1
|
Junaid M, Lee EJ, Lim SB. Single-cell and spatial omics: exploring hypothalamic heterogeneity. Neural Regen Res 2025; 20:1525-1540. [PMID: 38993130 PMCID: PMC11688568 DOI: 10.4103/nrr.nrr-d-24-00231] [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: 02/26/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Elucidating the complex dynamic cellular organization in the hypothalamus is critical for understanding its role in coordinating fundamental body functions. Over the past decade, single-cell and spatial omics technologies have significantly evolved, overcoming initial technical challenges in capturing and analyzing individual cells. These high-throughput omics technologies now offer a remarkable opportunity to comprehend the complex spatiotemporal patterns of transcriptional diversity and cell-type characteristics across the entire hypothalamus. Current single-cell and single-nucleus RNA sequencing methods comprehensively quantify gene expression by exploring distinct phenotypes across various subregions of the hypothalamus. However, single-cell/single-nucleus RNA sequencing requires isolating the cell/nuclei from the tissue, potentially resulting in the loss of spatial information concerning neuronal networks. Spatial transcriptomics methods, by bypassing the cell dissociation, can elucidate the intricate spatial organization of neural networks through their imaging and sequencing technologies. In this review, we highlight the applicative value of single-cell and spatial transcriptomics in exploring the complex molecular-genetic diversity of hypothalamic cell types, driven by recent high-throughput achievements.
Collapse
Affiliation(s)
- Muhammad Junaid
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Eun Jeong Lee
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| |
Collapse
|
2
|
Pipicelli F, Villalba A, Hippenmeyer S. How radial glia progenitor lineages generate cell-type diversity in the developing cerebral cortex. Curr Opin Neurobiol 2025; 93:103046. [PMID: 40383049 DOI: 10.1016/j.conb.2025.103046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/05/2025] [Accepted: 04/18/2025] [Indexed: 05/20/2025]
Abstract
The cerebral cortex is arguably the most complex organ in humans. The cortical architecture is characterized by a remarkable diversity of neuronal and glial cell types that make up its neuronal circuits. Following a precise temporally ordered program, radial glia progenitor (RGP) cells generate all cortical excitatory projection neurons and glial cell-types. Cortical excitatory projection neurons are produced either directly or via intermediate progenitors, through indirect neurogenesis. How the extensive cortical cell-type diversity is generated during cortex development remains, however, a fundamental open question. How do RGPs quantitatively and qualitatively generate all the neocortical neurons? How does direct and indirect neurogenesis contribute to the establishment of neuronal and lineage heterogeneity? Whether RGPs represent a homogeneous and/or multipotent progenitor population, or if RGPs consist of heterogeneous groups is currently also not known. In this review, we will summarize the latest findings that contributed to a deeper insight into the above key questions.
Collapse
Affiliation(s)
- Fabrizia Pipicelli
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Ana Villalba
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
3
|
Qian X, Coleman K, Jiang S, Kriz AJ, Marciano JH, Luo C, Cai C, Manam MD, Caglayan E, Lai A, Exposito-Alonso D, Otani A, Ghosh U, Shao DD, Andersen RE, Neil JE, Johnson R, LeFevre A, Hecht JL, Micali N, Sestan N, Rakic P, Miller MB, Sun L, Stringer C, Li M, Walsh CA. Spatial transcriptomics reveals human cortical layer and area specification. Nature 2025:10.1038/s41586-025-09010-1. [PMID: 40369074 DOI: 10.1038/s41586-025-09010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 04/04/2025] [Indexed: 05/16/2025]
Abstract
The human cerebral cortex is composed of six layers and dozens of areas that are molecularly and structurally distinct1-4. Although single-cell transcriptomic studies have advanced the molecular characterization of human cortical development, a substantial gap exists owing to the loss of spatial context during cell dissociation5-8. Here we used multiplexed error-robust fluorescence in situ hybridization (MERFISH)9, augmented with deep-learning-based nucleus segmentation, to examine the molecular, cellular and cytoarchitectural development of the human fetal cortex with spatially resolved single-cell resolution. Our extensive spatial atlas, encompassing more than 18 million single cells, spans eight cortical areas across seven developmental time points. We uncovered the early establishment of the six-layer structure, identifiable by the laminar distribution of excitatory neuron subtypes, 3 months before the emergence of cytoarchitectural layers. Notably, we discovered two distinct modes of cortical areal specification during mid-gestation: (1) a continuous, gradual transition observed across most cortical areas along the anterior-posterior axis and (2) a discrete, abrupt boundary specifically identified between the primary (V1) and secondary (V2) visual cortices as early as gestational week 20. This sharp binary transition in V1-V2 neuronal subtypes challenges the notion that mid-gestation cortical arealization involves only gradient-like transitions6,10. Furthermore, integrating single-nucleus RNA sequencing with MERFISH revealed an early upregulation of synaptogenesis in V1-specific layer 4 neurons. Collectively, our findings underscore the crucial role of spatial relationships in determining the molecular specification of cortical layers and areas. This study establishes a spatially resolved single-cell analysis paradigm and paves the way for the construction of a comprehensive developmental atlas of the human brain.
Collapse
Affiliation(s)
- Xuyu Qian
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kyle Coleman
- Statistical Center for Single-Cell and Spatial Genomics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shunzhou Jiang
- Statistical Center for Single-Cell and Spatial Genomics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrea J Kriz
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jack H Marciano
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunyu Luo
- Statistical Center for Single-Cell and Spatial Genomics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chunhui Cai
- Research Computing, Department of Information Technology, Boston Children's Hospital, Boston, MA, USA
| | - Monica Devi Manam
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emre Caglayan
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abbe Lai
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Exposito-Alonso
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aoi Otani
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Urmi Ghosh
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Diane D Shao
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rebecca E Andersen
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jennifer E Neil
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert Johnson
- University of Maryland Brain and Tissue Bank, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexandra LeFevre
- University of Maryland Brain and Tissue Bank, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicola Micali
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Michael B Miller
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Neuropathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Liang Sun
- Research Computing, Department of Information Technology, Boston Children's Hospital, Boston, MA, USA
| | - Carsen Stringer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Mingyao Li
- Statistical Center for Single-Cell and Spatial Genomics, Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Christopher A Walsh
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Wu Y, Wu BZ, Ellenbogen Y, Kant JBY, Yu P, Li X, Caloren L, Sotov V, Tran C, Restrepo M, Kushida M, Ayyadhury S, Kossinna P, Lau R, Habibi P, Mansouri S, Regala J, Durbic T, Aboualizadeh F, Tsao J, Ketela T, Pugh T, Butler MO, Wang BX, Dirks PB, Gao A, Zadeh G, Gaiti F. Neurodevelopmental hijacking of oligodendrocyte lineage programs drives glioblastoma infiltration. Dev Cell 2025:S1534-5807(25)00260-6. [PMID: 40381621 DOI: 10.1016/j.devcel.2025.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 02/06/2025] [Accepted: 04/25/2025] [Indexed: 05/20/2025]
Abstract
Glioblastoma (GBM) is an aggressive brain tumor with a highly invasive nature. Despite the clinical relevance of this behavior, the molecular underpinnings of infiltrating GBM cells in the peritumoral zone remain underexplored in patients. Here, we show that peritumoral progenitor-like GBM cells activate transcriptional programs associated with increased invasivity, synaptic activity, and NOTCH signaling. These cells spatially colocalize with neurons and exhibit an increased propensity for neuronal crosstalk. The epigenetic encoding of these infiltrative cells mirrors that of uncommitted oligodendrocyte progenitor cells (OPCs) in the developing human brain, a neurodevelopmental state marked by increased synaptic and migratory potential. Functional perturbation of a nominated regulatory factor, ZEB1, confirmed its role in maintaining the invasive and uncommitted developmental potential of infiltrative GBM cells. Our findings provide insights into the neurodevelopmental hijacking that drives GBM infiltration in patients, rationalizing further investigation into targeting differentiation potential as a therapeutic strategy.
Collapse
Affiliation(s)
- Yiyan Wu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Benson Z Wu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Yosef Ellenbogen
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada; MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Joan B Y Kant
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Pengcheng Yu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Xuyao Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Loïc Caloren
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Valentin Sotov
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Christine Tran
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michelle Restrepo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Michelle Kushida
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shamini Ayyadhury
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Pathum Kossinna
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ruth Lau
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Parnian Habibi
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Sheila Mansouri
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Johanna Regala
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Tanja Durbic
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | - Julissa Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Troy Ketela
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Trevor Pugh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Marcus O Butler
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Ben X Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Peter B Dirks
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada; Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Developmental and Stem Cell Biology Department, The Hospital for Sick Children, Toronto, ON, Canada
| | - Andrew Gao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gelareh Zadeh
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada; MacFeeters Hamilton Neuro-Oncology Program, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, ON, Canada; Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.
| | - Federico Gaiti
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Ontario Institute for Cancer Research, Toronto, ON, Canada; Vector Institute, Toronto, ON, Canada.
| |
Collapse
|
5
|
Nano PR, Fazzari E, Azizad D, Martija A, Nguyen CV, Wang S, Giang V, Kan RL, Yoo J, Wick B, Haeussler M, Bhaduri A. Integrated analysis of molecular atlases unveils modules driving developmental cell subtype specification in the human cortex. Nat Neurosci 2025; 28:949-963. [PMID: 40259073 PMCID: PMC12081304 DOI: 10.1038/s41593-025-01933-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/27/2025] [Indexed: 04/23/2025]
Abstract
Human brain development requires generating diverse cell types, a process explored by single-cell transcriptomics. Through parallel meta-analyses of the human cortex in development (seven datasets) and adulthood (16 datasets), we generated over 500 gene co-expression networks that can describe mechanisms of cortical development, centering on peak stages of neurogenesis. These meta-modules show dynamic cell subtype specificities throughout cortical development, with several developmental meta-modules displaying spatiotemporal expression patterns that allude to potential roles in cell fate specification. We validated the expression of these modules in primary human cortical tissues. These include meta-module 20, a module elevated in FEZF2+ deep layer neurons that includes TSHZ3, a transcription factor associated with neurodevelopmental disorders. Human cortical chimeroid experiments validated that both FEZF2 and TSHZ3 are required to drive module 20 activity and deep layer neuron specification but through distinct modalities. These studies demonstrate how meta-atlases can engender further mechanistic analyses of cortical fate specification.
Collapse
Grants
- UM1 MH130991 NIMH NIH HHS
- T32 NS048004 NINDS NIH HHS
- R01MH132689 U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- RF1 MH132662 NIMH NIH HHS
- T32 GM008243 NIGMS NIH HHS
- R00 NS111731 NINDS NIH HHS
- R00NS111731 U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 MH132689 NIMH NIH HHS
- T32 GM145388 NIGMS NIH HHS
- U24 HG002371 NHGRI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- We would like to thank the members of the Bhaduri Lab for their insightful advice and comments on the study. We would like to thank the Broad Stem Cell Research Center Flow Cytometry core for their help in isolating cells for this project, Charina Julian for help with running sequencing, and Dr. Laurent Fasano for generously sharing the antibody against TSHZ3. The work performed in the manuscript was generously funded by R00NS111731 from the NIH (NINDS), R01MH132689 from the NIH (NIMH), the Young Investigator Award from the Brain & Behavior Research Foundation, the Alfred P. Sloan Foundation, the Rose Hills Foundation, and the Klingenstein-Simons Fellowship from the Esther A. & Joseph Klingenstein Fund and the Simons Foundation (to A.B.). Additional funding was provided to P.R.N. (UCLA Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Training Program, UCLA Intercampus Medical Genetics Training Program (USHHS Ruth L. Kirschstein Institutional National Research Service Award # T32GM008243)), C.V.N. (T32 NS048004, Predoctoral Fellowship in association with the Training Grant in Neurobehavioral Genetics), and R.K. (T32 GM145388, Cell and Molecular Biology Training Program), and M.H. (NIMH BRAIN NIMH RF1MH132662, NHGRI U24HG002371, CIRM DISC0-14514 (with A.B.)).
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elisa Fazzari
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daria Azizad
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Antoni Martija
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Claudia V Nguyen
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sean Wang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vanna Giang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ryan L Kan
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Juyoun Yoo
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Brittney Wick
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
7
|
Erickson AW, Tan H, Hendrikse LD, Millman J, Thomson Z, Golser J, Khan O, He G, Bach K, Mishra AS, Kopic J, Krsnik Z, Encha-Razavi F, Petrilli G, Guimiot F, Silvestri E, Aldinger KA, Taylor MD, Millen KJ, Haldipur P. Mapping the developmental profile of ventricular zone-derived neurons in the human cerebellum. Proc Natl Acad Sci U S A 2025; 122:e2415425122. [PMID: 40249772 PMCID: PMC12054822 DOI: 10.1073/pnas.2415425122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/11/2025] [Indexed: 04/20/2025] Open
Abstract
The cerebellar ventricular zone (VZ) is the primary source of progenitors that generate cerebellar GABAergic neurons, including Purkinje cells (PCs) and interneurons (INs). This study provides detailed characterization of human cerebellar GABAergic neurogenesis using transcriptomic and histopathological analyses and reveals conserved and unique features compared to rodents. We show that the sequential progression of neurogenesis is conserved and occurs before 8 postconception weeks. Notably, PC differentiation occurs in the outer subventricular zone (SVZ), a region absent in the mouse cerebellum. Human PCs are generated during a compact two-week period before the onset of cerebral cortex histogenesis. A subset of human PCs retain proliferative marker expression weeks after leaving the VZ, another feature not observed in rodents. Human PC maturation is protracted with an extensive migration and reorganization throughout development with dendritic arborization developing in late gestation. We define a continuous transcriptional cascade of PC development from neuroepithelial cells to mature PCs. In contrast, while human interneuronal progenitors are born beginning in early fetal development, they exhibit an even more protracted differentiation across late gestation and into postnatal ages. These findings show dynamic developmental process for human cerebellar GABAergic neurons and underscore the importance of the embryonic environment, with early disruptions having potentially significant impacts.
Collapse
Affiliation(s)
- Anders W. Erickson
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, ONM5G0A4, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ONM5G0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S3K3, Canada
| | - Henry Tan
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Liam D. Hendrikse
- The Arthur and Sonia Labatt Brain Tumor Research Centre, The Hospital for Sick Children, Toronto, ONM5G0A4, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ONM5G0A4, Canada
| | - Jake Millman
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Zachary Thomson
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Joseph Golser
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Omar Khan
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Guanyi He
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Kathleen Bach
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| | - Arpit Suresh Mishra
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA98101
| | - Janja Kopic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb10000, Croatia
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb10000, Croatia
| | - Ferechte Encha-Razavi
- Assistance Publique Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Paris75015, France
| | | | - Fabien Guimiot
- Hôpital Robert-Debré, INSERM UMR 1141, Paris75019, France
| | - Evelina Silvestri
- Surgical Pathology Unit, San Camillo Forlanini Hospital, Rome00152, Italy
| | - Kimberly A. Aldinger
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
- Department of Neurology, University of Washington, Seattle, WA98195
- Department of Pediatrics, University of Washington, Seattle, WA98195
| | - Michael D. Taylor
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ONM5S3K3, Canada
- Texas Children’s Cancer and Hematology Center, Houston, TX77030
- Department of Pediatrics—Hematology/Oncology, Baylor College of Medicine, Houston, TX77030
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX77030
- Department of Neurosurgery, Texas Children’s Hospital, Houston, TX77030
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX77030
- The Arthur and Sonia Labatt Brain Tumour Research Centre and the Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ONM5G0A4, Canada
- Department of Surgery, University of Toronto, Toronto, ONM5S3K3, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ONM5S3K3, Canada
| | - Kathleen J. Millen
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
- Department of Pediatrics, University of Washington, Seattle, WA98195
| | - Parthiv Haldipur
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA98101
| |
Collapse
|
8
|
Scuderi S, Kang TY, Jourdon A, Nelson A, Yang L, Wu F, Anderson GM, Mariani J, Tomasini L, Sarangi V, Abyzov A, Levchenko A, Vaccarino FM. Specification of human brain regions with orthogonal gradients of WNT and SHH in organoids reveals patterning variations across cell lines. Cell Stem Cell 2025:S1934-5909(25)00141-9. [PMID: 40315847 DOI: 10.1016/j.stem.2025.04.006] [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: 05/17/2024] [Revised: 03/10/2025] [Accepted: 04/09/2025] [Indexed: 05/04/2025]
Abstract
The repertoire of neurons and their progenitors depends on their location along the antero-posterior and dorso-ventral axes of the neural tube. To model these axes, we designed the Dual Orthogonal-Morphogen Assisted Patterning System (Duo-MAPS) diffusion device to expose spheres of induced pluripotent stem cells (iPSCs) to concomitant orthogonal gradients of a posteriorizing and a ventralizing morphogen, activating WNT and SHH signaling, respectively. Comparison with single-cell transcriptomes from the fetal human brain revealed that Duo-MAPS-patterned organoids generated an extensive diversity of neuronal lineages from the forebrain, midbrain, and hindbrain. WNT and SHH crosstalk translated into early patterns of gene expression programs associated with the generation of specific brain lineages with distinct functional networks. Human iPSC lines showed substantial interindividual and line-to-line variations in their response to morphogens, highlighting that genetic and epigenetic variations may influence regional specification. Morphogen gradients promise to be a key approach to model the brain in its entirety.
Collapse
Affiliation(s)
- Soraya Scuderi
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Tae-Yun Kang
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Alexandre Jourdon
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Alex Nelson
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Liang Yang
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Feinan Wu
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA
| | | | - Jessica Mariani
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Livia Tomasini
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA
| | - Vivekananda Sarangi
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Alexej Abyzov
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Levchenko
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA.
| | - Flora M Vaccarino
- Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT 06520, USA; Child Study Center, Yale University, New Haven, CT 06520, USA; Department of Neuroscience, Yale University, New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
9
|
Jiang H, Xiao Z, Saleem K, Zhong P, Li L, Chhetri G, Li P, Jiang Z, Yan Z, Feng J. Generation of human induced pluripotent stem cell-derived cortical neurons expressing the six tau isoforms. J Alzheimers Dis 2025:13872877251334831. [PMID: 40267294 DOI: 10.1177/13872877251334831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe alternative splicing (AS) of MAPT, which encodes Tau, in the adult human brain produces six major isoforms that play critical roles in the pathogenesis of tauopathies including Alzheimer's disease. Previous efforts have failed to differentiate human induced pluripotent stem cells (hiPSCs) to cortical neurons expressing the six isoforms of Tau.ObjectiveWe aim to develop a differentiation method capable of producing the six Tau isoforms in hiPSC-derived cortical neurons.MethodsWe searched for the optimal concentration, duration and treatment window of morphogens in the differentiation of hiPSCs through embryoid bodies (EBs) to dorsal forebrain neuroepithelial cells then to cortical neurons.ResultsThe combined inhibition of WNT, SHH, and SMAD signaling in EBs generated neuroepithelial cells expressing appropriate dorsal forebrain markers, while suppressing ventral, midbrain, and hindbrain genes. Further differentiation in neurogenic and neurotrophic factors produced MAP2+ neurons at day 18. The iPSC-derived neurons expressed markers of all cortical layers and exhibited synapse formation and synaptic physiology. In addition, MAP2+ neurons and mitotic cells expressing radial glial markers formed aggregates that could be dissociated to produce mature neurons with similar properties. Most importantly, the six Tau isoforms were expressed from day 80 in a developmentally regulated manner, modeling the situation in human brains on an accelerated timeline.ConclusionsThis chemically defined differentiation method produces a key hallmark of mature human cortical neurons by expressing the six main splicing isoforms of Tau. It will greatly facilitate disease modeling and therapeutic discovery for many human brain disorders involving cortical neurons.
Collapse
Affiliation(s)
- Houbo Jiang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zichun Xiao
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Komal Saleem
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Li Li
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Gaurav Chhetri
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Pei Li
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhongjiao Jiang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jian Feng
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
10
|
Binder N, Khavaran A, Sankowski R. Primer on machine learning applications in brain immunology. FRONTIERS IN BIOINFORMATICS 2025; 5:1554010. [PMID: 40313869 PMCID: PMC12043695 DOI: 10.3389/fbinf.2025.1554010] [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: 01/07/2025] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Single-cell and spatial technologies have transformed our understanding of brain immunology, providing unprecedented insights into immune cell heterogeneity and spatial organisation within the central nervous system. These methods have uncovered complex cellular interactions, rare cell populations, and the dynamic immune landscape in neurological disorders. This review highlights recent advances in single-cell "omics" data analysis and discusses their applicability for brain immunology. Traditional statistical techniques, adapted for single-cell omics, have been crucial in categorizing cell types and identifying gene signatures, overcoming challenges posed by increasingly complex datasets. We explore how machine learning, particularly deep learning methods like autoencoders and graph neural networks, is addressing these challenges by enhancing dimensionality reduction, data integration, and feature extraction. Newly developed foundation models present exciting opportunities for uncovering gene expression programs and predicting genetic perturbations. Focusing on brain development, we demonstrate how single-cell analyses have resolved immune cell heterogeneity, identified temporal maturation trajectories, and uncovered potential therapeutic links to various pathologies, including brain malignancies and neurodegeneration. The integration of single-cell and spatial omics has elucidated the intricate cellular interplay within the developing brain. This mini-review is intended for wet lab biologists at all career stages, offering a concise overview of the evolving landscape of single-cell omics in the age of widely available artificial intelligence.
Collapse
Affiliation(s)
| | | | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Albertson AJ, Winkler EA, Yang AC, Buckwalter MS, Dingman AL, Fan H, Herson PS, McCullough LD, Perez-Pinzon M, Sansing LH, Sun D, Alkayed NJ. Single-Cell Analysis in Cerebrovascular Research: Primed for Breakthroughs and Clinical Impact. Stroke 2025; 56:1082-1091. [PMID: 39772596 PMCID: PMC11932790 DOI: 10.1161/strokeaha.124.049001] [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: 01/11/2025]
Abstract
Data generated using single-cell RNA-sequencing has the potential to transform understanding of the cerebral circulation and advance clinical care. However, the high volume of data, sometimes generated and presented without proper pathophysiological context, can be difficult to interpret and integrate into current understanding of the cerebral circulation and its disorders. Furthermore, heterogeneity in the representation of brain regions and vascular segments makes it difficult to compare results across studies. There are currently no standards for tissue collection and processing that allow easy comparisons across studies and analytical platforms. There are no standards either for single-cell data analysis and presentation. This topical review introduces single-cell RNA-sequencing to physicians and scientists in the cerebrovascular field, with the goals of highlighting opportunities and challenges of applying this technology in the cerebrovascular field and discussing key concepts and knowledge gaps that can be addressed by single-cell RNA-sequencing.
Collapse
Affiliation(s)
- Asher J. Albertson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Ethan A. Winkler
- Department of Neurological Surgery, University of California San Francisco, CA
| | - Andrew C. Yang
- Gladstone Institute of Neurological Disease and Department of Neurology, University of California San Francisco, CA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA
| | - Andra L. Dingman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Huihui Fan
- Department of Neurology, University of Texas Health Science Center, Houston, TX
| | - Paco S. Herson
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, OH
| | | | | | - Lauren H. Sansing
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Dandan Sun
- Department of Neurology and Pittsburgh Institute of Neurological Degeneration Diseases, University of Pittsburgh, Pittsburgh, PA
| | - Nabil J. Alkayed
- Department of Anesthesiology & Perioperative Medicine and Knight Cardiovascular Institute Portland, OR
| |
Collapse
|
14
|
Simard S, Matosin N, Mechawar N. Adult Hippocampal Neurogenesis in the Human Brain: Updates, Challenges, and Perspectives. Neuroscientist 2025; 31:141-158. [PMID: 38757781 DOI: 10.1177/10738584241252581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
The existence of neurogenesis in the adult human hippocampus has been under considerable debate within the past three decades due to the diverging conclusions originating mostly from immunohistochemistry studies. While some of these reports conclude that hippocampal neurogenesis in humans occurs throughout physiologic aging, others indicate that this phenomenon ends by early childhood. More recently, some groups have adopted next-generation sequencing technologies to characterize with more acuity the extent of this phenomenon in humans. Here, we review the current state of research on adult hippocampal neurogenesis in the human brain with an emphasis on the challenges and limitations of using immunohistochemistry and next-generation sequencing technologies for its study.
Collapse
Affiliation(s)
- Sophie Simard
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Natalie Matosin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, Canada
- Department of Psychiatry, McGill University, Montréal, Canada
| |
Collapse
|
15
|
Han L, Liu Z, Jing Z, Liu Y, Peng Y, Chang H, Lei J, Wang K, Xu Y, Liu W, Wu Z, Li Q, Shi X, Zheng M, Wang H, Deng J, Zhong Y, Pan H, Lin J, Zhang R, Chen Y, Wu J, Xu M, Ren B, Cheng M, Yu Q, Song X, Lu Y, Tang Y, Yuan N, Sun S, An Y, Ding W, Sun X, Wei Y, Zhang S, Dou Y, Zhao Y, Han L, Zhu Q, Xu J, Wang S, Wang D, Bai Y, Liang Y, Liu Y, Chen M, Xie C, Bo B, Li M, Zhang X, Ting W, Chen Z, Fang J, Li S, Jiang Y, Tan X, Zuo G, Xie Y, Li H, Tao Q, Li Y, Liu J, Liu Y, Hao M, Wang J, Wen H, Liu J, Yan Y, Zhang H, Sheng Y, Yu S, Liao X, Jiang X, Wang G, Liu H, Wang C, Feng N, Liu X, Ma K, Xu X, Han T, Cao H, Zheng H, Chen Y, Lu H, Yu Z, Zhang J, Wang B, Wang Z, Xie Q, Pan S, Liu C, Xu C, Cui L, Li Y, Liu S, Liao S, Chen A, Wu QF, et alHan L, Liu Z, Jing Z, Liu Y, Peng Y, Chang H, Lei J, Wang K, Xu Y, Liu W, Wu Z, Li Q, Shi X, Zheng M, Wang H, Deng J, Zhong Y, Pan H, Lin J, Zhang R, Chen Y, Wu J, Xu M, Ren B, Cheng M, Yu Q, Song X, Lu Y, Tang Y, Yuan N, Sun S, An Y, Ding W, Sun X, Wei Y, Zhang S, Dou Y, Zhao Y, Han L, Zhu Q, Xu J, Wang S, Wang D, Bai Y, Liang Y, Liu Y, Chen M, Xie C, Bo B, Li M, Zhang X, Ting W, Chen Z, Fang J, Li S, Jiang Y, Tan X, Zuo G, Xie Y, Li H, Tao Q, Li Y, Liu J, Liu Y, Hao M, Wang J, Wen H, Liu J, Yan Y, Zhang H, Sheng Y, Yu S, Liao X, Jiang X, Wang G, Liu H, Wang C, Feng N, Liu X, Ma K, Xu X, Han T, Cao H, Zheng H, Chen Y, Lu H, Yu Z, Zhang J, Wang B, Wang Z, Xie Q, Pan S, Liu C, Xu C, Cui L, Li Y, Liu S, Liao S, Chen A, Wu QF, Wang J, Liu Z, Sun Y, Mulder J, Yang H, Wang X, Li C, Yao J, Xu X, Liu L, Shen Z, Wei W, Sun YG. Single-cell spatial transcriptomic atlas of the whole mouse brain. Neuron 2025:S0896-6273(25)00133-3. [PMID: 40132589 DOI: 10.1016/j.neuron.2025.02.015] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 10/24/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
A comprehensive atlas of genes, cell types, and their spatial distribution across a whole mammalian brain is fundamental for understanding the function of the brain. Here, using single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq techniques, we generated a mouse brain atlas with spatial information for 308 cell clusters at single-cell resolution, involving over 4 million cells, as well as for 29,655 genes. We have identified cell clusters exhibiting preference for cortical subregions and explored their associations with brain-related diseases. Additionally, we pinpointed 155 genes with distinct regional expression patterns within the brainstem and unveiled 513 long non-coding RNAs showing region-enriched expression in the adult brain. Parcellation of brain regions based on spatial transcriptomic information revealed fine structure for several brain areas. Furthermore, we have uncovered 411 transcription factor regulons showing distinct spatiotemporal dynamics during neurodevelopment. Thus, we have constructed a single-cell-resolution spatial transcriptomic atlas of the mouse brain with genome-wide coverage.
Collapse
Affiliation(s)
- Lei Han
- BGI Research, Hangzhou 310030, China
| | - Zhen Liu
- Lingang Laboratory, Shanghai 200031, China; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxuan Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | - Junjie Lei
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kexin Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanfang Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Liu
- Lingang Laboratory, Shanghai 200031, China
| | - Zihan Wu
- Tencent AI Lab, Shenzhen 518057, China
| | - Qian Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI Research, Shenzhen 518083, China
| | - Xiaoxue Shi
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingyuan Zheng
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - He Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yanqing Zhong
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Junkai Lin
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ruiyi Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinhua Wu
- Lingang Laboratory, Shanghai 200031, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Biyu Ren
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Qian Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinxiang Song
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanbing Lu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanchun Tang
- BGI Research, Hangzhou 310030, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Nini Yuan
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Suhong Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingjie An
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenqun Ding
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xing Sun
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanrong Wei
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuzhen Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yannong Dou
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun Zhao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luyao Han
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Junfeng Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiwen Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yinqi Bai
- BGI Research, Hangzhou 310030, China
| | - Yikai Liang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengni Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chun Xie
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Binshi Bo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mei Li
- BGI Research, Shenzhen 518083, China
| | - Xinyan Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wang Ting
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiao Fang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xing Tan
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guolong Zuo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yue Xie
- BGI Research, Shenzhen 518083, China
| | - Huanhuan Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Quyuan Tao
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianfeng Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuyang Liu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingkun Hao
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingjing Wang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huiying Wen
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiabing Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Hui Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yifan Sheng
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shui Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xuyin Jiang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangling Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Congcong Wang
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning Feng
- BGI Research, Shenzhen 518083, China
| | - Xin Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xiangjie Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Huateng Cao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Haorong Lu
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Zixian Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Bo Wang
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | | | - Qing Xie
- BGI Research, Shenzhen 518083, China
| | | | - Chuanyu Liu
- BGI Research, Shenzhen 518083, China; Shenzhen Proof-of-Concept Center of Digital Cytopathology, BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Chan Xu
- BGI Research, Qingdao 266555, China
| | - Luman Cui
- BGI Research, Shenzhen 518083, China
| | - Yuxiang Li
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Ao Chen
- BGI Research, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China; Department of Biology, University of Copenhagen, Copenhagen 2200, Denmark
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Wang
- BGI Research, Shenzhen 518083, China; China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China
| | - Yidi Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jan Mulder
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
| | | | - Xiaofei Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China.
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| | - Zhiming Shen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China.
| | - Wu Wei
- Lingang Laboratory, Shanghai 200031, China; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
16
|
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
|
17
|
Mil J, Soto JA, Matulionis N, Krall A, Day F, Stiles L, Montales KP, Azizad DJ, Gonzalez CE, Nano PR, Martija AA, Perez-Ramirez CA, Nguyen CV, Kan RL, Andrews MG, Christofk HR, Bhaduri A. Metabolic Atlas of Early Human Cortex Identifies Regulators of Cell Fate Transitions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642470. [PMID: 40161647 PMCID: PMC11952424 DOI: 10.1101/2025.03.10.642470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Characterization of cell type emergence during human cortical development, which enables unique human cognition, has focused primarily on anatomical and transcriptional characterizations. Metabolic processes in the human brain that allow for rapid expansion, but contribute to vulnerability to neurodevelopmental disorders, remain largely unexplored. We performed a variety of metabolic assays in primary tissue and stem cell derived cortical organoids and observed dynamic changes in core metabolic functions, including an unexpected increase in glycolysis during late neurogenesis. By depleting glucose levels in cortical organoids, we increased outer radial glia, astrocytes, and inhibitory neurons. We found the pentose phosphate pathway (PPP) was impacted in these experiments and leveraged pharmacological and genetic manipulations to recapitulate these radial glia cell fate changes. These data identify a new role for the PPP in modulating radial glia cell fate specification and generate a resource for future exploration of additional metabolic pathways in human cortical development.
Collapse
Affiliation(s)
- Jessenya Mil
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jose A. Soto
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nedas Matulionis
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Abigail Krall
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Francesca Day
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California, USA
| | - Katrina P. Montales
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Daria J. Azizad
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Carlos E. Gonzalez
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Patricia R. Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Antoni A. Martija
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cesar A. Perez-Ramirez
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Claudia V. Nguyen
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ryan L. Kan
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Madeline G. Andrews
- School of Biological and Health Systems Engineering, Arizona State University, Phoenix, AZ, United States
| | - Heather R. Christofk
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
18
|
Holm Nygaard A, Schörling AL, Abay-Nørgaard Z, Hänninen E, Li Y, Ramón Santonja A, Rathore GS, Salvador A, Rusimbi C, Lauritzen KB, Zhang Y, Kirkeby A. Patterning effects of FGF17 and cAMP on generation of dopaminergic progenitors for cell replacement therapy in Parkinson's disease. Stem Cells 2025; 43:sxaf004. [PMID: 40071608 PMCID: PMC11976395 DOI: 10.1093/stmcls/sxaf004] [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: 07/08/2024] [Accepted: 01/06/2025] [Indexed: 04/09/2025]
Abstract
Cell replacement therapies using human pluripotent stem cell-derived ventral midbrain (VM) dopaminergic (DA) progenitors are currently in clinical trials for treatment of Parkinson's disease (PD). Recapitulating developmental patterning cues, such as fibroblast growth factor 8 (FGF8), secreted at the midbrain-hindbrain boundary (MHB), is critical for the in vitro production of authentic VM DA progenitors. Here, we explored the application of alternative MHB-secreted FGF-family members, FGF17 and FGF18, for VM DA progenitor patterning. We show that while FGF17 and FGF18 both recapitulated VM DA progenitor patterning events, FGF17 induced expression of key VM DA progenitor markers at higher levels than FGF8 and transplanted FGF17-patterned progenitors fully reversed motor deficits in a rat PD model. Early activation of the cAMP pathway mimicked FGF17-induced patterning, although strong cAMP activation came at the expense of EN1 expression. In summary, we identified FGF17 as a promising alternative FGF candidate for robust VM DA progenitor patterning.
Collapse
Affiliation(s)
- Amalie Holm Nygaard
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alrik L Schörling
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Zehra Abay-Nørgaard
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Erno Hänninen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yuan Li
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Adrian Ramón Santonja
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Gaurav Singh Rathore
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Alison Salvador
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Charlotte Rusimbi
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Katrine Bech Lauritzen
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Yu Zhang
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, SE-221 84 Lund, Sweden
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Clarence T, Bendl J, Cao X, Wang X, Zheng S, Hoffman GE, Kozlenkov A, Hong A, Iskhakova M, Jaiswal MK, Murphy S, Yu A, Haroutunian V, Dracheva S, Akbarian S, Fullard JF, Yuan GC, Lee D, Roussos P. Multiomic single-cell profiling identifies critical regulators of postnatal brain. Nat Genet 2025; 57:591-603. [PMID: 39962241 DOI: 10.1038/s41588-025-02083-8] [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: 02/25/2024] [Accepted: 01/08/2025] [Indexed: 03/15/2025]
Abstract
Human brain development spans from embryogenesis to adulthood, with dynamic gene expression controlled by cell-type-specific cis-regulatory element activity and three-dimensional genome organization. To advance our understanding of postnatal brain development, we simultaneously profiled gene expression and chromatin accessibility in 101,924 single nuclei from four brain regions across ten donors, covering five key postnatal stages from infancy to late adulthood. Using this dataset and chromosome conformation capture data, we constructed enhancer-based gene regulatory networks to identify cell-type-specific regulators of brain development and interpret genome-wide association study loci for ten main brain disorders. Our analysis connected 2,318 cell-specific loci to 1,149 unique genes, representing 41% of loci linked to the investigated traits, and highlighted 55 genes influencing several disease phenotypes. Pseudotime analysis revealed distinct stages of postnatal oligodendrogenesis and their regulatory programs. These findings provide a comprehensive dataset of cell-type-specific gene regulation at critical timepoints in postnatal brain development.
Collapse
Affiliation(s)
- Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuan Cao
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xinyi Wang
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexey Kozlenkov
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aram Hong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Iskhakova
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manoj K Jaiswal
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Sarah Murphy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Yu
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Stella Dracheva
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
20
|
Yang Z. The Principle of Cortical Development and Evolution. Neurosci Bull 2025; 41:461-485. [PMID: 39023844 PMCID: PMC11876516 DOI: 10.1007/s12264-024-01259-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Human's robust cognitive abilities, including creativity and language, are made possible, at least in large part, by evolutionary changes made to the cerebral cortex. This paper reviews the biology and evolution of mammalian cortical radial glial cells (primary neural stem cells) and introduces the concept that a genetically step wise process, based on a core molecular pathway already in use, is the evolutionary process that has molded cortical neurogenesis. The core mechanism, which has been identified in our recent studies, is the extracellular signal-regulated kinase (ERK)-bone morphogenic protein 7 (BMP7)-GLI3 repressor form (GLI3R)-sonic hedgehog (SHH) positive feedback loop. Additionally, I propose that the molecular basis for cortical evolutionary dwarfism, exemplified by the lissencephalic mouse which originated from a larger gyrencephalic ancestor, is an increase in SHH signaling in radial glia, that antagonizes ERK-BMP7 signaling. Finally, I propose that: (1) SHH signaling is not a key regulator of primate cortical expansion and folding; (2) human cortical radial glial cells do not generate neocortical interneurons; (3) human-specific genes may not be essential for most cortical expansion. I hope this review assists colleagues in the field, guiding research to address gaps in our understanding of cortical development and evolution.
Collapse
Affiliation(s)
- Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
21
|
Jiu J, Liu H, Li D, Li X, Zhang J, Yan L, Fan Z, Li S, Du G, Li JJ, Wu A, Liu W, Du Y, Zhao B, Wang B. 3D Mechanical Response Stem Cell Complex Repairs Spinal Cord Injury by Promoting Neurogenesis and Regulating Tissue Homeostasis. Adv Healthc Mater 2025; 14:e2404925. [PMID: 39853962 DOI: 10.1002/adhm.202404925] [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: 12/08/2024] [Indexed: 01/26/2025]
Abstract
Spinal cord injury (SCI) leads to acute tissue damage that disrupts the microenvironmental homeostasis of the spinal cord, inhibiting cell survival and function, and thereby undermining treatment efficacy. Traditional stem cell therapies have limited success in SCI, due to the difficulties in maintaining cell survival and inducing sustained differentiation into neural lineages. A new solution may arise from controlling the fate of stem cells by creating an appropriate mechanical microenvironment. In this study, mechanical response stem cell complex (MRSCC) is created as an innovative therapeutic strategy for SCI, utilizing 3D bioprinting technology and gelatin microcarriers (GM) loaded with mesenchymal stem cells (MSCs). GM creates an optimal microenvironment for MSCs growth and paracrine activity. Meanwhile, 3D bioprinting allows accurate control of spatial pore architecture and mechanical characteristics of the cell construct to encourage neuroregeneration. The mechanical microenvironment created by MRSCC is found to activate the Piezo1 channel and prevent excessive nuclear translocation of YAP, thereby increasing neural-related gene expression in MSCs. Transplanting MRSCC in rats with spinal cord injuries boosts sensory and motor recovery, reduces inflammation, and stimulates the regeneration of neurons and glial cells. The MRSCC offers a new tissue engineering solution that can promote spinal cord repair.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dijun Li
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xiaoke Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, China
| | - Lei Yan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zijuan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Guangyuan Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei Liu
- Development of Research, Beijing Hua Niche Biotechnology Co., LTD, Beijing, 100084, China
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bin Zhao
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| |
Collapse
|
22
|
Venkatesan S, Werner JM, Li Y, Gillis J. Cell Type-Agnostic Transcriptomic Signatures Enable Uniform Comparisons of Neurodevelopment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639936. [PMID: 40060479 PMCID: PMC11888278 DOI: 10.1101/2025.02.24.639936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Single-cell transcriptomics has revolutionized our understanding of neurodevelopmental cell identities, yet, predicting a cell type's developmental state from its transcriptome remains a challenge. We perform a meta-analysis of developing human brain datasets comprising over 2.8 million cells, identifying both tissue-level and cell-autonomous predictors of developmental age. While tissue composition predicts age within individual studies, it fails to generalize, whereas specific cell type proportions reliably track developmental time across datasets. Training regularized regression models to infer cell-autonomous maturation, we find that a cell type-agnostic model achieves the highest accuracy (error = 2.6 weeks), robustly capturing developmental dynamics across diverse cell types and datasets. This model generalizes to human neural organoids, accurately predicting normal developmental trajectories (R = 0.91) and disease-induced shifts in vitro. Furthermore, it extends to the developing mouse brain, revealing an accelerated developmental tempo relative to humans. Our work provides a unified framework for comparing neurodevelopment across contexts, model systems, and species.
Collapse
Affiliation(s)
- Sridevi Venkatesan
- Department of Physiology, University of Toronto, Canada
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Jonathan M Werner
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
| | - Yun Li
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Canada
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Canada
| |
Collapse
|
23
|
Dony L, Krontira AC, Kaspar L, Ahmad R, Demirel IS, Grochowicz M, Schäfer T, Begum F, Sportelli V, Raimundo C, Koedel M, Labeur M, Cappello S, Theis FJ, Cruceanu C, Binder EB. Chronic exposure to glucocorticoids amplifies inhibitory neuron cell fate during human neurodevelopment in organoids. SCIENCE ADVANCES 2025; 11:eadn8631. [PMID: 39951527 PMCID: PMC11827642 DOI: 10.1126/sciadv.adn8631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025]
Abstract
Disruptions in the tightly regulated process of human brain development have been linked to increased risk for brain and mental illnesses. While the genetic contribution to these diseases is well established, important environmental factors have been less studied at molecular and cellular levels. Here, we used single-cell and cell type-specific techniques to investigate the effect of glucocorticoid (GC) exposure, a mediator of antenatal environmental risk, on gene regulation and lineage specification in unguided human neural organoids. We characterized the transcriptional response to chronic GC exposure during neural differentiation and studied the underlying gene regulatory networks by integrating single-cell transcriptomics with chromatin accessibility data. We found lasting cell type-specific changes that included autism risk genes and several transcription factors associated with neurodevelopment. Chronic GC exposure influenced lineage specification primarily by priming the inhibitory neuron lineage through transcription factors like PBX3. We provide evidence for convergence of genetic and environmental risk factors through a common mechanism of altering lineage specification.
Collapse
Affiliation(s)
- Leander Dony
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
| | - Anthi C. Krontira
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Lea Kaspar
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), 80804 Munich, Germany
| | - Ruhel Ahmad
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Ilknur Safak Demirel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Tim Schäfer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Fatema Begum
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Vincenza Sportelli
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
| | - Catarina Raimundo
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Maik Koedel
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Marta Labeur
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Silvia Cappello
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- Physiological Genomics, Biomedical Center (BMC), LMU Munich Faculty of Medicine, 82152 Planegg-Martinsried, Germany
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, 85764 Neuherberg, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, 85748 Garching bei München, Germany
| | - Cristiana Cruceanu
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Elisabeth B. Binder
- Department Genes and Environment, Max Planck Institute of Psychiatry, 80804 Munich, Germany
- German Center for Mental Health (DZPG), partner site Munich, Munich, Germany
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
24
|
Perochon T, Krsnik Z, Massimo M, Ruchiy Y, Romero AL, Mohammadi E, Li X, Long KR, Parkkinen L, Blomgren K, Lagache T, Menassa DA, Holcman D. Unraveling microglial spatial organization in the developing human brain with DeepCellMap, a deep learning approach coupled with spatial statistics. Nat Commun 2025; 16:1577. [PMID: 39948387 PMCID: PMC11825940 DOI: 10.1038/s41467-025-56560-z] [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: 02/26/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Mapping cellular organization in the developing brain presents significant challenges due to the multidimensional nature of the data, characterized by complex spatial patterns that are difficult to interpret without high-throughput tools. Here, we present DeepCellMap, a deep-learning-assisted tool that integrates multi-scale image processing with advanced spatial and clustering statistics. This pipeline is designed to map microglial organization during normal and pathological brain development and has the potential to be adapted to any cell type. Using DeepCellMap, we capture the morphological diversity of microglia, identify strong coupling between proliferative and phagocytic phenotypes, and show that distinct spatial clusters rarely overlap as human brain development progresses. Additionally, we uncover an association between microglia and blood vessels in fetal brains exposed to maternal SARS-CoV-2. These findings offer insights into whether various microglial phenotypes form networks in the developing brain to occupy space, and in conditions involving haemorrhages, whether microglia respond to, or influence changes in blood vessel integrity. DeepCellMap is available as an open-source software and is a powerful tool for extracting spatial statistics and analyzing cellular organization in large tissue sections, accommodating various imaging modalities. This platform opens new avenues for studying brain development and related pathologies.
Collapse
Affiliation(s)
- Theo Perochon
- Group of Data Modeling and Computational Biology, IBENS, École Normale Supérieure, Paris, France
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, University of Zagreb, Zagreb, Croatia
| | - Marco Massimo
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Yana Ruchiy
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | | | - Elyas Mohammadi
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Xiaofei Li
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Katherine R Long
- Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Laura Parkkinen
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, United Kingdom
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Thibault Lagache
- BioImage Analysis Unit, CNRS UMR3691, Institut Pasteur, Université Paris Cité, Paris, France.
| | - David A Menassa
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
- Department of Neuropathology and The Queen's College, University of Oxford, Oxford, United Kingdom.
| | - David Holcman
- Group of Data Modeling and Computational Biology, IBENS, École Normale Supérieure, Paris, France.
- DAMPT, University of Cambridge, DAMPT and Churchill College, Cambridge, United Kingdom.
| |
Collapse
|
25
|
Chen J, Bai Y, He X, Xiao W, Chen L, Wong YK, Wang C, Gao P, Cheng G, Xu L, Yang C, Liao F, Han G, Sun J, Xu C, Wang J. The spatiotemporal transcriptional profiling of murine brain during cerebral malaria progression and after artemisinin treatment. Nat Commun 2025; 16:1540. [PMID: 39934099 PMCID: PMC11814382 DOI: 10.1038/s41467-024-52223-7] [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: 09/04/2023] [Accepted: 08/28/2024] [Indexed: 02/13/2025] Open
Abstract
Cerebral malaria (CM) is a severe encephalopathy caused by Plasmodium parasite infection, resulting in thousands of annual deaths and neuro-cognitive sequelae even after anti-malarial drugs treatment. Despite efforts to dissect the mechanism, the cellular transcriptomic reprogramming within the spatial context remains elusive. Here, we constructed single-cell and spatial transcriptome atlases of experimental CM (ECM) male murine brain tissues with or without artesunate (ART) treatment. We identified activated inflammatory endothelial cells during ECM, characterized by a disrupted blood-brain barrier, increased antigen presentation, and leukocyte adhesion. We also observed that inflammatory microglia enhance antigen presentation pathway such as MHC-I to CD8+ cytotoxic T cells. The latter underwent an inflammatory state transition with up-regulated cytokine expression and cytotoxic activity. Multi-omics analysis revealed that the activated interferon-gamma response of injured neurons during ECM and persisted after ART treatment. Overall, our research provides valuable resources for understanding malaria parasite-host interaction mechanisms and adjuvant therapy development.
Collapse
Affiliation(s)
- Jiayun Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Yunmeng Bai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Xueling He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Xiao
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Lina Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yin Kwan Wong
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Chen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Peng Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Liting Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chuanbin Yang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Fulong Liao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Guang Han
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Jichao Sun
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Chengchao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China.
| |
Collapse
|
26
|
Lemoine C, Da Veiga MA, Rogister B, Piette C, Neirinckx V. An integrated perspective on single-cell and spatial transcriptomic signatures in high-grade gliomas. NPJ Precis Oncol 2025; 9:44. [PMID: 39934275 DOI: 10.1038/s41698-025-00830-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/01/2025] [Indexed: 02/13/2025] Open
Abstract
High-grade gliomas (HGG) are incurable brain malignancies in children and adults. Breakthrough advances in transcriptomic technologies unveiled the intricate diversity of cellular states and their spatial organization within HGGs. We qualitatively integrated 55 neoplastic transcriptomic signatures described in 17 single-cell and spatial RNA sequencing-based studies. Our review delineates a spectrum of cellular states, represented by the expression of specific genes, which can be conceptualized along a "reactive-developmental programs" axis. Additionally, we discussed the potential cues influencing these cellular states, including how spatial organization may impact transcriptomic dynamics. Leveraging these insightful discoveries, we discussed a novel, evolutive way to integrate the different transcriptomic signatures in two or three dimensions, incorporating developmental states, their proliferative capacity, and their possible transition towards reactive states. This integrated analysis illuminates the diverse cellular landscape of HGGs and provides a valuable resource for further elucidation of malignant mechanisms, and for the design of therapeutic endeavors.
Collapse
Affiliation(s)
- Célia Lemoine
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
| | - Marc-Antoine Da Veiga
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
- Department of Neurology, CHU of Liège, 4000, Liège, Belgium
| | - Caroline Piette
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium
- Department of Pediatrics, Division of Hematology-Oncology, CHU Liège, 4000, Liège, Belgium
| | - Virginie Neirinckx
- Laboratory of Nervous System Disorders and Therapy, GIGA Institute, University of Liège, 4000, Liège, Belgium.
| |
Collapse
|
27
|
Blair JD, Hartman A, Zenk F, Wahle P, Brancati G, Dalgarno C, Treutlein B, Satija R. Phospho-seq: integrated, multi-modal profiling of intracellular protein dynamics in single cells. Nat Commun 2025; 16:1346. [PMID: 39905064 PMCID: PMC11794950 DOI: 10.1038/s41467-025-56590-7] [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: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
Cell signaling plays a critical role in neurodevelopment, regulating cellular behavior and fate. While multimodal single-cell sequencing technologies are rapidly advancing, scalable and flexible profiling of cell signaling states alongside other molecular modalities remains challenging. Here we present Phospho-seq, an integrated approach that aims to quantify cytoplasmic and nuclear proteins, including those with post-translational modifications, and to connect their activity with cis-regulatory elements and transcriptional targets. We utilize a simplified benchtop antibody conjugation method to create large custom neuro-focused antibody panels for simultaneous protein and scATAC-seq profiling on whole cells, alongside both experimental and computational strategies to incorporate transcriptomic measurements. We apply our workflow to cell lines, induced pluripotent stem cells, and months-old retinal and brain organoids to demonstrate its broad applicability. We show that Phospho-seq can provide insights into cellular states and trajectories, shed light on gene regulatory relationships, and help explore the causes and effects of diverse cell signaling in neurodevelopment.
Collapse
Affiliation(s)
- John D Blair
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | | | | | | | | | | | | | - Rahul Satija
- New York Genome Center, New York, NY, USA.
- Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
28
|
Caporale N, Castaldi D, Rigoli MT, Cheroni C, Valenti A, Stucchi S, Lessi M, Bulgheresi D, Trattaro S, Pezzali M, Vitriolo A, Lopez-Tobon A, Bonfanti M, Ricca D, Schmid KT, Heinig M, Theis FJ, Villa CE, Testa G. Multiplexing cortical brain organoids for the longitudinal dissection of developmental traits at single-cell resolution. Nat Methods 2025; 22:358-370. [PMID: 39653820 PMCID: PMC11810796 DOI: 10.1038/s41592-024-02555-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/31/2024] [Indexed: 12/20/2024]
Abstract
Dissecting human neurobiology at high resolution and with mechanistic precision requires a major leap in scalability, given the need for experimental designs that include multiple individuals and, prospectively, population cohorts. To lay the foundation for this, we have developed and benchmarked complementary strategies to multiplex brain organoids by pooling cells from different pluripotent stem cell (PSC) lines either during organoid generation (mosaic models) or before single-cell RNA sequencing (scRNA-seq) library preparation (downstream multiplexing). We have also developed a new computational method, SCanSNP, and a consensus call to deconvolve cell identities, overcoming current criticalities in doublets and low-quality cell identification. We validated both multiplexing methods for charting neurodevelopmental trajectories at high resolution, thus linking specific individuals' trajectories to genetic variation. Finally, we modeled their scalability across different multiplexing combinations and showed that mosaic organoids represent an enabling method for high-throughput settings. Together, this multiplexing suite of experimental and computational methods provides a highly scalable resource for brain disease and neurodiversity modeling.
Collapse
Affiliation(s)
- Nicolò Caporale
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Davide Castaldi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Marco Tullio Rigoli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | | | - Alessia Valenti
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Sarah Stucchi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Manuel Lessi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | | | | | - Martina Pezzali
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | | | | | | | | | - Katharina T Schmid
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technical University Munich, Munich, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technical University Munich, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technical University Munich, Munich, Germany
| | | | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
- Human Technopole, Milan, Italy.
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
29
|
Hoffman GE, Roussos P. Fast, flexible analysis of differences in cellular composition with crumblr. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635498. [PMID: 39975411 PMCID: PMC11838391 DOI: 10.1101/2025.01.29.635498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Changes in cell type composition play an important role in human health and disease. Recent advances in single-cell technology have enabled the measurement of cell type composition at increasing cell lineage resolution across large cohorts of individuals. Yet this raises new challenges for statistical analysis of these compositional data to identify changes in cell type frequency. We introduce crumblr (DiseaseNeurogenomics.github.io/crumblr), a scalable statistical method for analyzing count ratio data using precision-weighted linear mixed models incorporating random effects for complex study designs. Uniquely, crumblr performs statistical testing at multiple levels of the cell lineage hierarchy using a multivariate approach to increase power over tests of one cell type. In simulations, crumblr increases power compared to existing methods while controlling the false positive rate. We demonstrate the application of crumblr to published single-cell RNA-seq datasets for aging, tuberculosis infection in T cells, bone metastases from prostate cancer, and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Gabriel E. Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, New York
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, New York
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| |
Collapse
|
30
|
Vasan L, Chinchalongporn V, Saleh F, Zinyk D, Ke C, Suresh H, Ghazale H, Belfiore L, Touahri Y, Oproescu AM, Patel S, Rozak M, Amemiya Y, Han S, Moffat A, Black SE, McLaurin J, Near J, Seth A, Goubran M, Reiner O, Gillis J, Wang C, Okawa S, Schuurmans C. Examining the NEUROG2 lineage and associated gene expression in human cortical organoids. Development 2025; 152:dev202703. [PMID: 39680368 PMCID: PMC11829764 DOI: 10.1242/dev.202703] [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: 01/11/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Proneural genes are conserved drivers of neurogenesis across the animal kingdom. How their functions have adapted to guide human-specific neurodevelopmental features is poorly understood. Here, we mined transcriptomic data from human fetal cortices and generated from human embryonic stem cell-derived cortical organoids (COs) to show that NEUROG1 and NEUROG2 are most highly expressed in basal neural progenitor cells, with pseudotime trajectory analyses indicating that NEUROG1-derived lineages predominate early and NEUROG2 lineages later. Using ChIP-qPCR, gene silencing and overexpression studies in COs, we show that NEUROG2 is necessary and sufficient to directly transactivate known target genes (NEUROD1, EOMES, RND2). To identify new targets, we engineered NEUROG2-mCherry knock-in human embryonic stem cells for CO generation. The mCherry-high CO cell transcriptome is enriched in extracellular matrix-associated genes, and two genes associated with human-accelerated regions: PPP1R17 and FZD8. We show that NEUROG2 binds COL1A1, COL3A1 and PPP1R17 regulatory elements, and induces their ectopic expression in COs, although NEUROG2 is not required for this expression. Neurog2 similarly induces Col3a1 and Ppp1r17 in murine P19 cells. These data are consistent with a conservation of NEUROG2 function across mammalian species.
Collapse
Affiliation(s)
- Lakshmy Vasan
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Vorapin Chinchalongporn
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Fermisk Saleh
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dawn Zinyk
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Cao Ke
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Immunology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hamsini Suresh
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Hussein Ghazale
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lauren Belfiore
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yacine Touahri
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ana-Maria Oproescu
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shruti Patel
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Matthew Rozak
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Yutaka Amemiya
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sisu Han
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alexandra Moffat
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sandra E. Black
- Dr. Sandra Black Centre for Brain Resilience & Recovery, LC Campbell Cognitive Neurology Unit, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Hurvitz Brain Sciences Program
- Department of Medicine (Neurology) (SEB), University of Toronto, Toronto, ON M5S 3H2, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jamie Near
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Arun Seth
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Maged Goubran
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Chao Wang
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Immunology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Satoshi Okawa
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
31
|
Feng X, Gao Y, Chu F, Shan Y, Liu M, Wang Y, Zhu Y, Lu Q, Li M. Cortical arealization of interneurons defines shared and distinct molecular programs in developing human and macaque brains. Nat Commun 2025; 16:672. [PMID: 39809789 PMCID: PMC11733295 DOI: 10.1038/s41467-025-56058-8] [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: 02/08/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Cortical interneurons generated from ganglionic eminence via a long-distance journey of tangential migration display evident cellular and molecular differences across brain regions, which seeds the heterogeneous cortical circuitry in primates. However, whether such regional specifications in interneurons are intrinsically encoded or gained through interactions with the local milieu remains elusive. Here, we recruit 685,692 interneurons from cerebral cortex and subcortex including ganglionic eminence within the developing human and macaque species. Our integrative and comparative analyses reveal that less transcriptomic alteration is accompanied by interneuron migration within the ganglionic eminence subdivisions, in contrast to the dramatic changes observed in cortical tangential migration, which mostly characterize the transcriptomic specification for different destinations and for species divergence. Moreover, the in-depth survey of temporal regulation illustrates species differences in the developmental dynamics of cell types, e.g., the employment of CRH in primate interneurons during late-fetal stage distinguishes from their postnatal emergence in mice, and our entropy quantifications manifest the interneuron diversities gradually increase along the developmental ages in human and macaque cerebral cortices. Overall, our analyses depict the spatiotemporal features appended to cortical interneurons, providing a new proxy for understanding the relationship between cellular diversity and functional progression.
Collapse
Affiliation(s)
- Xiangling Feng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingjie Gao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Chu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwen Shan
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meicheng Liu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaoyi Wang
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ying Zhu
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingfeng Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.
- Innovation center for Brain Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Manning E, Chinnaiya K, Furley C, Kim DW, Blackshaw S, Placzek M, Place E. Resolving forebrain developmental organisation by analysis of differential growth patterns. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632351. [PMID: 39829908 PMCID: PMC11741420 DOI: 10.1101/2025.01.10.632351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The forebrain is the most complex region of the vertebrate CNS, and its developmental organisation is controversial. We fate-mapped the embryonic chick forebrain using lipophilic dyes and Cre-recombination lineage tracing, and built a 4D model of brain growth. We reveal modular patterns of anisotropic growth, ascribed to progenitor regions through multiplex HCR. Morphogenesis is dominated by directional growth towards the eye, more isometric expansion of the prethalamus and dorsal telencephalon, and anterior movement of ventral cells into the hypothalamus. Fate conversion experiments in chick and comparative gene expression analysis in chick and mouse support placement of the hypothalamus ventral to structures extending from the telencephalon up to and including the zona limitans intrathalamica (ZLI), with the dorsoventral axis becoming distorted at the base of the ZLI. Our findings challenge the widely accepted prosomere model of forebrain organisation, and we propose an alternative 'tripartite hypothalamus' model.
Collapse
|
33
|
Zhuo L, Wang M, Song T, Zhong S, Zeng B, Liu Z, Zhou X, Wang W, Wu Q, He S, Wang X. MAPbrain: a multi-omics atlas of the primate brain. Nucleic Acids Res 2025; 53:D1055-D1065. [PMID: 39420633 PMCID: PMC11701655 DOI: 10.1093/nar/gkae911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The brain is the central hub of the entire nervous system. Its development is a lifelong process guided by a genetic blueprint. Understanding how genes influence brain development is critical for deciphering the formation of human cognitive functions and the underlying mechanisms of neurological disorders. Recent advances in multi-omics techniques have now made it possible to explore these aspects comprehensively. However, integrating and analyzing extensive multi-omics data presents significant challenges. Here, we introduced MAPbrain (http://bigdata.ibp.ac.cn/mapBRAIN/), a multi-omics atlas of the primate brain. This repository integrates and normalizes both our own lab's published data and publicly available multi-omics data, encompassing 21 million brain cells from 38 key brain regions and 436 sub-regions across embryonic and adult stages, with 164 time points in humans and non-human primates. MAPbrain offers a unique, robust, and interactive platform that includes transcriptomics, epigenomics, and spatial transcriptomics data, facilitating a comprehensive exploration of brain development. The platform enables the exploration of cell type- and time point-specific markers, gene expression comparison between brain regions and species, joint analyses across transcriptome and epigenome, and navigation of cell types across species, brain regions, and development stages. Additionally, MAPbrain provides an online integration module for users to navigate and analyze their own data within the platform.
Collapse
Affiliation(s)
- Liangchen Zhuo
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengdi Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingrui Song
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Suijuan Zhong
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing 100875, China
| | - Bo Zeng
- Changping Laboratory, Beijing 102206, China
| | - Zeyuan Liu
- Changping Laboratory, Beijing 102206, China
| | - Xin Zhou
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing 100875, China
| | - Wei Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing 100875, China
| | - Shunmin He
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoqun Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing 100875, China
- Changping Laboratory, Beijing 102206, China
| |
Collapse
|
34
|
Kim TW, Piao J, Bocchi VD, Koo SY, Choi SJ, Chaudhry F, Yang D, Cho HS, Hergenreder E, Perera LR, Joshi S, Mrad ZA, Claros N, Donohue SA, Frank AK, Walsh R, Mosharov EV, Betel D, Tabar V, Studer L. Enhanced yield and subtype identity of hPSC-derived midbrain dopamine neuron by modulation of WNT and FGF18 signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631400. [PMID: 39829874 PMCID: PMC11741396 DOI: 10.1101/2025.01.06.631400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
While clinical trials are ongoing using human pluripotent stem cell-derived midbrain dopamine (mDA) neuron precursor grafts in Parkinson's disease (PD), current protocols to derive mDA neurons remain suboptimal. In particular, the yield of TH+ mDA neurons after in vivo grafting and the expression of some mDA neuron and subtype-specific markers can be further improved. For example, characterization of mDA grafts by single cell transcriptomics has yielded only a small proportion of mDA neurons and a considerable fraction of contaminating cell populations. Here we present an optimized mDA neuron differentiation strategy that builds on our clinical grade ("Boost") protocol but includes the addition of FGF18 and IWP2 treatment ("Boost+") at the mDA neurogenesis stage. We demonstrate that Boost+ mDA neurons show higher expression of EN1, PITX3 and ALDH1A1. Improvements in both mDA neurons yield and transcriptional similarity to primary mDA neurons is observed both in vitro and in grafts. Furthermore, grafts are enriched in authentic A9 mDA neurons by single nucSeq. Functional studies in vitro demonstrate increased dopamine production and release and improved electrophysiological properties. In vivo analyses show increased percentages of TH+ mDA neurons resulting in efficient rescue of amphetamine induced rotation behavior in the 6-OHDA rat model and rescue of some motor deficits in non-drug induced assays, including the ladder rung assay that is not improved by Boost mDA neurons. The Boost+ conditions present an optimized protocol with advantages for disease modeling and mDA neuron grafting paradigms.
Collapse
|
35
|
Wang Q, Zhu H, Deng L, Xu S, Xie W, Li M, Wang R, Tie L, Zhan L, Yu G. Spatial Transcriptomics: Biotechnologies, Computational Tools, and Neuroscience Applications. SMALL METHODS 2025:e2401107. [PMID: 39760243 DOI: 10.1002/smtd.202401107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Spatial transcriptomics (ST) represents a revolutionary approach in molecular biology, providing unprecedented insights into the spatial organization of gene expression within tissues. This review aims to elucidate advancements in ST technologies, their computational tools, and their pivotal applications in neuroscience. It is begun with a historical overview, tracing the evolution from early image-based techniques to contemporary sequence-based methods. Subsequently, the computational methods essential for ST data analysis, including preprocessing, cell type annotation, spatial clustering, detection of spatially variable genes, cell-cell interaction analysis, and 3D multi-slices integration are discussed. The central focus of this review is the application of ST in neuroscience, where it has significantly contributed to understanding the brain's complexity. Through ST, researchers advance brain atlas projects, gain insights into brain development, and explore neuroimmune dysfunctions, particularly in brain tumors. Additionally, ST enhances understanding of neuronal vulnerability in neurodegenerative diseases like Alzheimer's and neuropsychiatric disorders such as schizophrenia. In conclusion, while ST has already profoundly impacted neuroscience, challenges remain issues such as enhancing sequencing technologies and developing robust computational tools. This review underscores the transformative potential of ST in neuroscience, paving the way for new therapeutic insights and advancements in brain research.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hongyuan Zhu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin Deng
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuangbin Xu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rui Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liang Tie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
36
|
Thompson JR, Nelson ED, Tippani M, Ramnauth AD, Divecha HR, Miller RA, Eagles NJ, Pattie EA, Kwon SH, Bach SV, Kaipa UM, Yao J, Hou C, Kleinman JE, Collado-Torres L, Han S, Maynard KR, Hyde TM, Martinowich K, Page SC, Hicks SC. An integrated single-nucleus and spatial transcriptomics atlas reveals the molecular landscape of the human hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590643. [PMID: 38712198 PMCID: PMC11071618 DOI: 10.1101/2024.04.26.590643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The hippocampus contains many unique cell types, which serve the structure's specialized functions, including learning, memory and cognition. These cells have distinct spatial organization, morphology, physiology, and connectivity, highlighting the importance of transcriptome-wide profiling strategies that retain cytoarchitectural organization. Here, we generated spatially-resolved transcriptomics (SRT) and single-nucleus RNA-sequencing (snRNA-seq) data from adjacent tissue sections of the anterior human hippocampus in ten adult neurotypical donors to define molecular profiles for hippocampal cell types and spatial domains. Using non-negative matrix factorization (NMF) and label transfer, we integrated these data by defining gene expression patterns within the snRNA-seq data and inferring their expression in the SRT data. We identified NMF patterns that captured transcriptional variation across neuronal cell types and indicated that the response of excitatory and inhibitory postsynaptic specializations were prioritized in different SRT spatial domains. We used the NMF and label transfer approach to leverage existing rodent datasets, identifying patterns of activity-dependent transcription and subpopulations of dentate gyrus granule cells in our SRT dataset that may be predisposed to participate in learning and memory ensembles. Finally, we characterized the spatial organization of NMF patterns corresponding to non-cornu ammonis pyramidal neurons and identified snRNA-seq clusters mapping to distinct regions of the retrohippocampus, to three subiculum layers, and to a population of presubiculum neurons. To make this comprehensive molecular atlas accessible to the scientific community, both raw and processed data are freely available, including through interactive web applications.
Collapse
Affiliation(s)
- Jacqueline R. Thompson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Erik D. Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Anthony D. Ramnauth
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Ryan A. Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Nicholas J. Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Elizabeth A. Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Svitlana V. Bach
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Uma M. Kaipa
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Jianing Yao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Hou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Shizhong Han
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, MD, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Stephanie C. Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
37
|
Martin N, Olsen P, Quon J, Campos J, Cuevas NV, Nagra J, VanNess M, Maltzer Z, Gelfand EC, Oyama A, Gary A, Wang Y, Alaya A, Ruiz A, Reynoldson C, Bielstein C, Pom CA, Huang C, Slaughterbeck C, Liang E, Alexander J, Ariza J, Malone J, Melchor J, Colbert K, Brouner K, Shulga L, Reding M, Latimer P, Sanchez R, Barta S, Egdorf T, Madigan Z, Pagan CM, Close JL, Long B, Kunst M, Lein ES, Zeng H, McMillen D, Waters J. MerQuaCo: a computational tool for quality control in image-based spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626766. [PMID: 39677693 PMCID: PMC11643037 DOI: 10.1101/2024.12.04.626766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Image-based spatial transcriptomics platforms are powerful tools often used to identify cell populations and describe gene expression in intact tissue. Spatial experiments return large, high-dimension datasets and several open-source software packages are available to facilitate analysis and visualization. Spatial results are typically imperfect. For example, local variations in transcript detection probability are common. Software tools to characterize imperfections and their impact on downstream analyses are lacking so the data quality is assessed manually, a laborious and often a subjective process. Here we describe imperfections in a dataset of 641 fresh-frozen adult mouse brain sections collected using the Vizgen MERSCOPE. Common imperfections included the local loss of tissue from the section, tissue outside the imaging volume due to detachment from the coverslip, transcripts missing due to dropped images, varying detection probability through space, and differences in transcript detection probability between experiments. We describe the incidence of each imperfection and the likely impact on the accuracy of cell type labels. We develop MerQuaCo, open-source code that detects and quantifies imperfections without user input, facilitating the selection of sections for further analysis with existing packages. Together, our results and MerQuaCo facilitate rigorous, objective assessment of the quality of spatial transcriptomics results.
Collapse
Affiliation(s)
| | | | - Jacob Quon
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jazmin Campos
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | - Josh Nagra
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Marshall VanNess
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Zoe Maltzer
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Emily C Gelfand
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Alana Oyama
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Amanda Gary
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Yimin Wang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Angela Alaya
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Augustin Ruiz
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Cade Reynoldson
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | | | - Cindy Huang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | - Elizabeth Liang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jason Alexander
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jeanelle Ariza
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jocelin Malone
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jose Melchor
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Kaity Colbert
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Krissy Brouner
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Lyudmila Shulga
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Melissa Reding
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Patrick Latimer
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Raymond Sanchez
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Stuard Barta
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Tom Egdorf
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Zachary Madigan
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Chelsea M Pagan
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jennie L Close
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Brian Long
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Michael Kunst
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Ed S Lein
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Hongkui Zeng
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Delissa McMillen
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jack Waters
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| |
Collapse
|
38
|
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
|
39
|
Cassim A, Dun MD, Gallego-Ortega D, Valdes-Mora F. EZHIP's role in diffuse midline glioma: echoes of oncohistones? Trends Cancer 2024; 10:1095-1105. [PMID: 39343635 DOI: 10.1016/j.trecan.2024.09.002] [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: 04/27/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
The enhancer of zeste inhibitory protein (EZHIP) is typically expressed during germ cell development and has been classified as a cancer-testis antigen (CTA) in various cancers. In 2020, 4% of diffuse midline gliomas (DMGs) were shown to aberrantly express EZHIP, mirroring the DMG hallmark histone H3 K27M (H3K27M) oncohistone mutation. Similar to H3K27M, EZHIP is a negative regulator of polycomb repressive complex 2 (PRC2), leading to global epigenomic remodeling. In this opinion, we explore the similarities and disparities between H3K27M- and EZHIP-DMGs with a focus on their shared functional hallmark of PRC2 inhibition, their genetic and epigenomic landscapes, plausible differences in the cell of origin, and therapeutic avenues. Upcoming research on EZHIP will help better understand its role in gliomagenesis and DMG therapy.
Collapse
Affiliation(s)
- Afraah Cassim
- Cancer Epigenetic Biology and Therapeutics Laboratory, Children's Cancer Institute, Lowy Cancer Centre, Kensington, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine, and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia; Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia; Paediatric Stream, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine, and Wellbeing, Callaghan, New South Wales, Australia
| | - David Gallego-Ortega
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales Sydney, New South Wales, Australia; Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics Laboratory, Children's Cancer Institute, Lowy Cancer Centre, Kensington, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, New South Wales, Australia; School of Clinical Medicine, Faculty of Medicine & Health, University of New South Wales Sydney, New South Wales, Australia; Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia.
| |
Collapse
|
40
|
Werner JM, Gillis J. Meta-analysis of single-cell RNA sequencing co-expression in human neural organoids reveals their high variability in recapitulating primary tissue. PLoS Biol 2024; 22:e3002912. [PMID: 39621752 PMCID: PMC11637388 DOI: 10.1371/journal.pbio.3002912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/12/2024] [Accepted: 10/24/2024] [Indexed: 12/14/2024] Open
Abstract
Human neural organoids offer an exciting opportunity for studying inaccessible human-specific brain development; however, it remains unclear how precisely organoids recapitulate fetal/primary tissue biology. We characterize field-wide replicability and biological fidelity through a meta-analysis of single-cell RNA-sequencing data for first and second trimester human primary brain (2.95 million cells, 51 data sets) and neural organoids (1.59 million cells, 173 data sets). We quantify the degree primary tissue cell type marker expression and co-expression are recapitulated in organoids across 10 different protocol types. By quantifying gene-level preservation of primary tissue co-expression, we show neural organoids lie on a spectrum ranging from virtually no signal to co-expression indistinguishable from primary tissue, demonstrating a high degree of variability in biological fidelity among organoid systems. Our preserved co-expression framework provides cell type-specific measures of fidelity applicable to diverse neural organoids, offering a powerful tool for uncovering unifying axes of variation across heterogeneous neural organoid experiments.
Collapse
Affiliation(s)
- Jonathan M. Werner
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Jesse Gillis
- The Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
- Physiology Department and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Baumgartner M, Ji Y, Noonan JP. Reconstructing human-specific regulatory functions in model systems. Curr Opin Genet Dev 2024; 89:102259. [PMID: 39270593 PMCID: PMC11588545 DOI: 10.1016/j.gde.2024.102259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024]
Abstract
Uniquely human physical traits, such as an expanded cerebral cortex and changes in limb morphology that allow us to use tools and walk upright, are in part due to human-specific genetic changes that altered when, where, and how genes are expressed during development. Over 20 000 putative regulatory elements with potential human-specific functions have been discovered. Understanding how these elements contributed to human evolution requires identifying candidates most likely to have shaped human traits, then studying them in genetically modified animal models. Here, we review the progress and challenges in generating and studying such models and propose a pathway for advancing the field. Finally, we highlight that large-scale collaborations across multiple research domains are essential to decipher what makes us human.
Collapse
Affiliation(s)
| | - Yu Ji
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - James P Noonan
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510 USA; Wu Tsai Institute, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
42
|
Lederer AR, Leonardi M, Talamanca L, Bobrovskiy DM, Herrera A, Droin C, Khven I, Carvalho HJF, Valente A, Dominguez Mantes A, Mulet Arabí P, Pinello L, Naef F, La Manno G. Statistical inference with a manifold-constrained RNA velocity model uncovers cell cycle speed modulations. Nat Methods 2024; 21:2271-2286. [PMID: 39482463 PMCID: PMC11621032 DOI: 10.1038/s41592-024-02471-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/15/2024] [Indexed: 11/03/2024]
Abstract
Across biological systems, cells undergo coordinated changes in gene expression, resulting in transcriptome dynamics that unfold within a low-dimensional manifold. While low-dimensional dynamics can be extracted using RNA velocity, these algorithms can be fragile and rely on heuristics lacking statistical control. Moreover, the estimated vector field is not dynamically consistent with the traversed gene expression manifold. To address these challenges, we introduce a Bayesian model of RNA velocity that couples velocity field and manifold estimation in a reformulated, unified framework, identifying the parameters of an explicit dynamical system. Focusing on the cell cycle, we implement VeloCycle to study gene regulation dynamics on one-dimensional periodic manifolds and validate its ability to infer cell cycle periods using live imaging. We also apply VeloCycle to reveal speed differences in regionally defined progenitors and Perturb-seq gene knockdowns. Overall, VeloCycle expands the single-cell RNA sequencing analysis toolkit with a modular and statistically consistent RNA velocity inference framework.
Collapse
Affiliation(s)
- Alex R Lederer
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Maxine Leonardi
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lorenzo Talamanca
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Daniil M Bobrovskiy
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Antonio Herrera
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Colas Droin
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Irina Khven
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Hugo J F Carvalho
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alessandro Valente
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Albert Dominguez Mantes
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Laboratory of Bioimage Analysis and Computational Microscopy, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pau Mulet Arabí
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Luca Pinello
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Felix Naef
- Laboratory of Computational and Systems Biology, Institute of Bioengineering, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| | - Gioele La Manno
- Laboratory of Brain Development and Biological Data Science, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
43
|
Dong P, Song L, Bendl J, Misir R, Shao Z, Edelstien J, Davis DA, Haroutunian V, Scott WK, Acker S, Lawless N, Hoffman GE, Fullard JF, Roussos P. A multi-regional human brain atlas of chromatin accessibility and gene expression facilitates promoter-isoform resolution genetic fine-mapping. Nat Commun 2024; 15:10113. [PMID: 39578476 PMCID: PMC11584674 DOI: 10.1038/s41467-024-54448-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 11/08/2024] [Indexed: 11/24/2024] Open
Abstract
Brain region- and cell-specific transcriptomic and epigenomic features are associated with heritability for neuropsychiatric traits, but a systematic view, considering cortical and subcortical regions, is lacking. Here, we provide an atlas of chromatin accessibility and gene expression profiles in neuronal and non-neuronal nuclei across 25 distinct human cortical and subcortical brain regions from 6 neurotypical controls. We identified extensive gene expression and chromatin accessibility differences across brain regions, including variation in alternative promoter-isoform usage and enhancer-promoter interactions. Genes with distinct promoter-isoform usage across brain regions were strongly enriched for neuropsychiatric disease risk variants. Moreover, we built enhancer-promoter interactions at promoter-isoform resolution across different brain regions and highlighted the contribution of brain region-specific and promoter-isoform-specific regulation to neuropsychiatric disorders. Including promoter-isoform resolution uncovers additional distal elements implicated in the heritability of diseases, thereby increasing the power to fine-map risk genes. Our results provide a valuable resource for studying molecular regulation across multiple regions of the human brain and underscore the importance of considering isoform information in gene regulation.
Collapse
Affiliation(s)
- Pengfei Dong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Liting Song
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth Misir
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhiping Shao
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Edelstien
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David A Davis
- Brain Endowment Bank, Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA
| | - William K Scott
- Brain Endowment Bank, Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
- John P. Hussman Institute for Human Genomics and Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Susanne Acker
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany
| | - Nathan Lawless
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach, Germany
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
44
|
Montserrat-Ayuso T, Esteve-Codina A. High content of nuclei-free low-quality cells in reference single-cell atlases: a call for more stringent quality control using nuclear fraction. BMC Genomics 2024; 25:1124. [PMID: 39574015 PMCID: PMC11580415 DOI: 10.1186/s12864-024-11015-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/08/2024] [Indexed: 11/25/2024] Open
Abstract
The advent of droplet-based single-cell RNA-sequencing (scRNA-seq) has dramatically increased data throughput, enabling the release of a diverse array of tissue cell atlases to the public. However, we will show that prominent initiatives such as the Human Cell Atlas [1], the Tabula Sapiens [2] and the Tabula Muris [3] contain a significant amount of contamination products (frequently affecting the whole organ) in their data portals due to suboptimal quality filtering. Our work addresses a critical gap by advocating for more stringent quality filtering, highlighting the imperative for a shift from existing standards, which currently lean towards greater permissiveness. We will show the importance of incorporating cell intronic fraction in quality control -or MALAT1 expression otherwise- showcasing its informative nature and potential to elevate cell atlas data reliability. In summary, here, we unveil the hidden intronic landscape of every tissue and highlight the importance of more rigorous single-cell RNA-sequencing quality assessment in cell atlases to enhance their applicability in diverse downstream analyses.
Collapse
Affiliation(s)
- Tomàs Montserrat-Ayuso
- Centre Nacional d'Anàlisi Genòmica (CNAG), Baldiri Reixac 4, Barcelona, 08028, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| | - Anna Esteve-Codina
- Centre Nacional d'Anàlisi Genòmica (CNAG), Baldiri Reixac 4, Barcelona, 08028, Spain.
- Universitat de Barcelona (UB), Barcelona, Spain.
| |
Collapse
|
45
|
Nolbrant S, Wallace JL, Ding J, Zhu T, Sevetson JL, Kajtez J, Baldacci IA, Corrigan EK, Hoglin K, McMullen R, Schmitz MT, Breevoort A, Swope D, Wu F, Pavlovic BJ, Salama SR, Kirkeby A, Huang H, Schaefer NK, Pollen AA. INTERSPECIES ORGANOIDS REVEAL HUMAN-SPECIFIC MOLECULAR FEATURES OF DOPAMINERGIC NEURON DEVELOPMENT AND VULNERABILITY. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623592. [PMID: 39605599 PMCID: PMC11601475 DOI: 10.1101/2024.11.14.623592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The disproportionate expansion of telencephalic structures during human evolution involved tradeoffs that imposed greater connectivity and metabolic demands on midbrain dopaminergic neurons. Despite the central role of dopaminergic neurons in human-enriched disorders, molecular specializations associated with human-specific features and vulnerabilities of the dopaminergic system remain unexplored. Here, we establish a phylogeny-in-a-dish approach to examine gene regulatory evolution by differentiating pools of human, chimpanzee, orangutan, and macaque pluripotent stem cells into ventral midbrain organoids capable of forming long-range projections, spontaneous activity, and dopamine release. We identify human-specific gene expression changes related to axonal transport of mitochondria and reactive oxygen species buffering and candidate cis- and trans-regulatory mechanisms underlying gene expression divergence. Our findings are consistent with a model of evolved neuroprotection in response to tradeoffs related to brain expansion and could contribute to the discovery of therapeutic targets and strategies for treating disorders involving the dopaminergic system.
Collapse
Affiliation(s)
- Sara Nolbrant
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Jenelle L. Wallace
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Jingwen Ding
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- These authors contributed equally
| | - Tianjia Zhu
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Jess L. Sevetson
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, CA, United States of America
| | - Janko Kajtez
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW)), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Isabella A. Baldacci
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Emily K. Corrigan
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Kaylynn Hoglin
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Reed McMullen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew T. Schmitz
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Arnar Breevoort
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Dani Swope
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Fengxia Wu
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Anatomy and Neurobiology, Shandong University, Jinan, Shandong Province, China
| | - Bryan J. Pavlovic
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Sofie R. Salama
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, CA, United States of America
- Genomics Institute, University of California Santa Cruz, CA, United States of America
| | - Agnete Kirkeby
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW)), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Experimental Medical Sciences, Wallenberg Center for Molecular Medicine (WCMM) and Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Hao Huang
- Department of Radiology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Nathan K. Schaefer
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Alex A. Pollen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
- Lead contact
| |
Collapse
|
46
|
Kinreich S, Bialer-Tsypin A, Viner-Breuer R, Keshet G, Suhler R, Lim PSL, Golan-Lev T, Yanuka O, Turjeman A, Ram O, Meshorer E, Egli D, Yilmaz A, Benvenisty N. Genome-wide screening reveals essential roles for HOX genes and imprinted genes during caudal neurogenesis of human embryonic stem cells. Stem Cell Reports 2024; 19:1598-1619. [PMID: 39486407 PMCID: PMC11589199 DOI: 10.1016/j.stemcr.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 11/04/2024] Open
Abstract
Mapping the essential pathways for neuronal differentiation can uncover new therapeutics and models for neurodevelopmental disorders. We thus utilized a genome-wide loss-of-function library in haploid human embryonic stem cells, differentiated into caudal neuronal cells. We show that essential genes for caudal neurogenesis are enriched for secreted and membrane proteins and that a large group of neurological conditions, including neurodegenerative disorders, manifest early neuronal phenotypes. Furthermore, essential transcription factors are enriched with homeobox (HOX) genes demonstrating synergistic regulation and surprising non-redundant functions between HOXA6 and HOXB6 paralogs. Moreover, we establish the essentialome of imprinted genes during neurogenesis, demonstrating that maternally expressed genes are non-essential in pluripotent cells and their differentiated germ layers, yet several are essential for neuronal development. These include Beckwith-Wiedemann syndrome- and Angelman syndrome-related genes, for which we suggest a novel regulatory pathway. Overall, our work identifies essential pathways for caudal neuronal differentiation and stage-specific phenotypes of neurological disorders.
Collapse
Affiliation(s)
- Shay Kinreich
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Anna Bialer-Tsypin
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Ruth Viner-Breuer
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Gal Keshet
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Roni Suhler
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Patrick Siang Lin Lim
- Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Tamar Golan-Lev
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Ofra Yanuka
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Adi Turjeman
- The Center for Genomic Technologies, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Oren Ram
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel
| | - Eran Meshorer
- Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; The Edmond and Lily Center for Brain Sciences (ELSC), The Hebrew University, Jerusalem 91904, Israel
| | - Dieter Egli
- Naomi Berrie Diabetes Center & Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Atilgan Yilmaz
- Leuven Stem Cell Institute, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium.
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel; Department of Genetics, Institute of Life Sciences, The Hebrew University, Jerusalem 91904, Israel.
| |
Collapse
|
47
|
Chen X, Ma Y, Shi Y, Fu Y, Nan M, Ren Q, Gao J. Population-Level Cell Trajectory Inference Based on Gaussian Distributions. Biomolecules 2024; 14:1396. [PMID: 39595573 PMCID: PMC11592043 DOI: 10.3390/biom14111396] [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: 10/09/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
In the past decade, inferring developmental trajectories from single-cell data has become a significant challenge in bioinformatics. RNA velocity, with its incorporation of directional dynamics, has significantly advanced the study of single-cell trajectories. However, as single-cell RNA sequencing technology evolves, it generates complex, high-dimensional data with high noise levels. Existing trajectory inference methods, which overlook cell distribution characteristics, may perform inadequately under such conditions. To address this, we introduce CPvGTI, a Gaussian distribution-based trajectory inference method. CPvGTI utilizes a Gaussian mixture model, optimized by the Expectation-Maximization algorithm, to construct new cell populations in the original data space. By integrating RNA velocity, CPvGTI employs Gaussian Process Regression to analyze the differentiation trajectories of these cell populations. To evaluate the performance of CPvGTI, we assess CPvGTI's performance against several state-of-the-art methods using four structurally diverse simulated datasets and four real datasets. The simulation studies indicate that CPvGTI excels in pseudo-time prediction and structural reconstruction compared to existing methods. Furthermore, the discovery of new branch trajectories in human forebrain and mouse hematopoiesis datasets confirms CPvGTI's superior performance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jie Gao
- School of Science, Jiangnan University, Wuxi 214122, China; (X.C.); (Y.M.); (Y.S.); (Y.F.); (M.N.); (Q.R.)
| |
Collapse
|
48
|
He Z, Dony L, Fleck JS, Szałata A, Li KX, Slišković I, Lin HC, Santel M, Atamian A, Quadrato G, Sun J, Pașca SP, Camp JG, Theis FJ, Treutlein B. An integrated transcriptomic cell atlas of human neural organoids. Nature 2024; 635:690-698. [PMID: 39567792 PMCID: PMC11578878 DOI: 10.1038/s41586-024-08172-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 10/08/2024] [Indexed: 11/22/2024]
Abstract
Human neural organoids, generated from pluripotent stem cells in vitro, are useful tools to study human brain development, evolution and disease. However, it is unclear which parts of the human brain are covered by existing protocols, and it has been difficult to quantitatively assess organoid variation and fidelity. Here we integrate 36 single-cell transcriptomic datasets spanning 26 protocols into one integrated human neural organoid cell atlas totalling more than 1.7 million cells1-26. Mapping to developing human brain references27-30 shows primary cell types and states that have been generated in vitro, and estimates transcriptomic similarity between primary and organoid counterparts across protocols. We provide a programmatic interface to browse the atlas and query new datasets, and showcase the power of the atlas to annotate organoid cell types and evaluate new organoid protocols. Finally, we show that the atlas can be used as a diverse control cohort to annotate and compare organoid models of neural disease, identifying genes and pathways that may underlie pathological mechanisms with the neural models. The human neural organoid cell atlas will be useful to assess organoid fidelity, characterize perturbed and diseased states and facilitate protocol development.
Collapse
Affiliation(s)
- Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| | - Leander Dony
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jonas Simon Fleck
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Artur Szałata
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- School of Computation, Information, and Technology, Technical University of Munich, Munich, Germany
| | - Katelyn X Li
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
| | - Irena Slišković
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Department Genes and Environment, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Hsiu-Chuan Lin
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Malgorzata Santel
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Alexander Atamian
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jieran Sun
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neurosciences Institute and Bio-X, Stanford, CA, USA
| | - J Gray Camp
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Fabian J Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- School of Computation, Information, and Technology, Technical University of Munich, Munich, Germany.
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
49
|
Bartels T, Rowitch DH, Bayraktar OA. Generation of Mammalian Astrocyte Functional Heterogeneity. Cold Spring Harb Perspect Biol 2024; 16:a041351. [PMID: 38692833 PMCID: PMC11529848 DOI: 10.1101/cshperspect.a041351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Mammalian astrocytes have regional roles within the brain parenchyma. Indeed, the notion that astrocytes are molecularly heterogeneous could help explain how the central nervous system (CNS) retains embryonic positional information through development into specialized regions into adulthood. A growing body of evidence supports the concept of morphological and molecular differences between astrocytes in different brain regions, which might relate to their derivation from regionally patterned radial glia and/or local neuron inductive cues. Here, we review evidence for regionally encoded functions of astrocytes to provide an integrated concept on lineage origins and heterogeneity to understand regional brain organization, as well as emerging technologies to identify and further investigate novel roles for astrocytes.
Collapse
Affiliation(s)
- Theresa Bartels
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - David H Rowitch
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
50
|
Bosone C, Castaldi D, Burkard TR, Guzman SJ, Wyatt T, Cheroni C, Caporale N, Bajaj S, Bagley JA, Li C, Sorre B, Villa CE, Testa G, Krenn V, Knoblich JA. A polarized FGF8 source specifies frontotemporal signatures in spatially oriented cell populations of cortical assembloids. Nat Methods 2024; 21:2147-2159. [PMID: 39294368 PMCID: PMC11541204 DOI: 10.1038/s41592-024-02412-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 08/12/2024] [Indexed: 09/20/2024]
Abstract
Organoids generating major cortical cell types in distinct compartments are used to study cortical development, evolution and disorders. However, the lack of morphogen gradients imparting cortical positional information and topography in current systems hinders the investigation of complex phenotypes. Here, we engineer human cortical assembloids by fusing an organizer-like structure expressing fibroblast growth factor 8 (FGF8) with an elongated organoid to enable the controlled modulation of FGF8 signaling along the longitudinal organoid axis. These polarized cortical assembloids mount a position-dependent transcriptional program that in part matches the in vivo rostrocaudal gene expression patterns and that is lost upon mutation in the FGFR3 gene associated with temporal lobe malformations and intellectual disability. By producing spatially oriented cell populations with signatures related to frontal and temporal area identity within individual assembloids, this model recapitulates in part the early transcriptional divergence embedded in the protomap and enables the study of cortical area-relevant alterations underlying human disorders.
Collapse
Affiliation(s)
- Camilla Bosone
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Davide Castaldi
- Human Technopole, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Thomas Rainer Burkard
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Segundo Jose Guzman
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Tom Wyatt
- Laboratoire "Matière et Systèmes Complexes" (MSC), UMR 7057 CNRS, University of Paris, Paris, France
| | | | - Nicolò Caporale
- Human Technopole, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sunanjay Bajaj
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Joshua Adam Bagley
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Chong Li
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Benoit Sorre
- Laboratoire "Matière et Systèmes Complexes" (MSC), UMR 7057 CNRS, University of Paris, Paris, France
- Physics of Cells and Cancer, Institut Curie, Université PSL, Sorbonne University, CNRS UMR168, Paris, France
| | | | - Giuseppe Testa
- Human Technopole, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - Veronica Krenn
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Biotechnology and Bioscience, University of Milan-Bicocca, Milan, Italy.
| | - Jürgen Arthur Knoblich
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|