1
|
Sakaguchi H. Self-organization and applications of neural organoids. Eur J Cell Biol 2025; 104:151496. [PMID: 40451097 DOI: 10.1016/j.ejcb.2025.151496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/29/2025] [Accepted: 05/13/2025] [Indexed: 06/16/2025] Open
Abstract
Organoid technology has become a field that attract many researcher's attention and involvement. "Organoid" is a coined word which means organ like-tissue (Organ+oid), and organoid is determined as stem cell-derived three-dimensional (3D) tissues that recapitulate developmental processes and tissue specific function in vivo. Generally, they are derived from pluripotent stem cells (PSCs) including induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs), or from tissue stem cells. The first report that created human 3D cerebral tissue arose in 2008 which is currently considered as the pioneering work of "neural organoid" (Eiraku et al., 2008, Sasai 2013a, Sasai 2013b). The neural organoids provide living human neural tissues that bring opportunities to study human development, human neuroscience, neurological and psychiatric disorders, and evolutions. The neural organoid can be said as "cut & paste" of developmental biological process into a dish. Thus, understanding the background of neural organoid needs developmental knowledge, but current organoid researches looks to use organoid as a tool to study the aim that the researchers want to focus. This leads the organoid research more methodological, and the improvement or sophistication of organoid methods has still been difficult for most of new coming researchers. For this problem, this review provide insights of how to assemble organoid methods from viewpoints of development especially from morphological/structual changes. In this review, I start from the brief history of how neural organoid research emerged from developmental biology. Then I introduce some interesting aspects of neural organoid generation focusing on self-organization of regions and structures. From the viewpoint of a developer of this field, this review also show how to think and adjust the methods to generate novel regional organoids taking hippocampal organoids as an example. Regarding structural self-organization I will introduce cerebral organoid for an example of layer organization in a dish. By showing background knowledge with scientific achievements and interesting aspects, this review will help researchers who want to create novel neural organoids.
Collapse
Affiliation(s)
- Hideya Sakaguchi
- RIKEN Center for Biosystems Dynamics Research, BDR-Otsuka Pharmaceutical Collaboration Center, Neural Organogenesis Laboratory, Research Leader, Japan.
| |
Collapse
|
2
|
Colinet M, Chiver I, Bonafina A, Masset G, Almansa D, Di Valentin E, Twizere JC, Nguyen L, Espuny-Camacho I. SARS-CoV2 infection triggers inflammatory conditions and astrogliosis-related gene expression in long-term human cortical organoids. Stem Cells 2025; 43:sxaf010. [PMID: 40103011 PMCID: PMC12121356 DOI: 10.1093/stmcls/sxaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
SARS-CoV2, severe acute respiratory syndrome coronavirus 2, is frequently associated with neurological manifestations. Despite the presence of mild to severe CNS-related symptoms in a cohort of patients, there is no consensus whether the virus can infect directly brain tissue or if the symptoms in patients are a consequence of peripheral infectivity of the virus. Here, we use long-term human stem cell-derived cortical organoids to assess SARS-CoV2 infectivity of brain cells and unravel the cell-type tropism and its downstream pathological effects. Our results show consistent and reproducible low levels of SARS-CoV2 infection of astrocytes, deep projection neurons, upper callosal neurons, and inhibitory neurons in 6 months of human cortical organoids. Interestingly, astrocytes showed the highest infection rate among all infected cell populations which led to changes in their morphology and upregulation of SERPINA3, CD44, and S100A10 astrogliosis markers. Further, transcriptomic analysis revealed overall changes in expression of genes related to cell metabolism, astrogliosis and, inflammation and further, upregulation of cell survival pathways. Thus, local and minor infectivity of SARS-CoV2 in the brain may induce widespread adverse effects and lead to the resilience of dysregulated neurons and astrocytes within an inflammatory environment.
Collapse
Affiliation(s)
- Mathilde Colinet
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Ioana Chiver
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Antonela Bonafina
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Gérald Masset
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Daniel Almansa
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Emmanuel Di Valentin
- GIGA Viral Vector Platform, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Jean-Claude Twizere
- Laboratory of Viral Interactomes, Unit of Molecular Biology of Diseases, GIGA Institute, University of Liège, Liège 4000, Belgium
| | - Laurent Nguyen
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
- WELBIO Department, WEL Research Institute, Wavre 1300, Belgium
| | - Ira Espuny-Camacho
- Laboratory of Molecular Regulation of Neurogenesis, GIGA Institute, University of Liège, Liège 4000, Belgium
- GIGA HIPS, GIGA Institute, University of Liège, Liège 4000, Belgium
| |
Collapse
|
3
|
Park SB, Jo JH, Kim SS, Jung WH, Bae MA, Koh B, Kim KY. Microplastics Accumulation Induces Kynurenine-Derived Neurotoxicity in Cerebral Organoids and Mouse Brain. Biomol Ther (Seoul) 2025; 33:447-457. [PMID: 40181595 PMCID: PMC12059365 DOI: 10.4062/biomolther.2024.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 04/05/2025] Open
Abstract
Microplastics (MP) are pervasive environmental pollutants with potential adverse effects on human health, particularly concerning neurotoxicity. This study investigates the accumulation and neurotoxic effects of MP in cerebral organoids and mouse brains. Utilizing in vitro cerebral organoids and in vivo mouse models, we examined the penetration of MP, revealing that smaller MP (50 nm) infiltrated deeper into the organoids compared to larger ones (100 nm). Exposure to 50 nm MP resulted in a significant reduction in organoid viability. Furthermore, total RNA sequencing indicated substantial alterations in neurotoxicity-related gene expression. In vivo, MP-treated mice exhibited notable DNA fragmentation in the hippocampus and cortex, alongside elevated levels of inflammatory markers and neurotoxic metabolites, such as kynurenine (KYN) and 3-hydroxykynurenine (3-HK). Our findings suggest that MP may promote neurotoxicity through the kynurenine pathway, leading to heightened levels of neurotoxic compounds like quinolinic acid. This research highlights the potential for MP to induce neuroinflammatory responses and disrupt normal brain function, underscoring the need for further investigation into the long-term effects of MP exposure on neurological health.
Collapse
Affiliation(s)
- Sung Bum Park
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Jeong Hyeon Jo
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Seong Soon Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Won Hoon Jung
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Myung-Ae Bae
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Byumseok Koh
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Medicinal Chemistry & Pharmacology, University of Science & Technology (UST), Daejeon 34113, Republic of Korea
| | - Ki Young Kim
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| |
Collapse
|
4
|
Dolgin E. Brain tissues, assemble! Inside the push to build better brain models. Nature 2025; 641:809-812. [PMID: 40369142 DOI: 10.1038/d41586-025-01468-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
|
5
|
Birtele M, Lancaster M, Quadrato G. Modelling human brain development and disease with organoids. Nat Rev Mol Cell Biol 2025; 26:389-412. [PMID: 39668188 DOI: 10.1038/s41580-024-00804-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 12/14/2024]
Abstract
Organoids are systems derived from pluripotent stem cells at the interface between traditional monolayer cultures and in vivo animal models. The structural and functional characteristics of organoids enable the modelling of early stages of brain development in a physiologically relevant 3D environment. Moreover, organoids constitute a tool with which to analyse how individual genetic variation contributes to the susceptibility and progression of neurodevelopmental disorders. This Roadmap article describes the features of brain organoids, focusing on the neocortex, and their advantages and limitations - in comparison with other model systems - for the study of brain development, evolution and disease. We highlight avenues for enhancing the physiological relevance of brain organoids by integrating bioengineering techniques and unbiased high-throughput analyses, and discuss future applications. As organoids advance in mimicking human brain functions, we address the ethical and societal implications of this technology.
Collapse
Affiliation(s)
- Marcella Birtele
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Madeline Lancaster
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Sun Y, Ikeuchi Y, Guo F, Hyun I, Ming GL, Fu J. Bioengineering innovations for neural organoids with enhanced fidelity and function. Cell Stem Cell 2025; 32:689-709. [PMID: 40315834 PMCID: PMC12052258 DOI: 10.1016/j.stem.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/19/2025] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Neural organoids have been utilized to recapitulate different aspects of the developing nervous system. While hailed as promising experimental tools for studying human neural development and neuropathology, current neural organoids do not fully recapitulate the anatomy or microcircuitry-level functionality of the developing brain, spinal cord, or peripheral nervous system. In this review, we discuss emerging bioengineering approaches that control morphogen signals and biophysical microenvironments, which have improved the efficiency, fidelity, and utility of neural organoids. Furthermore, advancements in bioengineered tools have facilitated more sophisticated analyses of neural organoid functions and applications, including improved neural-bioelectronic interfaces and organoid-based information processing. Emerging bioethical issues associated with advanced neural organoids are also discussed. Future opportunities of neural organoid research lie in enhancing their fidelity, maturity, and complexity and expanding their applications in a scalable manner.
Collapse
Affiliation(s)
- Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA.
| | - Yoshiho Ikeuchi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-8654, Japan
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, Bloomington, IN 47408, USA
| | - Insoo Hyun
- Center for Life Sciences and Public Learning, Museum of Science, Boston, MA 02114, USA; Center for Bioethics, Harvard Medical School, Boston, MA 02115, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School of Medicine, Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Marín O. Development of GABAergic Interneurons in the Human Cerebral Cortex. Eur J Neurosci 2025; 61:e70136. [PMID: 40356226 PMCID: PMC12069972 DOI: 10.1111/ejn.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025]
Abstract
GABAergic interneurons are critical regulators of information processing in the cerebral cortex. They constitute a heterogeneous group of neurons with unique spatial and temporal capabilities to control information flow and influence neural network dynamics through inhibitory and disinhibitory mechanisms. Interneuron diversity is largely conserved between rodents and primates, which indicates that the addition of new types of GABAergic neurons is not the most critical innovation of the primate cortex. In contrast, interneurons are much more abundant and seem more widely interconnected in the cerebral cortex of primates than in rodents, suggesting selective evolutionary pressure in the mechanisms regulating the generation, survival and maturation of cortical interneurons. Recent studies are beginning to shed light on the cellular and molecular mechanisms controlling the development of cortical interneurons in humans, from their generation in the embryonic telencephalon to their early integration in cortical networks. These studies identified many features in the development of human cortical interneurons that are shared with other mammals, along with distinctive features that seem characteristic of the primate brain, such as a previously unrecognised protracted period of neurogenesis and migration that extends the earliest stages of interneuron development into the first months of postnatal life in humans.
Collapse
Affiliation(s)
- Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- Medical Research Council Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| |
Collapse
|
8
|
Li CV, Knoblich JA. Advancing autism research: Insights from brain organoid modeling. Curr Opin Neurobiol 2025; 92:103030. [PMID: 40279814 DOI: 10.1016/j.conb.2025.103030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
Autism Spectrum Disorders (ASD) are characterized by a variety of behavioral symptoms and a complex genetic architecture, posing significant challenges in understanding the mechanistic processes underlying their pathology. Despite extensive research, the mechanisms linking genetic variations to the phenotypic outcomes associated with ASD remain elusive. Consistent evidence indicates disruptions in early brain development among individuals with ASD. The advent of brain organoids offers a unique opportunity for uncovering, how brain development changes in ASD patients. Brain organoids are three-dimensional in vitro model systems derived from pluripotent stem cells that recapitulate early human brain development across multiple biological levels. They have become an invaluable tool for studying human-specific brain development processes and neurodevelopmental disorders. In this review, we discuss recent findings using brain organoid technologies to model ASD and discuss, how these new technologies can enhance our understanding of ASD genetics and pathology at the molecular, cellular, and tissue levels.
Collapse
Affiliation(s)
- Chong V Li
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna, Austria; Beijing Institute for Brain Research, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China; Chinese Institute for Brain Research, Beijing, PR China.
| | - Jürgen A Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna, Austria; Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Mostajo-Radji MA, Leon WRM, Breevoort A, Gonzalez-Ferrer J, Schweiger HE, Lehrer J, Zhou L, Schmitz MT, Perez Y, Mukhtar T, Robbins A, Chu J, Andrews MG, Sullivan FN, Tejera D, Choy EC, Paredes MF, Teodorescu M, Kriegstein AR, Alvarez-Buylla A, Pollen AA. Fate plasticity of interneuron specification. iScience 2025; 28:112295. [PMID: 40264797 PMCID: PMC12013500 DOI: 10.1016/j.isci.2025.112295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/21/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Neuronal subtype generation in the mammalian central nervous system is governed by competing genetic programs. The medial ganglionic eminence (MGE) produces two major cortical interneuron (IN) populations, somatostatin (Sst) and parvalbumin (Pvalb), which develop on different timelines. The extent to which external signals influence these identities remains unclear. Pvalb-positive INs are crucial for cortical circuit regulation but challenging to model in vitro. We grafted mouse MGE progenitors into diverse 2D and 3D co-culture systems, including mouse and human cortical, MGE, and thalamic models. Strikingly, only 3D human corticogenesis models promoted efficient, non-autonomous Pvalb differentiation, characterized by upregulation of Pvalb maturation markers, downregulation of Sst-specific markers, and the formation of perineuronal nets. Additionally, lineage-traced postmitotic Sst-positive INs upregulated Pvalb when grafted onto human cortical models. These findings reveal unexpected fate plasticity in MGE-derived INs, suggesting that their identities can be dynamically shaped by the environment.
Collapse
Affiliation(s)
- Mohammed A. Mostajo-Radji
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Walter R. Mancia Leon
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arnar Breevoort
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jesus Gonzalez-Ferrer
- 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
| | - 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
| | - Julian Lehrer
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Li Zhou
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew T. Schmitz
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yonatan Perez
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tanzila Mukhtar
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, 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
| | - Julia Chu
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Madeline G. Andrews
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Dario Tejera
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric C. Choy
- 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
| | - Mercedes F. Paredes
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, 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
| | - Arnold R. Kriegstein
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arturo Alvarez-Buylla
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alex A. Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
10
|
Wu SR, Nowakowski TJ. Exploring human brain development and disease using assembloids. Neuron 2025; 113:1133-1150. [PMID: 40107269 PMCID: PMC12022838 DOI: 10.1016/j.neuron.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/10/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
How the human brain develops and what goes awry in neurological disorders represent two long-lasting questions in neuroscience. Owing to the limited access to primary human brain tissue, insights into these questions have been largely gained through animal models. However, there are fundamental differences between developing mouse and human brain, and neural organoids derived from human pluripotent stem cells (hPSCs) have recently emerged as a robust experimental system that mimics self-organizing and multicellular features of early human brain development. Controlled integration of multiple organoids into assembloids has begun to unravel principles of cell-cell interactions. Moreover, patient-derived or genetically engineered hPSCs provide opportunities to investigate phenotypic correlates of neurodevelopmental disorders and to develop therapeutic hypotheses. Here, we outline the advances in technologies that facilitate studies by using assembloids and summarize their applications in brain development and disease modeling. Lastly, we discuss the major roadblocks of the current system and potential solutions.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
11
|
Maisumu G, Willerth S, Nestor M, Waldau B, Schülke S, Nardi FV, Ahmed O, Zhou Y, Durens M, Liang B, Yakoub AM. Brain organoids: building higher-order complexity and neural circuitry models. Trends Biotechnol 2025:S0167-7799(25)00046-0. [PMID: 40221251 DOI: 10.1016/j.tibtech.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/09/2024] [Accepted: 02/07/2025] [Indexed: 04/14/2025]
Abstract
Brain organoids are 3D tissue models of the human brain that are derived from pluripotent stem cells (PSCs). They have enabled studies that were previously stymied by the inaccessibility of human brain tissue or the limitations of mouse models of some brain diseases. Despite their enormous potential, brain organoids have had significant limitations that prevented them from recapitulating the full complexity of the human brain and reduced their utility in disease studies. We describe recent progress in addressing these limitations, especially building complex organoids that recapitulate the interactions between multiple brain regions, and reconstructing in vitro the neural circuitry present in in vivo. These major advances in the human brain organoid technology will remarkably facilitate brain disease modeling and neuroscience research.
Collapse
Affiliation(s)
- Gulimiheranmu Maisumu
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA; Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - Stephanie Willerth
- Department of Biomedical Engineering, University of Victoria, Victoria, BC, Canada
| | - Michael Nestor
- National Academies of Sciences, Engineering, and Medicine, Washington, DC, USA
| | - Ben Waldau
- Department of Neurological Surgery, University of California Davis, Sacramento, CA, USA
| | - Stefan Schülke
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany; Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Francesco V Nardi
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA; Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - Osama Ahmed
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA; Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - You Zhou
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA
| | - Madel Durens
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bo Liang
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND, USA
| | - Abraam M Yakoub
- Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Peng T, Ma X, Hua W, Wang C, Chu Y, Sun M, Fermi V, Hamelmann S, Lindner K, Shao C, Zaman J, Tian W, Zhuo Y, Harim Y, Stöffler N, Hammann L, Xiao Q, Jin X, Warta R, Lotsch C, Zhuang X, Feng Y, Fu M, Zhang X, Zhang J, Xu H, Qiu F, Xie L, Zhang Y, Zhu W, Du Z, Salgueiro L, Schneider M, Eichhorn F, Lefevre A, Pusch S, Grinevich V, Ratliff M, Loges S, Bunse L, Sahm F, Xiang Y, Unterberg A, von Deimling A, Platten M, Herold-Mende C, Wu Y, Liu HK, Mao Y. Individualized patient tumor organoids faithfully preserve human brain tumor ecosystems and predict patient response to therapy. Cell Stem Cell 2025; 32:652-669.e11. [PMID: 39938519 DOI: 10.1016/j.stem.2025.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/27/2024] [Accepted: 01/03/2025] [Indexed: 02/14/2025]
Abstract
Tumor organoids are important tools for cancer research, but current models have drawbacks that limit their applications for predicting response to therapy. Here, we developed a fast, efficient, and complex culture system (IPTO, individualized patient tumor organoid) that accurately recapitulates the cellular and molecular pathology of human brain tumors. Patient-derived tumor explants were cultured in induced pluripotent stem cell (iPSC)-derived cerebral organoids, thus enabling culture of a wide range of human tumors in the central nervous system (CNS), including adult, pediatric, and metastatic brain cancers. Histopathological, genomic, epigenomic, and single-cell RNA sequencing (scRNA-seq) analyses demonstrated that the IPTO model recapitulates cellular heterogeneity and molecular features of original tumors. Crucially, we showed that the IPTO model predicts patient-specific drug responses, including resistance mechanisms, in a prospective patient cohort. Collectively, the IPTO model represents a major breakthrough in preclinical modeling of human cancers, which provides a path toward personalized cancer therapy.
Collapse
Affiliation(s)
- Tianping Peng
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiujian Ma
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Changwen Wang
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Valentina Fermi
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Stefan Hamelmann
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Katharina Lindner
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Chunxuan Shao
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Julia Zaman
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Weili Tian
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Yue Zhuo
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Yassin Harim
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Nadja Stöffler
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Linda Hammann
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Qungen Xiao
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Xiaoliang Jin
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Catharina Lotsch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Feng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Xin Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Jinsen Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Fufang Qiu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Liqian Xie
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Yi Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China
| | - Zunguo Du
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lorena Salgueiro
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim 68167, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Mark Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University, Heidelberg 69120, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Florian Eichhorn
- Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, Roentgenstrasse 1, Heidelberg 69126, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Arthur Lefevre
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Stefan Pusch
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Miriam Ratliff
- DKTK Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Research Center (DKFZ), Department of Neurosurgery, University Hospital Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Sonja Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim 68167, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Personalized Oncology, University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany; Translational Lung Research Center Heidelberg (TRLC), German Center for Lung Research (DZL), Heidelberg 69120, Germany
| | - Lukas Bunse
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany
| | - Felix Sahm
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Andreas Unterberg
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Andreas von Deimling
- Deptment of Neuropathology, University Hospital Heidelberg, CCU Neuropathology, German Cancer Research Center (DKFZ), University Heidelberg, Heidelberg 69120, Germany
| | - Michael Platten
- DKTK Clinical Cooperation Unit (CCU) Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Helmholtz Institute of Translational Oncology Mainz (HI-TRON Mainz) - a Helmholtz Institute of the DKFZ, Mainz 55131, Germany; Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Tanslational Neuroscience (MCTN), Heidelberg University, Heidelberg 69120, Germany; Immune Monitoring Unit, National Center for Tumor Diseases (NCT), Heidelberg 69120, Germany; German Cancer Consortium (DKTK), DKFZ, Core Center, Heidelberg 69120, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, Heidelberg 69120, Germany
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| | - Hai-Kun Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University; Shanghai Clinical Research and Trial Center, Shanghai 201210, China; Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Im Neuenheimer Feld 581, Heidelberg 69120, Germany.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University; National Center for Neurological Disorders, Shanghai 200040, China.
| |
Collapse
|
13
|
Choe MS, Lo C, Park IH. Modeling forebrain regional development and connectivity by human brain organoids. Curr Opin Genet Dev 2025; 91:102324. [PMID: 39983347 DOI: 10.1016/j.gde.2025.102324] [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: 11/05/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/23/2025]
Abstract
The forebrain is one of the most important brain structures for modern human existence, which houses the uniquely sophisticated social and cognitive functions that distinguish our species. Therefore, modeling the forebrain development by using human cells is especially critical for our understanding of the intricacies of human development and devising treatments for related diseases. Recent advancements in brain organoid fields have offered unprecedented tools to investigate forebrain development from studies on specific regions to exploring tract formation and connectivity between different regions of the forebrain. In this review, we discuss the developmental biology of the forebrain and diverse methods for modeling its development by using organoids.
Collapse
Affiliation(s)
- Mu Seog Choe
- Interdepartmental Neuroscience Program, Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States
| | - Cynthia Lo
- Interdepartmental Neuroscience Program, Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States
| | - In-Hyun Park
- Interdepartmental Neuroscience Program, Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Wu Tsai Institute, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
14
|
Kaiser VM, Gonzalez-Cordero A. Organoids - the future of pre-clinical development of AAV gene therapy for CNS disorders. Gene Ther 2025:10.1038/s41434-025-00527-8. [PMID: 40148593 DOI: 10.1038/s41434-025-00527-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Advancements in our understanding of genetic disease and adeno-associated virus has prompted great excitement into the field of AAV-mediated gene therapy, particularly for genetic diseases of the central nervous system, including retinal disorders. Despite significant progress, exemplified by the approval of therapies such as Luxturna® and Zolgensma®, a substantial number of therapies remain in pre-clinical or early clinical stages, with many failing to advance to later phases. Whilst the use of animal models to test safety and delivery route efficacy of AAV treatments is imperative, differences in tissue structure and physiology between humans and animal models has restricted precise disease modelling and gene therapy development for many CNS disorders. Alongside the FDA push for non-animal alternative models, researchers are increasingly turning to human-based models, including stem cell-derived organoids, which can offer a more accurate representation of human cellular microenvironments and niches. As such, this review explores the advantages and limitations of brain and retinal organoids as pre-clinical models of disease, with a primary focus on their utility in identifying novel AAV capsids, cell-specific promoters, and their role in recent pre-clinical AAV gene therapy studies.
Collapse
Affiliation(s)
- Vivienne M Kaiser
- Stem Cell Medicine Unit, Children's Medical Research Institute, Westmead, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Unit, Children's Medical Research Institute, Westmead, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Lin Z, Wang W, Liu R, Li Q, Lee J, Hirschler C, Liu J. Cyborg organoids integrated with stretchable nanoelectronics can be functionally mapped during development. Nat Protoc 2025:10.1038/s41596-025-01147-7. [PMID: 40140634 DOI: 10.1038/s41596-025-01147-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/31/2024] [Indexed: 03/28/2025]
Abstract
Organoids are in vitro miniaturized cellular models of organs that offer opportunities for studying organ development, disease mechanisms and drug screening. Understanding the complex processes governing organoid development and function requires methods suitable for the continuous, long-term monitoring of cell activities (for example, electrophysiological and mechanical activity) at single-cell resolution throughout the entire three-dimensional (3D) structure. Cyborg organoid technology addresses this need by seamlessly integrating stretchable mesh nanoelectronics with tissue-like properties, such as tissue-level flexibility, subcellular feature size and mesh-like networks, into 3D organoids through a 2D-to-3D tissue reconfiguration process during organogenesis. This approach enables longitudinal, tissue-wide, single-cell functional mapping, thereby overcoming the limitations of existing techniques including recording duration, spatial coverage, and the ability to maintain stable contact with the tissue during organoid development. This protocol describes the fabrication and characterization of stretchable mesh nanoelectronics, their electrical performance, their integration with organoids and the acquisition of long-term functional organoid activity requiring multimodal data analysis techniques. Cyborg organoid technology represents a transformative tool for investigating organoid development and function, with potential for improving in vitro disease models, drug screening and personalized medicine. The procedure is suitable for users within a multidisciplinary team with expertise in stem cell biology, tissue engineering, nanoelectronics fabrication, electrophysiology and data science.
Collapse
Affiliation(s)
- Zuwan Lin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wenbo Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Qiang Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jaeyong Lee
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Charles Hirschler
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA.
| |
Collapse
|
16
|
Kataoka M, Niikawa T, Nagaishi N, Lee TL, Erler A, Savulescu J, Sawai T. Beyond consciousness: Ethical, legal, and social issues in human brain organoid research and application. Eur J Cell Biol 2025; 104:151470. [PMID: 39729735 DOI: 10.1016/j.ejcb.2024.151470] [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: 09/02/2024] [Revised: 11/09/2024] [Accepted: 12/15/2024] [Indexed: 12/29/2024] Open
Abstract
This study aims to provide a comprehensive review of the ethical, legal and social issues in human brain organoid research, with a view to different types of research and applications: in vitro research, transplantation into non-human animals, and biocomputing. Despite the academic and societal attention on the possibility that human brain organoids may be conscious, we have identified diverse issues in human brain organoid research and applications. To guide the complex terrain of human brain organoid research and applications, a multidisciplinary approach that integrates ethical, legal, and social perspectives is essential.
Collapse
Affiliation(s)
- Masanori Kataoka
- Uehiro Division for Applied Ethics, Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, Japan
| | - Takuya Niikawa
- Graduate School of Humanities, Kobe University, Hyogo, Japan
| | - Naoya Nagaishi
- Graduate School of Interdisciplinary Information Studies, The University of Tokyo, Tokyo, Japan
| | - Tsung-Ling Lee
- Graduate Institute of Health and Biotechnology Law, Taipei Medical University, Taipei, Taiwan
| | - Alexandre Erler
- Institute of Philosophy of Mind and Cognition, National Yang Ming Chiao Tung University, Taiwan
| | - Julian Savulescu
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Oxford Uehiro Centre for Practical Ethics, Faculty of Philosophy, University of Oxford, Oxford, UK; Biomedical Ethics Research Group, Murdoch Children's Research Institute, Australia; Melbourne Law School, The University of Melbourne, Australia
| | - Tsutomu Sawai
- Uehiro Division for Applied Ethics, Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, Japan; Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Graduate School of Humanities and Social Sciences, Hiroshima University, Hiroshima, Japan; Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
| |
Collapse
|
17
|
Nishimura M, Kodera T, Adachi S, Sato AY, Takeuchi RF, Nonaka H, Hamachi I, Osakada F. Conversion of silent synapses to AMPA receptor-mediated functional synapses in human cortical organoids. Neurosci Res 2025; 212:20-30. [PMID: 39724942 DOI: 10.1016/j.neures.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
Despite the crucial role of synaptic connections and neural activity in the development and organization of cortical circuits, the mechanisms underlying the formation of functional synaptic connections in the developing human cerebral cortex remain unclear. We investigated the development of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR)-mediated synaptic transmission using human cortical organoids (hCOs) derived from induced pluripotent stem cells. Two-photon Ca2⁺ imaging revealed an increase in the frequency and amplitude of spontaneous activity in hCOs on day 80 compared to day 50. Additionally, spontaneous neural activity in late-stage hCOs, but not in early-stage hCOs, was blocked by N-methyl-D-aspartate receptor (NMDAR) and AMPAR antagonists. However, transsynaptic circuit tracing with G-deleted rabies viral vectors indicated a similar number of synaptic connections in early- and late-stage hCOs. Notably, chemical labeling demonstrated a significant increase in AMPAR expression on the postsynaptic membrane and colocalization with NMDARs in late-stage hCOs. These results suggest that hCOs progressively organize excitatory synaptic transmission, concurrent with the transition from silent synapses lacking AMPARs to functional synapses containing NMDARs and AMPARs. This in vitro model of human cortical circuits derived from induced pluripotent stem cells reflects the developmental programs underlying physiological transitions, providing valuable insights into human corticogenesis and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Masatoshi Nishimura
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Tomoki Kodera
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Shota Adachi
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Akinori Y Sato
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Ryosuke F Takeuchi
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan
| | - Hiroshi Nonaka
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya, Japan; Laboratory of Neural Information Processing, Institute for Advanced Research, Nagoya University, Nagoya, Japan; PRESTO/CREST, Japan Science and Technology Agency, Saitama, Japan.
| |
Collapse
|
18
|
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
|
19
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2025; 60:101036. [PMID: 39567282 PMCID: PMC11825288 DOI: 10.1016/j.dmpk.2024.101036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding its neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUD using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUD in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA.
| |
Collapse
|
20
|
Guo D, Yao B, Shao W, Zuo J, Chang Z, Shi J, Hu N, Bao S, Chen M, Fan X, Li X. The Critical Role of YAP/BMP/ID1 Axis on Simulated Microgravity-Induced Neural Tube Defects in Human Brain Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410188. [PMID: 39656892 PMCID: PMC11792043 DOI: 10.1002/advs.202410188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/05/2024] [Indexed: 12/17/2024]
Abstract
Integrated biochemical and biophysical signals regulate embryonic development. Correct neural tube formation is critical for the development of central nervous system. However, the role of microgravity in neurodevelopment and its underlying molecular mechanisms remain unclear. In this study, the effects of stimulated microgravity (SMG) on the development of human brain organoids are investigated. SMG impairs N-cadherin-based adherens junction formation, leading to neural tube defects associated with dysregulated self-renewal capacity and neuroepithelial disorganization in human brain organoids. Bulk gene expression analyses reveal that SMG alters Hippo and BMP signaling in brain organoids. The neuropathological deficits in SMG-treated organoids can be rescued by regulating YAP/BMP/ID1 axis. Furthermore, sing-cell RNA sequencing data show that SMG results in perturbations in the number and function of neural stem and progenitor cell subpopulations. One of these subpopulations senses SMG cues and transmits BMP signals to the subpopulation responsible for tube morphogenesis, ultimately affecting the proliferating cell population. Finally, SMG intervention leads to persistent neurologic damage even after returning to normal gravity conditions. Collectively, this study reveals molecular and cellular abnormalities associated with SMG during human brain development, providing opportunities for countermeasures to maintain normal neurodevelopment in space.
Collapse
Affiliation(s)
- Di Guo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Bin Yao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Wen‐Wei Shao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jia‐Chen Zuo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Zhe‐Han Chang
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Jian‐Xin Shi
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Nan Hu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Shuang‐Qing Bao
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Meng‐Meng Chen
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiu Fan
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| | - Xiao‐Hong Li
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- State Key Laboratory of Advanced Medical Materials and DevicesTianjin300072China
- Haihe Laboratory of Brain‐Computer Interaction and Human‐Machine IntegrationTianjin300072China
| |
Collapse
|
21
|
Tsai YC, Ozaki H, Morikawa A, Shiraiwa K, Pin AP, Salem AG, Phommahasay KA, Sugita BK, Vu CH, Hammad SM, Kamei KI, Watanabe M. Proof of concept for brain organoid-on-a-chip to create multiple domains in forebrain organoids. RSC Adv 2025; 15:3749-3755. [PMID: 39911547 PMCID: PMC11797153 DOI: 10.1039/d4ra04194a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/14/2025] [Indexed: 02/07/2025] Open
Abstract
Brain organoids are three-dimensionally reconstructed brain tissue derived from pluripotent stem cells in vitro. 3D tissue cultures have opened new avenues for exploring development and disease modeling. However, many physiological conditions, including signaling gradients in 3D cultures, have not yet been easily achieved. Here, we introduce brain organoid-on-a-chip platforms that generate signaling gradients that in turn enable the induction of topographic forebrain organoids. This creates a more continuous spectrum of brain regions and provides a more complete mimic of the human brain for evaluating neurodevelopment and disease in unprecedented detail.
Collapse
Affiliation(s)
- Yuan-Chen Tsai
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | - Hajime Ozaki
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | - Ango Morikawa
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University Institute for Advanced Study Yoshida-Ushinomiya-cho, Sakyo-ku Kyoto 606-8501 Japan
| | - Kaori Shiraiwa
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | - Andy Prosvey Pin
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | - Aya Galal Salem
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | | | - Bret Kiyoshi Sugita
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | - Christine Hein Vu
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
| | - Saba Mamoun Hammad
- Programs of Biology and Bioengineering, Divisions of Science and Engineering, New York University Abu Dhabi Abu Dhabi UAE
| | - Ken-Ichiro Kamei
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University Institute for Advanced Study Yoshida-Ushinomiya-cho, Sakyo-ku Kyoto 606-8501 Japan
- Programs of Biology and Bioengineering, Divisions of Science and Engineering, New York University Abu Dhabi Abu Dhabi UAE
- Department of Biomedical Engineering, Tandon School of Engineering, New York University Brooklyn NY 11201 USA
| | - Momoko Watanabe
- Department of Anatomy and Neurobiology, University of California Irvine CA 92697 USA
- Sue & Bill Gross Stem Cell Research Center, School of Medicine, University of California Irvine CA 92697 USA
| |
Collapse
|
22
|
Moss SP, Bakirci E, Feinberg AW. Engineering the 3D structure of organoids. Stem Cell Reports 2025; 20:102379. [PMID: 39706178 PMCID: PMC11784486 DOI: 10.1016/j.stemcr.2024.11.009] [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: 12/27/2023] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024] Open
Abstract
Organoids form through the sel f-organizing capabilities of stem cells to produce a variety of differentiated cell and tissue types. Most organoid models, however, are limited in terms of the structure and function of the tissues that form, in part because it is difficult to regulate the cell type, arrangement, and cell-cell/cell-matrix interactions within these systems. In this article, we will discuss the engineering approaches to generate more complex organoids with improved function and translational relevance, as well as their advantages and disadvantages. Additionally, we will explore how biofabrication strategies can manipulate the cell composition, 3D organization, and scale-up of organoids, thus improving their utility for disease modeling, drug screening, and regenerative medicine applications.
Collapse
Affiliation(s)
- Samuel P Moss
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
23
|
Dionne O, Sabatié S, Laurent B. Deciphering the physiopathology of neurodevelopmental disorders using brain organoids. Brain 2025; 148:12-26. [PMID: 39222411 PMCID: PMC11706293 DOI: 10.1093/brain/awae281] [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: 03/30/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Neurodevelopmental disorders (NDD) encompass a range of conditions marked by abnormal brain development in conjunction with impaired cognitive, emotional and behavioural functions. Transgenic animal models, mainly rodents, traditionally served as key tools for deciphering the molecular mechanisms driving NDD physiopathology and significantly contributed to the development of pharmacological interventions aimed at treating these disorders. However, the efficacy of these treatments in humans has proven to be limited, due in part to the intrinsic constraint of animal models to recapitulate the complex development and structure of the human brain but also to the phenotypic heterogeneity found between affected individuals. Significant advancements in the field of induced pluripotent stem cells (iPSCs) offer a promising avenue for overcoming these challenges. Indeed, the development of advanced differentiation protocols for generating iPSC-derived brain organoids gives an unprecedented opportunity to explore human neurodevelopment. This review provides an overview of how 3D brain organoids have been used to investigate various NDD (i.e. Fragile X syndrome, Rett syndrome, Angelman syndrome, microlissencephaly, Prader-Willi syndrome, Timothy syndrome, tuberous sclerosis syndrome) and elucidate their pathophysiology. We also discuss the benefits and limitations of employing such innovative 3D models compared to animal models and 2D cell culture systems in the realm of personalized medicine.
Collapse
Affiliation(s)
- Olivier Dionne
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Salomé Sabatié
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
| | - Benoit Laurent
- Research Center on Aging, Centre Intégré Universitaire de Santé et Services Sociaux de l'Estrie-Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 4C4, Canada
- Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5H4, Canada
| |
Collapse
|
24
|
Imani Farahani N, Lin L, Nazir S, Naderi A, Rokos L, McIntosh AR, Julian LM. Advances in physiological and clinical relevance of hiPSC-derived brain models for precision medicine pipelines. Front Cell Neurosci 2025; 18:1478572. [PMID: 39835290 PMCID: PMC11743572 DOI: 10.3389/fncel.2024.1478572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Precision, or personalized, medicine aims to stratify patients based on variable pathogenic signatures to optimize the effectiveness of disease prevention and treatment. This approach is favorable in the context of brain disorders, which are often heterogeneous in their pathophysiological features, patterns of disease progression and treatment response, resulting in limited therapeutic standard-of-care. Here we highlight the transformative role that human induced pluripotent stem cell (hiPSC)-derived neural models are poised to play in advancing precision medicine for brain disorders, particularly emerging innovations that improve the relevance of hiPSC models to human physiology. hiPSCs derived from accessible patient somatic cells can produce various neural cell types and tissues; current efforts to increase the complexity of these models, incorporating region-specific neural tissues and non-neural cell types of the brain microenvironment, are providing increasingly relevant insights into human-specific neurobiology. Continued advances in tissue engineering combined with innovations in genomics, high-throughput screening and imaging strengthen the physiological relevance of hiPSC models and thus their ability to uncover disease mechanisms, therapeutic vulnerabilities, and tissue and fluid-based biomarkers that will have real impact on neurological disease treatment. True physiological understanding, however, necessitates integration of hiPSC-neural models with patient biophysical data, including quantitative neuroimaging representations. We discuss recent innovations in cellular neuroscience that can provide these direct connections through generative AI modeling. Our focus is to highlight the great potential of synergy between these emerging innovations to pave the way for personalized medicine becoming a viable option for patients suffering from neuropathologies, particularly rare epileptic and neurodegenerative disorders.
Collapse
Affiliation(s)
- Negin Imani Farahani
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa Lin
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Shama Nazir
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Alireza Naderi
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Leanne Rokos
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Rotman Research Institute, Baycrest Health Sciences, University of Toronto, Toronto, ON, Canada
| | - Anthony Randal McIntosh
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Lisa M. Julian
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
- Institute for Neuroscience and Neurotechnology, Simon Fraser University, Burnaby, BC, Canada
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
25
|
Yin Y, Zhou W, Zhu J, Chen Z, Jiang L, Zhuang X, Chen J, Wei J, Lu X, Liu Y, Pang W, Zhang Q, Cao Y, Li Z, Zhu Y, Xiang Y. Generation of self-organized neuromusculoskeletal tri-tissue organoids from human pluripotent stem cells. Cell Stem Cell 2025; 32:157-171.e8. [PMID: 39657678 DOI: 10.1016/j.stem.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/26/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024]
Abstract
The human body function requires crosstalk between different tissues. An essential crosstalk is in the neuromusculoskeletal (NMS) axis involving neural, muscular, and skeletal tissues, which is challenging to model using human cells. Here, we describe the generation of three-dimensional, NMS tri-tissue organoids (hNMSOs) from human pluripotent stem cells through a co-development strategy. Staining, single-nucleus RNA sequencing, and spatial transcriptome profiling revealed the co-emergence and self-organization of neural, muscular, and skeletal lineages within individual organoids, and the neural domains of hNMSOs obtained a ventral-specific identity and produced motor neurons innervating skeletal muscles. The neural, muscular, and skeletal regions of hNMSOs exhibited maturation and established functional connections during development. Notably, structural, functional, and transcriptomic analyses revealed that skeletal support in hNMSOs benefited human muscular development. Modeling with hNMSOs also unveiled the neuromuscular alterations following pathological skeletal degeneration. Together, our study provides an accessible experimental model for future studies of human NMS crosstalk and abnormality.
Collapse
Affiliation(s)
- Yao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinkui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziling Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jia Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoxiang Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yantong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Pang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qinzhi Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yajing Cao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhuoya Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuyan Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
26
|
Mathis K, Meckes B. Precision Cell-Cell Assembly Through Light-Mediated DNA Interactions. Methods Mol Biol 2025; 2902:173-182. [PMID: 40029603 DOI: 10.1007/978-1-0716-4402-7_11] [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: 03/05/2025]
Abstract
Complex interactions between diverse cell populations influence processes ranging from cell division to programmed cell death. Replicating this complexity with reproducibility remains challenging. We introduce an innovative approach that combines DNA-mediated interactions with photolithography to achieve controlled cell-cell interactions. By coating cells with DNA sequences responsive to light, we enable meticulous spatial organization through light activation, facilitating rapid and programmable construction of intricate cell structures. This method not only addresses the challenges of orchestrating cell arrangements in vitro, particularly in three-dimensional settings but also offers a new avenue for the on-demand creation of complex cellular architectures.
Collapse
Affiliation(s)
- Katelyn Mathis
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
- BioDiscovery Institute, University of North Texas, Denton, TX, USA
| | - Brian Meckes
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA.
- BioDiscovery Institute, University of North Texas, Denton, TX, USA.
| |
Collapse
|
27
|
Lu R, Xu Y, Li H, Xiong M, Zhou W, Feng W, Zhao R. Identifying the Pathogenicity of a Novel NPRL3 Missense Mutation Using Personalized Cortical Organoid Model of Focal Cortical Dysplasia. J Mol Neurosci 2024; 75:3. [PMID: 39729176 DOI: 10.1007/s12031-024-02304-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Focal cortical dysplasia (FCD) II is a cortical malformation characterized by cortical architectural abnormalities, dysmorphic neurons, with or without balloon cells. Here, we systematically explored the pathophysiological role of the GATOR1 subunit NPRL3 variants including a novel mutation from iPSCs derived from one FCD II patient. Three FCD II children aged 0.5-7 years who underwent cerebral lesion resection in our hospital from March 2019 to October 2019 were included in this study. We generated patient-derived iPSCs and performed whole-exome sequencing to accurately identify somatic cells with mutations. The effect of the newly identified NPRL3 mutation found in one of our FCD II patients was evaluated using the personalized cortical organoid model and the NPRL3 knockout HEK293T cells. Whole-exome sequencing of iPSCs derived from FCD II patients revealed a novel NPRL3 C.767G > C (p.R256P) heterozygous mutation. Cortical organoids generated from iPSCs of FCD II patients were larger than control iPSCs, with increased number of p-S6+ cells and NeuN+ neurons. In NPRL3 knockout HEK293T cells, overexpression of NPRL3 together with NPRL2 protein is necessary to reduce p-S6 level upon amino acid starvation. The reduced binding between NPRL3 Arg256Pro and NPRL2 protein leads to downregulation of the relative total protein amount of both proteins in the cell. Our study describes a novel cortical organoid model generated from iPSCs of the FCD patients to investigate the underlying mechanism of NPRL3-related epilepsy. The mutation of NPRL3 Arg256Pro impaired the function of NPRL3 protein via affecting the binding with NPRL2 protein, which resulted in unstable protein monomer.
Collapse
Affiliation(s)
- Rongrong Lu
- Department of Neurosurgery, National Children's Medical Center (Shanghai), Children's Hospital of Fudan University, No.399 Wan Yuan Avenue, Minhang District, Shanghai, 201102, China
| | - Ying Xu
- Institute of Pediatrics, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Children's Hospital of Fudan University, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Hao Li
- Department of Neurosurgery, National Children's Medical Center (Shanghai), Children's Hospital of Fudan University, No.399 Wan Yuan Avenue, Minhang District, Shanghai, 201102, China
- Department of Neurosurgery, Xiamen Children's Hospital, Children's Hospital of Fudan University at Xiamen, Xiamen, 361006, China
| | - Man Xiong
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wenhao Zhou
- Division of Neonatology and Center for Newborn Care, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Weijun Feng
- Institute of Pediatrics, Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Children's Hospital of Fudan University, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Fujian Key Laboratory of Neonatal Diseases, Xiamen Key Laboratory of Neonatal Diseases, Xiamen Children's Hospital, Children's Hospital of Fudan University at Xiamen, Xiamen, 361006, China.
| | - Rui Zhao
- Department of Neurosurgery, National Children's Medical Center (Shanghai), Children's Hospital of Fudan University, No.399 Wan Yuan Avenue, Minhang District, Shanghai, 201102, China.
- Department of Neurosurgery, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China.
| |
Collapse
|
28
|
Boutom SM, Silva TP, Palecek SP, Shusta EV, Fernandes TG, Ashton RS. Central nervous system vascularization in human embryos and neural organoids. Cell Rep 2024; 43:115068. [PMID: 39693224 PMCID: PMC11975460 DOI: 10.1016/j.celrep.2024.115068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
In recent years, neural organoids derived from human pluripotent stem cells (hPSCs) have offered a transformative pre-clinical platform for understanding central nervous system (CNS) development, disease, drug effects, and toxicology. CNS vasculature plays an important role in all these scenarios; however, most published studies describe CNS organoids that lack a functional vasculature or demonstrate rudimentary incorporation of endothelial cells or blood vessel networks. Here, we review the existing knowledge of vascularization during the development of different CNS regions, including the brain, spinal cord, and retina, and compare it to vascularized CNS organoid models. We highlight several areas of contrast where further bioengineering innovation is needed and discuss potential applications of vascularized neural organoids in modeling human CNS development, physiology, and disease.
Collapse
Affiliation(s)
- Sarah M Boutom
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Teresa P Silva
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tiago G Fernandes
- Department of Bioengineering and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.
| | - Randolph S Ashton
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
29
|
Werner JM, Gillis J. Meta-analysis of single-cell RNA sequencing co-expression in human neural organoids reveals their high variability in recapitulating primary tissue. PLoS Biol 2024; 22:e3002912. [PMID: 39621752 PMCID: PMC11637388 DOI: 10.1371/journal.pbio.3002912] [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: 11/19/2023] [Revised: 12/12/2024] [Accepted: 10/24/2024] [Indexed: 12/14/2024] Open
Abstract
Human neural organoids offer an exciting opportunity for studying inaccessible human-specific brain development; however, it remains unclear how precisely organoids recapitulate fetal/primary tissue biology. We characterize field-wide replicability and biological fidelity through a meta-analysis of single-cell RNA-sequencing data for first and second trimester human primary brain (2.95 million cells, 51 data sets) and neural organoids (1.59 million cells, 173 data sets). We quantify the degree primary tissue cell type marker expression and co-expression are recapitulated in organoids across 10 different protocol types. By quantifying gene-level preservation of primary tissue co-expression, we show neural organoids lie on a spectrum ranging from virtually no signal to co-expression indistinguishable from primary tissue, demonstrating a high degree of variability in biological fidelity among organoid systems. Our preserved co-expression framework provides cell type-specific measures of fidelity applicable to diverse neural organoids, offering a powerful tool for uncovering unifying axes of variation across heterogeneous neural organoid experiments.
Collapse
Affiliation(s)
- Jonathan M. Werner
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Jesse Gillis
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
- Physiology Department and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| |
Collapse
|
30
|
Ryu JR, Ko K, Sun W. Polarization of organoids by bioengineered symmetry breaking. IBRO Neurosci Rep 2024; 17:22-31. [PMID: 38881849 PMCID: PMC11176950 DOI: 10.1016/j.ibneur.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/08/2024] [Indexed: 06/18/2024] Open
Abstract
Symmetry breaking leading to axis formation and spatial patterning is crucial for achieving more accurate recapitulation of human development in organoids. While these processes can occur spontaneously by self-organizing capabilities of pluripotent stem cells, they can often result in variation in structure and composition of cell types within organoids. To address this limitation, bioengineering techniques that utilize geometric, topological and stiffness factors are increasingly employed to enhance control and consistency. Here, we review how spontaneous manners and engineering tools such as micropattern, microfluidics, biomaterials, etc. can facilitate the process of symmetry breaking leading to germ layer patterning and the formation of anteroposterior and dorsoventral axes in blastoids, gastruloids, neuruloids and neural organoids. Furthermore, brain assembloids, which are composed of multiple brain regions through fusion processes are discussed. The overview of organoid polarization in terms of patterning tools can offer valuable insights for enhancing the physiological relevance of organoid system.
Collapse
Affiliation(s)
- Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Kahee Ko
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
31
|
Colonna M, Konopka G, Liddelow SA, Nowakowski T, Awatramani R, Bateup HS, Cadwell CR, Caglayan E, Chen JL, Gillis J, Kampmann M, Krienen F, Marsh SE, Monje M, O'Dea MR, Patani R, Pollen AA, Quintana FJ, Scavuzzo M, Schmitz M, Sloan SA, Tesar PJ, Tollkuhn J, Tosches MA, Urbanek ME, Werner JM, Bayraktar OA, Gokce O, Habib N. Implementation and validation of single-cell genomics experiments in neuroscience. Nat Neurosci 2024; 27:2310-2325. [PMID: 39627589 DOI: 10.1038/s41593-024-01814-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 10/15/2024] [Indexed: 12/13/2024]
Abstract
Single-cell or single-nucleus transcriptomics is a powerful tool for identifying cell types and cell states. However, hypotheses derived from these assays, including gene expression information, require validation, and their functional relevance needs to be established. The choice of validation depends on numerous factors. Here, we present types of orthogonal and functional validation experiment to strengthen preliminary findings obtained using single-cell and single-nucleus transcriptomics as well as the challenges and limitations of these approaches.
Collapse
Affiliation(s)
- Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Genevieve Konopka
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Tomasz Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
| | - Rajeshwar Awatramani
- Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| | - Helen S Bateup
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Cathryn R Cadwell
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Emre Caglayan
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jerry L Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Center for Neurophotonics, Boston University, Boston, MA, USA
- Department of Biology, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Jesse Gillis
- Department of Physiology and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Fenna Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Samuel E Marsh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Michael R O'Dea
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marissa Scavuzzo
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, OH, USA
- Institute for Glial Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matthew Schmitz
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, OH, USA
- Institute for Glial Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Madeleine E Urbanek
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan M Werner
- Department of Physiology and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Ozgun Gokce
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn, Bonn, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
32
|
Shan Z, Zhao Y, Chen X, Zhan G, Huang J, Yang X, Xu C, Guo N, Xiong Z, Wu F, Liu Y, Liu H, Chen B, Chen B, Sun J, He J, Guo Y, Cao S, Wu K, Mao R, Wu G, Lin L, Zou X, Wang J, Chen J. KMT2D deficiency leads to cellular developmental disorders and enhancer dysregulation in neural-crest-containing brain organoids. Sci Bull (Beijing) 2024; 69:3533-3546. [PMID: 39327125 DOI: 10.1016/j.scib.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 05/15/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024]
Abstract
KMT2D, a H3K4me1 methyltransferase primarily regulating enhancers, is a leading cause of KABUKI syndrome. This multisystem disorder leads to craniofacial and cognitive abnormalities, possibly through neural crest and neuronal lineages. However, the impacted cell-of-origin and molecular mechanism of KMT2D during the development of KABUKI disease remains unknown. Here we have optimized a brain organoid model to investigate neural crest and neuronal differentiation. To pinpoint KMT2D's enhancer target, we developed a genome-wide cis-regulatory element explorer (GREE) based on single-cell multiomic integration. Single cell RNA-seq revealed that KMT2D-knockout (KO) and patient-derived organoids exhibited neural crest deformities and GABAergic overproduction. Mechanistically, GREE identified that KMT2D targets a roof-plate-like niche cell and activates the niche cell-specific WNT3A enhancer, providing the microenvironment for neural crest and neuronal development. Interestingly, KMT2D-mutated mice displayed decreased WNT3A expression in the diencephalon roof plate, indicating impaired niche cell function. Deleting the WNT3A enhancer in the organoids presented phenotypic similarities to KMT2D-depletion, emphasizing the WNT3A enhancer as the predominant target of KMT2D. Conversely, reactivating WNT signaling in KMT2D-KO rescued the lineage defects by restoring the microenvironment. Overall, our discovery of KMT2D's primary target provides insights for reconciling complex phenotypes of KABUKI syndrome and establishes a new paradigm for dissecting the mechanisms of genetic disorders from genotype to phenotype.
Collapse
Affiliation(s)
- Ziyun Shan
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingying Zhao
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China
| | - Xiuyu Chen
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guodong Zhan
- Child Development and Behavior Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Junju Huang
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Xuejie Yang
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Chongshen Xu
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ning Guo
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China
| | - Zhi Xiong
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Laboratory, Guangzhou 510005, China
| | - Fang Wu
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yujian Liu
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - He Liu
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou 510005, China; The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou 510700, China
| | - Biyuan Chen
- Child Development and Behavior Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Bingqiu Chen
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Jiangping He
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou 510005, China; Guangzhou Laboratory, Guangzhou 510005, China
| | - Yiping Guo
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | | | - Kaixin Wu
- Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou 510005, China
| | - Rui Mao
- Animal Research Center, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | | | - Lihui Lin
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xiaobing Zou
- Child Development and Behavior Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Jie Wang
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| | - Jiekai Chen
- Center for Cell Lineage and Development, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; University of Chinese Academy of Sciences, Beijing 100049, China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China; Center for Cell Lineage and Atlas (CCLA), Bioland Laboratory, Guangzhou 510005, China; Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China; Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
33
|
Walsh RM, Crabtree GW, Kalpana K, Jubierre L, Koo SY, Ciceri G, Gogos JA, Kruglikov I, Studer L. Cortical assembloids support the development of fast-spiking human PVALB+ cortical interneurons and uncover schizophrenia-associated defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.624368. [PMID: 39651135 PMCID: PMC11623588 DOI: 10.1101/2024.11.26.624368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Disruption of parvalbumin positive (PVALB+) cortical interneurons is implicated in the pathogenesis of schizophrenia. However, how these defects emerge during brain development remains poorly understood. The protracted maturation of these cells during postnatal life has made their derivation from human pluripotent stem cells (hPSCs) extremely difficult, precluding hPSC-based disease modeling of their role in neuropsychiatric disease. Here we present a cortical assembloid system that supports the development of PVALB+ cortical interneurons which match the molecular profiles of primary PVALB+ interneurons and display their distinctive electrophysiological features. Further, we characterized cortical interneuron development in a series of CRISPR-generated isogenic structural variants associated with schizophrenia and identified variant-specific phenotypes affecting cortical interneuron migration and the molecular profile of PVALB+ cortical interneurons. These findings offer plausible mechanisms on how the disruption of cortical interneuron development may impact schizophrenia risk and provide the first human experimental platform to study of PVALB+ cortical interneurons.
Collapse
|
34
|
Hongen T, Sakai K, Ito T, Qin XY, Sone H. Human-Induced Pluripotent Stem Cell-Derived Neural Organoids as a Novel In Vitro Platform for Developmental Neurotoxicity Assessment. Int J Mol Sci 2024; 25:12523. [PMID: 39684235 DOI: 10.3390/ijms252312523] [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: 09/29/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
There has been a recent drive to replace in vivo studies with in vitro studies in the field of toxicity testing. Therefore, instead of conventional animal or planar cell culture models, there is an urgent need for in vitro systems whose conditions can be strictly controlled, including cell-cell interactions and sensitivity to low doses of chemicals. Neural organoids generated from human-induced pluripotent stem cells (iPSCs) are a promising in vitro platform for modeling human brain development. In this study, we developed a new tool based on various iPSCs to study and predict chemical-induced toxicity in humans. The model displayed several neurodevelopmental features and showed good reproducibility, comparable to that of previously published models. The results revealed that basic fibroblast growth factor plays a key role in the formation of the embryoid body, as well as complex neural networks and higher-order structures such as layered stacking. Using organoid models, pesticide toxicities were assessed. Cells treated with low concentrations of rotenone underwent apoptosis to a greater extent than those treated with high concentrations of rotenone. Morphological changes associated with the development of neural progenitor cells were observed after exposure to low doses of chlorpyrifos. These findings suggest that the neuronal organoids developed in this study mimic the developmental processes occurring in the brain and nerves and are a useful tool for evaluating drug efficacy, safety, and toxicity.
Collapse
Affiliation(s)
- Tsunehiko Hongen
- Environmental Health and Prevention Research Unit, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
| | - Kenta Sakai
- Environmental Health and Prevention Research Unit, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
| | - Tomohiro Ito
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan
| | - Xian-Yang Qin
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro, Tsurumi, Yokohama 230-0045, Japan
| | - Hideko Sone
- Environmental Health and Prevention Research Unit, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama 245-0066, Japan
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba 305-8506, Japan
| |
Collapse
|
35
|
Street JS, Zourray C, Lignani G. Organoids as a model of status epilepticus. Epilepsy Behav 2024; 161:110145. [PMID: 39556970 DOI: 10.1016/j.yebeh.2024.110145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/20/2024]
Abstract
Status epilepticus (SE) is a neurological emergency that can be studied in animal models, particularly mice. However, these models are labour-intensive and require large numbers of animals, which raises ethical and logistical challenges. Additionally, rodent-based models could lack direct relevance to human physiology. While reduced models offer some insights, they fail to replicate the full complexity of brain connectivity and interactions with other organs. To address this, human forebrain assembloids, formed by both cortical excitatory and subpallial inhibitory neurons, could be an alternative SE model. Assembloids offer a middle ground, enabling high-throughput screening of potential treatments while maintaining relevant human cell biology. This approach could serve as an intermediate step before transitioning to animal models, ultimately reducing the time and number of animals required for SE research. This paper is based on a presentation made at the 9thLondon-Innsbruck Colloquium on Status Epilepticus and Acute Seizures in April 2024.
Collapse
Affiliation(s)
- J S Street
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, United Kingdom
| | - C Zourray
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, United Kingdom
| | - G Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, United Kingdom.
| |
Collapse
|
36
|
Han Y, Yu Z, Chen Y, Guo X, Liu Y, Zhang H, Li Z, Chen L. PM2.5 induces developmental neurotoxicity in cortical organoids. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 361:124913. [PMID: 39245199 DOI: 10.1016/j.envpol.2024.124913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/10/2024]
Abstract
There is mounting evidence implicating the potential neurotoxic effects of PM2.5 during brain development, as it has been observed to traverse both the placental barrier and the fetal blood-brain barrier. However, the current utilization of 2D cell culture and animal models falls short in providing an accurate representation of human brain development. Consequently, the precise mechanisms underlying PM2.5-induced developmental neurotoxicity in humans remain obscure. To address this research gap, we constructed three-dimensional (3D) cortical organoids that faithfully recapitulate the initial stages of human cerebral cortex development. Our goal is to investigate the mechanisms of PM2.5-induced neurotoxicity using 3D brain organoids that express cortical layer proteins. Our findings demonstrate that exposure to PM2.5 concentrations of 5 μg/mL and 50 μg/mL induces neuronal apoptosis and disrupts normal neural differentiation, thereby suggesting a detrimental impact on neurodevelopment. Furthermore, transcriptomic analysis revealed PM2.5 exposure induced aberrations in mitochondrial complex I functionality, which is reminiscent of Parkinson's syndrome, potentially mediated by misguided axon guidance and compromised synaptic maintenance. This study is a pioneering assessment of the neurotoxicity of PM2.5 pollution on human brain tissues based on 3D cortical organoids, and the results are of great significance in guiding the formulation of the next air pollution prevention and control policies in China to achieve the sustainable improvement of air quality and to formulate pollution abatement strategies that can maximize the benefits to public health.
Collapse
Affiliation(s)
- Yuqing Han
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| | - Zhenjie Yu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| | - Yue Chen
- Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin Institute of Urology, Tianjin, 300211, China.
| | - Xiaoyu Guo
- Academy of Medical Engineering and Translational Medicine, Department of Medicine, Tianjin University, Tianjin, 300072, China.
| | - Yeming Liu
- Academy of Medical Engineering and Translational Medicine, Department of Medicine, Tianjin University, Tianjin, 300072, China.
| | - Hao Zhang
- Shanxi Meijin Coal Coking Co. Ltd., Shanxi, 030400, China.
| | - Zhiqing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| | - Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Department of Medicine, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
37
|
Huang R, Zhu Y, Chen H, Yu L, Liu Z, Liu Y, Wang Z, He X, Yang L, Xu X, Bai Y, Chen B, Zhu R. Progress in spinal cord organoid research: advancing understanding of neural development, disease modelling, and regenerative medicine. BIOMATERIALS TRANSLATIONAL 2024; 5:355-371. [PMID: 39872925 PMCID: PMC11764192 DOI: 10.12336/biomatertransl.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 01/30/2025]
Abstract
Stem cell-derived spinal cord organoids (SCOs) have revolutionised the study of spinal cord development and disease mechanisms, offering a three-dimensional model that recapitulates the complexity of native tissue. This review synthesises recent advancements in SCO technology, highlighting their role in modelling spinal cord morphogenesis and their application in neurodegenerative disease research. We discuss the methodological breakthroughs in inducing regional specification and cellular diversity within SCOs, which have enhanced their predictive ability for drug screening and their relevance in mimicking pathological conditions such as neurodegenerative diseases and neuromuscular disorders. Despite these strides, challenges in achieving vascularisation and mature neuronal integration persist. The future of SCOs lies in addressing these limitations, potentially leading to transformative impactions in regenerative medicine and therapeutic development.
Collapse
Affiliation(s)
- Ruiqi Huang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yanjing Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| | - Haokun Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Liqun Yu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Zhibo Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yuchen Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xiaolie He
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Li Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xu Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yuxin Bai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Bairu Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital affiliated to Tongji University, School of Life Science and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, China
| |
Collapse
|
38
|
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
|
39
|
Hong H, Jun Y, Yoon SB, Park S, Lee J, Jang JW, Nam HJ, Cho H. Manufacturing Uniform Cerebral Organoids for Neurological Disease Modeling and Drug Evaluation. Biomater Res 2024; 28:0104. [PMID: 39507522 PMCID: PMC11538552 DOI: 10.34133/bmr.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Human cerebral organoids are promising tools for investigating brain development and the pathogenesis underlying neurological disorders. To use organoids for drug effectiveness and safety screening, the organoids dispensed into each well must be prepared under precisely the same conditions as the cells. Despite decades of extensive research on approaches to improve organoid generation, various challenges remain, such as low yields and heterogeneity in size and differentiation both within and between batches. Here, we newly established uniform cerebral organoids (UCOs) derived from induced pluripotent stem cells by optimizing organoid size and performing real-time monitoring of telencephalic differentiation marker expression. These organoids exhibited morphological uniformity and consistent expression of FOXG1 during telencephalic differentiation, with high productivity. Moreover, UCOs faithfully recapitulated early corticogenesis, concomitant with the establishment of neuroepithelial populations, cortical plate neurons, and glial cells. Furthermore, UCOs systematically developed neural networks and exhibited both excitatory and inhibitory electrophysiological signals when exposed to neurotransmission blockers. Neurodevelopmental disease models derived from UCOs manifested neurite outgrowth defects, which could be ameliorated with targeted drug treatment. We propose UCOs as an advanced platform with low organoid variations and high reproducibility for modeling both brain development and neurological diseases.
Collapse
Affiliation(s)
- Hyowon Hong
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Yesl Jun
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Sae-Bom Yoon
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Seoyoon Park
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jaemeun Lee
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jeong Woon Jang
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Hye Jin Nam
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medicinal Chemistry and Pharmacology,
University of Science and Technology, Daejeon, Republic of Korea
| | - Heeyeong Cho
- Therapeutics & Biotechnology Division,
Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
- Medicinal Chemistry and Pharmacology,
University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
40
|
Teles E Silva AL, Yokota-Moreno BY, Branquinho MS, Salles GR, de Souza TC, de Carvalho RA, Batista G, Varella Branco E, Griesi-Oliveira K, Passos Bueno MR, Porcionatto MA, Herai RH, Gamarra LF, Sertié AL. Generation and characterization of cortical organoids from iPSC-derived dental pulp stem cells using traditional and innovative approaches. Neurochem Int 2024; 180:105854. [PMID: 39241808 DOI: 10.1016/j.neuint.2024.105854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cortical organoids derived from human induced pluripotent stem cells (hiPSCs) represent a powerful in vitro experimental system to investigate human brain development and disease, often inaccessible to direct experimentation. However, despite steady progress in organoid technology, several limitations remain, including high cost and variability, use of hiPSCs derived from tissues harvested invasively, unexplored three-dimensional (3D) structural features and neuronal connectivity. Here, using a cost-effective and reproducible protocol as well as conventional two-dimensional (2D) immunostaining, we show that cortical organoids generated from hiPSCs obtained by reprogramming stem cells from human exfoliated deciduous teeth (SHED) recapitulate key aspects of human corticogenesis, such as polarized organization of neural progenitor zones with the presence of outer radial glial stem cells, and differentiation of superficial- and deep-layer cortical neurons and glial cells. We also show that 3D bioprinting and magnetic resonance imaging of intact cortical organoids are alternative and complementary approaches to unravel critical features of the 3D architecture of organoids. Finally, extracellular electrical recordings in whole organoids showed functional neuronal networks. Together, our findings suggest that SHED-derived cortical organoids constitute an attractive model of human neurodevelopment, and support the notion that a combination of 2D and 3D techniques to analyze organoid structure and function may help improve this promising technology.
Collapse
Affiliation(s)
| | | | | | - Geisa Rodrigues Salles
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Ronald Almeida de Carvalho
- Pontifícia Universidade Católica Do Paraná, Escola de Medicina, Laboratório de Bioinformática e Neurogenética, Curitiba, Paraná, Brazil
| | - Gabriel Batista
- Pontifícia Universidade Católica Do Paraná, Escola de Medicina, Laboratório de Bioinformática e Neurogenética, Curitiba, Paraná, Brazil
| | - Elisa Varella Branco
- Centro de Estudos Do Genoma Humano e Células Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Maria Rita Passos Bueno
- Centro de Estudos Do Genoma Humano e Células Tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Roberto Hirochi Herai
- Pontifícia Universidade Católica Do Paraná, Escola de Medicina, Laboratório de Bioinformática e Neurogenética, Curitiba, Paraná, Brazil
| | | | | |
Collapse
|
41
|
Sebastian R, Song Y, Pak C. Probing the molecular and cellular pathological mechanisms of schizophrenia using human induced pluripotent stem cell models. Schizophr Res 2024; 273:4-23. [PMID: 35835709 PMCID: PMC9832179 DOI: 10.1016/j.schres.2022.06.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 01/13/2023]
Abstract
With recent advancements in psychiatric genomics, as a field, "stem cell-based disease modelers" were given the exciting yet daunting task of translating the extensive list of disease-associated risks into biologically and clinically relevant information in order to deliver therapeutically meaningful leads and insights. Despite their limitations, human induced pluripotent stem cell (iPSCs) based models have greatly aided our understanding of the molecular and cellular mechanisms underlying the complex etiology of brain disorders including schizophrenia (SCZ). In this review, we summarize the major findings from studies in the past decade which utilized iPSC models to investigate cell type-specific phenotypes relevant to idiopathic SCZ and disease penetrant alleles. Across cell type differences, several biological themes emerged, serving as potential neurodevelopmental mechanisms of SCZ, including oxidative stress and mitochondrial dysfunction, depletion of progenitor pools and insufficient differentiation potential of these progenitors, and structural and functional deficits of neurons and other supporting cells. Here, we discuss both the recent progress as well as challenges and improvements needed for future studies utilizing iPSCs as a model for SCZ and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rebecca Sebastian
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA; Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Yoonjae Song
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
42
|
Bosone C, Castaldi D, Burkard TR, Guzman SJ, Wyatt T, Cheroni C, Caporale N, Bajaj S, Bagley JA, Li C, Sorre B, Villa CE, Testa G, Krenn V, Knoblich JA. A polarized FGF8 source specifies frontotemporal signatures in spatially oriented cell populations of cortical assembloids. Nat Methods 2024; 21:2147-2159. [PMID: 39294368 PMCID: PMC11541204 DOI: 10.1038/s41592-024-02412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 08/12/2024] [Indexed: 09/20/2024]
Abstract
Organoids generating major cortical cell types in distinct compartments are used to study cortical development, evolution and disorders. However, the lack of morphogen gradients imparting cortical positional information and topography in current systems hinders the investigation of complex phenotypes. Here, we engineer human cortical assembloids by fusing an organizer-like structure expressing fibroblast growth factor 8 (FGF8) with an elongated organoid to enable the controlled modulation of FGF8 signaling along the longitudinal organoid axis. These polarized cortical assembloids mount a position-dependent transcriptional program that in part matches the in vivo rostrocaudal gene expression patterns and that is lost upon mutation in the FGFR3 gene associated with temporal lobe malformations and intellectual disability. By producing spatially oriented cell populations with signatures related to frontal and temporal area identity within individual assembloids, this model recapitulates in part the early transcriptional divergence embedded in the protomap and enables the study of cortical area-relevant alterations underlying human disorders.
Collapse
Affiliation(s)
- Camilla Bosone
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Davide Castaldi
- Human Technopole, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Thomas Rainer Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Segundo Jose Guzman
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Tom Wyatt
- Laboratoire "Matière et Systèmes Complexes" (MSC), UMR 7057 CNRS, University of Paris, Paris, France
| | | | - Nicolò Caporale
- Human Technopole, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sunanjay Bajaj
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Joshua Adam Bagley
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Chong Li
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Benoit Sorre
- Laboratoire "Matière et Systèmes Complexes" (MSC), UMR 7057 CNRS, University of Paris, Paris, France
- Physics of Cells and Cancer, Institut Curie, Université PSL, Sorbonne University, CNRS UMR168, Paris, France
| | | | - Giuseppe Testa
- Human Technopole, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - Veronica Krenn
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Biotechnology and Bioscience, University of Milan-Bicocca, Milan, Italy.
| | - Jürgen Arthur Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
43
|
Yu D, Jain S, Wangzhou A, De Florencio S, Zhu B, Kim JY, Choi JJY, Paredes MF, Nowakowski TJ, Huang EJ, Piao X. Microglia regulate GABAergic neurogenesis in prenatal human brain through IGF1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619180. [PMID: 39464051 PMCID: PMC11507959 DOI: 10.1101/2024.10.19.619180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
GABAergic neurons are an essential cellular component of neural circuits. Their abundance and diversity have enlarged significantly in the human brain, contributing to the expanded cognitive capacity of humans. However, the developmental mechanism of the extended production of GABAergic neurons in the human brain remains elusive. Here, we use single-cell transcriptomics, bioinformatics, and histological analyses to uncover microglial regulation of the sustained proliferation of GABAergic progenitors and neuroblasts in the human medial ganglionic eminence (hMGE). We show that insulin-like growth factor 1 (IGF1) and its receptor IGR1R as the top ligand-receptor pair underlying microglia-progenitor communication in the prenatal human brain. Using our newly developed neuroimmune hMGE organoids, which mimics hMGE cytoarchitecture and developmental trajectory, we demonstrate that microglia-derived IGF1 promotes progenitor proliferation and the production of GABAergic neurons. Conversely, IGF1-neutralizing antibodies and IGF1 knockout human embryonic stem cells (hESC)-induced microglia (iMG) completely abolished iMG-mediated progenitor proliferation. Together, these findings reveal a previously unappreciated role of microglia-derived IGF1 in promoting proliferation of neural progenitors and the development of GABAergic neurons.
Collapse
|
44
|
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
|
45
|
Niu W, Yu S, Li X, Wang Z, Chen R, Michalski C, Jahangiri A, Zohdy Y, Chern JJ, Whitworth TJ, Wang J, Xu J, Zhou Y, Qin Z, Li B, Gambello MJ, Peng J, Wen Z. Longitudinal multi-omics reveals pathogenic TSC2 variants disrupt developmental trajectories of human cortical organoids derived from Tuberous Sclerosis Complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617121. [PMID: 39416123 PMCID: PMC11482767 DOI: 10.1101/2024.10.07.617121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Tuberous Sclerosis Complex (TSC), an autosomal dominant condition, is caused by heterozygous mutations in either the TSC1 or TSC2 genes, manifesting in systemic growth of benign tumors. In addition to brain lesions, neurologic sequelae represent the greatest morbidity in TSC patients. Investigations utilizing TSC1/2-knockout animal or human stem cell models suggest that TSC deficiency-causing hyper-activation of mTOR signaling might precipitate anomalous neurodevelopmental processes. However, how the pathogenic variants of TSC1/2 genes affect the longitudinal trajectory of human brain development remains largely unexplored. Here, we employed 3-dimensional cortical organoids derived from induced pluripotent stem cells (iPSCs) from TSC patients harboring TSC2 variants, alongside organoids from age- and sex-matched healthy individuals as controls. Through comprehensively longitudinal molecular and cellular analyses of TSC organoids, we found that TSC2 pathogenic variants dysregulate neurogenesis, synaptogenesis, and gliogenesis, particularly for reactive astrogliosis. The altered developmental trajectory of TSC organoids significantly resembles the molecular signatures of neuropsychiatric disorders, including autism spectrum disorders, epilepsy, and intellectual disability. Intriguingly, single cell transcriptomic analyses on TSC organoids revealed that TSC2 pathogenic variants disrupt the neuron/reactive astrocyte crosstalk within the NLGN-NRXN signaling network. Furthermore, cellular and electrophysiological assessments of TSC cortical organoids, along with proteomic analyses of synaptosomes, demonstrated that the TSC2 variants precipitate perturbations in synaptic transmission, neuronal network activity, mitochondrial translational integrity, and neurofilament formation. Notably, similar perturbations were observed in surgically resected cortical specimens from TSC patients. Collectively, our study illustrates that disease-associated TSC2 variants disrupt the neurodevelopmental trajectories through perturbations of gene regulatory networks during early cortical development, leading to mitochondrial dysfunction, aberrant neurofilament formation, impaired synaptic formation and neuronal network activity.
Collapse
Affiliation(s)
- Weibo Niu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- These authors contributed equally
| | - Shaojun Yu
- Department of Computer Science, Emory University, Atlanta, GA 30322, USA
- These authors contributed equally
| | - Xiangru Li
- College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Zhen Wang
- Department of Structural Biology, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Rui Chen
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Christina Michalski
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Arman Jahangiri
- Department of Neurological Surgery, Emory University, Atlanta, GA 30322, USA
- Pediatric Neurosurgery Associates at Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
| | - Youssef Zohdy
- Department of Neurological Surgery, Emory University, Atlanta, GA 30322, USA
- Pediatric Neurosurgery Associates at Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
| | - Joshua J Chern
- Department of Neurological Surgery, Emory University, Atlanta, GA 30322, USA
- Pediatric Neurosurgery Associates at Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
| | - Ted J Whitworth
- Robert P. Apkarian Integrated Electron Microscopy Core, Emory University, Atlanta, GA 30322, USA
| | - Jianjun Wang
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jie Xu
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ying Zhou
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhaohui Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Bingshan Li
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael J Gambello
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Junmin Peng
- Department of Structural Biology, Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
46
|
Pang W, Zhu J, Yang K, Zhu X, Zhou W, Jiang L, Zhuang X, Liu Y, Wei J, Lu X, Yin Y, Chen Z, Xiang Y. Generation of human region-specific brain organoids with medullary spinal trigeminal nuclei. Cell Stem Cell 2024; 31:1501-1512.e8. [PMID: 39208804 DOI: 10.1016/j.stem.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/16/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Brain organoids with nucleus-specific identities provide unique platforms for studying human brain development and diseases at a finer resolution. Despite its essential role in vital body functions, the medulla of the hindbrain has seen a lack of in vitro models, let alone models resembling specific medullary nuclei, including the crucial spinal trigeminal nucleus (SpV) that relays peripheral sensory signals to the thalamus. Here, we report a method to differentiate human pluripotent stem cells into region-specific brain organoids resembling the dorsal domain of the medullary hindbrain. Importantly, organoids specifically recapitulated the development of the SpV derived from the dorsal medulla. We also developed an organoid system to create the trigeminothalamic projections between the SpV and the thalamus by fusing these organoids, namely human medullary SpV-like organoids (hmSpVOs), with organoids representing the thalamus (hThOs). Our study provides a platform for understanding SpV development, nucleus-based circuit organization, and related disorders in the human brain.
Collapse
Affiliation(s)
- Wei Pang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinkui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaona Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yantong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoxiang Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziling Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
47
|
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
|
48
|
Yan Y, Yang Z, Chen L. High-quality models for assessing the effects of environmental pollutants on the nervous system: 3D brain organoids. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116876. [PMID: 39146594 DOI: 10.1016/j.ecoenv.2024.116876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The prevalence of environmental problems and the increasing risk of human exposure to environmental pollutants have become a global concern. The increasing environmental pollution is one of the main reasons for the rising incidence of most neurological-related diseases in recent years. However, the ethical constraints of direct human research and the racial limitations of animal models have slowed the progress of research in this area. The purpose of this study is to review the neurotoxicity of different environmental pollutants on the brain using brain organoids as a new model and to conclude that brain organoids may play a key role in assessing the mechanisms by which environmental pollutants affect neurogenesis and cause neurological pathogenesis. To accurately determine the negative effects of environmental pollutants on the nervous system, self-organizing brain organoids that are highly similar to the developing brain have become a new model system for studying the effects of environmental pollutants on human brain development and disease. This study uses brain organoids as a model to summarize the neurotoxicity of different environmental pollutants on the nervous system, including structural changes in brain organoids, inhibition of neuronal differentiation and migration, impairment of mitochondrial function, damage to cellular cilia, and influence on signaling pathways. In conclusion, exposure to environmental pollutants may cause different neurotoxicity to the nervous system. Therefore, it is crucial to understand how to use brain organoids to ameliorate neurological disorders caused by environmental pollution.
Collapse
Affiliation(s)
- Yu Yan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Ziye Yang
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China
| | - Liqun Chen
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
49
|
Pan T, Jaklic DC, Vaid S, Lin G, VanHeyningen D, Dang LT. A Multi-Electrode Array Platform for Modeling Epilepsy Using Human Pluripotent Stem Cell-Derived Brain Assembloids. J Vis Exp 2024:10.3791/67396. [PMID: 39400160 PMCID: PMC11730284 DOI: 10.3791/67396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Human brain organoids are three-dimensional (3D) structures derived from human pluripotent stem cells (hPSCs) that recapitulate aspects of fetal brain development. The fusion of dorsal with ventral regionally specified brain organoids in vitro generates assembloids, which have functionally integrated microcircuits with excitatory and inhibitory neurons. Due to their structural complexity and diverse population of neurons, assembloids have become a useful in vitro tool for studying aberrant network activity. Multi-electrode array (MEA) recordings serve as a method for capturing electrical field potentials, spikes, and longitudinal network dynamics from a population of neurons without compromising cell membrane integrity. However, adhering assembloids onto the electrodes for long-term recordings can be challenging due to their large size and limited contact surface area with the electrodes. Here, we demonstrate a method to plate assembloids onto MEA plates for recording electrophysiological activity over a 2-month span. Although the current protocol utilizes human cortical organoids, it can be broadly adapted to organoids differentiated to model other brain regions. This protocol establishes a robust, longitudinal, electrophysiological assay for studying the development of a neuronal network, and this platform has the potential to be used in drug screening for therapeutic development in epilepsy.
Collapse
Affiliation(s)
- Tong Pan
- Division of Pediatric Neurology, Department of Pediatrics, Michigan Medicine, Ann Arbor; Michigan Neuroscience Institute, University of Michigan, Ann Arbor
| | - Daniel C Jaklic
- Division of Pediatric Neurology, Department of Pediatrics, Michigan Medicine, Ann Arbor; Michigan Neuroscience Institute, University of Michigan, Ann Arbor
| | - Shivanshi Vaid
- Division of Pediatric Neurology, Department of Pediatrics, Michigan Medicine, Ann Arbor; Michigan Neuroscience Institute, University of Michigan, Ann Arbor
| | - Grace Lin
- Division of Pediatric Neurology, Department of Pediatrics, Michigan Medicine, Ann Arbor; Michigan Neuroscience Institute, University of Michigan, Ann Arbor
| | - Debora VanHeyningen
- Division of Pediatric Neurology, Department of Pediatrics, Michigan Medicine, Ann Arbor; Michigan Neuroscience Institute, University of Michigan, Ann Arbor
| | - Louis T Dang
- Division of Pediatric Neurology, Department of Pediatrics, Michigan Medicine, Ann Arbor; Michigan Neuroscience Institute, University of Michigan, Ann Arbor;
| |
Collapse
|
50
|
Pavon N, Sun Y, Pak C. Cell type specification and diversity in subpallial organoids. Front Genet 2024; 15:1440583. [PMID: 39391063 PMCID: PMC11465425 DOI: 10.3389/fgene.2024.1440583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Neural organoids have emerged as valuable tools for studying the developing brain, sparking enthusiasm and driving their adoption in disease modeling, drug screening, and investigating fetal neural development. The increasing popularity of neural organoids as models has led to a wide range of methodologies aimed at continuous improvement and refinement. Consequently, research groups often improve and reconfigure protocols to create region-specific organoids, resulting in diverse phenotypes, including variations in morphology, gene expression, and cell populations. While these improvements are exciting, routine adoptions of such modifications and protocols in the research laboratories are often challenging due to the reiterative empirical testing necessary to validate the cell types generated. To address this challenge, we systematically compare the similarities and differences that exist across published protocols that generates subpallial-specific organoids to date. In this review, we focus specifically on exploring the production of major GABAergic neuronal subtypes, especially Medium Spiny Neurons (MSNs) and Interneurons (INs), from multiple subpallial organoid protocols. Importantly, we look to evaluate the cell type diversity and the molecular pathways manipulated to generate them, thus broadening our understanding of the existing subpallial organoids as well as assessing the in vitro applicability of specific patterning factors. Lastly, we discuss the current challenges and outlook on the improved patterning of region-specific neural organoids. Given the critical roles MSN and IN dysfunction play in neurological disorders, comprehending the GABAergic neurons generated by neural organoids will undoubtedly facilitate clinical translation.
Collapse
Affiliation(s)
- Narciso Pavon
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Neuroscience and Behavior, University of Massachusetts Amherst, Amherst, MA, United States
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, United States
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|