1
|
Zhao Y, Wang T, Liu J, Wang Z, Lu Y. Emerging brain organoids: 3D models to decipher, identify and revolutionize brain. Bioact Mater 2025; 47:378-402. [PMID: 40026825 PMCID: PMC11869974 DOI: 10.1016/j.bioactmat.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Brain organoids are an emerging in vitro 3D brain model that is integrated from pluripotent stem cells. This model mimics the human brain's developmental process and disease-related phenotypes to a certain extent while advancing the development of human brain-based biological intelligence. However, many limitations of brain organoid culture (e.g., lacking a functional vascular system, etc.) prevent in vitro-cultured organoids from truly replicating the human brain in terms of cell type and structure. To improve brain organoids' scalability, efficiency, and stability, this paper discusses important contributions of material biology and microprocessing technology in solving the related limitations of brain organoids and applying the latest imaging technology to make real-time imaging of brain organoids possible. In addition, the related applications of brain organoids, especially the development of organoid intelligence combined with artificial intelligence, are analyzed, which will help accelerate the rational design and guidance of brain organoids.
Collapse
Affiliation(s)
- Yuli Zhao
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Ting Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Jiajun Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze Wang
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
2
|
Tzekaki E, Bekiari C, Pantazaki A, Tsantarliotou M, Tsolaki M, Lavrentiadou SN. A new protocol for the development of organoids based on molecular mechanisms in the developing newborn rat brain: Prospective applications in the study of Alzheimer's disease. J Neurosci Methods 2025; 417:110404. [PMID: 39978482 DOI: 10.1016/j.jneumeth.2025.110404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Brain organoids have emerged as powerful models for studying brain development and neurological disorders COMPARISON WITH EXISTING METHODS: Current models rely on stem cell isolation and differentiation using different growth factors. Thus, their composition varies according to the protocol followed. NEW METHOD We developed a simple protocol to generate organoids from newborn rat whole brain. It is a one-step procedure that yields organoids of consistent composition. The whole brains from 3-day old pups were digested enzymatically. All isolated cells were seeded in culture plates using a basement membrane extract (BME) matrix as a scaffold and cultured in the presence of the appropriate medium. RESULTS Hematoxylin-eosin staining of 28-day-old cultured domes revealed their structural integrity, while immunohistochemistry confirmed the presence of neurons, astrocytes, microglia, and progenitor stem cells in the structures. To assess whether these organoids can serve as a model to study brain physiopathology, and in particular neurodegenerative diseases such as Alzheimer's disease (AD), we determined how these organoids respond upon their exposure to lipopolysaccharides (LPS), a potent neuroinflammatory factor. LPS-induced amyloid precursor protein (APP), tau protein and glial fibrillary acidic protein (GFAP) expression. Moreover, the intracellular levels of IL-1β and the extracellular levels of amyloid-beta (Aβ) were also elevated. CONCLUSIONS Therefore, this simple protocol results in the generation of functional brain organoids with a consistent structure, that requires no use of varying factors that may affect the structure and function of the produced organoids, thus providing a valuable tool for the study of the physiopathology of neurodegenerative disorders.
Collapse
Affiliation(s)
- Eleni Tzekaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Chryssa Bekiari
- Laboratory of Anatomy and Histology School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| | - Anastasia Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece; Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Maria Tsantarliotou
- Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| | - Magda Tsolaki
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece.
| | - Sophia N Lavrentiadou
- Center for Interdisciplinary Research and Innovation, Laboratory of Neurodegenerative Diseases (LND), 57001, Thermi, Thessaloniki, Greece; Laboratory of Animal Physiology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Greece.
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 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
|
4
|
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
|
5
|
Berlind JE, Lai JD, Lie C, Vicente J, Lam K, Guo S, Chang J, Yu V, Ichida JK. KCTD20 suppression mitigates excitotoxicity in tauopathy patient organoids. Neuron 2025; 113:1169-1189.e7. [PMID: 40049159 PMCID: PMC12005969 DOI: 10.1016/j.neuron.2025.02.001] [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: 12/13/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025]
Abstract
Excitotoxicity is a major pathologic mechanism in patients with tauopathy and other neurodegenerative diseases. However, the key neurotoxic drivers and the most effective strategies for mitigating these degenerative processes are unclear. Here, we show that glutamate treatment of induced pluripotent stem cell (iPSC)-derived cerebral organoids induces tau oligomerization and neurodegeneration and that these phenotypes are enhanced in organoids derived from tauopathy patients. Using a genome-wide CRISPR interference (CRISPRi) screen, we find that the suppression of KCTD20 potently ameliorates tau pathology and neurodegeneration in glutamate-treated organoids and mice, as well as in transgenic mice overexpressing mutant human tau. KCTD20 suppression reduces oligomeric tau and improves neuron survival by activating lysosomal exocytosis, which clears pathological tau. Our results show that glutamate signaling can induce neuronal tau pathology and identify KCTD20 suppression and lysosomal exocytosis as effective strategies for clearing neurotoxic tau species.
Collapse
Affiliation(s)
- Joshua E Berlind
- 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, Los Angeles, CA, USA
| | - Jesse D Lai
- 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, Los Angeles, CA, USA; Department of Neuroscience, Amgen Inc., Cambridge, MA, USA; Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA.
| | - Cecilia Lie
- 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, Los Angeles, CA, USA
| | - Jokabeth Vicente
- 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, Los Angeles, CA, USA
| | - Kelsey Lam
- 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, Los Angeles, CA, USA
| | - Sheron Guo
- 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, Los Angeles, CA, USA
| | - Jonathan Chang
- 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, Los Angeles, CA, USA
| | - Violeta Yu
- Neurological & Rare Diseases, Dewpoint Therapeutics, Boston, MA, USA
| | - Justin K Ichida
- 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, Los Angeles, CA, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Kang M, Choi J, Han J, Araki T, Kim SW, Ryu HH, Kim MG, Kim S, Jang H, Kim SY, Hwang KD, Kim S, Yoo M, Lee J, Kim K, Park P, Choi JE, Han DH, Kim Y, Kim J, Chang S, Kaang BK, Ko JM, Cheon KA, An JY, Kim SJ, Park H, Neel BG, Kim CH, Lee YS. Aberrant ERK signaling in astrocytes impairs learning and memory in RASopathy-associated BRAF mutant mouse models. J Clin Invest 2025; 135:e176631. [PMID: 39964758 PMCID: PMC11996877 DOI: 10.1172/jci176631] [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: 10/17/2023] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
RAS/MAPK pathway mutations often induce RASopathies with overlapping features, such as craniofacial dysmorphology, cardiovascular defects, dermatologic abnormalities, and intellectual disabilities. Although B-Raf proto-oncogene (BRAF) mutations are associated with cardio-facio-cutaneous (CFC) syndrome and Noonan syndrome, it remains unclear how these mutations impair cognition. Here, we investigated the underlying neural mechanisms using several mouse models harboring a gain-of-function BRAF mutation (K499E) discovered in RASopathy patients. We found expressing BRAF K499E (KE) in neural stem cells under the control of a Nestin-Cre promoter (Nestin;BRAFKE/+) induced hippocampal memory deficits, but expressing it in excitatory or inhibitory neurons did not. BRAF KE expression in neural stem cells led to aberrant reactive astrogliosis, increased astrocytic Ca2+ fluctuations, and reduced hippocampal long-term depression (LTD) in mice. Consistently, 3D human cortical spheroids expressing BRAF KE also showed reactive astrogliosis. Astrocyte-specific adeno-associated virus-BRAF KE (AAV-BRAF KE) delivery induced memory deficits and reactive astrogliosis and increased astrocytic Ca2+ fluctuations. Notably, reducing extracellular signal-regulated kinase (ERK) activity in astrocytes rescued the memory deficits and altered astrocytic Ca2+ activity of Nestin;BRAFKE/+ mice. Furthermore, reducing astrocyte Ca2+ activity rescued the spatial memory impairments of BRAF KE-expressing mice. Our results demonstrate that ERK hyperactivity contributes to astrocyte dysfunction associated with Ca2+ dysregulation, leading to the memory deficits of BRAF-associated RASopathies.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jihye Choi
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeongho Han
- Research Group of Neurovascular Unit, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Toshiyuki Araki
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA
| | - Soo-Whee Kim
- Department of Integrated Biomedical and Life Science, and
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Republic of Korea
| | | | - Min-Gyun Kim
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seoyeon Kim
- Department of Integrated Biomedical and Life Science, and
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Hanbyul Jang
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sun Yong Kim
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Doo Hwang
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soobin Kim
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Myeongjong Yoo
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jaegeon Lee
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kitae Kim
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Pojeong Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ja Eun Choi
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dae Hee Han
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yujin Kim
- Department of Integrated Biomedical and Life Science, and
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Jeongyeon Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Sunghoe Chang
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jung Min Ko
- Department of Pediatrics, Division of Clinical Genetics, Seoul National University College of Medicine, Seoul, Republic of Korea
- Rare Disease Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Keun-Ah Cheon
- Department of Child and Adolescent Psychiatry, Severance Hospital, and
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, and
| | - Sang Jeong Kim
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyungju Park
- Research Group of Neurovascular Unit, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Benjamin G. Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA
| | - Chul Hoon Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Seok Lee
- Department of Physiology, and
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
7
|
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
|
8
|
Kim JI, Imaizumi K, Jurjuț O, Kelley KW, Wang D, Thete MV, Hudacova Z, Amin ND, Levy RJ, Scherrer G, Pașca SP. Human assembloid model of the ascending neural sensory pathway. Nature 2025:10.1038/s41586-025-08808-3. [PMID: 40205039 DOI: 10.1038/s41586-025-08808-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2025] [Indexed: 04/11/2025]
Abstract
Somatosensory pathways convey crucial information about pain, touch, itch and body part movement from peripheral organs to the central nervous system1,2. Despite substantial needs to understand how these pathways assemble and to develop pain therapeutics, clinical translation remains challenging. This is probably related to species-specific features and the lack of in vitro models of the polysynaptic pathway. Here we established a human ascending somatosensory assembloid (hASA), a four-part assembloid generated from human pluripotent stem cells that integrates somatosensory, spinal, thalamic and cortical organoids to model the spinothalamic pathway. Transcriptomic profiling confirmed the presence of key cell types of this circuit. Rabies tracing and calcium imaging showed that sensory neurons connect to dorsal spinal cord neurons, which further connect to thalamic neurons. Following noxious chemical stimulation, calcium imaging of hASA demonstrated a coordinated response. In addition, extracellular recordings and imaging revealed synchronized activity across the assembloid. Notably, loss of the sodium channel NaV1.7, which causes pain insensitivity, disrupted synchrony across hASA. By contrast, a gain-of-function SCN9A variant associated with extreme pain disorder induced hypersynchrony. These experiments demonstrated the ability to functionally assemble the essential components of the human sensory pathway, which could accelerate our understanding of sensory circuits and facilitate therapeutic development.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kent Imaizumi
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Ovidiu Jurjuț
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Dong Wang
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Zuzana Hudacova
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA
| | - Rebecca J Levy
- Department of Neurology & Neurological Sciences, Division of Child Neurology, Stanford University, Stanford, CA, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute & Bio-X, Stanford, CA, USA.
| |
Collapse
|
9
|
Bessetti RN, Cobb M, Lilley RM, Johnson NZ, Perez DA, Koonce VM, McCoy K, Litwa KA. Sulforaphane protects developing neural networks from VPA-induced synaptic alterations. Mol Psychiatry 2025:10.1038/s41380-025-02967-5. [PMID: 40175519 DOI: 10.1038/s41380-025-02967-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/13/2025] [Accepted: 03/20/2025] [Indexed: 04/04/2025]
Abstract
Prenatal brain development is particularly sensitive to chemicals that can disrupt synapse formation and cause neurodevelopmental disorders. In most cases, such chemicals increase cellular oxidative stress. For example, prenatal exposure to the anti-epileptic drug valproic acid (VPA), induces oxidative stress and synaptic alterations, promoting autism spectrum disorders (ASD) in humans and autism-like behaviors in rodents. Using VPA to model chemically induced ASD, we tested whether activation of cellular mechanisms that increase antioxidant gene expression would be sufficient to prevent VPA-induced synaptic alterations. As a master regulator of cellular defense pathways, the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) promotes expression of detoxification enzymes and antioxidant gene products. To increase NRF2 activity, we used the phytochemical and potent NRF2 activator, sulforaphane (SFN). In our models of human neurodevelopment, SFN activated NRF2, increasing expression of antioxidant genes and preventing oxidative stress. SFN also enhanced expression of genes associated with synapse formation. Consistent with these gene expression profiles, SFN protected developing neural networks from VPA-induced reductions in synapse formation. Furthermore, in mouse cortical neurons, SFN rescued VPA-induced reductions in neural activity. These results demonstrate the ability of SFN to protect developing neural networks during the vulnerable period of synapse formation, while also identifying molecular signatures of SFN-mediated neuroprotection that could be relevant for combatting other environmental toxicants.
Collapse
Affiliation(s)
- Riley N Bessetti
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Michelle Cobb
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Rosario M Lilley
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Noah Z Johnson
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Daisy A Perez
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA
| | - Virginia M Koonce
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA
| | | | - Karen A Litwa
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University (ECU), Greenville, NC, USA.
- East Carolina Diabetes and Obesity Institute at ECU, Greenville, NC, USA.
| |
Collapse
|
10
|
Howard CE, Cheenath M, Crouch E. The promise of cerebral organoids for neonatology. Curr Opin Pediatr 2025; 37:182-190. [PMID: 40013913 PMCID: PMC11902893 DOI: 10.1097/mop.0000000000001446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Applying discoveries from basic research to patients in the neonatal intensive care unit (NICU) is challenging given the difficulty of modeling this population in animal models, lack of translational relevance from animal models to humans, and scarcity of primary human tissue. Human cell-derived cerebral organoid models are an appealing way to address some of these gaps. In this review, we will touch on previous work to model neonatal conditions in cerebral organoids, some limitations of this approach, and recent strategies that have attempted to address these limitations. RECENT FINDINGS While modeling of neurodevelopmental disorders has been an application of cerebral organoids since their initial description, recent studies have dramatically expanded the types of brain regions and disease models available. Additionally, work to increase the complexity of organoid models by including immune and vascular cells, as well as modeling human heterogeneity with mixed donor organoids will provide new opportunities to model neonatal pathologies. SUMMARY Organoids are an attractive model to study human neurodevelopmental pathologies relevant to patients in the neonatal ICU. New technologies will broaden the applicability of these models to neonatal research and their usefulness as a drug screening platform.
Collapse
Affiliation(s)
- Clare E Howard
- Division of Newborn Medicine, Boston Children’s Hospital
| | - Manju Cheenath
- Department of Obstetrics and Gynecology, University of California, San Francisco
| | - Elizabeth Crouch
- Department of Pediatrics, University of California, San Francisco
| |
Collapse
|
11
|
Jeong Y, Son S, Park J, Kim CY, Kim J. Antidepressant aripiprazole induces adverse effects on neural development during cortex organoid generation. Reprod Toxicol 2025; 133:108862. [PMID: 39971107 DOI: 10.1016/j.reprotox.2025.108862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
A significant number of women experience anxiety and depressive symptoms during pregnancy, leading to the prescription of antidepressants, including aripiprazole. However, although a few animal studies have reported its developmental toxicity, there is a lack of research on the potential risks aripiprazole may pose to the fetus, particularly regarding neural development, as well as an absence of appropriate models to verify these effects. Therefore, this study investigates the impact of aripiprazole on neural development using cortex organoids, which can effectively model human brain development and function while overcoming interspecies differences. Cortex organoids were generated and exposed to aripiprazole at concentrations of 0.3-9 µM over 4 weeks. We assessed morphological changes, cell viability, gene expression, immunofluorescence staining, and electrophysiological function. The results revealed that aripiprazole led to significant reductions in organoid size and increased cell death, particularly at higher concentrations. Immunofluorescence analysis showed abnormalities in the expression patterns of neural stem cells and neuronal markers. Additionally, real-time PCR demonstrated decreased expression of genes related to neural stem cells, neural differentiation and migration, maturation, synaptogenesis, and gliogenesis, along with increased apoptosis-related gene expression. Electrophysiological recordings indicated impaired neural activity, evidenced by reduced mean firing rates. Our study is the first to demonstrate that aripiprazole induces adverse effects on neural development across functional, molecular, and morphological aspects. The findings will aid in a better understanding of the risks associated with antidepressant use during pregnancy in terms of neural development and suggest that cortex organoids are a valuable model for evaluating potential neurodevelopmental toxicants.
Collapse
Affiliation(s)
- Youngin Jeong
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Suil Son
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jiyun Park
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - C-Yoon Kim
- College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Jin Kim
- Department of Physiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea.
| |
Collapse
|
12
|
Saade M, Martí E. Early spinal cord development: from neural tube formation to neurogenesis. Nat Rev Neurosci 2025; 26:195-213. [PMID: 39915695 DOI: 10.1038/s41583-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 03/26/2025]
Abstract
As one of the simplest and most evolutionarily conserved parts of the vertebrate nervous system, the spinal cord serves as a key model for understanding the principles of nervous system construction. During embryonic development, the spinal cord originates from a population of bipotent stem cells termed neuromesodermal progenitors, which are organized within a transient embryonic structure known as the neural tube. Neural tube morphogenesis differs along its anterior-to-posterior axis: most of the neural tube (including the regions that will develop into the brain and the anterior spinal cord) forms via the bending and dorsal fusion of the neural groove, but the establishment of the posterior region of the neural tube involves de novo formation of a lumen within a solid medullary cord. The early spinal cord primordium consists of highly polarized neural progenitor cells organized into a pseudostratified epithelium. Tight regulation of the cell division modes of these progenitors drives the embryonic growth of the neural tube and initiates primary neurogenesis. A rich history of observational and functional studies across various vertebrate models has advanced our understanding of the cellular events underlying spinal cord development, and these foundational studies are beginning to inform our knowledge of human spinal cord development.
Collapse
Affiliation(s)
- Murielle Saade
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| |
Collapse
|
13
|
Karam M, Ortega-Gascó A, Tornero D. Emerging Insights into Brain Inflammation: Stem-Cell-Based Approaches for Regenerative Medicine. Int J Mol Sci 2025; 26:3275. [PMID: 40244116 PMCID: PMC11989304 DOI: 10.3390/ijms26073275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Neuroinflammation is a complex immune response triggered by brain injury or pathological stimuli, and is highly exacerbated in neurodegenerative diseases. It plays a dual role in the central nervous system, promoting repair in acute stages while aggravating disease progression by contributing to neuronal loss, synaptic dysfunction, and glial dysregulation in chronic phases. Inflammatory responses are mainly orchestrated by microglia and infiltrated monocytes, which, when dysregulated, not only harm existing neurons, but also impair the survival and differentiation of neural stem and progenitor cells in the affected brain regions. Modulating neuroinflammation is crucial for harnessing its protective functions while minimizing its detrimental effects. Current therapeutic strategies focus on fine-tuning inflammatory responses through pharmacological agents, bioactive molecules, and stem cell-based therapies. These approaches aim to restore immune homeostasis, support neuroprotection, and promote regeneration in various neurological disorders. However, animal models sometimes fail to reproduce human-specific inflammatory responses in the brain. In this context, stem-cell-derived models provide a powerful tool to study neuroinflammatory mechanisms in a patient-specific and physiologically relevant context. These models facilitate high-throughput screening, personalized medicine, and the development of targeted therapies while addressing the limitations of traditional animal models, paving the way for more targeted and effective treatments.
Collapse
Affiliation(s)
- Marie Karam
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Alba Ortega-Gascó
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Daniel Tornero
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
14
|
Sun Z, Zhu H, He X, Lendemeijer B, Wang Z, Fan J, Sun Y, Zhang Z, Markx S, Kushner SA, Xu B, Gogos JA. Genomic and Transcriptomic Signatures of SETD1A Disruption in Human Excitatory Neuron Development and Psychiatric Disease Risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645419. [PMID: 40196527 PMCID: PMC11974865 DOI: 10.1101/2025.03.26.645419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Genetic disruption of SETD1A markedly increases the risk for schizophrenia. To elucidate the underlying mechanisms, we generated isogenic organoid models of the developing human cerebral cortex harboring a SETD1A loss-of-function schizophrenia risk mutation. Employing chromatin profiling combined with RNA sequencing, we identified high-confidence SETD1A target genes, analyzed the impact of the mutation on SETD1A binding and transcriptional regulation and validated key findings with orthogonal approaches. Disruption of SETD1A function disturbs the finely tuned temporal gene expression in the excitatory neuron lineage, yielding an aberrant transcriptional program that compromises key regulatory and metabolic pathways essential for neurodevelopmental transitions. Although overall SETD1A binding remains unchanged in mutant neurons, we identified localized alterations in SETD1A binding that correlate with shifts in H3K4me3 levels and gene expression. These changes are enriched at enhancer regions, suggesting that enhancer-regulated genes are especially vulnerable to SETD1A reduction. Notably, target genes with enhancer-bound SETD1A are primarily linked to neuronal functions while those with promoter-bound SETD1A are enriched for basic cellular functions. By mapping the SETD1A binding landscape in excitatory neurons of the human fetal frontal cortex and integrating multimodal neuroimaging and genetic datasets, we demonstrate that the genomic context of SETD1A binding differentially correlates with macroscale brain organization and establish a link between SETD1A-bound enhancers, schizophrenia-associated brain alterations and genetic susceptibility. Our study advances our understanding of the role of SETD1A binding patterns in schizophrenia pathogenesis, offering insights that may guide future therapeutic strategies.
Collapse
Affiliation(s)
- Zhixiong Sun
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Huixiang Zhu
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Xiaofu He
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Bas Lendemeijer
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Zanxu Wang
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jack Fan
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Yan Sun
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Zhiguo Zhang
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Sander Markx
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Steven A. Kushner
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Joseph A. Gogos
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| |
Collapse
|
15
|
SoRelle ED, Luftig MA. Multiple sclerosis and infection: history, EBV, and the search for mechanism. Microbiol Mol Biol Rev 2025; 89:e0011923. [PMID: 39817754 PMCID: PMC11948499 DOI: 10.1128/mmbr.00119-23] [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: 01/18/2025] Open
Abstract
SUMMARYInfection has long been hypothesized as the cause of multiple sclerosis (MS), and recent evidence for Epstein-Barr virus (EBV) as the trigger of MS is clear and compelling. This clarity contrasts with yet uncertain viral mechanisms and their relation to MS neuroinflammation and demyelination. As long as this disparity persists, it will invigorate virologists, molecular biologists, immunologists, and clinicians to ascertain how EBV potentiates MS onset, and possibly the disease's chronic activity and progression. Such efforts should take advantage of the diverse body of basic and clinical research conducted over nearly two centuries since the first clinical descriptions of MS plaques. Defining the contribution of EBV to the complex and multifactorial pathology of MS will also require suitable experimental models and techniques. Such efforts will broaden our understanding of virus-driven neuroinflammation and specifically inform the development of EBV-targeted therapies for MS management and, ultimately, prevention.
Collapse
Affiliation(s)
- Elliott D. SoRelle
- Department of Molecular Genetics & Microbiology, Center for Virology, Duke University, Durham, North Carolina, USA
| | - Micah A. Luftig
- Department of Molecular Genetics & Microbiology, Center for Virology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
16
|
Wu X, Xiong D, Liu R, Lai X, Tian Y, Xie Z, Chen L, Hu L, Duan J, Gao X, Zeng X, Dong W, Xu T, Fu F, Yang X, Cheng X, Plewczynski D, Kim M, Xin W, Wang T, Xiang AP, Tang Z. Evolutionary divergence in CTCF-mediated chromatin topology drives transcriptional innovation in humans. Nat Commun 2025; 16:2941. [PMID: 40140405 PMCID: PMC11947266 DOI: 10.1038/s41467-025-58275-7] [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: 07/26/2024] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Chromatin topology can impact gene regulation, but how evolutionary divergence in chromatin topology has shaped gene regulatory landscapes for distinctive human traits remains poorly understood. CTCF sites determine chromatin topology by forming domains and loops. Here, we show evolutionary divergence in CTCF-mediated chromatin topology at the domain and loop scales during primate evolution, elucidating distinct mechanisms for shaping regulatory landscapes. Human-specific divergent domains lead to a broad rewiring of transcriptional landscapes. Divergent CTCF loops concord with species-specific enhancer activity, influencing enhancer connectivity to target genes in a concordant yet constrained manner. Under this concordant mechanism, we establish the role of human-specific CTCF loops in shaping transcriptional isoform diversity, with functional implications for disease susceptibility. Furthermore, we validate the function of these human-specific CTCF loops using human forebrain organoids. This study advances our understanding of genetic evolution from the perspective of genome architecture.
Collapse
Affiliation(s)
- Xia Wu
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Dan Xiong
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Rong Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangdong, China
| | - Xingqiang Lai
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangdong, China
| | - Yuhan Tian
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Ziying Xie
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Li Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Lanqi Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Jingjing Duan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Xinyu Gao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Xian Zeng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Wei Dong
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Ting Xu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Fang Fu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Xin Yang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Xinlai Cheng
- Buchmann Institute for Molecular Life Sciences, Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Minji Kim
- Department of Computational Medicine and Bioinformatics, University of Michigan, Michigan, MI, USA
| | - Wenjun Xin
- Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Key Laboratory for Neuroscience, Ministry of Education of China & National Health Commission of China, Beijing, China
- Autism Research Center, Peking University Health Science Center, Beijing, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangdong, China
| | - Zhonghui Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.
| |
Collapse
|
17
|
O'Laughlin R, Cheng F, Song H, Ming GL. Bioengineering tools for next-generation neural organoids. Curr Opin Neurobiol 2025; 92:103011. [PMID: 40132519 DOI: 10.1016/j.conb.2025.103011] [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: 10/15/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Human stem cell-derived neural organoids were recently introduced as powerful in vitro 3D experimental model systems that innately undergo critical steps of organogenesis in culture and exhibit molecular, cellular, and structural features similar to the fetal human nervous system. These organoids have yielded new insights into human neurodevelopment and associated disorders. However, neural organoids have some crucial limitations that arise from the loosely controlled conditions for their development, an inability to maintain their spatial orientation in culture and a lack of technologies for taking long-term measurements on their morphology and electrical activity. Here, we review recent progress in using bioengineering methods to improve neural organoid formation and analysis by leveraging microfabrication, biomaterials, 3D printing, and flexible electrodes. We discuss how the applications of each technique can help to address critical limitations with standard neural organoid models. We conclude with a perspective on future applications of bioengineered next-generation neural organoids.
Collapse
Affiliation(s)
- Richard O'Laughlin
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fangyi Cheng
- Graduate Program in Bioengineering, School of Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Program in Bioengineering, School of Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Program in Bioengineering, School of Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
González-Sastre R, Coronel R, Mateos-Martínez P, Maeso L, Llorente-Beneyto E, Luque A, Anta B, López-Alonso V, Liste I. Protocol for generating human cerebral organoids from two-dimensional cultures of pluripotent stem cells bypassing embryoid body aggregation. STAR Protoc 2025; 6:103678. [PMID: 40073022 PMCID: PMC11950768 DOI: 10.1016/j.xpro.2025.103678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/10/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Human cerebral organoids (hCOs) provide an excellent model for the study of human brain development and disease. Here, we present a protocol to obtain hCOs directly from two-dimensional (2D) pluripotent stem cell (PSC) cultures, avoiding cell dissociation and posterior embryoid body (EB) aggregation. We describe steps for subjecting 2D cultures to a neural fate and subsequently developing hCOs. We then detail the evaluation of different cellular types. For complete details on the use and execution of this protocol, please refer to González-Sastre et al.1.
Collapse
Affiliation(s)
- Rosa González-Sastre
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain; Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain; Doctoranda en la Escuela Internacional de Doctorado de la Universidad Nacional de Educación a Distancia (UNED), Programa en Ciencias Biomédicas y Salud Pública
| | - Raquel Coronel
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain.
| | - Patricia Mateos-Martínez
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain; Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain; Doctoranda en la Escuela Internacional de Doctorado de la Universidad Nacional de Educación a Distancia (UNED), Programa en Ciencias Biomédicas y Salud Pública
| | - Laura Maeso
- Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain
| | - Elena Llorente-Beneyto
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain; Doctoranda en la Escuela Internacional de Doctorado de la Universidad Nacional de Educación a Distancia (UNED), Programa en Ciencias Biomédicas y Salud Pública
| | - Alfonso Luque
- Unidad de Endotelio Funcional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain
| | - Berta Anta
- Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain
| | - Victoria López-Alonso
- Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain.
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid 28220, Spain.
| |
Collapse
|
19
|
Santos JLDS, Paredes BD, Adanho CSA, Nonaka CKV, da Silva KN, Santos IM, Loiola EC, Silva VAO, Rocha CAG, Souza BSDF. Generation and characterization of human-induced pluripotent stem cell lines from patients with autism spectrum disorder and SCN2A variants. Hum Cell 2025; 38:74. [PMID: 40111547 DOI: 10.1007/s13577-025-01199-7] [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: 07/02/2024] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Autism spectrum disorders (ASD) comprise a group of complex neurodevelopmental disorders that affect communication and social interactions. Over a thousand genes have been associated with ASD, with SCN2A standing out due to its critical role in neuronal function and development. Induced pluripotent stem cells (iPSCs) derived from individuals with ASD have become invaluable in vitro models for investigating the cellular and molecular mechanisms underlying the disorder. In this study, we generated and characterized four iPSC clones from peripheral blood mononuclear cells (PBMCs) of two ASD patients carrying loss-of-function variants in the SCN2A gene. These iPSC lines underwent comprehensive characterization through multiple assays. Reverse transcription polymerase chain reaction (RT-PCR), flow cytometry, and immunofluorescence analyses confirmed the presence of pluripotency markers. An embryoid body formation assay demonstrated their potential to differentiate into the three germ layers. Sequencing analysis confirmed the SCN2A variants, while short tandem repeat (STR) analysis authenticated the cell lines, and karyotype analysis ensured chromosomal integrity. The iPSCs exhibited typical morphologic characteristics, including large nuclei with prominent nucleoli, a high nucleus-to-cytoplasm ratio, densely packed cells, and well-defined borders. These cells maintained pluripotency markers, demonstrated the ability to differentiate into the three germ layers, and showed a normal karyotype. Furthermore, we successfully generated cerebral organoids from these cells. Our study establishes a robust platform for further exploration of the pathophysiological mechanisms of ASD, particularly those involving SCN2A.
Collapse
Affiliation(s)
| | - Bruno Diaz Paredes
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Corynne Stephanie Ahouefa Adanho
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
- Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | - Carolina Kymie Vasques Nonaka
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Katia Nunes da Silva
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Ian Marinho Santos
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Erick Correia Loiola
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Viviane Aline Oliveira Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Clarissa Araújo Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador, Brazil.
- D'Or Institute for Research and Education (IDOR), Salvador, Brazil.
- Pioneer Science Initiative, D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Li TL, Blair JD, Yoo T, Grant GA, Hockemeyer D, Porter BE, Bateup HS. mTORC1 activation drives astrocyte reactivity in cortical tubers and brain organoid models of TSC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640914. [PMID: 40093155 PMCID: PMC11908165 DOI: 10.1101/2025.02.28.640914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Tuberous Sclerosis Complex (TSC) is a genetic neurodevelopmental disorder associated with early onset epilepsy, intellectual disability and neuropsychiatric disorders. A hallmark of the disorder is cortical tubers, which are focal malformations of brain development containing dysplastic cells with hyperactive mTORC1 signaling. One barrier to developing therapeutic approaches and understanding the origins of tuber cells is the lack of a model system that recapitulates this pathology. To address this, we established a genetically mosaic cortical organoid system that models a somatic "second-hit" mutation, which is thought to drive the formation of tubers in TSC. With this model, we find that loss of TSC2 cell-autonomously promotes the differentiation of astrocytes, which exhibit features of a disease-associated reactive state. TSC2 -/- astrocytes have pronounced changes in morphology and upregulation of proteins that are risk factors for neurodegenerative diseases, such as clusterin and APOE. Using multiplexed immunofluorescence in primary tubers from TSC patients, we show that tuber cells with hyperactive mTORC1 activity also express reactive astrocyte proteins, and we identify a unique population of cells with expression profiles that match those observed in organoids. Together, this work reveals that reactive astrogliosis is a primary feature of TSC that arises early in cortical development. Dysfunctional glia are therefore poised to be drivers of pathophysiology, nominating a potential therapeutic target for treating TSC and related mTORopathies.
Collapse
Affiliation(s)
- Thomas L. Li
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
| | - John D. Blair
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Taesun Yoo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
| | - Gerald A. Grant
- Department of Neurosurgery, Lucile Packard Children’s Hospital and Stanford University Medical Center, Stanford, CA, USA
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Brenda E. Porter
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Helen S. Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
21
|
Busquets O, Li H, Syed KM, Jerez PA, Dunnack J, Bu RL, Verma Y, Pangilinan GR, Martin A, Straub J, Du Y, Simon VM, Poser S, Bush Z, Diaz J, Sahagun A, Gao J, Hong S, Hernandez DG, Levine KS, Booth EO, Blanchette M, Bateup HS, Rio DC, Blauwendraat C, Hockemeyer D, Soldner F. iSCORE-PD: an isogenic stem cell collection to research Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.12.579917. [PMID: 38405931 PMCID: PMC10888955 DOI: 10.1101/2024.02.12.579917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by complex genetic and environmental factors. Genome-edited human pluripotent stem cells (hPSCs) offer a unique experimental platform to advance our understanding of PD etiology by enabling the generation of disease-relevant cell types carrying patient mutations along with isogenic control cells. To facilitate this approach, we generated a collection of 65 human stem cell lines genetically engineered to harbor high risk or causal variants in genes associated with PD (SNCA A53T, SNCA A30P, PRKN Ex3del, PINK1 Q129X, DJ1/PARK7 Ex1-5del, LRRK2 G2019S, ATP13A2 FS, FBXO7 R498X/FS, DNAJC6 c.801 A>G/FS, SYNJ1 R258Q/FS, VPS13C A444P/FS, VPS13C W395C/FS, GBA1 IVS2+1/FS). All mutations were introduced into a fully characterized and sequenced female human embryonic stem cell (hESC) line (WIBR3; NIH approval number NIHhESC-10-0079) using different genome editing techniques. To ensure the genetic integrity of these cell lines, we implemented rigorous quality controls, including whole-genome sequencing of each line. Our analysis of the genetic variation in this cell line collection revealed that while genome editing, particularly using CRISPR/Cas9, can introduce rare off-target mutations, the predominant source of genetic variants arises from routine cell culture and are fixed in cell lines during clonal isolation. The observed genetic variation was minimal compared to that typically found in patient-derived iPSC experiments and predominantly affected non-coding regions of the genome. Importantly, our analysis outlines strategies for effectively managing genetic variation through stringent quality control measures and careful experimental design. This systematic approach ensures the high quality of our stem cell collection, highlights advantages of prime editing over conventional CRISPR/Cas9 methods and provides a roadmap for the generation of gene-edited hPSC collections at scale in an academic setting. Our iSCORE-PD collection represents an easily accessible and valuable platform to study PD, which can be used by investigators to understand the molecular pathophysiology of PD in a human cellular setting.
Collapse
Affiliation(s)
- Oriol Busquets
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY 10461, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY 10461, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- These authors contributed equally
| | - Hanqin Li
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- These authors contributed equally
| | - Khaja Mohieddin Syed
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- These authors contributed equally
| | - Pilar Alvarez Jerez
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- These authors contributed equally
| | - Jesse Dunnack
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- These authors contributed equally
| | - Riana Lo Bu
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY 10461, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Yogendra Verma
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Gabriella R. Pangilinan
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Annika Martin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jannes Straub
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - YuXin Du
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Vivien M. Simon
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Steven Poser
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY 10461, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Zipporiah Bush
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY 10461, USA
| | - Jessica Diaz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY 10461, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Atehsa Sahagun
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jianpu Gao
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Samantha Hong
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dena G. Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kristin S. Levine
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ezgi O. Booth
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Helen S. Bateup
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| | - Donald C. Rio
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer’s and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dirk Hockemeyer
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| | - Frank Soldner
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Albert Einstein College of Medicine, 1410 Pelham Parkway South, Bronx, NY 10461, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY 10461, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Ave., Bronx, NY 10461, USA
- Lead contact
| |
Collapse
|
22
|
Jin Y, Guo Y, Li Q, Wu L, Ge Y, Zhao J. Non-Invasive and Long-Term Electrophysiological Monitoring Sensors for Cerebral Organoids Differentiation. BIOSENSORS 2025; 15:173. [PMID: 40136970 PMCID: PMC11940203 DOI: 10.3390/bios15030173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
Cerebral organoids derived from human induced pluripotent stem cells (iPSCs) have emerged as powerful in vitro models for studying human brain development and neurological disorders. Understanding the electrophysiological properties of these organoids is crucial for evaluating their functional maturity and potential applications. However, the differentiation and maturation of stem cells into cerebral organoids is a long, slow, and error-prone process. Hence, it is vitally crucial to establish a non-invasive method of monitoring the process over a long period of time. In this study, a planar microelectrode array (MEA) with platinum (Pt) black electroplating is designed to monitor the electrophysiological activities and pharmacological responses of cerebral organoids using an external neural signal acquisition system interfaced with the MEA. The planar MEA with Pt black electroplating has a significantly reduced electrode impedance and exhibits a robust capability for the real-time detection of spontaneous neural activities, including extracellular spikes and local field potentials. Distinct electrophysiological signal strengths in cerebral organoids were observed at early and late developmental stages. Further pharmacological stimulations showed that 30 mM KCl would induce a marked increase in spike rate, indicating an enhancement of neuronal depolarization and an elevation of network excitability. This robust response to KCl stimulation in mature networks serves as a reliable indicator of neural maturity in cerebral organoids and underscores the platform's potential for drug screening applications. This work highlights the integration of MEA technology with cerebral organoids, offering a powerful platform for real-time electrophysiological monitoring. It provides new insights into the functional maturation of neural networks and establishes a reliable system for drug screening and disease modeling, facilitating future research into human brain physiology and pathology.
Collapse
Affiliation(s)
- Yan Jin
- College of Sciences, Shanghai Institute of Technology, Shanghai 201418, China; (Y.J.); (Y.G.)
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Yixun Guo
- College of Sciences, Shanghai Institute of Technology, Shanghai 201418, China; (Y.J.); (Y.G.)
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Qiushi Li
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Lei Wu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Yuqing Ge
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
| | - Jianlong Zhao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, China; (Q.L.); (L.W.)
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Frontier Innovation Research Institute, Shanghai 201108, China
| |
Collapse
|
23
|
Duenki T, Ikeuchi Y. Insulative Compression of Neuronal Tissues on Microelectrode Arrays by Perfluorodecalin Enhances Electrophysiological Measurements. Adv Healthc Mater 2025; 14:e2403771. [PMID: 39757474 PMCID: PMC11874680 DOI: 10.1002/adhm.202403771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Indexed: 01/07/2025]
Abstract
Microelectrode array (MEA) techniques provide a powerful method for exploration of neural network dynamics. A critical challenge is to interface 3D neural tissues including neural organoids with the flat MEAs surface, as it is essential to place neurons near to the electrodes for recording weak extracellular signals of neurons. To enhance performance of MEAs, most research have focused on improving their surface treatment, while little attention has been given to improve the tissue-MEA interactions from the medium side. Here, a strategy is introduced to augment MEA measurements by overlaying perfluorodecalin (PFD), a biocompatible fluorinated solvent, over neural tissues. Laying PFD over cerebral organoids insulates and compresses the tissues on MEA, which significantly enhances electrophysiological recordings. Even subtle signals such as the propagation of action potentials in bundled axons of motor nerve organoids can be detected with the technique. Moreover, PFD stabilizes tissues in acute recordings and its transparency allows optogenetic manipulations. This research highlights the potential of PFD as a tool for refining electrophysiological measurements of in vitro neuronal cultures. This can open new avenues to leverage precision of neuroscientific investigations and expanding the toolkit for in vitro studies of neural function and connectivity.
Collapse
Affiliation(s)
- Tomoya Duenki
- Institute of Industrial ScienceThe University of TokyoMeguroTokyo153‐8505Japan
- Institute for AI and BeyondThe University of TokyoBunkyoTokyo113‐8655Japan
- Department of Chemistry and BiotechnologyThe University of TokyoBunkyoTokyo113‐8655Japan
- LIMMSCNRS‐Institute of Industrial ScienceThe University of TokyoIRL 2820MeguroTokyo153‐8505Japan
| | - Yoshiho Ikeuchi
- Institute of Industrial ScienceThe University of TokyoMeguroTokyo153‐8505Japan
- Institute for AI and BeyondThe University of TokyoBunkyoTokyo113‐8655Japan
- Department of Chemistry and BiotechnologyThe University of TokyoBunkyoTokyo113‐8655Japan
- LIMMSCNRS‐Institute of Industrial ScienceThe University of TokyoIRL 2820MeguroTokyo153‐8505Japan
| |
Collapse
|
24
|
Wu X, Kreutz A, Dixon D, Tokar EJ. Engineering human cerebral organoids to explore mechanisms of arsenic-induced developmental neurotoxicity. Toxicol Appl Pharmacol 2025; 496:117230. [PMID: 39842615 PMCID: PMC11846691 DOI: 10.1016/j.taap.2025.117230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/26/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025]
Abstract
Modeling brain development and function is challenging due to complexity of the organ. Human pluripotent stem cell (PSC)-derived brain-like organoids provide new tools to study the human brain. Compared with traditional in vivo toxicological studies, these 3D models, together with 2D cellular assays, enhance our understanding of the mechanisms of developmental neurotoxicity (DNT) during the early stages of neurogenesis and offer numerous advantages including a rapid, cost-effective approach for understanding compound mechanisms and assessing chemical safety. Arsenic (As) exposure is associated with DNT, although the mechanisms involved are not well-defined. Here, we used 3D PSC-derived embryoid bodies (EBs) to recapitulate events involved in embryogenesis and neurogenesis before neural induction, and EB-derived cerebral organoids to mimic neural development in vivo. As (0.5 μM; 35 ppb) increased ectoderm differentiation within the EBs by upregulating genes (PAX6, SOX1) critical for embryonic development. Histological staining of EBs showed As disrupted neural rosette structures. qPCR and RNA-seq showed As inhibited expression of markers of mature neural cells (MAP2+/vGLUT2+) and astrocytes (GFAP+). In organoids, Ingenuity Pathway Analysis was used to identify the top 5 pathways affected by As exposure, and Gene Ontology enrichment analysis found several key signaling pathways to be inhibited by As exposure. These data provide insights into pathways contributing to As-induced inhibition of neurite outgrowth and disrupted neural rosette structures in the 2D neurite outgrowth assay and in organoids, respectively. Results herein show this multipronged 2D/3D approach can provide valuable insights into cellular events and molecular mechanisms of As-induced DNT.
Collapse
Affiliation(s)
- Xian Wu
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Anna Kreutz
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Darlene Dixon
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America
| | - Erik J Tokar
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, United States of America.
| |
Collapse
|
25
|
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
|
26
|
Mattingly Z, Chetty S. Untangling the Molecular Mechanisms Contributing to Autism Spectrum Disorder Using Stem Cells. Autism Res 2025; 18:476-485. [PMID: 39989339 DOI: 10.1002/aur.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Autism spectrum disorder (ASD) is a complex neuro developmental condition characterized by significant genetic and phenotypic variability, making diagnosis and treatment challenging. The heterogeneity of ASD-associated genetic variants and the absence of clear causal factors in many cases complicate personalized care. Traditional models, such as postmortem brain tissue and animal studies, have provided valuable insights but are limited in capturing the dynamic processes and human-specific aspects of ASD pathology. Recent advances in human induced pluripotent stem cell (iPSC) technology have transformed ASD research by enabling the generation of patient-derived neural cells in both two-dimensional cultures and three-dimensional brain organoid models. These models retain the donor's genetic background, allowing researchers to investigate disease-specific cellular and molecular mechanisms while identifying potential therapeutic targets tailored to individual patients. This commentary highlights how stem cell-based approaches are advancing our understanding of ASD and paving the way for more personalized diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zoe Mattingly
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sundari Chetty
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Park SH, Sun W. Toxicity assessment using neural organoids: innovative approaches and challenges. Toxicol Res 2025; 41:91-103. [PMID: 40013084 PMCID: PMC11850696 DOI: 10.1007/s43188-025-00279-y] [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: 10/21/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 02/28/2025] Open
Abstract
Assessment of toxicity and efficacy in the nervous system is essential to ensure the safety of compounds and the efficacy of neurotherapeutics. Recently, technologies using neural organoids to mimic the structural and functional properties of human brain tissue have been developed to improve our understanding of human-specific brain development and to model neurodevelopmental disorders. This approach offers the potential for standardized toxicity testing and large-scale drug screening at the organ level. Here, we review recent advances in neural organoids and explore the possibility of establishing more accurate and efficient systems for toxicological screening applications. Our review provides insights into toxicity and efficacy assessment research using neural organoids.
Collapse
Affiliation(s)
- Si-Hyung Park
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21 Plus Program for Biomedical Science, College of Medicine, Korea University, 73, Goryeodae-ro, Seongbuk-gu, Seoul, 02841 Republic of Korea
| |
Collapse
|
28
|
Kim E, Jeong E, Hong YM, Jeong I, Kim J, Kwon YW, Park YG, Lee J, Choi S, Kim JY, Lee JH, Cho SW, Park JU. Magnetically reshapable 3D multi-electrode arrays of liquid metals for electrophysiological analysis of brain organoids. Nat Commun 2025; 16:2011. [PMID: 40016200 PMCID: PMC11868496 DOI: 10.1038/s41467-024-55752-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 12/23/2024] [Indexed: 03/01/2025] Open
Abstract
To comprehend the volumetric neural connectivity of a brain organoid, it is crucial to monitor the spatiotemporal electrophysiological signals within the organoid, known as intra-organoid signals. However, previous methods risked damaging the three-dimensional (3D) cytoarchitecture of organoids, either through sectioning or inserting rigid needle-like electrodes. Also, the limited numbers of electrodes in fixed positions with non-adjustable electrode shapes were insufficient for examining the complex neural activity throughout the organoid. Herein, we present a magnetically reshapable 3D multi-electrode array (MEA) using direct printing of liquid metals for electrophysiological analysis of brain organoids. The adaptable distribution and the softness of these printed electrodes facilitate the spatiotemporal recording of intra-organoid signals. Furthermore, the unique capability to reshape these soft electrodes within the organoid using magnetic fields allows a single electrode in the MEA to record from multiple points, effectively increasing the recording site density without the need for additional electrodes.
Collapse
Affiliation(s)
- Enji Kim
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yeon-Mi Hong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Inhea Jeong
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Junghoon Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yong Won Kwon
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Young-Geun Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
| | - Jiin Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suah Choi
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ju-Young Kim
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea.
| | - Seung-Woo Cho
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea.
| | - Jang-Ung Park
- Department of Materials Science and Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, 03722, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Yonsei, Republic of Korea.
- Department of Neurosurgery, Yonsei University College of Medicine, Yonsei, Republic of Korea.
- Yonsei-KIST Convergence Research Institute, Seoul, 03722, Republic of Korea.
| |
Collapse
|
29
|
Załuska-Ogryzek K, Wróblewska-Łuczka P, Góralczyk A, Łuszczki JJ. Isobolographic interactions of cannabidiol and AM 1172 with cisplatin in human neuroblastoma and glioblastoma cell lines: An in vitro study. Chem Biol Interact 2025; 408:111392. [PMID: 39828184 DOI: 10.1016/j.cbi.2025.111392] [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: 09/10/2024] [Revised: 12/10/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
Glioblastoma is the most aggressive brain cancer in humans with very poor prognosis and high mortality rate. Despite advances in treatment, glioblastoma almost always recurs and new therapeutic methods are urgently needed. This study aimed at assessing the cytotoxic and antiproliferative effects of AM 1172 and cannabidiol (two cannabinoid receptor ligands) in vitro, when used alone and in combination with cisplatin (a standard cytotoxic drug), in various human neuroblastoma (CHP-134, KELLY), human glioblastoma (U-87MG and T98G) and rat glioblastoma (C6) cell lines. Our experiments showed that AM 1172 and cannabidiol inhibited cell proliferation with IC50 values in the range of 2.29-17.21 μM (for AM 1172) and 11.61-20.35 μM (for cannabidiol), respectively. The selectivity index for AM 1172 ranged from 0.61 to 4.60 and that for cannabidiol ranged from 1.45 to 2.55 in the studied glioblastoma and neuroblastoma cell lines. With isobolographic analysis, it was found that AM 1172 combined with cisplatin exerted a synergistic interaction in the CHP-134 cell line (p < 0.01). In contrast, AM 1172 when combined with cisplatin produced an antagonistic interaction in the C6 cell line (p < 0.01). The remaining combinations of AM 1172 with cisplatin in the U-87MG, KELLY and T98G cell lines were additive. In case of cannabidiol, its combination with cisplatin produced an antagonistic interaction in the T98G cell line (p < 0.0001), whereas the combinations of cannabidiol with cisplatin in the CHP-134, U-87MG, KELLY, and C6 cell lines were additive in nature. The synergistic and additive interactions for the combination of AM 1172 and cannabidiol with cisplatin seem to be a promising direction in glioblastoma therapy. Unfortunately, the combinations producing antagonistic interactions (AM 1172+cisplatin in C6, and cannabidiol + cisplatin in T98G cell lines) should be avoided due to the antagonistic antiproliferative effect of two-drug mixtures.
Collapse
Affiliation(s)
- Katarzyna Załuska-Ogryzek
- Department of Pathophysiology, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Paula Wróblewska-Łuczka
- Department of Occupational Medicine, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Agnieszka Góralczyk
- Department of Occupational Medicine, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland
| | - Jarogniew J Łuszczki
- Department of Occupational Medicine, Medical University of Lublin, Ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| |
Collapse
|
30
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [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: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
31
|
Rubio AD, Hamilton L, Bausch M, Jin M, Papetti A, Jiang P, Yelamanchili SV. A Comprehensive Review on Utilizing Human Brain Organoids to Study Neuroinflammation in Neurological Disorders. J Neuroimmune Pharmacol 2025; 20:23. [PMID: 39987404 PMCID: PMC11846768 DOI: 10.1007/s11481-025-10181-x] [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: 07/15/2024] [Accepted: 01/26/2025] [Indexed: 02/24/2025]
Abstract
Most current information about neurological disorders and diseases is derived from direct patient and animal studies. However, patient studies in many cases do not allow replication of the early stages of the disease and, therefore, offer limited opportunities to understand disease progression. On the other hand, although the use of animal models allows us to study the mechanisms of the disease, they present significant limitations in developing drugs for humans. Recently, 3D-cultured in vitro models derived from human pluripotent stem cells have surfaced as a promising system. They offer the potential to connect findings from patient studies with those from animal models. In this comprehensive review, we discuss their application in modeling neurodevelopmental conditions such as Down Syndrome or Autism, neurodegenerative diseases such as Alzheimer's or Parkinson's, and viral diseases like Zika virus or HIV. Furthermore, we will discuss the different models used to study prenatal exposure to drugs of abuse, as well as the limitations and challenges that must be met to transform the landscape of research on human brain disorders.
Collapse
Affiliation(s)
- Adrian Domene Rubio
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | - Luke Hamilton
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | - Mark Bausch
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Ava Papetti
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Sowmya V Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA.
| |
Collapse
|
32
|
Tian C, Ao Z, Cerneckis J, Cai H, Chen L, Niu H, Takayama K, Kim J, Shi Y, Gu M, Kanekiyo T, Guo F. Understanding monocyte-driven neuroinflammation in Alzheimer's disease using human brain organoid microphysiological systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638539. [PMID: 40027735 PMCID: PMC11870548 DOI: 10.1101/2025.02.16.638539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) pathogenesis strongly correlates with neuroinflammation. Peripheral monocytes are crucial components of the human immune system that may play a role in neuroinflammation, but their contribution to AD pathogenesis is largely understudied partially due to the lack of appropriate human models. Here, we present human cortical organoid microphysiological systems (hCO-MPSs) for modeling dynamic AD neuroinflammation mediated by monocytes. By incorporating 3D printed devices into an existing cortical organoid protocol, 96 hCO-MPSs can be established with significantly reduced necrosis and hypoxia as well as enhanced viability within a commonly used 96 well plate, and each hCO-MPS consists of a doughnut-shaped hCO and a 3D printed device per well. Using this approach, monocytes from AD patients exhibit higher infiltration, decreased amyloid-beta (Aβ) clearance, and stronger inflammatory responses compared to monocytes from age-matched control donors. Moreover, pro-inflammatory effects such as elevated astrocyte activation and neuronal apoptosis were observed to be induced by AD monocytes. Furthermore, the significant increase in the expression of IL1B and CCL3, both at the transcriptional and protein levels, indicated the pivotal role of these cytokine and chemokine in monocyte-mediated AD neuroinflammation. Our findings provide insight for understanding monocytes' role in AD pathogenesis, and the user-friendly MPS models we present are compatible with existing laboratory settings, highlighting their potential for modeling neuroinflammation and developing new therapeutics for various neuroinflammatory diseases.
Collapse
|
33
|
Akkouh IA, Osete JR, Szabo A, Andreassen OA, Djurovic S. Neurobiological Perturbations in Bipolar Disorder Compared With Schizophrenia: Evidence From Cell Cultures and Brain Organoids. Biol Psychiatry 2025:S0006-3223(25)00110-6. [PMID: 39983953 DOI: 10.1016/j.biopsych.2025.02.012] [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: 10/16/2024] [Revised: 01/06/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
Bipolar disorder (BD) and schizophrenia (SCZ) are uniquely human disorders with a complex pathophysiology that involves adverse neuropathological events in brain development. High disease polygenicity and limited access to live human brain tissue make these disorders exceedingly challenging to study mechanistically. Cellular cultures and brain organoids generated from human-derived pluripotent stem cells preserve the genetic background of the donor cells and recapitulate some of the defining characteristics of human brain architecture and early spatiotemporal development. These model systems have already proven successful in deciphering some of the neuropathological perturbations in BD and SCZ, and methodological advancements, such as the functional integration of 2 or more region-specific organoids and organoid transplantation in animals, promise to deliver increasingly refined insights. Here, we review a selection of recent discoveries achieved by stem cell-based models, with a particular focus on patterns of cellular and molecular convergence and divergence between BD and SCZ. First, we provide a brief overview of the evidence from glial and neuronal cell cultures and brain organoids, centering our discussion on several key functional domains, including neuroinflammation, neuronal excitability, and mitochondrial function. Then, we review recent findings demonstrating the power of integrating stem cell-based systems with gene editing technologies to elucidate the functional consequences of risk variants identified through genetic association studies. We end with a discussion of current challenges and some promising avenues for future research.
Collapse
Affiliation(s)
- Ibrahim A Akkouh
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Jordi Requena Osete
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Attila Szabo
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
34
|
Acha C, George D, Diaz LC, Ouyang Z, Alam El Din DMM, Surlekar H, Moghadas B, Loftus E, Mangalvedhekar GM, Rayasam PSR, Lai YC, Smirnova L, Caffo BS, Johnson EC, Gracias DH. Neuromodulation in neural organoids with shell MEAs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.637712. [PMID: 40027665 PMCID: PMC11870477 DOI: 10.1101/2025.02.18.637712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Neural organoids (NOs) have emerged as important tissue engineering models for brain sciences and biocomputing. Establishing reliable relationships between stimulation and recording traces of electrical activity is essential to monitor the functionality of NOs, especially as it relates to realizing biocomputing paradigms such as reinforcement learning or stimulus discrimination. While researchers have demonstrated neuromodulation in NOs, they have primarily used 2D microelectrode arrays (MEAs) with limited access to the entire 3D contour of the NOs. Here, we report neuromodulation using tiny mimics of macroscale EEG caps or shell MEAs. Specifically, we observe that stimulating current within a specific range (20 to 30 µA) induced a statistically significant increase in neuron firing rate when comparing the activity five seconds before and after stimulation. We observed neuromodulatory behavior using both three- and 16-electrode shells and could generate 3D spatiotemporal maps of neuromodulatory activity around the surface of the NO. Our studies demonstrate a methodology for investigating 3D spatiotemporal neuromodulation in organoids of broad relevance to biomedical engineering and biocomputing. One-Sentence Summary Neuromodulation, an essential intelligence feature, was observed using 3D stimulation and recording from neural organoids.
Collapse
|
35
|
Noh S, Park Y, Kim B, Mun JY. Structural Analysis of Cerebral Organoids Using Confocal Microscopy and Transmission/Scanning Electron Microscopy. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2025; 31:ozae119. [PMID: 39999189 DOI: 10.1093/mam/ozae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/14/2024] [Accepted: 11/10/2024] [Indexed: 02/27/2025]
Abstract
Cerebral organoid cultures from human-induced pluripotent stem cells are widely used to study complex human brain development; however, there is still limited ultrastructural information regarding the development. In this study, we examined the structural details of cerebral organoids using various microscopy techniques. Two protocols were chosen as representative methods for the development of brain organoids: the classic whole-cerebral organoid (Whole-CO) culture technique, and the air-liquid interface-cerebral organoid (ALI-CO) culture technique. Immunostained confocal laser scanning microscopy (CLSM) revealed the formation of the CTIP2- and TBR1-positive cortical deep layer on days 90 and 150, depending on the developmental progress of both methods. Furthermore, the presence of astrocytes and oligodendrocytes was verified through immunostained CLSM utilizing two-dimensional and three-dimensional reconstruction images after a 150-day period. Transmission electron microscopy analysis revealed nanometer-resolution details of the cellular organelles and neuron-specific structures including synapses and myelin. Large-area scanning electron microscopy confirmed the well-developed neuronal connectivity from each culture method on day 150. Using those microscopy techniques, we clearly showed significant details within two representative culture protocols, the Whole-CO and ALI-CO culture methods. These multi-level images provide ultrastructural insight into the features of cerebral organoids depending on the developmental stage.
Collapse
Affiliation(s)
- Seulgi Noh
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Korea
| | - Yurim Park
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Beomsue Kim
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| | - Ji Young Mun
- Neural Circuits Research Group, Korea Brain Research Institute (KBRI), Daegu, Korea
| |
Collapse
|
36
|
Charles S, Jackson-Holmes E, Sun G, Zhou Y, Siciliano B, Niu W, Han H, Nikitina A, Kemp ML, Wen Z, Lu H. Non-Invasive Quality Control of Organoid Cultures Using Mesofluidic CSTR Bioreactors and High-Content Imaging. ADVANCED MATERIALS TECHNOLOGIES 2025; 10:2400473. [PMID: 40248044 PMCID: PMC12002419 DOI: 10.1002/admt.202400473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Indexed: 04/19/2025]
Abstract
Human brain organoids produce anatomically relevant cellular structures and recapitulate key aspects of in vivo brain function, which holds great potential to model neurological diseases and screen therapeutics. However, the long growth time of 3D systems complicates the culturing of brain organoids and results in heterogeneity across samples hampering their applications. We developed an integrated platform to enable robust and long-term culturing of 3D brain organoids. We designed a mesofluidic bioreactor device based on a reaction-diffusion scaling theory, which achieves robust media exchange for sufficient nutrient delivery in long-term culture. We integrated this device with longitudinal tracking and machine learning-based classification tools to enable non-invasive quality control of live organoids. This integrated platform allows for sample pre-selection for downstream molecular analysis. Transcriptome analyses of organoids revealed that our mesofluidic bioreactor promoted organoid development while reducing cell death. Our platform thus offers a generalizable tool to establish reproducible culture standards for 3D cellular systems for a variety of applications beyond brain organoids.
Collapse
Affiliation(s)
- Seleipiri Charles
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Emily Jackson-Holmes
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Gongchen Sun
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Ying Zhou
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Benjamin Siciliano
- Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, 615 Michael Street, Atlanta, GA, 30322, U.S.A
| | - Weibo Niu
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Haejun Han
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Arina Nikitina
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| | - Melissa L Kemp
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, 615 Michael Street, Atlanta, Georgia 30322, U.S.A
| | - Hang Lu
- Interdisciplinary Program in Bioengineering, Georgia Institute of Technology, 311 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, Georgia 30332, U.S.A
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive NW Atlanta, Georgia 30332, U.S.A
| |
Collapse
|
37
|
Sojka C, Wang HLV, Bhatia TN, Li Y, Chopra P, Sing A, Voss A, King A, Wang F, Joseph K, Ravi VM, Olson J, Hoang K, Nduom E, Corces VG, Yao B, Sloan SA. Mapping the developmental trajectory of human astrocytes reveals divergence in glioblastoma. Nat Cell Biol 2025; 27:347-359. [PMID: 39779941 DOI: 10.1038/s41556-024-01583-9] [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: 06/03/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Glioblastoma (GBM) is defined by heterogeneous and resilient cell populations that closely reflect neurodevelopmental cell types. Although it is clear that GBM echoes early and immature cell states, identifying the specific developmental programmes disrupted in these tumours has been hindered by a lack of high-resolution trajectories of glial and neuronal lineages. Here we delineate the course of human astrocyte maturation to uncover discrete developmental stages and attributes mirrored by GBM. We generated a transcriptomic and epigenomic map of human astrocyte maturation using cortical organoids maintained in culture for nearly 2 years. Through this approach, we chronicled a multiphase developmental process. Our time course of human astrocyte maturation includes a molecularly distinct intermediate period that serves as a lineage commitment checkpoint upstream of mature quiescence. This intermediate stage acts as a site of developmental deviation separating IDH-wild-type neoplastic astrocyte-lineage cells from quiescent astrocyte populations. Interestingly, IDH1-mutant tumour astrocyte-lineage cells are the exception to this developmental perturbation, where immature properties are suppressed as a result of D-2-hydroxyglutarate oncometabolite exposure. We propose that this defiance is a consequence of IDH1-mutant-associated epigenetic dysregulation, and we identified biased DNA hydroxymethylation (5hmC) in maturation genes as a possible mechanism. Together, this study illustrates a distinct cellular state aberration in GBM astrocyte-lineage cells and presents developmental targets for experimental and therapeutic exploration.
Collapse
Affiliation(s)
- Caitlin Sojka
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Hsiao-Lin V Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Tarun N Bhatia
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Yangping Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Pankaj Chopra
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anson Sing
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Anna Voss
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexia King
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Feng Wang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Kevin Joseph
- Department of Neurosurgery, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vidhya M Ravi
- Department of Neurosurgery, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jeffrey Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Kimberly Hoang
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Edjah Nduom
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Victor G Corces
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Bing Yao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
38
|
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
|
39
|
You JH, Kim NY, Choi YY, Choi HW, Chung BG. Dual-stimuli-responsive nanoparticles for the co-delivery of small molecules to promote neural differentiation of human iPSCs. NANOSCALE 2025; 17:2506-2519. [PMID: 39815767 DOI: 10.1039/d4nr04413d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The differentiation of human induced pluripotent stem cells (hiPSCs) into neural progenitor cells (NPCs) is a promising approach for the treatment of neurodegenerative diseases and regenerative medicine. Dual-SMAD inhibition using small molecules has been identified as a key strategy for directing the differentiation of hiPSCs into NPCs by regulating specific cell signaling pathways. However, conventional culture methods are time-consuming and exhibit low differentiation efficiency in neural differentiation. Nanocarriers can address these obstacles as an efficient platform for the controlled release and accurate delivery of small molecules. In this paper, we developed calcium phosphate-coated mesoporous silica nanoparticles capable of delivering multiple small molecules, including LDN193189 as a bone morphogenetic protein (BMP) inhibitor and SB431542 as a transforming growth factor (TGF)-beta inhibitor, for direct differentiation of hiPSC-mediated NPCs. Our results demonstrated that this nanocarrier-mediated small molecule release system not only enhanced the in vitro formation of neural rosettes but also modulated the expression levels of key markers. In particular, it downregulated OCT4, a marker of pluripotency, while upregulating PAX6, a critical marker for the neuroectoderm. These findings suggest that this controlled small molecule release system holds significant potential for therapeutic applications in neural development and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jeong Hyun You
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea.
| | - Na Yeon Kim
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea.
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| | - Hyung Woo Choi
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
| | - Bong Geun Chung
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea.
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
- Institute of Smart Biosensor, Sogang University, Seoul 04107, Korea
| |
Collapse
|
40
|
Park S, Min CH, Choi E, Choi JS, Park K, Han S, Choi W, Jang HJ, Cho KO, Kim M. Long-term tracking of neural and oligodendroglial development in large-scale human cerebral organoids by noninvasive volumetric imaging. Sci Rep 2025; 15:2536. [PMID: 39833280 PMCID: PMC11747076 DOI: 10.1038/s41598-025-85455-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025] Open
Abstract
Human cerebral organoids serve as a quintessential model for deciphering the complexities of brain development in a three-dimensional milieu. However, imaging these organoids, particularly when they exceed several millimeters in size, has been curtailed by the technical impediments such as phototoxicity, slow imaging speeds, and inadequate resolution and imaging depth. Addressing these pivotal challenges, our study has pioneered a high-speed scanning microscope, synergistically coupled with advanced computational image processing. This ensemble has empowered us to monitor the intricate dynamics of neuron and oligodendrocyte development within cerebral organoids across a trajectory of approximately two months. Line-shaped illumination mitigates photodamage and, alongside refined spatial gating, maximizes signal collection through integrating with computational processing. The integration of deconvolution and compressive sensing has improved image contrast by 6-fold, elucidating fine features of the neurites. Thus, noninvasive imaging enabled us to perform long-term tracking of neural and oligodendroglial development in the large-scale human cerebral organoid. Furthermore, our sophisticated volumetric segmentation algorithm has yielded a robust four-dimensional quantitative analysis, encapsulating both neuronal and oligodendroglial maturation. Collectively, these advances mark a significant advancement in the field of neurodevelopment, providing a powerful tool for in-depth study of complex brain organoid systems.
Collapse
Affiliation(s)
- Sangjun Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Cheol Hong Min
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Eunjin Choi
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jeong-Sun Choi
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Kyungjin Park
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedical Engineering, UNIST, Ulsan, 44919, Korea
| | - Seokyoung Han
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Mechanical Engineering, University of Louisville, Louisville, KY, 40208, USA
| | - Wonjun Choi
- Park Systems Corp, Suwon, 16229, Gyeonggi-do, Korea
| | - Hyun-Jong Jang
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, Seoul, 06591, Korea
| | - Kyung-Ok Cho
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Catholic Neuroscience Institute, Institute for Aging and Metabolic Diseases, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, Seoul, 06591, Korea.
| | - Moonseok Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Medical Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- CMC Institute for Basic Medical Science, The Catholic Medical Center of The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
41
|
Pagliaro A, Artegiani B, Hendriks D. Emerging approaches to enhance human brain organoid physiology. Trends Cell Biol 2025:S0962-8924(24)00254-X. [PMID: 39826996 DOI: 10.1016/j.tcb.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Brain organoids are important 3D models for studying human brain development, disease, and evolution. To overcome some of the existing limitations that affect organoid quality, reproducibility, characteristics, and in vivo resemblance, current efforts are directed to improve their physiological relevance by exploring different, yet interconnected, routes. In this review, these approaches and their latest developments are discussed, including stem cell optimization, refining morphogen administration strategies, altering the extracellular matrix (ECM) niche, and manipulating tissue architecture to mimic in vivo brain morphogenesis. Additionally, strategies to increase cell diversity and enhance organoid maturation, such as establishing co-cultures, assembloids, and organoid in vivo xenotransplantation, are reviewed. We explore how these various factors can be tuned and intermingled and speculate on future avenues towards even more physiologically-advanced brain organoids.
Collapse
Affiliation(s)
- Anna Pagliaro
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | | | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
42
|
Winden KD, Gisser I, Sahin M. Using cortical organoids to understand the pathogenesis of malformations of cortical development. Front Neurosci 2025; 18:1522652. [PMID: 39881808 PMCID: PMC11774837 DOI: 10.3389/fnins.2024.1522652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025] Open
Abstract
Malformations of cortical development encompass a broad range of disorders associated with abnormalities in corticogenesis. Widespread abnormalities in neuronal formation or migration can lead to small head size or microcephaly with disorganized placement of cell types. Specific, localized malformations are termed focal cortical dysplasias (FCD). Neurodevelopmental disorders are common in all types of malformations of cortical development with the most prominent being refractory epilepsy, behavioral disorders such as autism spectrum disorder (ASD), and learning disorders. Several genetic pathways have been associated with these disorders from control of cell cycle and cytoskeletal dynamics in global malformations to variants in growth factor signaling pathways, especially those interacting with the mechanistic target of rapamycin (mTOR), in FCDs. Despite advances in understanding these disorders, the underlying developmental pathways that lead to lesion formation and mechanisms through which defects in cortical development cause specific neurological symptoms often remains unclear. One limitation is the difficulty in modeling these disorders, as animal models frequently do not faithfully mirror the human phenotype. To circumvent this obstacle, many investigators have turned to three-dimensional human stem cell models of the brain, known as organoids, because they recapitulate early neurodevelopmental processes. High throughput analysis of these organoids presents a promising opportunity to model pathophysiological processes across the breadth of malformations of cortical development. In this review, we highlight advances in understanding the pathophysiology of brain malformations using organoid models.
Collapse
Affiliation(s)
| | | | - Mustafa Sahin
- Department of Neurology, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Chen X, Sun G, Feng L, Tian E, Shi Y. Human iPSC-derived microglial cells protect neurons from neurodegeneration in long-term cultured adhesion brain organoids. Commun Biol 2025; 8:30. [PMID: 39789340 PMCID: PMC11718079 DOI: 10.1038/s42003-024-07401-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
Brain organoid models have greatly facilitated our understanding of human brain development and disease. However, key brain cell types, such as microglia, are lacking in most brain organoid models. Because microglia have been shown to play important roles in brain development and pathologies, attempts have been made to add microglia to brain organoids through co-culture. However, only short-term microglia-organoid co-cultures can be established, and it remains challenging to have long-lasting survival of microglia in organoids to mimic long-term residency of microglia in the brain. In this study, we developed an adhesion brain organoid (ABO) platform that allows prolonged culture of brain organoids (greater than a year). Moreover, the long-term (LT)-ABO system contains abundant astrocytes and can support prolonged survival and ramification of microglia. Furthermore, we showed that microglia in the LT-ABO could protect neurons from neurodegeneration by increasing synaptic density and reducing p-Tau level and cell death in the LT-ABO. Therefore, the microglia-containing LT-ABO platform generated in this study provides a promising human cellular model for studying neuron-glia and glia-glia interactions in brain development and the pathogenesis of neurodegenerative diseases such as Alzheimer's disease.
Collapse
Affiliation(s)
- Xianwei Chen
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, 200092, China
| | - Guoqiang Sun
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Lizhao Feng
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - E Tian
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, 1500 E. Duarte Rd, Duarte, CA, 91010, USA.
| |
Collapse
|
44
|
Yang Z, Teaney NA, Buttermore ED, Sahin M, Afshar-Saber W. Harnessing the potential of human induced pluripotent stem cells, functional assays and machine learning for neurodevelopmental disorders. Front Neurosci 2025; 18:1524577. [PMID: 39844857 PMCID: PMC11750789 DOI: 10.3389/fnins.2024.1524577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Neurodevelopmental disorders (NDDs) affect 4.7% of the global population and are associated with delays in brain development and a spectrum of impairments that can lead to lifelong disability and even mortality. Identification of biomarkers for accurate diagnosis and medications for effective treatment are lacking, in part due to the historical use of preclinical model systems that do not translate well to the clinic for neurological disorders, such as rodents and heterologous cell lines. Human-induced pluripotent stem cells (hiPSCs) are a promising in vitro system for modeling NDDs, providing opportunities to understand mechanisms driving NDDs in human neurons. Functional assays, including patch clamping, multielectrode array, and imaging-based assays, are popular tools employed with hiPSC disease models for disease investigation. Recent progress in machine learning (ML) algorithms also presents unprecedented opportunities to advance the NDD research process. In this review, we compare two-dimensional and three-dimensional hiPSC formats for disease modeling, discuss the applications of functional assays, and offer insights on incorporating ML into hiPSC-based NDD research and drug screening.
Collapse
Affiliation(s)
- Ziqin Yang
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Nicole A. Teaney
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Elizabeth D. Buttermore
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Human Neuron Core, Boston Children’s Hospital, Boston, MA, United States
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- Human Neuron Core, Boston Children’s Hospital, Boston, MA, United States
| | - Wardiya Afshar-Saber
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
45
|
Estudillo E, Castillo-Arellano JI, Martínez E, Rangel-López E, López-Ornelas A, Magaña-Maldonado R, Adalid-Peralta L, Velasco I, Escobedo-Ávila I. Modeling the Effect of Cannabinoid Exposure During Human Neurodevelopment Using Bidimensional and Tridimensional Cultures. Cells 2025; 14:70. [PMID: 39851498 PMCID: PMC11763397 DOI: 10.3390/cells14020070] [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: 11/07/2024] [Revised: 12/18/2024] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
Our knowledge about the consumption of cannabinoids during pregnancy lacks consistent evidence to determine whether it compromises neurodevelopment. Addressing this task is challenging and complex since pregnant women display multiple confounding factors that make it difficult to identify the real effect of cannabinoids' consumption. Recent studies shed light on this issue by using pluripotent stem cells of human origin, which can recapitulate human neurodevelopment. These revolutionary platforms allow studying how exogenous cannabinoids could alter human neurodevelopment without ethical concerns and confounding factors. Here, we review the information to date on the clinical studies about the impact of exogenous cannabinoid consumption on human brain development and how exogenous cannabinoids alter nervous system development in humans using cultured pluripotent stem cells as 2D and 3D platforms to recapitulate brain development.
Collapse
Affiliation(s)
- Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Jorge Iván Castillo-Arellano
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Emilio Martínez
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Edgar Rangel-López
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Adolfo López-Ornelas
- División de Investigación, Hospital Juárez de México, Mexico City 07760, Mexico;
- Hospital Nacional Homeopático, Hospitales Federales de Referencia, Mexico City 06800, Mexico
| | - Roxana Magaña-Maldonado
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Laura Adalid-Peralta
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
| | - Iván Velasco
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.I.C.-A.); (E.M.); (E.R.-L.); (R.M.-M.); (L.A.-P.); (I.V.)
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Itzel Escobedo-Ávila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
46
|
Kavakli E, Gul N, Begentas OC, Kiris E. Astrocytes in Primary Familial Brain Calcification (PFBC): Emphasis on the Importance of Induced Pluripotent Stem Cell-Derived Human Astrocyte Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:19-38. [PMID: 39841380 DOI: 10.1007/5584_2024_840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Primary familial brain calcification (PFBC) is a rare, progressive central nervous system (CNS) disorder without a cure, and the current treatment methodologies primarily aim to relieve neurological and psychiatric symptoms of the patients. The disease is characterized by abnormal bilateral calcifications in the brain, however, our mechanistic understanding of the biology of the disease is still limited. Determining the roles of the specific cell types and molecular mechanisms involved in the pathophysiological processes of the disease is of great importance for the development of novel and effective treatment methodologies. There is a growing interest in the involvement of astrocytes in PFBC, as recent studies have suggested that astrocytes play a central role in the disease and that functional defects in these cells are critical for the development and progression of the disease. This review aims to discuss recent findings on the roles of astrocytes in PFBC pathophysiology, with a focus on known expression and roles of PFBC genes in astrocytes. Additionally, we discuss the importance of human astrocytes for PFBC disease modeling, and astrocytes as a potential therapeutic target in PFBC. Utilization of species-specific and physiologically relevant PFBC model systems can open new avenues for basic research, drug development, and regenerative medicine.
Collapse
Affiliation(s)
- Ebru Kavakli
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Nazli Gul
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Onur Can Begentas
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Erkan Kiris
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye.
| |
Collapse
|
47
|
Shi J, Chen Q, Lai J, Zhu J, Zhang R, Mazid MA, Li D, Su H, Qin D. Impact of c-JUN deficiency on thalamus development in mice and human neural models. Cell Biosci 2024; 14:149. [PMID: 39707500 DOI: 10.1186/s13578-024-01303-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/04/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND c-Jun is a key regulator of gene expression. Through the formation of homo- or heterodimers, c-JUN binds to DNA and regulates gene transcription. While c-Jun plays a crucial role in embryonic development, its impact on nervous system development in higher mammals, especially for some deep structures, for example, thalamus in diencephalon, remains unclear. METHODS To investigate the influence of c-JUN on early nervous system development, c-Jun knockout (KO) mice and c-JUN KO H1 embryonic stem cells (ESCs)-derived neural progenitor cells (NPCs), cerebral organoids (COs), and thalamus organoids (ThOs) models were used. We detected the dysplasia via histological examination and immunofluorescence staining, omics analysis, and loss/gain of function analysis. RESULTS At embryonic day 14.5, c-Jun knockout (KO) mice exhibited sparseness of fibers in the brain ventricular parenchyma and malformation of the thalamus in the diencephalon. The absence of c-JUN accelerated the induction of NPCs but impaired the extension of fibers in human neuronal cultures. COs lacking c-JUN displayed a robust PAX6+/NESTIN+ exterior layer but lacked a fibers-connected core. Moreover, the subcortex-like areas exhibited defective thalamus characteristics with transcription factor 7 like 2-positive cells. Notably, in guided ThOs, c-JUN KO led to inadequate thalamus patterning with sparse internal nerve fibers. Chromatin accessibility analysis confirmed a less accessible chromatin state in genes related to the thalamus. Overexpression of c-JUN rescued these defects. RNA-seq identified 18 significantly down-regulated genes including RSPO2, WNT8B, MXRA5, HSPG2 and PLAGL1 while 24 genes including MSX1, CYP1B1, LMX1B, NQO1 and COL2A1 were significantly up-regulated. CONCLUSION Our findings from in vivo and in vitro experiments indicate that c-JUN depletion impedes the extension of nerve fibers and renders the thalamus susceptible to dysplasia during early mouse embryonic development and human ThO patterning. Our work provides evidence for the first time that c-JUN is a key transcription regulator that play important roles in the thalamus/diencephalon development.
Collapse
Affiliation(s)
- Jiantao Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qing Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jianheng Lai
- Guangdong Engineering Technology Research Center of Biological Targeting Diagnosis, Therapy and Rehabilitation, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jieying Zhu
- CAS Key Laboratory of Regenerative Biology, Center for Cell Lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ran Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Md Abdul Mazid
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dongwei Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Dajiang Qin
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Guangdong Engineering Technology Research Center of Biological Targeting Diagnosis, Therapy and Rehabilitation, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- Guangdong Engineering Research Center of Early Clinical Trials of Biotechnology Drugs, The Fifth Affiliated Hospital,, Guangzhou Medical University, Guangzhou, China.
- Bioland Laboratory Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| |
Collapse
|
48
|
Harary PM, Rajaram S, Chen MS, Hori YS, Park DJ, Chang SD. Genomic predictors of radiation response: recent progress towards personalized radiotherapy for brain metastases. Cell Death Discov 2024; 10:501. [PMID: 39695143 DOI: 10.1038/s41420-024-02270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/03/2024] [Accepted: 12/09/2024] [Indexed: 12/20/2024] Open
Abstract
Radiotherapy remains a key treatment modality for both primary and metastatic brain tumors. Significant technological advances in precision radiotherapy, such as stereotactic radiosurgery and intensity-modulated radiotherapy, have contributed to improved clinical outcomes. Notably, however, molecular genetics is not yet widely used to inform brain radiotherapy treatment. By comparison, genetic testing now plays a significant role in guiding targeted therapies and immunotherapies, particularly for brain metastases (BM) of lung cancer, breast cancer, and melanoma. Given increasing evidence of the importance of tumor genetics to radiation response, this may represent a currently under-utilized means of enhancing treatment outcomes. In addition, recent studies have shown potentially actionable mutations in BM which are not present in the primary tumor. Overall, this suggests that further investigation into the pathways mediating radiation response variability is warranted. Here, we provide an overview of key mechanisms implicated in BM radiation resistance, including intrinsic and acquired resistance and intratumoral heterogeneity. We then discuss advances in tumor sampling methods, such as a collection of cell-free DNA and RNA, as well as progress in genomic analysis. We further consider how these tools may be applied to provide personalized radiotherapy for BM, including patient stratification, detection of radiotoxicity, and use of radiosensitization agents. In addition, we describe recent developments in preclinical models of BM and consider their relevance to investigating radiation response. Given the increase in clinical trials evaluating the combination of radiotherapy and targeted therapies, as well as the rising incidence of BM, it is essential to develop genomically informed approaches to enhance radiation response.
Collapse
Affiliation(s)
- Paul M Harary
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjeeth Rajaram
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Maggie S Chen
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Yusuke S Hori
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David J Park
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| | - Steven D Chang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
49
|
Kim JI, Miura Y, Li MY, Revah O, Selvaraj S, Birey F, Meng X, Thete MV, Pavlov SD, Andersen J, Pașca AM, Porteus MH, Huguenard JR, Pașca SP. Human assembloids reveal the consequences of CACNA1G gene variants in the thalamocortical pathway. Neuron 2024; 112:4048-4059.e7. [PMID: 39419023 DOI: 10.1016/j.neuron.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Abnormalities in thalamocortical crosstalk can lead to neuropsychiatric disorders. Variants in CACNA1G, which encodes the α1G subunit of the thalamus-enriched T-type calcium channel, are associated with absence seizures, intellectual disability, and schizophrenia, but the cellular and circuit consequences of these genetic variants in humans remain unknown. Here, we developed a human assembloid model of the thalamocortical pathway to dissect the contribution of genetic variants in T-type calcium channels. We discovered that the M1531V CACNA1G variant associated with seizures led to changes in T-type currents in thalamic neurons, as well as correlated hyperactivity of thalamic and cortical neurons in assembloids. By contrast, CACNA1G loss, which has been associated with risk of schizophrenia, resulted in abnormal thalamocortical connectivity that was related to both increased spontaneous thalamic activity and aberrant axonal projections. These results illustrate the utility of multi-cellular systems for interrogating human genetic disease risk variants at both cellular and circuit level.
Collapse
Affiliation(s)
- Ji-Il Kim
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Min-Yin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA; Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sridhar Selvaraj
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Fikri Birey
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Xiangling Meng
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Mayuri Vijay Thete
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Sergey D Pavlov
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Jimena Andersen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA
| | - Anca M Pașca
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Stanford Brain Organogenesis, Wu Tsai Neuroscience Institute, Stanford, CA 94305, USA.
| |
Collapse
|
50
|
Amin ND, Kelley KW, Kaganovsky K, Onesto M, Hao J, Miura Y, McQueen JP, Reis N, Narazaki G, Li T, Kulkarni S, Pavlov S, Pașca SP. Generating human neural diversity with a multiplexed morphogen screen in organoids. Cell Stem Cell 2024; 31:1831-1846.e9. [PMID: 39642864 DOI: 10.1016/j.stem.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 07/31/2024] [Accepted: 10/28/2024] [Indexed: 12/09/2024]
Abstract
Morphogens choreograph the generation of remarkable cellular diversity in the developing nervous system. Differentiation of stem cells in vitro often relies upon the combinatorial modulation of these signaling pathways. However, the lack of a systematic approach to understand morphogen-directed differentiation has precluded the generation of many neural cell populations, and the general principles of regional specification and maturation remain incomplete. Here, we developed an arrayed screen of 14 morphogen modulators in human neural organoids cultured for over 70 days. Deconvolution of single-cell-multiplexed RNA sequencing data revealed design principles of brain region specification. We tuned neural subtype diversity to generate a tachykinin 3 (TAC3)-expressing striatal interneuron type within assembloids. To circumvent limitations of in vitro neuronal maturation, we used a neonatal rat transplantation strategy that enabled human Purkinje neurons to develop their hallmark complex dendritic branching. This comprehensive platform yields insights into the factors influencing stem cell-derived neural diversification and maturation.
Collapse
Affiliation(s)
- Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Kevin W Kelley
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Konstantin Kaganovsky
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Massimo Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Jin Hao
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - James P McQueen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Noah Reis
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Genta Narazaki
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Tommy Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Shravanti Kulkarni
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Sergey Pavlov
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA; Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X, Stanford, CA, USA.
| |
Collapse
|