric disorders including autism. Using chemogenetics to activate this midbrain-striatal circuit, we observed increased calcium signaling in microglia involving GABAB receptors. Leveraging this model, we examined microglial responses within neural circuits harboring an SCN2A nonsense (C959X) mutation associated with profound autism. Remarkably, microglia displayed heightened calcium responses to SCN2A mutation-mediated neuronal hyperactivity, and engaged in excessive synaptic pruning. These pathological effects were reversed by pharmacological inhibition of microglial GABAB receptors. Collectively, our findings establish an advanced platform to dissect human neuroimmune interactions in sub-cortical regions, highlighting the important role of microglia in shaping critical circuitry related to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jiaxiang Wu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- These authors contributed equally
| | - Xiaoling Chen
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- These authors contributed equally
| | - Jingliang Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- These authors contributed equally
| | - Kyle Wettschurack
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- These authors contributed equally
| | - Morgan Robinson
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Davidson School of Chemical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Weihao Li
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Yuanrui Zhao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Ye-Eun Yoo
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Brody A. Deming
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Akila D. Abeyaratna
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Zhefu Que
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Dongshu Du
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Matthew Tegtmeyer
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, College of Engineering, Purdue University, West Lafayette, IN 47907, USA
| | | | - Jean-Christophe Rochet
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Long-Jun Wu
- Center for Neuroimmunology and Glial Biology, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yang Yang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Lead contact
| |
Collapse
|
3
|
Yang H, Zhang J, Li Y, Zhong Z, Li W, Luo H, Liu Y, Ouyang L, Jiang Z, Sun Y, Sun H, Liu L, Yang H, Wang Y, Yang N, Ma W, Mao Y. Multiscale Organization of Neural Networks in a 3D Bioprinted Matrix. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04455. [PMID: 40434038 DOI: 10.1002/advs.202504455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/24/2025] [Indexed: 05/29/2025]
Abstract
The efficient establishment of in vitro neural models that accurately mimic the structural and functional connectivity of neural networks is critical in neuroscience research. 3D bioprinting shows great potential for constructing sophisticated in vitro models with high freedom of design. However, mature neurons are delicate and susceptible to manipulation. Here, extrusion-based 3D bioprinting is employed to fabricate gelatin methacryloyl (GelMA)-based constructs containing embryonic day 18 (E18) rat cortical neurons, referred to as 3D neuMatrix. 3D neuMatrix displays favorable neuronal viability, with the progressive formation of a 3D brain-like neural network with local and long-range functional axon connections. Compared with 2D cultured neurons, 3D neuMatrix is more similar to the E18 cortex according to the bulk transcriptomic profile, with a recreation of cellular components in the cerebral cortex. The 3D neuMatrix is employed to establish a disease model of ischemic stroke, with a faithful recapitulation of the viability, function, and transcriptomic features of rats with middle cerebral artery occlusion/reperfusion (MCAO/R). These findings demonstrate the formation of multiscale neural circuits within 3D neuMatrix and its valuable potential in the study of neurodevelopment, disease modeling with drug screening, and in vitro intelligence.
Collapse
Affiliation(s)
- Huiyu Yang
- Department of Neurosurgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
- Eight-Year Medical Doctor Program, CAMS & PUMC, Beijing, 100730, China
| | - Jiangang Zhang
- Department of Liver Surgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
- Eight-Year Medical Doctor Program, CAMS & PUMC, Beijing, 100730, China
| | - Yiran Li
- Institute of Clinical Medicine, Translational Medicine Center, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Zihan Zhong
- Department of Neurosurgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
- Eight-Year Medical Doctor Program, CAMS & PUMC, Beijing, 100730, China
| | - Wenhua Li
- Department of Pharmacology, Institute of Basic Medical Sciences, CAMS & PUMC, Beijing, 100005, China
| | - Haojun Luo
- Department of Pharmacology, Institute of Basic Medical Sciences, CAMS & PUMC, Beijing, 100005, China
| | - Yanyong Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, CAMS & PUMC, Beijing, 100005, China
| | - Liujian Ouyang
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310003, China
| | - Zhuoran Jiang
- Department of Liver Surgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Yuning Sun
- Department of Liver Surgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Hang Sun
- Department of Liver Surgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Lulu Liu
- Center for Biomedical Technology of National Infrastructures for Translational Medicine, State Key Laboratory of Complex, Severe, and Rare Diseases in Peking Union Medical College Hospital, Beijing, 100730, China
| | - Huayu Yang
- Department of Liver Surgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Yu Wang
- Department of Neurosurgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Nan Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, CAMS & PUMC, Beijing, 100005, China
| | - Wenbin Ma
- Department of Neurosurgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| | - Yilei Mao
- Department of Liver Surgery, PUMCH, PUMC & CAMS, Beijing, 100730, China
| |
Collapse
|
4
|
Zhai Y, Gao F, Shi S, Zhong Q, Zhang J, Fang J, He F, Zhang Y, Li Y, Liu F, Xue B, Gu Y, Li S. Noninvasive Optogenetics Realized by iPSC-Derived Tentacled Carrier in Alzheimer's Disease Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2419768. [PMID: 40434197 DOI: 10.1002/adma.202419768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/21/2025] [Indexed: 05/29/2025]
Abstract
Neural-activated optogenetics technique contributing to the "restart" of degenerative neurons offers hope for the treatment of several neurodegenerative diseases. However, the limitations of persistent invasiveness and inadequate repair of the pathological environment strongly hinder its further application. Here, a concept of differentiating stem cells is proposed to produce functional materials to enhance the therapeutic applicability of optogenetics. Induced pluripotent stem cells (iPSCs) are differentiated to generate the "tentacled" stem cells TenSCs. Their "tentacled" vesicles TenSCev, upon inheriting the biological functions of the parent cell, will possess both neural targeting capacity and pathological environment repair ability. Hence, TenSCev are utilized as functional carrier to deliver optogenetics elements for completely non-traumatic and controllable neuron activation, while also facilitating the comprehensive restoration of the pathological environment, thus effectively halted disease progression and significantly improved cognitive function in Alzheimer's disease or aged mice. Further, the concept of generating specialized biomaterials from differentiated stem cells as functional carriers holds the potential to broaden the applicability of various neuroregulatory techniques in the treatment of neurological disorders.
Collapse
Affiliation(s)
- Yuewen Zhai
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Fan Gao
- Department of Biomedical Engineering, School of Automation, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun street, Jiangning District, Nanjing, Jiangsu Province, 211106, China
| | - Shihao Shi
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Qifeng Zhong
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Jinnan Zhang
- Department of Neurosurgery, China-Japan union Hospital, Jilin University, No. 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Ji Fang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Fang He
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Yanqin Zhang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Yu Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Fei Liu
- Department of Biomedical Engineering, Sichuan University, No.24, Wangjiang Road, Wuhou District, Chengdu, Sichuan Province, 610000, China
| | - Bing Xue
- Department of Biomedical Engineering, Sichuan University, No.24, Wangjiang Road, Wuhou District, Chengdu, Sichuan Province, 610000, China
| | - Yueqing Gu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| | - Siwen Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, No. 639 Longmian Avenue, Jiangning District, Nanjing, Jiangsu Province, 211198, China
| |
Collapse
|
5
|
Pan T, Lin G, Li X, VanHeyningen D, Walker JC, Kohli S, Saravanan A, Kondur A, Jaklic DC, Pantoja-Gutierrez S, Vaid S, Sturza J, Inoki K, Imamichi T, Chang W, Dang LT. Delayed forebrain excitatory and inhibitory neurogenesis in STRADA-related megalencephaly via mTOR hyperactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.13.653911. [PMID: 40462897 PMCID: PMC12132401 DOI: 10.1101/2025.05.13.653911] [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: 06/16/2025]
Abstract
Biallelic pathogenic variants in STRADA , an upstream regulator of the mechanistic target of rapamycin (mTOR) pathway, result in megalencephaly, drug-resistant epilepsy, and severe intellectual disability. This study explores how mTOR pathway hyperactivity alters cell fate specification in dorsal and ventral forebrain development using STRADA knock-out human stem cell derived brain organoids. In both dorsal and ventral forebrain STRADA knock-out organoids, neurogenesis is delayed, with a predilection for progenitor renewal and proliferation and an increase in outer radial glia. Ventrally, interneuron subtypes shift to an increase in neuropeptide-Y expressing cells. Inhibition of the mTOR pathway with rapamycin results in rescue for most phenotypes. When mTOR pathway variants are present in all cells of the developing brain, overproduction of interneurons and altered interneuron cell fate may underlie mechanisms of megalencephaly, epilepsy, and cognitive impairment. Our findings suggest mTOR inhibition during fetal brain development as a potential therapeutic strategy in STRADA deficiency.
Collapse
Affiliation(s)
- Tong Pan
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Grace Lin
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xuan Li
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Debora VanHeyningen
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - John Clay Walker
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sahej Kohli
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Aiswarya Saravanan
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Amrita Kondur
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Daniel C. Jaklic
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Saul Pantoja-Gutierrez
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Shivanshi Vaid
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Julie Sturza
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Departments of Molecular and Integrative Physiology and Internal Medicine, Michigan Medicine, Ann Arbor, MI, USA
| | - Tomozumi Imamichi
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Weizhong Chang
- Laboratory of Human Retrovirology and Immunoinformatics, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Louis T Dang
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Lead contact
| |
Collapse
|
6
|
Zhao Y, Wang T, Liu J, Wang Z, Lu Y. Emerging brain organoids: 3D models to decipher, identify and revolutionize brain. Bioact Mater 2025; 47:378-402. [PMID: 40026825 PMCID: PMC11869974 DOI: 10.1016/j.bioactmat.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Brain organoids are an emerging in vitro 3D brain model that is integrated from pluripotent stem cells. This model mimics the human brain's developmental process and disease-related phenotypes to a certain extent while advancing the development of human brain-based biological intelligence. However, many limitations of brain organoid culture (e.g., lacking a functional vascular system, etc.) prevent in vitro-cultured organoids from truly replicating the human brain in terms of cell type and structure. To improve brain organoids' scalability, efficiency, and stability, this paper discusses important contributions of material biology and microprocessing technology in solving the related limitations of brain organoids and applying the latest imaging technology to make real-time imaging of brain organoids possible. In addition, the related applications of brain organoids, especially the development of organoid intelligence combined with artificial intelligence, are analyzed, which will help accelerate the rational design and guidance of brain organoids.
Collapse
Affiliation(s)
- Yuli Zhao
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Ting Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Jiajun Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze Wang
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
7
|
Andreatta F, Hendriks D, Artegiani B. Human Organoids as an Emerging Tool for Genome Screenings. Annu Rev Biomed Eng 2025; 27:157-183. [PMID: 40310889 DOI: 10.1146/annurev-bioeng-103023-122327] [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: 05/03/2025]
Abstract
Over the last decade, a plethora of organoid models have been generated to recapitulate aspects of human development, disease, tissue homeostasis, and repair. Organoids representing multiple tissues have emerged and are typically categorized based on their origin. Tissue-derived organoids are established directly from tissue-resident stem/progenitor cells of either adult or fetal origin. Starting from pluripotent stem cells (PSCs), PSC-derived organoids instead recapitulate the developmental trajectory of a given organ. Gene editing technologies, particularly the CRISPR-Cas toolbox, have greatly facilitated gene manipulation experiments with considerable ease and scalability, revolutionizing organoid-based human biology research. Here, we review the recent adaptation of CRISPR-based screenings in organoids. We examine the strategies adopted to perform CRISPR screenings in organoids, discuss different screening scopes and readouts, and highlight organoid-specific challenges. We then discuss individual organoid-based genome screening studies that have uncovered novel genes involved in a variety of biological processes. We close by providing an outlook on how widespread adaptation of CRISPR screenings across the organoid field may be achieved, to ultimately leverage our understanding of human biology.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; ,
| | | |
Collapse
|
8
|
Kim JI, Imaizumi K, Jurjuț O, Kelley KW, Wang D, Thete MV, Hudacova Z, Amin ND, Levy RJ, Scherrer G, Pașca SP. Human assembloid model of the ascending neural sensory pathway. Nature 2025:10.1038/s41586-025-08808-3. [PMID: 40205039 DOI: 10.1038/s41586-025-08808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Somatosensory pathways convey crucial information about pain, touch, itch and body part movement from peripheral organs to the central nervous system1,2. Despite substantial needs to understand how these pathways assemble and to develop pain therapeutics, clinical translation remains challenging. This is probably related to species-specific features and the lack of in vitro models of the polysynaptic pathway. Here we established a human ascending somatosensory assembloid (hASA), a four-part assembloid generated from human pluripotent stem cells that integrates somatosensory, spinal, thalamic and cortical organoids to model the spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types of this circuit. Rabies tracing and calcium imaging showed that sensory neurons connect to dorsal spinal cord neurons, which further connect to thalamic neurons. Following noxious chemical stimulation, calcium imaging of hASA demonstrated a coordinated response. In addition, extracellular recordings and imaging revealed synchronized activity across the assembloid. Notably, loss of the sodium channel NaV1.7, which causes pain insensitivity, disrupted synchrony across hASA. By contrast, a gain-of-function SCN9A variant associated with extreme pain disorder induced hypersynchrony. These experiments demonstrated the ability to functionally assemble the essential components of the human sensory pathway, which could accelerate our understanding of sensory circuits and facilitate therapeutic development.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kent Imaizumi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Ovidiu Jurjuț
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Dong Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Zuzana Hudacova
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Rebecca J Levy
- Department of Neurology & Neurological Sciences, Division of Child Neurology, Stanford University, Stanford, CA, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA.
| |
Collapse
|
9
|
Howard CE, Cheenath M, Crouch E. The promise of cerebral organoids for neonatology. Curr Opin Pediatr 2025; 37:182-190. [PMID: 40013913 PMCID: PMC11902893 DOI: 10.1097/mop.0000000000001446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Applying discoveries from basic research to patients in the neonatal intensive care unit (NICU) is challenging given the difficulty of modeling this population in animal models, lack of translational relevance from animal models to humans, and scarcity of primary human tissue. Human cell-derived cerebral organoid models are an appealing way to address some of these gaps. In this review, we will touch on previous work to model neonatal conditions in cerebral organoids, some limitations of this approach, and recent strategies that have attempted to address these limitations. RECENT FINDINGS While modeling of neurodevelopmental disorders has been an application of cerebral organoids since their initial description, recent studies have dramatically expanded the types of brain regions and disease models available. Additionally, work to increase the complexity of organoid models by including immune and vascular cells, as well as modeling human heterogeneity with mixed donor organoids will provide new opportunities to model neonatal pathologies. SUMMARY Organoids are an attractive model to study human neurodevelopmental pathologies relevant to patients in the neonatal ICU. New technologies will broaden the applicability of these models to neonatal research and their usefulness as a drug screening platform.
Collapse
Affiliation(s)
- Clare E Howard
- Division of Newborn Medicine, Boston Children’s Hospital
| | - Manju Cheenath
- Department of Obstetrics and Gynecology, University of California, San Francisco
| | - Elizabeth Crouch
- Department of Pediatrics, University of California, San Francisco
| |
Collapse
|
10
|
Guan Y, Jia Z, Xiong X, He R, Ouyang Y, Liu H, Liang L, Meng X, Zhang R, Guan C, Wang S, Li D, Cui Y, Bai J, Zhao J, Meng H, Peng J, Wang Y. Tissue-specific extracellular matrix for the larger-scaled expansion of spinal cord organoids. Mater Today Bio 2025; 31:101561. [PMID: 40083838 PMCID: PMC11904521 DOI: 10.1016/j.mtbio.2025.101561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Spinal cord organoids (SCOs) are in vitro models that faithfully recapitulate the basic tissue architecture and cell types of the spinal cord and play a crucial role in developmental studies, disease modeling, and drug screening. Physiological cues are required for proliferation and differentiation during SCO culture. However, commonly used basement membrane matrix products, such as Matrigel®, lack tissue-specific biophysical signals. The current study utilizes decellularization process to fabricate tissue-derived hydrogel from porcine spinal cord tissue that retain intrinsic matrix components. This gel system supported an expanded neuroepithelial scale and enhanced ventral recognition patterns during SCO cultivation. Based on the characteristics of the enlarged aggregate size, a technical system for SCO cutting and subculture are proposed to improve the economic feasibility. Finally, the advantage of S-gel in maintaining neurite outgrowth are also found, which suggests its potential application in neural-related microphysiological systems.
Collapse
Affiliation(s)
- Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Zhibo Jia
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Xing Xiong
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Ruichao He
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Yiben Ouyang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Haolin Liu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Lijing Liang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiaoran Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Ranran Zhang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Congcong Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Sice Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Dongdong Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Yuhui Cui
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Jun Bai
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jinjuan Zhao
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Haoye Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, PR China
| | - Yu Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, PR China
| |
Collapse
|
11
|
Zhu ZW, Li G, Wu GG, Zhang YJ, Bai YR, Lai BQ, Ding Y, Zeng X, Ma YH, Liu S, Wang R, Liang JH, Xu YB, He B, Zeng YS. Transplantation of peripheral nerve tissueoid based on a decellularized optic nerve scaffold to restore rat hindlimb sensory and movement functions. Biomaterials 2025; 315:122949. [PMID: 39531746 DOI: 10.1016/j.biomaterials.2024.122949] [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: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Peripheral nerve injury (PNI) involving the loss of sensory and movement functions is challenging to repair. Although the gold standard of PNI repair is still the use of autologous nerve grafts, the destruction of the donor side is inevitable. In the present study, peripheral nerve tissueoids (PNTs) composed of a Schwann cell (SC)-based neurotrophin-3 (NT-3) delivery system and a decellularized optic nerve (DON) with naturally oriented channels were engineered to investigate the mechanism of PNTs in nerve regeneration. Proteomic analysis and mRNA sequencing revealed that PNTs have the advantage of promoting nerve regeneration by the three mechanisms of chemotaxis, adhesion and intrinsic mobilisation. The results demonstrated that a local NT-3-enriched pool was constructed by laminin γ3 (LAMC3) in PNTs, creating a niche for the colonization of TrkC-positive SCs, attraction of axons to the defect/graft area, and remyelination. In addition, LAMC3 in PNTs can rapidly promote axon adhesion through integrin aVβ6 and can precisely guide long projecting axons to target tissues. Furthermore, the interactions among the NT-3/TrkC, LAMC3/integrin aVβ6 and the scaffold synergistically activate the PI3K-AKT signalling pathway in damaged neurons, further stimulating the intrinsic regenerative drive within the neurons to ultimately achieve the rapid reinnervation and the improvement of sensory and movement functions in the hindlimb.
Collapse
Affiliation(s)
- Zhao-Wei Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Ge Li
- Medical Research Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Guang-Geng Wu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu-Jing Zhang
- The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Yu-Rong Bai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Shu Liu
- Department of Anatomy, Anhui Medical University, Hefei, 230032, China
| | - Rui Wang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Jing-Hua Liang
- Medical Research Institute, Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, China
| | - Yang-Bin Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Bo He
- Joint and Orthopaedics Trauma, Department of Orthopedics, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China.
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
12
|
Zhang J, Eaton M, Chen X, Zhao Y, Kant S, Deming BA, Harish K, Nguyen HP, Shu Y, Lai S, Wu J, Que Z, Wettschurack KW, Zhang Z, Xiao T, Halurkar MS, Olivero-Acosta MI, Yoo YE, Lanman NA, Koss WA, Skarnes WC, Yang Y. Restoration of excitation/inhibition balance enhances neuronal signal-to-noise ratio and rescues social deficits in autism-associated Scn2a-deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641498. [PMID: 40093153 PMCID: PMC11908182 DOI: 10.1101/2025.03.04.641498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Social behavior is critical for survival and adaptation, which is profoundly disrupted in autism spectrum disorders (ASD). Social withdrawal due to information overload was often described in ASD, and it was suspected that increased basal noise, i.e., excessive background neuronal activities in the brain could be a disease mechanism. However, experimental test of this hypothesis is limited. Loss-of-function mutations (deficiency) in SCN2A, which encodes the voltage-gated sodium channel NaV1.2, have been revealed as a leading monogenic cause of profound ASD. Here, we revealed that Scn2a deficiency results in robust and multifaceted social impairments in mice. Scn2a-deficient neurons displayed an increased excitation-inhibition (E/I) ratio, contributing to elevated basal neuronal noise and diminished signal-to-noise ratio (SNR) during social interactions. Notably, the restoration of Scn2a expression in adulthood is able to rescue both SNR and social deficits. By balancing the E/I ratio and reducing basal neuronal firing, an FDA-approved GABAA receptor-positive allosteric modulator improves sociability in Scn2a-deficient mice and normalizes neuronal activities in translationally relevant human brain organoids carrying autism-associated SCN2A nonsense mutation. Collectively, our findings revealed a critical role of the NaV1.2 channel in the regulation of social behaviors, and identified molecular, cellular, and circuitry mechanisms underlying SCN2A-associated disorders.
Collapse
Affiliation(s)
- Jingliang Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Muriel Eaton
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Xiaoling Chen
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Yuanrui Zhao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Shivam Kant
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Brody A. Deming
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Kothandaraman Harish
- Department of Comparative Pathobiology, Purdue University
- Purdue University Center for Cancer Research, Purdue University
| | - Huynhvi P. Nguyen
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Yue Shu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Shirong Lai
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Jiaxiang Wu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Zhefu Que
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Kyle W. Wettschurack
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Zaiyang Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| | - Tiange Xiao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University
| | - Manasi S. Halurkar
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Maria I. Olivero-Acosta
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Ye-Eun Yoo
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| | - Nadia A. Lanman
- Department of Comparative Pathobiology, Purdue University
- Purdue University Center for Cancer Research, Purdue University
| | - Wendy A. Koss
- Purdue Institute for Integrative Neuroscience, Purdue University
- Office of the Executive Vice President for Research and Partnerships, Purdue University
| | | | - Yang Yang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University
- Purdue Institute for Integrative Neuroscience, Purdue University
| |
Collapse
|
13
|
Cappelletti G, Brambilla L, Strizzi S, Limanaqi F, Melzi V, Rizzuti M, Nizzardo M, Saulle I, Trabattoni D, Corti S, Clerici M, Biasin M. iPSC-derived human cortical organoids display profound alterations of cellular homeostasis following SARS-CoV-2 infection and Spike protein exposure. FASEB J 2025; 39:e70396. [PMID: 39950320 PMCID: PMC11826378 DOI: 10.1096/fj.202401604rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025]
Abstract
COVID-19 commonly leads to respiratory issues, yet numerous patients also exhibit a diverse range of neurological conditions, suggesting a detrimental impact of SARS-CoV-2 or the viral Spike protein on the central nervous system. Nonetheless, the molecular pathway behind neurological pathology and the presumed neurotropism of SARS-CoV-2 remains largely unexplored. We generated human cortical organoids (HCOs) derived from human induced pluripotent stem cells (hiPSC) to assess: (1) the expression of SARS-CoV-2 main entry factors; (2) their vulnerability to SARS-CoV-2 infection; and (3) the impact of SARS-CoV-2 infection and exposure to the Spike protein on their transcriptome. Results proved that (1) HCOs express the main SARS-CoV-2 receptors and co-receptors; (2) HCOs may be productively infected by SARS-CoV-2; (3) the viral particles released by SARS-CoV-2-infected HCOs are able to re-infect another cellular line; and (4) the infection resulted in the activation of apoptotic and stress pathways, along with inflammatory processes. Notably, these effects were recapitulated when HCOs were exposed to the Spike protein alone. The data obtained demonstrate that SARS-CoV-2 likely infects HCOs probably through the binding of ACE2, CD147, and NRP1 entry factors. Furthermore, exposure to the Spike protein alone proved sufficient to disrupt their homeostasis and induce neurotoxic effects, potentially contributing to the onset of long-COVID symptoms.
Collapse
Affiliation(s)
- Gioia Cappelletti
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Lorenzo Brambilla
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Sergio Strizzi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Fiona Limanaqi
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Valentina Melzi
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mafalda Rizzuti
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Monica Nizzardo
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Irma Saulle
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
| | - Daria Trabattoni
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| | - Stefania Corti
- Neurology UnitFoundation IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience SectionUniversity of MilanMilanItaly
- Neuromuscular and Rare Diseases Unit, Department of NeuroscienceFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mario Clerici
- Department of Pathophysiology and TransplantationUniversity of MilanMilanItaly
- Don C. Gnocchi FoundationIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) FoundationMilanItaly
| | - Mara Biasin
- Department of Biomedical and Clinical SciencesUniversity of MilanMilanItaly
| |
Collapse
|
14
|
Acha C, George D, Diaz LC, Ouyang Z, Alam El Din DMM, Surlekar H, Moghadas B, Loftus E, Mangalvedhekar GM, Rayasam PSR, Lai YC, Smirnova L, Caffo BS, Johnson EC, Gracias DH. Neuromodulation in neural organoids with shell MEAs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.637712. [PMID: 40027665 PMCID: PMC11870477 DOI: 10.1101/2025.02.18.637712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Neural organoids (NOs) have emerged as important tissue engineering models for brain sciences and biocomputing. Establishing reliable relationships between stimulation and recording traces of electrical activity is essential to monitor the functionality of NOs, especially as it relates to realizing biocomputing paradigms such as reinforcement learning or stimulus discrimination. While researchers have demonstrated neuromodulation in NOs, they have primarily used 2D microelectrode arrays (MEAs) with limited access to the entire 3D contour of the NOs. Here, we report neuromodulation using tiny mimics of macroscale EEG caps or shell MEAs. Specifically, we observe that stimulating current within a specific range (20 to 30 µA) induced a statistically significant increase in neuron firing rate when comparing the activity five seconds before and after stimulation. We observed neuromodulatory behavior using both three- and 16-electrode shells and could generate 3D spatiotemporal maps of neuromodulatory activity around the surface of the NO. Our studies demonstrate a methodology for investigating 3D spatiotemporal neuromodulation in organoids of broad relevance to biomedical engineering and biocomputing. One-Sentence Summary Neuromodulation, an essential intelligence feature, was observed using 3D stimulation and recording from neural organoids.
Collapse
|
15
|
Li D, Wang Y, Ma L, Wang Y, Cheng L, Liu Y, Shi W, Lu Y, Wang H, Gao C, Erichsen CT, Zhang Y, Yang Z, Eickhoff SB, Chen CH, Jiang T, Chu C, Fan L. Topographic Axes of Wiring Space Converge to Genetic Topography in Shaping the Human Cortical Layout. J Neurosci 2025; 45:e1510242024. [PMID: 39824638 PMCID: PMC11823343 DOI: 10.1523/jneurosci.1510-24.2024] [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: 08/09/2024] [Revised: 10/25/2024] [Accepted: 12/04/2024] [Indexed: 01/20/2025] Open
Abstract
Genetic information is involved in the gradual emergence of cortical areas since the neural tube begins to form, shaping the heterogeneous functions of neural circuits in the human brain. Informed by invasive tract-tracing measurements, the cortex exhibits marked interareal variation in connectivity profiles, revealing the heterogeneity across cortical areas. However, it remains unclear about the organizing principles possibly shared by genetics and cortical wiring to manifest the spatial heterogeneity across the cortex. Instead of considering a complex one-to-one mapping between genetic coding and interareal connectivity, we hypothesized the existence of a more efficient way that the organizing principles are embedded in genetic profiles to underpin the cortical wiring space. Leveraging vertex-wise tractography in diffusion-weighted MRI, we derived the global connectopies (GCs) in both female and male to reliably index the organizing principles of interareal connectivity variation in a low-dimensional space, which captured three dominant topographic patterns along the dorsoventral, rostrocaudal, and mediolateral axes of the cortex. More importantly, we demonstrated that the GCs converge with the gradients of a vertex-by-vertex genetic correlation matrix on the phenotype of cortical morphology and the cortex-wide spatiomolecular gradients. By diving into the genetic profiles, we found that the critical role of genes scaffolding the GCs was related to brain morphogenesis and enriched in radial glial cells before birth and excitatory neurons after birth. Taken together, our findings demonstrated the existence of a genetically determined space that encodes the interareal connectivity variation, which may give new insights into the links between cortical connections and arealization.
Collapse
Affiliation(s)
- Deying Li
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yufan Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liang Ma
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaping Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Luqi Cheng
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin 541004, China
- Zhejiang Lab, Hangzhou 311121, China
| | - Yinan Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiyang Shi
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Yuheng Lu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haiyan Wang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Chaohong Gao
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Camilla T Erichsen
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, Aarhus University, Aarhus 8000, Denmark
| | - Yu Zhang
- Zhejiang Lab, Hangzhou 311121, China
| | - Zhengyi Yang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Research Centre Jülich, Jülich 52425, Germany
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Chi-Hua Chen
- Department of Radiology, University of California San Diego, La Jolla, California 92093
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China
- Xiaoxiang Institute for Brain Health and Yongzhou Central Hospital, Yongzhou 425000, China
| | - Congying Chu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Lingzhong Fan
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100190, China
- School of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266000, China
- Shandong Key Lab of Complex Medical Intelligence and Aging, Binzhou Medical University, Yantai, Shandong 264003, PR China
| |
Collapse
|
16
|
Popson P, Knowles JK. Zebrafish Provide Critical Insights in a Sea of Genes. Epilepsy Curr 2025; 25:58-60. [PMID: 39703932 PMCID: PMC11653375 DOI: 10.1177/15357597241301510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
Zebrafish Models of Candidate Human Epilepsy-Associated Genes Provide Evidence of Hyperexcitability LaCoursiere CM, Ullmann JFP, Koh HY, Turner L, Baker CM, Robens B, Shao W, Rotenberg A, McGraw CM, Poduri AH. iScience . 2024 Jun 5;27(7):110172. doi: 10.1016/j.isci.2024.110172. PMID: 39021799; PMCID: PMC11253282. Hundreds of novel candidate human epilepsy-associated genes have been identified thanks to advancements in next-generation sequencing and large genome-wide association studies, but establishing genetic etiology requires functional validation. We generated a list of >2200 candidate epilepsy-associated genes, of which 48 were developed into stable loss-of-function zebrafish models. Of those 48, evidence of seizure-like behavior was present in 5 (arfgef1, kcnd2, kcnv1, ubr5, and wnt8b). Further characterization provided evidence for epileptiform activity via electrophysiology in kcnd2 and wnt8b mutants. Additionally, arfgef1 and wnt8b mutants showed a decrease in the number of inhibitory interneurons in the optic tectum of larval animals. Further, RNA sequencing revealed convergent transcriptional abnormalities between mutant lines, consistent with their developmental defects and hyperexcitable phenotypes. These zebrafish models provide strongest experimental evidence supporting the role of ARFGEF1, KCND2, and WNT8B in human epilepsy and further demonstrate the utility of this model system for evaluating candidate human epilepsy genes.
Collapse
Affiliation(s)
- Pierce Popson
- Stanford University Ringgold Standard Institution-Neurology
| | | |
Collapse
|
17
|
Kim JI, Miura Y, Li MY, Revah O, Selvaraj S, Birey F, Meng X, Thete MV, Pavlov SD, Andersen J, Pașca AM, Porteus MH, Huguenard JR, Pașca SP. Human assembloids reveal the consequences of CACNA1G gene variants in the thalamocortical pathway. Neuron 2024; 112:4048-4059.e7. [PMID: 39419023 DOI: 10.1016/j.neuron.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Abnormalities in thalamocortical crosstalk can lead to neuropsychiatric disorders. Variants in CACNA1G, which encodes the α1G subunit of the thalamus-enriched T-type calcium channel, are associated with absence seizures, intellectual disability, and schizophrenia, but the cellular and circuit consequences of these genetic variants in humans remain unknown. Here, we developed a human assembloid model of the thalamocortical pathway to dissect the contribution of genetic variants in T-type calcium channels. We discovered that the M1531V CACNA1G variant associated with seizures led to changes in T-type currents in thalamic neurons, as well as correlated hyperactivity of thalamic and cortical neurons in assembloids. By contrast, CACNA1G loss, which has been associated with risk of schizophrenia, resulted in abnormal thalamocortical connectivity that was related to both increased spontaneous thalamic activity and aberrant axonal projections. These results illustrate the utility of multi-cellular systems for interrogating human genetic disease risk variants at both cellular and circuit level.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Min-Yin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sridhar Selvaraj
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Fikri Birey
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Xiangling Meng
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Sergey D Pavlov
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Jimena Andersen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Anca M Pașca
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA.
| |
Collapse
|
18
|
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
|
19
|
Lin CC'J, Tian Y, Tanzi RE, Jorfi M. Approaches for studying neuroimmune interactions in Alzheimer's disease. Trends Immunol 2024; 45:971-986. [PMID: 39537528 PMCID: PMC11624993 DOI: 10.1016/j.it.2024.10.002] [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: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Peripheral immune cells play an important role in the pathology of Alzheimer's disease (AD), impacting processes such as amyloid and tau protein aggregation, glial activation, neuronal integrity, and cognitive decline. Here, we examine cutting-edge strategies - encompassing animal and cellular models - used to investigate the roles of peripheral immune cells in AD. Approaches such as antibody-mediated depletion, genetic ablation, and bone marrow chimeras in mouse models have been instrumental in uncovering T, B, and innate immune cell disease-modifying functions. However, challenges such as specificity, off-target effects, and differences between human and mouse immune systems underscore the need for more human-relevant models. Emerging multicellular models replicating critical aspects of human brain tissue and neuroimmune interactions increasingly offer fresh insights into the role of immune cells in AD pathogenesis. Refining these methodologies can deepen our understanding of immune cell contributions to AD and support the development of novel immune-related therapeutic interventions.
Collapse
Affiliation(s)
- Chih-Chung 'Jerry' Lin
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yuyao Tian
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
20
|
Kelley KW, Revah O, Gore F, Kaganovsky K, Chen X, Deisseroth K, Pașca SP. Host circuit engagement of human cortical organoids transplanted in rodents. Nat Protoc 2024; 19:3542-3567. [PMID: 39075308 DOI: 10.1038/s41596-024-01029-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/22/2024] [Indexed: 07/31/2024]
Abstract
Human neural organoids represent promising models for studying neural function; however, organoids grown in vitro lack certain microenvironments and sensory inputs that are thought to be essential for maturation. The transplantation of patient-derived neural organoids into animal hosts helps overcome some of these limitations and offers an approach for neural organoid maturation and circuit integration. Here, we describe a method for transplanting human stem cell-derived cortical organoids (hCOs) into the somatosensory cortex of newborn rats. The differentiation of human induced pluripotent stem cells into hCOs occurs over 30-60 days, and the transplantation procedure itself requires ~0.5-1 hours per animal. The use of neonatal hosts provides a developmentally appropriate stage for circuit integration and allows the generation and experimental manipulation of a unit of human neural tissue within the cortex of a living animal host. After transplantation, animals can be maintained for hundreds of days, and transplanted hCO growth can be monitored by using brain magnetic resonance imaging. We describe the assessment of human neural circuit function in vivo by monitoring genetically encoded calcium responses and extracellular activity. To demonstrate human neuron-host functional integration, we also describe a procedure for engaging host neural circuits and for modulating animal behavior by using an optogenetic behavioral training paradigm. The transplanted human neurons can then undergo ex vivo characterization across modalities including dendritic morphology reconstruction, single-nucleus transcriptomics, optogenetic manipulation and electrophysiology. This approach may enable the discovery of cellular phenotypes from patient-derived cells and uncover mechanisms that contribute to human brain evolution from previously inaccessible developmental stages.
Collapse
Affiliation(s)
- Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Felicity Gore
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Konstantin Kaganovsky
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Karl Deisseroth
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
21
|
Bayati A, Ayoubi R, Aguila A, Zorca CE, Deyab G, Han C, Recinto SJ, Nguyen-Renou E, Rocha C, Maussion G, Luo W, Shlaifer I, Banks E, McDowell I, Del Cid Pellitero E, Ding XE, Sharif B, Séguéla P, Yaqubi M, Chen CXQ, You Z, Abdian N, McBride HM, Fon EA, Stratton JA, Durcan TM, Nahirney PC, McPherson PS. Modeling Parkinson's disease pathology in human dopaminergic neurons by sequential exposure to α-synuclein fibrils and proinflammatory cytokines. Nat Neurosci 2024; 27:2401-2416. [PMID: 39379564 DOI: 10.1038/s41593-024-01775-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/28/2024] [Indexed: 10/10/2024]
Abstract
Lewy bodies (LBs), α-synuclein-enriched intracellular inclusions, are a hallmark of Parkinson's disease (PD) pathology, yet a cellular model for LB formation remains elusive. Recent evidence indicates that immune dysfunction may contribute to the development of PD. In this study, we found that induced pluripotent stem cell (iPSC)-derived human dopaminergic (DA) neurons form LB-like inclusions after treatment with α-synuclein preformed fibrils (PFFs) but only when coupled to a model of immune challenge (interferon-γ or interleukin-1β treatment) or when co-cultured with activated microglia-like cells. Exposure to interferon-γ impairs lysosome function in DA neurons, contributing to LB formation. The knockdown of LAMP2 or the knockout of GBA in conjunction with PFF administration is sufficient for inclusion formation. Finally, we observed that the LB-like inclusions in iPSC-derived DA neurons are membrane bound, suggesting that they are not limited to the cytoplasmic compartment but may be formed due to dysfunctions in autophagy. Together, these data indicate that immune-triggered lysosomal dysfunction may contribute to the development of PD pathology.
Collapse
Affiliation(s)
- Armin Bayati
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Riham Ayoubi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Adriana Aguila
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Cornelia E Zorca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Ghislaine Deyab
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Chanshuai Han
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Sherilyn Junelle Recinto
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | | | - Cecilia Rocha
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Gilles Maussion
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Wen Luo
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Irina Shlaifer
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Emily Banks
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Ian McDowell
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Esther Del Cid Pellitero
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Xue Er Ding
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Behrang Sharif
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Philippe Séguéla
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Moein Yaqubi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Carol X-Q Chen
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Zhipeng You
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Narges Abdian
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Heidi M McBride
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Thomas M Durcan
- The Neuro's Early Drug Discovery Unit (EDDU), McGill University, Montreal, QC, Canada
| | - Patrick C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Peter S McPherson
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
22
|
Onesto MM, Kim JI, Pasca SP. Assembloid models of cell-cell interaction to study tissue and disease biology. Cell Stem Cell 2024; 31:1563-1573. [PMID: 39454582 DOI: 10.1016/j.stem.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/26/2024] [Accepted: 09/23/2024] [Indexed: 10/28/2024]
Abstract
Neurodevelopment involves the migration, projection, and integration of various cell types across different regions of the nervous system. Assembloids are self-organizing systems formed by the integration of multiple organoids or cell types. Here, we outline the generation and application of assembloids. We illustrate how assembloids recapitulate critical neurodevelopmental steps, like migration, axon projection, and circuit formation, and how they are starting to provide biological insights into neuropsychiatric disorders. Additionally, we review how assembloids can be used to study properties emerging from cell-cell interactions within non-neural tissues. Overall, assembloid platforms represent a powerful tool for discovering human biology and developing therapeutics.
Collapse
Affiliation(s)
- Massimo M Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA
| | - Sergiu P Pasca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute and Bio-X, Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
Li M, Yuan Y, Hou Z, Hao S, Jin L, Wang B. Human brain organoid: trends, evolution, and remaining challenges. Neural Regen Res 2024; 19:2387-2399. [PMID: 38526275 PMCID: PMC11090441 DOI: 10.4103/1673-5374.390972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 10/28/2023] [Indexed: 03/26/2024] Open
Abstract
Advanced brain organoids provide promising platforms for deciphering the cellular and molecular processes of human neural development and diseases. Although various studies and reviews have described developments and advancements in brain organoids, few studies have comprehensively summarized and analyzed the global trends in this area of neuroscience. To identify and further facilitate the development of cerebral organoids, we utilized bibliometrics and visualization methods to analyze the global trends and evolution of brain organoids in the last 10 years. First, annual publications, countries/regions, organizations, journals, authors, co-citations, and keywords relating to brain organoids were identified. The hotspots in this field were also systematically identified. Subsequently, current applications for brain organoids in neuroscience, including human neural development, neural disorders, infectious diseases, regenerative medicine, drug discovery, and toxicity assessment studies, are comprehensively discussed. Towards that end, several considerations regarding the current challenges in brain organoid research and future strategies to advance neuroscience will be presented to further promote their application in neurological research.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhan Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zongkun Hou
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
24
|
Xue J, Chu Y, Huang Y, Chen M, Sun M, Fan Z, Wu Y, Chen L. A tumorigenicity evaluation platform for cell therapies based on brain organoids. Transl Neurodegener 2024; 13:53. [PMID: 39472972 PMCID: PMC11520457 DOI: 10.1186/s40035-024-00446-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Tumorigenicity represents a critical challenge in stem cell-based therapies requiring rigorous monitoring. Conventional approaches for tumorigenicity evaluation are based on animal models and have numerous limitations. Brain organoids, which recapitulate the structural and functional complexity of the human brain, have been widely used in neuroscience research. However, the capacity of brain organoids for tumorigenicity evaluation needs to be further elucidated. METHODS A cerebral organoid model produced from human pluripotent stem cells (hPSCs) was employed. Meanwhile, to enhance the detection sensitivity for potential tumorigenic cells, we created a glioblastoma-like organoid (GBM organoid) model from TP53-/-/PTEN-/- hPSCs to provide a tumor microenvironment for injected cells. Midbrain dopamine (mDA) cells from human embryonic stem cells were utilized as a cell therapy product. mDA cells, hPSCs, mDA cells spiked with hPSCs, and immature mDA cells were then injected into the brain organoids and NOD SCID mice. The injected cells within the brain organoids were characterized, and compared with those injected in vivo to evaluate the capability of the brain organoids for tumorigenicity evaluation. Single-cell RNA sequencing was performed to identify the differential gene expression between the cerebral organoids and the GBM organoids. RESULTS Both cerebral organoids and GBM organoids supported maturation of the injected mDA cells. The hPSCs and immature mDA cells injected in the GBM organoids showed a significantly higher proliferative capacity than those injected in the cerebral organoids and in NOD SCID mice. Furthermore, the spiked hPSCs were detectable in both the cerebral organoids and the GBM organoids. Notably, the GBM organoids demonstrated a superior capacity to enhance proliferation and pluripotency of spiked hPSCs compared to the cerebral organoids and the mouse model. Kyoto Encyclopedia of Genes and Genomes analysis revealed upregulation of tumor-related metabolic pathways and cytokines in the GBM organoids, suggesting that these factors underlie the high detection sensitivity for tumorigenicity evaluation. CONCLUSIONS Our findings suggest that brain organoids could represent a novel and effective platform for evaluating the tumorigenic risk in stem cell-based therapies. Notably, the GBM organoids offer a superior platform that could complement or potentially replace traditional animal-based models for tumorigenicity evaluation.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yanwang Huang
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ming Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Zhiqin Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
25
|
Cheng JL, Cook AL, Talbot J, Perry S. How is Excitotoxicity Being Modelled in iPSC-Derived Neurons? Neurotox Res 2024; 42:43. [PMID: 39405005 PMCID: PMC11480214 DOI: 10.1007/s12640-024-00721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
Excitotoxicity linked either to environmental causes (pesticide and cyanotoxin exposure), excitatory neurotransmitter imbalance, or to intrinsic neuronal hyperexcitability, is a pathological mechanism central to neurodegeneration in amyotrophic lateral sclerosis (ALS). Investigation of excitotoxic mechanisms using in vitro and in vivo animal models has been central to understanding ALS mechanisms of disease. In particular, advances in induced pluripotent stem cell (iPSC) technologies now provide human cell-based models that are readily amenable to environmental and network-based excitotoxic manipulations. The cell-type specific differentiation of iPSC, combined with approaches to modelling excitotoxicity that include editing of disease-associated gene variants, chemogenetics, and environmental risk-associated exposures make iPSC primed to examine gene-environment interactions and disease-associated excitotoxic mechanisms. Critical to this is knowledge of which neurotransmitter receptor subunits are expressed by iPSC-derived neuronal cultures being studied, how their activity responds to antagonists and agonists of these receptors, and how to interpret data derived from multi-parameter electrophysiological recordings. This review explores how iPSC-based studies have contributed to our understanding of ALS-linked excitotoxicity and highlights novel approaches to inducing excitotoxicity in iPSC-derived neurons to further our understanding of its pathological pathways.
Collapse
Affiliation(s)
- Jan L Cheng
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Anthony L Cook
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Jana Talbot
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia
| | - Sharn Perry
- Wicking Dementia Research and Education Centre, College of Health and Medicine, University of Tasmania, 17 Liverpool Street, Hobart, TAS, Australia.
| |
Collapse
|
26
|
Miura Y, Kim JI, Jurjuț O, Kelley KW, Yang X, Chen X, Thete MV, Revah O, Cui B, Pachitariu M, Pașca SP. Assembloid model to study loop circuits of the human nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.617729. [PMID: 39463945 PMCID: PMC11507680 DOI: 10.1101/2024.10.13.617729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Neural circuits connecting the cerebral cortex, the basal ganglia and the thalamus are fundamental networks for sensorimotor processing and their dysfunction has been consistently implicated in neuropsychiatric disorders 1-9 . These recursive, loop circuits have been investigated in animal models and by clinical neuroimaging, however, direct functional access to developing human neurons forming these networks has been limited. Here, we use human pluripotent stem cells to reconstruct an in vitro cortico-striatal-thalamic-cortical circuit by creating a four-part loop assembloid. More specifically, we generate regionalized neural organoids that resemble the key elements of the cortico-striatal-thalamic-cortical circuit, and functionally integrate them into loop assembloids using custom 3D-printed biocompatible wells. Volumetric and mesoscale calcium imaging, as well as extracellular recordings from individual parts of these assembloids reveal the emergence of synchronized patterns of neuronal activity. In addition, a multi-step rabies retrograde tracing approach demonstrate the formation of neuronal connectivity across the network in loop assembloids. Lastly, we apply this system to study heterozygous loss of ASH1L gene associated with autism spectrum disorder and Tourette syndrome and discover aberrant synchronized activity in disease model assembloids. Taken together, this human multi-cellular platform will facilitate functional investigations of the cortico-striatal-thalamic-cortical circuit in the context of early human development and in disease conditions.
Collapse
|
27
|
Lulla V, Sridhar A. Understanding neurotropic enteric viruses: routes of infection and mechanisms of attenuation. Cell Mol Life Sci 2024; 81:413. [PMID: 39365457 PMCID: PMC11452578 DOI: 10.1007/s00018-024-05450-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/05/2024]
Abstract
The intricate connection between the gut and the brain involves multiple routes. Several viral families begin their infection cycle in the intestinal tract. However, amongst the long list of viral intestinal pathogens, picornaviruses, and astroviruses stand out for their ability to transition from the intestinal epithelia to central or peripheral nervous system cells. In immunocompromised, neonates and young children, these viral infections can manifest as severe diseases, such as encephalitis, meningitis, and acute flaccid paralysis. What confers this remarkable plasticity and makes them efficient in infecting cells of the gut and the brain axes? Here, we review the current understanding of the virus infection along the gut-brain axis for some enteric viruses and discuss the molecular mechanisms of their attenuation.
Collapse
Affiliation(s)
- Valeria Lulla
- Division of Virology, Department of Pathology, Addenbrooke's Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Adithya Sridhar
- OrganoVIR Labs, Department of Pediatric Infectious Diseases, Amsterdam UMC, location Academic Medical Center, Amsterdam Institute for Reproduction and Development, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
- OrganoVIR Labs, Department of Medical Microbiology, Amsterdam UMC, location Academic Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Meibergdreef 9, 1100 AZ, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Zhao HT, Schmidt ER. Human-specific genetic modifiers of cortical architecture and function. Curr Opin Genet Dev 2024; 88:102241. [PMID: 39111228 PMCID: PMC11547859 DOI: 10.1016/j.gde.2024.102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Evolution of the cerebral cortex is thought to have been critical for the emergence of our cognitive abilities. Major features of cortical evolution include increased neuron number and connectivity and altered morpho-electric properties of cortical neurons. Significant progress has been made in identifying human-specific genetic modifiers (HSGMs), some of which are involved in shaping these features of cortical architecture. But how did these evolutionary changes support the emergence of our cognitive abilities? Here, we highlight recent studies aimed at examining the impact of HSGMs on cortical circuit function and behavior. We also discuss the need for greater insight into the link between evolution of cortical architecture and the functional and computational properties of neuronal circuits, as we seek to provide a neurobiological foundation for human cognition.
Collapse
Affiliation(s)
- Hanzhi T Zhao
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA
| | - Ewoud Re Schmidt
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA.
| |
Collapse
|
29
|
Mierke CT. Bioprinting of Cells, Organoids and Organs-on-a-Chip Together with Hydrogels Improves Structural and Mechanical Cues. Cells 2024; 13:1638. [PMID: 39404401 PMCID: PMC11476109 DOI: 10.3390/cells13191638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
The 3D bioprinting technique has made enormous progress in tissue engineering, regenerative medicine and research into diseases such as cancer. Apart from individual cells, a collection of cells, such as organoids, can be printed in combination with various hydrogels. It can be hypothesized that 3D bioprinting will even become a promising tool for mechanobiological analyses of cells, organoids and their matrix environments in highly defined and precisely structured 3D environments, in which the mechanical properties of the cell environment can be individually adjusted. Mechanical obstacles or bead markers can be integrated into bioprinted samples to analyze mechanical deformations and forces within these bioprinted constructs, such as 3D organoids, and to perform biophysical analysis in complex 3D systems, which are still not standard techniques. The review highlights the advances of 3D and 4D printing technologies in integrating mechanobiological cues so that the next step will be a detailed analysis of key future biophysical research directions in organoid generation for the development of disease model systems, tissue regeneration and drug testing from a biophysical perspective. Finally, the review highlights the combination of bioprinted hydrogels, such as pure natural or synthetic hydrogels and mixtures, with organoids, organoid-cell co-cultures, organ-on-a-chip systems and organoid-organ-on-a chip combinations and introduces the use of assembloids to determine the mutual interactions of different cell types and cell-matrix interferences in specific biological and mechanical environments.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
30
|
Yoo S, Lee HJ. Spheroid-Hydrogel-Integrated Biomimetic System: A New Frontier in Advanced Three-Dimensional Cell Culture Technology. Cells Tissues Organs 2024; 214:128-147. [PMID: 39265553 PMCID: PMC11965833 DOI: 10.1159/000541416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Despite significant advances in three-dimensional (3D) cell culture technologies, creating accurate in vitro models that faithfully recapitulate complex in vivo environments remains a major challenge in biomedical research. Traditional culture methods often fail to simultaneously facilitate critical cell-cell and cell-extracellular matrix (ECM) interactions while providing control over mechanical and biochemical properties. SUMMARY This review introduces the spheroid-hydrogel-integrated biomimetic system (SHIBS), a groundbreaking approach that synergistically combines spheroid culture with tailored hydrogel technologies. SHIBS uniquely bridges the gap between traditional culture methods and physiological conditions by offering unprecedented control over both cellular interactions and environmental properties. We explore how SHIBS is revolutionizing fields ranging from drug discovery and disease modeling to regenerative medicine and basic biological research. The review discusses current challenges in SHIBS technology, including reproducibility, scalability, and high-resolution imaging, and outlines ongoing research addressing these issues. Furthermore, we envision the future evolution of SHIBS into more sophisticated organoid-hydrogel-integrated biomimetic systems and its integration with cutting-edge technologies such as microfluidics, 3D bioprinting, and artificial intelligence. KEY MESSAGES SHIBS represents a paradigm shift in 3D cell culture technology, offering a unique solution to recreate complex in vivo environments. Its potential to accelerate the development of personalized therapies across various biomedical fields is significant. While challenges persist, the ongoing advancements in SHIBS technology promise to overcome current limitations, paving the way for more accurate and reliable in vitro models. The future integration of SHIBS with emerging technologies may revolutionize biomimetic modeling, potentially reducing the need for animal testing and expediting drug discovery processes. This comprehensive review provides researchers and clinicians with a holistic understanding of SHIBS technology, its current capabilities, and its future prospects in advancing biomedical research and therapeutic innovations. BACKGROUND Despite significant advances in three-dimensional (3D) cell culture technologies, creating accurate in vitro models that faithfully recapitulate complex in vivo environments remains a major challenge in biomedical research. Traditional culture methods often fail to simultaneously facilitate critical cell-cell and cell-extracellular matrix (ECM) interactions while providing control over mechanical and biochemical properties. SUMMARY This review introduces the spheroid-hydrogel-integrated biomimetic system (SHIBS), a groundbreaking approach that synergistically combines spheroid culture with tailored hydrogel technologies. SHIBS uniquely bridges the gap between traditional culture methods and physiological conditions by offering unprecedented control over both cellular interactions and environmental properties. We explore how SHIBS is revolutionizing fields ranging from drug discovery and disease modeling to regenerative medicine and basic biological research. The review discusses current challenges in SHIBS technology, including reproducibility, scalability, and high-resolution imaging, and outlines ongoing research addressing these issues. Furthermore, we envision the future evolution of SHIBS into more sophisticated organoid-hydrogel-integrated biomimetic systems and its integration with cutting-edge technologies such as microfluidics, 3D bioprinting, and artificial intelligence. KEY MESSAGES SHIBS represents a paradigm shift in 3D cell culture technology, offering a unique solution to recreate complex in vivo environments. Its potential to accelerate the development of personalized therapies across various biomedical fields is significant. While challenges persist, the ongoing advancements in SHIBS technology promise to overcome current limitations, paving the way for more accurate and reliable in vitro models. The future integration of SHIBS with emerging technologies may revolutionize biomimetic modeling, potentially reducing the need for animal testing and expediting drug discovery processes. This comprehensive review provides researchers and clinicians with a holistic understanding of SHIBS technology, its current capabilities, and its future prospects in advancing biomedical research and therapeutic innovations.
Collapse
Affiliation(s)
- Seungyeop Yoo
- School of Chemical, Biological and Battery Engineering, Gachon University, Seongnam-si, Republic of Korea
| | - Hyun Jong Lee
- School of Chemical, Biological and Battery Engineering, Gachon University, Seongnam-si, Republic of Korea
| |
Collapse
|
31
|
Evangelisti C, Ramadan S, Orlacchio A, Panza E. Experimental Cell Models for Investigating Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9747. [PMID: 39273694 PMCID: PMC11396244 DOI: 10.3390/ijms25179747] [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: 08/16/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Experimental models play a pivotal role in biomedical research, facilitating the understanding of disease mechanisms and the development of novel therapeutics. This is particularly true for neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and motor neuron disease, which present complex challenges for research and therapy development. In this work, we review the recent literature about experimental models and motor neuron disease. We identified three main categories of models that are highly studied by scientists. In fact, experimental models for investigating these diseases encompass a variety of approaches, including modeling the patient's cell culture, patient-derived induced pluripotent stem cells, and organoids. Each model offers unique advantages and limitations, providing researchers with a range of tools to address complex biological questions. Here, we discuss the characteristics, applications, and recent advancements in terms of each model system, highlighting their contributions to advancing biomedical knowledge and translational research.
Collapse
Affiliation(s)
- Cecilia Evangelisti
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sherin Ramadan
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Antonio Orlacchio
- Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy
- Laboratory of Neurogenetics, European Center for Brain Research (CERC), IRCCS Santa Lucia Foundation, 00143 Rome, Italy
| | - Emanuele Panza
- Medical Genetics Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
32
|
Wu J, Zhang J, Chen X, Wettschurack K, Que Z, Deming BA, Olivero-Acosta MI, Cui N, Eaton M, Zhao Y, Li SM, Suzuki M, Chen I, Xiao T, Halurkar MS, Mandal P, Yuan C, Xu R, Koss WA, Du D, Chen F, Wu LJ, Yang Y. Microglial over-pruning of synapses during development in autism-associated SCN2A-deficient mice and human cerebral organoids. Mol Psychiatry 2024; 29:2424-2437. [PMID: 38499656 DOI: 10.1038/s41380-024-02518-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Autism spectrum disorder (ASD) is a major neurodevelopmental disorder affecting 1 in 36 children in the United States. While neurons have been the focus of understanding ASD, an altered neuro-immune response in the brain may be closely associated with ASD, and a neuro-immune interaction could play a role in the disease progression. As the resident immune cells of the brain, microglia regulate brain development and homeostasis via core functions including phagocytosis of synapses. While ASD has been traditionally considered a polygenic disorder, recent large-scale human genetic studies have identified SCN2A deficiency as a leading monogenic cause of ASD and intellectual disability. We generated a Scn2a-deficient mouse model, which displays major behavioral and neuronal phenotypes. However, the role of microglia in this disease model is unknown. Here, we reported that Scn2a-deficient mice have impaired learning and memory, accompanied by reduced synaptic transmission and lower spine density in neurons of the hippocampus. Microglia in Scn2a-deficient mice are partially activated, exerting excessive phagocytic pruning of post-synapses related to the complement C3 cascades during selective developmental stages. The ablation of microglia using PLX3397 partially restores synaptic transmission and spine density. To extend our findings from rodents to human cells, we established a microglia-incorporated human cerebral organoid model carrying an SCN2A protein-truncating mutation identified in children with ASD. We found that human microglia display increased elimination of post-synapse in cerebral organoids carrying the SCN2A mutation. Our study establishes a key role of microglia in multi-species autism-associated models of SCN2A deficiency from mouse to human cells.
Collapse
Affiliation(s)
- Jiaxiang Wu
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Jingliang Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiaoling Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Kyle Wettschurack
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Zhefu Que
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Brody A Deming
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Maria I Olivero-Acosta
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Ningren Cui
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Muriel Eaton
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Yuanrui Zhao
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Sophia M Li
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Matthew Suzuki
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Ian Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Tiange Xiao
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Manasi S Halurkar
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Purba Mandal
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Chongli Yuan
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Ranjie Xu
- College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA
| | - Wendy A Koss
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Dongshu Du
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yang Yang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
33
|
Wan S, Aregueta Robles U, Poole-Warren L, Esrafilzadeh D. Advances in 3D tissue models for neural engineering: self-assembled versus engineered tissue models. Biomater Sci 2024; 12:3522-3549. [PMID: 38829222 DOI: 10.1039/d4bm00317a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Neural tissue engineering has emerged as a promising field that aims to create functional neural tissue for therapeutic applications, drug screening, and disease modelling. It is becoming evident in the literature that this goal requires development of three-dimensional (3D) constructs that can mimic the complex microenvironment of native neural tissue, including its biochemical, mechanical, physical, and electrical properties. These 3D models can be broadly classified as self-assembled models, which include spheroids, organoids, and assembloids, and engineered models, such as those based on decellularized or polymeric scaffolds. Self-assembled models offer advantages such as the ability to recapitulate neural development and disease processes in vitro, and the capacity to study the behaviour and interactions of different cell types in a more realistic environment. However, self-assembled constructs have limitations such as lack of standardised protocols, inability to control the cellular microenvironment, difficulty in controlling structural characteristics, reproducibility, scalability, and lengthy developmental timeframes. Integrating biomimetic materials and advanced manufacturing approaches to present cells with relevant biochemical, mechanical, physical, and electrical cues in a controlled tissue architecture requires alternate engineering approaches. Engineered scaffolds, and specifically 3D hydrogel-based constructs, have desirable properties, lower cost, higher reproducibility, long-term stability, and they can be rapidly tailored to mimic the native microenvironment and structure. This review explores 3D models in neural tissue engineering, with a particular focus on analysing the benefits and limitations of self-assembled organoids compared with hydrogel-based engineered 3D models. Moreover, this paper will focus on hydrogel based engineered models and probe their biomaterial components, tuneable properties, and fabrication techniques that allow them to mimic native neural tissue structures and environment. Finally, the current challenges and future research prospects of 3D neural models for both self-assembled and engineered models in neural tissue engineering will be discussed.
Collapse
Affiliation(s)
- Shuqian Wan
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Ulises Aregueta Robles
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| | - Laura Poole-Warren
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
- Tyree Foundation Institute of Health Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
34
|
Serna C, Sandepudi K, Keate RL, Zhang SL, Cotton KY, De La Isla A, Murillo M, Bouricha Y, Domenighetti AA, Franz CK, Jordan SW. Incorporation of decellularized-ECM in graphene-based scaffolds enhances axonal outgrowth and branching in neuro-muscular co-cultures. Sci Prog 2024; 107:368504241281469. [PMID: 39314156 PMCID: PMC11423365 DOI: 10.1177/00368504241281469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Peripheral nerve and large-scale muscle injuries result in significant disability, necessitating the development of biomaterials that can restore functional deficits by promoting tissue regrowth in an electroactive environment. Among these materials, graphene is favored for its high conductivity, but its low bioactivity requires enhancement through biomimetic components. In this study, we extrusion printed graphene-poly(lactide-co-glycolide) (graphene) lattice scaffolds, aiming to increase bioactivity by incorporating decellularized extracellular matrix (dECM) derived from mouse pup skeletal muscle. We first evaluated these scaffolds using human-induced pluripotent stem cell (hiPSC)-derived motor neurons co-cultured with supportive glia, observing significant improvements in axon outgrowth. Next, we tested the scaffolds with C2C12 mouse and human primary myoblasts, finding no significant differences in myotube formation between dECM-graphene and graphene scaffolds. Finally, using a more complex hiPSC-derived 3D motor neuron spheroid model co-cultured with human myoblasts, we demonstrated that dECM-graphene scaffolds significantly improved axonal expansion towards peripheral myoblasts and increased axonal network density compared to graphene-only scaffolds. Features of early neuromuscular junction formation were identified near neuromuscular interfaces in both scaffold types. These findings suggest that dECM-graphene scaffolds are promising candidates for enhancing neuromuscular regeneration, offering robust support for the growth and development of diverse neuromuscular tissues.
Collapse
Affiliation(s)
- Carlos Serna
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Kirtana Sandepudi
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Rebecca L Keate
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Sophia L Zhang
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Kristen Y Cotton
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Alberto De La Isla
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Matias Murillo
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Yasmine Bouricha
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Andrea A Domenighetti
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colin K Franz
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Sumanas W Jordan
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| |
Collapse
|
35
|
Wyle Y, Lu N, Hepfer J, Sayal R, Martinez T, Wang A. The Role of Biophysical Factors in Organ Development: Insights from Current Organoid Models. Bioengineering (Basel) 2024; 11:619. [PMID: 38927855 PMCID: PMC11200479 DOI: 10.3390/bioengineering11060619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Biophysical factors play a fundamental role in human embryonic development. Traditional in vitro models of organogenesis focused on the biochemical environment and did not consider the effects of mechanical forces on developing tissue. While most human tissue has a Young's modulus in the low kilopascal range, the standard cell culture substrate, plasma-treated polystyrene, has a Young's modulus of 3 gigapascals, making it 10,000-100,000 times stiffer than native tissues. Modern in vitro approaches attempt to recapitulate the biophysical niche of native organs and have yielded more clinically relevant models of human tissues. Since Clevers' conception of intestinal organoids in 2009, the field has expanded rapidly, generating stem-cell derived structures, which are transcriptionally similar to fetal tissues, for nearly every organ system in the human body. For this reason, we conjecture that organoids will make their first clinical impact in fetal regenerative medicine as the structures generated ex vivo will better match native fetal tissues. Moreover, autologously sourced transplanted tissues would be able to grow with the developing embryo in a dynamic, fetal environment. As organoid technologies evolve, the resultant tissues will approach the structure and function of adult human organs and may help bridge the gap between preclinical drug candidates and clinically approved therapeutics. In this review, we discuss roles of tissue stiffness, viscoelasticity, and shear forces in organ formation and disease development, suggesting that these physical parameters should be further integrated into organoid models to improve their physiological relevance and therapeutic applicability. It also points to the mechanotransductive Hippo-YAP/TAZ signaling pathway as a key player in the interplay between extracellular matrix stiffness, cellular mechanics, and biochemical pathways. We conclude by highlighting how frontiers in physics can be applied to biology, for example, how quantum entanglement may be applied to better predict spontaneous DNA mutations. In the future, contemporary physical theories may be leveraged to better understand seemingly stochastic events during organogenesis.
Collapse
Affiliation(s)
- Yofiel Wyle
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
| | - Nathan Lu
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Jason Hepfer
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Rahul Sayal
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Taylor Martinez
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817, USA
| |
Collapse
|
36
|
Yan Y, Cho AN. Human Brain In Vitro Model for Pathogen Infection-Related Neurodegeneration Study. Int J Mol Sci 2024; 25:6522. [PMID: 38928228 PMCID: PMC11204318 DOI: 10.3390/ijms25126522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Recent advancements in stem cell biology and tissue engineering have revolutionized the field of neurodegeneration research by enabling the development of sophisticated in vitro human brain models. These models, including 2D monolayer cultures, 3D organoids, organ-on-chips, and bioengineered 3D tissue models, aim to recapitulate the cellular diversity, structural organization, and functional properties of the native human brain. This review highlights how these in vitro brain models have been used to investigate the effects of various pathogens, including viruses, bacteria, fungi, and parasites infection, particularly in the human brain cand their subsequent impacts on neurodegenerative diseases. Traditional studies have demonstrated the susceptibility of different 2D brain cell types to infection, elucidated the mechanisms underlying pathogen-induced neuroinflammation, and identified potential therapeutic targets. Therefore, current methodological improvement brought the technology of 3D models to overcome the challenges of 2D cells, such as the limited cellular diversity, incomplete microenvironment, and lack of morphological structures by highlighting the need for further technological advancements. This review underscored the significance of in vitro human brain cell from 2D monolayer to bioengineered 3D tissue model for elucidating the intricate dynamics for pathogen infection modeling. These in vitro human brain cell enabled researchers to unravel human specific mechanisms underlying various pathogen infections such as SARS-CoV-2 to alter blood-brain-barrier function and Toxoplasma gondii impacting neural cell morphology and its function. Ultimately, these in vitro human brain models hold promise as personalized platforms for development of drug compound, gene therapy, and vaccine. Overall, we discussed the recent progress in in vitro human brain models, their applications in studying pathogen infection-related neurodegeneration, and future directions.
Collapse
Affiliation(s)
- Yuwei Yan
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
| | - Ann-Na Cho
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
37
|
Suong DNA, Imamura K, Kato Y, Inoue H. Design of neural organoids engineered by mechanical forces. IBRO Neurosci Rep 2024; 16:190-195. [PMID: 38328799 PMCID: PMC10847990 DOI: 10.1016/j.ibneur.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/19/2024] [Indexed: 02/09/2024] Open
Abstract
Neural organoids consist of three-dimensional tissue derived from pluripotent stem cells that could recapitulate key features of the human brain. During the past decade, organoid technology has evolved in the field of human brain science by increasing the quality and applicability of its products. Among them, a novel approach involving the design of neural organoids engineered by mechanical forces has emerged. This review describes previous approaches for the generation of neural organoids, the engineering of neural organoids by mechanical forces, and future challenges for the application of mechanical forces in the design of neural organoids.
Collapse
Affiliation(s)
- Dang Ngoc Anh Suong
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Keiko Imamura
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical‑Risk Avoidance Based On iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE MultiMix Corporation, Saitama, Japan
| | - Haruhisa Inoue
- iPSC‑Based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical‑Risk Avoidance Based On iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| |
Collapse
|
38
|
Boylin K, Aquino GV, Purdon M, Abedi K, Kasendra M, Barrile R. Basic models to advanced systems: harnessing the power of organoids-based microphysiological models of the human brain. Biofabrication 2024; 16:032007. [PMID: 38749420 DOI: 10.1088/1758-5090/ad4c08] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 05/15/2024] [Indexed: 05/29/2024]
Abstract
Understanding the complexities of the human brain's function in health and disease is a formidable challenge in neuroscience. While traditional models like animals offer valuable insights, they often fall short in accurately mirroring human biology and drug responses. Moreover, recent legislation has underscored the need for more predictive models that more accurately represent human physiology. To address this requirement, human-derived cell cultures have emerged as a crucial alternative for biomedical research. However, traditional static cell culture models lack the dynamic tissue microenvironment that governs human tissue function. Advancedin vitrosystems, such as organoids and microphysiological systems (MPSs), bridge this gap by offering more accurate representations of human biology. Organoids, which are three-dimensional miniaturized organ-like structures derived from stem cells, exhibit physiological responses akin to native tissues, but lack essential tissue-specific components such as functional vascular structures and immune cells. Recent endeavors have focused on incorporating endothelial cells and immune cells into organoids to enhance vascularization, maturation, and disease modeling. MPS, including organ-on-chip technologies, integrate diverse cell types and vascularization under dynamic culture conditions, revolutionizing brain research by bridging the gap betweenin vitroandin vivomodels. In this review, we delve into the evolution of MPS, with a particular focus on highlighting the significance of vascularization in enhancing the viability, functionality, and disease modeling potential of organoids. By examining the interplay of vasculature and neuronal cells within organoids, we can uncover novel therapeutic targets and gain valuable insights into disease mechanisms, offering the promise of significant advancements in neuroscience and improved patient outcomes.
Collapse
Affiliation(s)
- Katherine Boylin
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Grace V Aquino
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Michael Purdon
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Kimia Abedi
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Magdalena Kasendra
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| | - Riccardo Barrile
- Department of Biomedical Engineering, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States of America
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States of America
| |
Collapse
|
39
|
Wu S, Hong Y, Chu C, Gan Y, Li X, Tao M, Wang D, Hu H, Zheng Z, Zhu Q, Han X, Zhu W, Xu M, Dong Y, Liu Y, Guo X. Construction of human 3D striato-nigral assembloids to recapitulate medium spiny neuronal projection defects in Huntington's disease. Proc Natl Acad Sci U S A 2024; 121:e2316176121. [PMID: 38771878 PMCID: PMC11145230 DOI: 10.1073/pnas.2316176121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/22/2024] [Indexed: 05/23/2024] Open
Abstract
The striato-nigral (Str-SN) circuit is composed of medium spiny neuronal projections that are mainly sent from the striatum to the midbrain substantial nigra (SN), which is essential for regulating motor behaviors. Dysfunction of the Str-SN circuitry may cause a series of motor disabilities that are associated with neurodegenerative disorders, such as Huntington's disease (HD). Although the etiology of HD is known as abnormally expanded CAG repeats of the huntingtin gene, treatment of HD remains tremendously challenging. One possible reason is the lack of effective HD model that resembles Str-SN circuitry deficits for pharmacological studies. Here, we first differentiated striatum-like organoids from human pluripotent stem cells (hPSCs), containing functional medium spiny neurons (MSNs). We then generated 3D Str-SN assembloids by assembling striatum-like organoids with midbrain SN-like organoids. With AAV-hSYN-GFP-mediated viral tracing, extensive MSN projections from the striatum to the SN are established, which formed synaptic connection with GABAergic neurons in SN organoids and showed the optically evoked inhibitory postsynaptic currents and electronic field potentials by labeling the striatum-like organoids with optogenetic virus. Furthermore, these Str-SN assembloids exhibited enhanced calcium activity compared to that of individual striatal organoids. Importantly, we further demonstrated the reciprocal projection defects in HD iPSC-derived assembloids, which could be ameliorated by treatment of brain-derived neurotrophic factor. Taken together, these findings suggest that Str-SN assembloids could be used for identifying MSN projection defects and could be applied as potential drug test platforms for HD.
Collapse
Affiliation(s)
- Shanshan Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yuan Hong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Chu Chu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yixia Gan
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, China
| | - Xinrui Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Mengdan Tao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- School of Biological Science and Medical Engineering Southeast University, Sipailou, Nanjing210096, China
| | - Da Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Hao Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Zhilong Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing211166, China
| | - Qian Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Xiao Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Wanying Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Min Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai200241, China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, Nanjing Medical University, Nanjing211166, China
- School of Biological Science and Medical Engineering Southeast University, Sipailou, Nanjing210096, China
| | - Xing Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing211166, China
- Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing211166, China
- Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu226001, China
| |
Collapse
|
40
|
Salzinger A, Ramesh V, Das Sharma S, Chandran S, Thangaraj Selvaraj B. Neuronal Circuit Dysfunction in Amyotrophic Lateral Sclerosis. Cells 2024; 13:792. [PMID: 38786016 PMCID: PMC11120636 DOI: 10.3390/cells13100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
The primary neural circuit affected in Amyotrophic Lateral Sclerosis (ALS) patients is the corticospinal motor circuit, originating in upper motor neurons (UMNs) in the cerebral motor cortex which descend to synapse with the lower motor neurons (LMNs) in the spinal cord to ultimately innervate the skeletal muscle. Perturbation of these neural circuits and consequent loss of both UMNs and LMNs, leading to muscle wastage and impaired movement, is the key pathophysiology observed. Despite decades of research, we are still lacking in ALS disease-modifying treatments. In this review, we document the current research from patient studies, rodent models, and human stem cell models in understanding the mechanisms of corticomotor circuit dysfunction and its implication in ALS. We summarize the current knowledge about cortical UMN dysfunction and degeneration, altered excitability in LMNs, neuromuscular junction degeneration, and the non-cell autonomous role of glial cells in motor circuit dysfunction in relation to ALS. We further highlight the advances in human stem cell technology to model the complex neural circuitry and how these can aid in future studies to better understand the mechanisms of neural circuit dysfunction underpinning ALS.
Collapse
Affiliation(s)
- Andrea Salzinger
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Vidya Ramesh
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Shreya Das Sharma
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Siddharthan Chandran
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic (ARRNC), University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Bhuvaneish Thangaraj Selvaraj
- UK Dementia Research Institute, University of Edinburgh, Edinburgh EH16 4SB, UK; (A.S.); (V.R.); (S.D.S.); (S.C.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Anne Rowling Regenerative Neurology Clinic (ARRNC), University of Edinburgh, Edinburgh EH16 4SB, UK
| |
Collapse
|
41
|
Sinha S, Huang MS, Mikos G, Bedi Y, Soto L, Lensch S, Ayushman M, Bintu L, Bhutani N, Heilshorn SC, Yang F. Laminin-associated integrins mediate Diffuse Intrinsic Pontine Glioma infiltration and therapy response within a neural assembloid model. Acta Neuropathol Commun 2024; 12:71. [PMID: 38706008 PMCID: PMC11070088 DOI: 10.1186/s40478-024-01765-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/24/2024] [Indexed: 05/07/2024] Open
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a highly aggressive and fatal pediatric brain cancer. One pre-requisite for tumor cells to infiltrate is adhesion to extracellular matrix (ECM) components. However, it remains largely unknown which ECM proteins are critical in enabling DIPG adhesion and migration and which integrin receptors mediate these processes. Here, we identify laminin as a key ECM protein that supports robust DIPG cell adhesion and migration. To study DIPG infiltration, we developed a DIPG-neural assembloid model, which is composed of a DIPG spheroid fused to a human induced pluripotent stem cell-derived neural organoid. Using this assembloid model, we demonstrate that knockdown of laminin-associated integrins significantly impedes DIPG infiltration. Moreover, laminin-associated integrin knockdown improves DIPG response to radiation and HDAC inhibitor treatment within the DIPG-neural assembloids. These findings reveal the critical role of laminin-associated integrins in mediating DIPG progression and drug response. The results also provide evidence that disrupting integrin receptors may offer a novel therapeutic strategy to enhance DIPG treatment outcomes. Finally, these results establish DIPG-neural assembloid models as a powerful tool to study DIPG disease progression and enable drug discovery.
Collapse
Affiliation(s)
- Sauradeep Sinha
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Georgios Mikos
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yudhishtar Bedi
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1254, Palo Alto, CA, 94305, USA
| | - Luis Soto
- Department of Radiation Oncology, Stanford University, Stanford, CA, 94305, USA
| | - Sarah Lensch
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Manish Ayushman
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Lacramioara Bintu
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Nidhi Bhutani
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1254, Palo Alto, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, 476 Lomita Mall, McCullough Building, Room 246, Palo Alto, CA, 94305, USA.
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA.
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, 240 Pasteur Dr., Biomedical Innovation Building 1254, Palo Alto, CA, 94305, USA.
| |
Collapse
|
42
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
43
|
Bai X. Use of induced pluripotent stem cell-derived brain cells, organoids, assembloids, and blood-brain barrier models in understanding alcohol and anesthetic-induced brain injuries: an emerging perspective. Neural Regen Res 2024; 19:953-954. [PMID: 37862185 PMCID: PMC10749634 DOI: 10.4103/1673-5374.385297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 10/22/2023] Open
Affiliation(s)
- Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
44
|
Layer PG. In a century from agitated cells to human organoids. J Neurosci Methods 2024; 405:110083. [PMID: 38387805 DOI: 10.1016/j.jneumeth.2024.110083] [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: 12/18/2023] [Revised: 02/09/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
Reaching back more than a century, suspension cultures have provided major insights into processes of histogenesis; e.g., cell communication, distinction of self/nonself, cell sorting and cell adhesion. Besides studies on lower animals, the vertebrate retina served as excellent reaggregate model to analyze 3D reconstruction of a complex neural laminar tissue. Methodologically, keeping cells under suspension is essential to achieve tissue organisation in vitro; thereby, the environmental conditions direct the emergent histotypic particulars. Recent progress in regenerative medicine is based to a large extent on human induced pluripotent stem cells (hiPSCs), which are cultured under suspension. Following their genetically directed differentiation into various histologic 3D structures, organoids provide excellent multipurpose in vitro assay models, as well as tissues for repair transplantations. Historically, a nearly fully laminated retinal spheroid from avian embryos was achieved already in 1984, foreshadowing the potential of culturing stem cells under suspension for tissue reconstruction purposes.
Collapse
Affiliation(s)
- Paul Gottlob Layer
- Technical University of Darmstadt, Developmental Biology & Neurogenetics, Schnittspahnstrasse 13, Darmstadt 64297, Germany.
| |
Collapse
|
45
|
Mencattini A, Daprati E, Della-Morte D, Guadagni F, Sangiuolo F, Martinelli E. Assembloid learning: opportunities and challenges for personalized approaches to brain functioning in health and disease. Front Artif Intell 2024; 7:1385871. [PMID: 38708094 PMCID: PMC11066156 DOI: 10.3389/frai.2024.1385871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Affiliation(s)
- Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
- Interdisciplinary Center of Advanced Study of Organ-on-Chip and Lab-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, Rome, Italy
| | - Elena Daprati
- Department of System Medicine and Centro di Biomedicina Spaziale (CBMS), University of Rome Tor Vergata, Rome, Italy
| | - David Della-Morte
- Interdisciplinary Center of Advanced Study of Organ-on-Chip and Lab-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, Rome, Italy
- San Raffaele Rome University, Rome, Italy
| | - Fiorella Guadagni
- San Raffaele Rome University, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
- Interdisciplinary Center of Advanced Study of Organ-on-Chip and Lab-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
46
|
Petersilie L, Heiduschka S, Nelson JS, Neu LA, Le S, Anand R, Kafitz KW, Prigione A, Rose CR. Cortical brain organoid slices (cBOS) for the study of human neural cells in minimal networks. iScience 2024; 27:109415. [PMID: 38523789 PMCID: PMC10957451 DOI: 10.1016/j.isci.2024.109415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/29/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Brain organoids derived from human pluripotent stem cells are a promising tool for studying human neurodevelopment and related disorders. Here, we generated long-term cultures of cortical brain organoid slices (cBOS) grown at the air-liquid interphase from regionalized cortical organoids. We show that cBOS host mature neurons and astrocytes organized in complex architecture. Whole-cell patch-clamp demonstrated subthreshold synaptic inputs and action potential firing of neurons. Spontaneous intracellular calcium signals turned into synchronous large-scale oscillations upon combined disinhibition of NMDA receptors and blocking of GABAA receptors. Brief metabolic inhibition to mimic transient energy restriction in the ischemic brain induced reversible intracellular calcium loading of cBOS. Moreover, metabolic inhibition induced a reversible decline in neuronal ATP as revealed by ATeam1.03YEMK. Overall, cBOS provide a powerful platform to assess morphological and functional aspects of human neural cells in intact minimal networks and to address the pathways that drive cellular damage during brain ischemia.
Collapse
Affiliation(s)
- Laura Petersilie
- 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
| | - Joel S.E. Nelson
- 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
| | - Stephanie Le
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Duesseldorf, 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
| | - 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
|
47
|
Ozgun A, Lomboni DJ, Aylsworth A, Macdonald A, Staines WA, Martina M, Schlossmacher MG, Tauskela JS, Woulfe J, Variola F. Unraveling the assembloid: Real-time monitoring of dopaminergic neurites in an inter-organoid pathway connecting midbrain and striatal regions. Mater Today Bio 2024; 25:100992. [PMID: 38371467 PMCID: PMC10873721 DOI: 10.1016/j.mtbio.2024.100992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/18/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024] Open
Abstract
Modern in vitro technologies for preclinical research, including organ-on-a-chip, organoids- and assembloid-based systems, have rapidly emerged as pivotal tools for elucidating disease mechanisms and assessing the efficacy of putative therapeutics. In this context, advanced in vitro models of Parkinson's Disease (PD) offer the potential to accelerate drug discovery by enabling effective platforms that recapitulate both physiological and pathological attributes of the in vivo environment. Although these systems often aim at replicating the PD-associated loss of dopaminergic (DA) neurons, only a few have modelled the degradation of dopaminergic pathways as a way to mimic the disruption of downstream regulation mechanisms that define the characteristic motor symptoms of the disease. To this end, assembloids have been successfully employed to recapitulate neuronal pathways between distinct brain regions. However, the investigation and characterization of these connections through neural tracing and electrophysiological analysis remain a technically challenging and time-consuming process. Here, we present a novel bioengineered platform consisting of surface-grown midbrain and striatal organoids at opposite sides of a self-assembled DA pathway. In particular, dopaminergic neurons and striatal GABAergic neurons spontaneously form DA connections across a microelectrode array (MEA), specifically integrated for the real-time monitoring of electrophysiological development and stimuli response. Calcium imaging data showed spiking synchronicity of the two organoids forming the inter-organoid pathways (IOPs) demonstrating that they are functionally connected. MEA recordings confirm a more robust response to the DA neurotoxin 6-OHDA compared to midbrain organoids alone, thereby validating the potential of this technology to generate highly tractable, easily extractable real-time functional readouts to investigate the dysfunctional dopaminergic network of PD patients.
Collapse
Affiliation(s)
- Alp Ozgun
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, Canada
| | - David J. Lomboni
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Amy Aylsworth
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
| | - Allison Macdonald
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, Canada
| | - William A. Staines
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Marzia Martina
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
| | - Michael G. Schlossmacher
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Joseph S. Tauskela
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, Canada
| | - John Woulfe
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Department of Pathology, The Ottawa Hospital, Ottawa, Canada
| | - Fabio Variola
- Department of Mechanical Engineering, Faculty of Engineering, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
- Children's Hospital of Eastern Ontario (CHEO), Ottawa, Canada
| |
Collapse
|
48
|
Zhao HH, Haddad G. Brain organoid protocols and limitations. Front Cell Neurosci 2024; 18:1351734. [PMID: 38572070 PMCID: PMC10987830 DOI: 10.3389/fncel.2024.1351734] [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: 12/06/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Stem cell-derived organoid technology is a powerful tool that revolutionizes the field of biomedical research and extends the scope of our understanding of human biology and diseases. Brain organoids especially open an opportunity for human brain research and modeling many human neurological diseases, which have lagged due to the inaccessibility of human brain samples and lack of similarity with other animal models. Brain organoids can be generated through various protocols and mimic whole brain or region-specific. To provide an overview of brain organoid technology, we summarize currently available protocols and list several factors to consider before choosing protocols. We also outline the limitations of current protocols and challenges that need to be solved in future investigation of brain development and pathobiology.
Collapse
Affiliation(s)
- Helen H. Zhao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
| | - Gabriel Haddad
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, United States
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
- The Rady Children's Hospital, San Diego, CA, United States
| |
Collapse
|
49
|
Kim JI, Imaizumi K, Thete MV, Hudacova Z, Jurjuţ O, Amin ND, Scherrer G, Paşca SP. Human assembloid model of the ascending neural sensory pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584539. [PMID: 38559133 PMCID: PMC10979925 DOI: 10.1101/2024.03.11.584539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The ascending somatosensory pathways convey crucial information about pain, touch, itch, and body part movement from peripheral organs to the central nervous system. Despite a significant need for effective therapeutics modulating pain and other somatosensory modalities, clinical translation remains challenging, which is likely related to species-specific features and the lack of in vitro models to directly probe and manipulate this polysynaptic pathway. Here, we established human ascending somatosensory assembloids (hASA)- a four-part assembloid completely generated from human pluripotent stem cells that integrates somatosensory, spinal, diencephalic, and cortical organoids to model the human ascending spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types in this circuit. Rabies tracing and calcium imaging showed that sensory neurons connected with dorsal spinal cord projection neurons, which ascending axons further connected to thalamic neurons. Following noxious chemical stimulation, single neuron calcium imaging of intact hASA demonstrated coordinated response, while four-part concomitant extracellular recordings and calcium imaging revealed synchronized activity across the assembloid. Loss of the sodium channel SCN9A, which causes pain insensitivity in humans, disrupted synchrony across the four-part hASA. Taken together, these experiments demonstrate the ability to functionally assemble the essential components of the human sensory pathway. These findings could both accelerate our understanding of human sensory circuits and facilitate therapeutic development.
Collapse
|
50
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|