101
|
Huang Y, Yu G, Yang Y. MIGGRI: A multi-instance graph neural network model for inferring gene regulatory networks for Drosophila from spatial expression images. PLoS Comput Biol 2023; 19:e1011623. [PMID: 37939200 PMCID: PMC10659162 DOI: 10.1371/journal.pcbi.1011623] [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: 04/02/2023] [Revised: 11/20/2023] [Accepted: 10/22/2023] [Indexed: 11/10/2023] Open
Abstract
Recent breakthrough in spatial transcriptomics has brought great opportunities for exploring gene regulatory networks (GRNs) from a brand-new perspective. Especially, the local expression patterns and spatio-temporal regulation mechanisms captured by spatial expression images allow more delicate delineation of the interplay between transcript factors and their target genes. However, the complexity and size of spatial image collections pose significant challenges to GRN inference using image-based methods. Extracting regulatory information from expression images is difficult due to the lack of supervision and the multi-instance nature of the problem, where a gene often corresponds to multiple images captured from different views. While graph models, particularly graph neural networks, have emerged as a promising method for leveraging underlying structure information from known GRNs, incorporating expression images into graphs is not straightforward. To address these challenges, we propose a two-stage approach, MIGGRI, for capturing comprehensive regulatory patterns from image collections for each gene and known interactions. Our approach involves a multi-instance graph neural network (GNN) model for GRN inference, which first extracts gene regulatory features from spatial expression images via contrastive learning, and then feeds them to a multi-instance GNN for semi-supervised learning. We apply our approach to a large set of Drosophila embryonic spatial gene expression images. MIGGRI achieves outstanding performance in the inference of GRNs for early eye development and mesoderm development of Drosophila, and shows robustness in the scenarios of missing image information. Additionally, we perform interpretable analysis on image reconstruction and functional subgraphs that may reveal potential pathways or coordinate regulations. By leveraging the power of graph neural networks and the information contained in spatial expression images, our approach has the potential to advance our understanding of gene regulation in complex biological systems.
Collapse
Affiliation(s)
- Yuyang Huang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, and Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, China
| | - Gufeng Yu
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, and Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, China
| | - Yang Yang
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, and Key Laboratory of Shanghai Education Commission for Intelligent Interaction and Cognitive Engineering, Shanghai, China
| |
Collapse
|
102
|
Bhattacherjee A, Zhang C, Watson BR, Djekidel MN, Moffitt JR, Zhang Y. Spatial transcriptomics reveals the distinct organization of mouse prefrontal cortex and neuronal subtypes regulating chronic pain. Nat Neurosci 2023; 26:1880-1893. [PMID: 37845544 PMCID: PMC10620082 DOI: 10.1038/s41593-023-01455-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 09/07/2023] [Indexed: 10/18/2023]
Abstract
The prefrontal cortex (PFC) is a complex brain region that regulates diverse functions ranging from cognition, emotion and executive action to even pain processing. To decode the cellular and circuit organization of such diverse functions, we employed spatially resolved single-cell transcriptome profiling of the adult mouse PFC. Results revealed that PFC has distinct cell-type composition and gene-expression patterns relative to neighboring cortical areas-with neuronal excitability-regulating genes differently expressed. These cellular and molecular features are further segregated within PFC subregions, alluding to the subregion-specificity of several PFC functions. PFC projects to major subcortical targets through combinations of neuronal subtypes, which emerge in a target-intrinsic fashion. Finally, based on these features, we identified distinct cell types and circuits in PFC underlying chronic pain, an escalating healthcare challenge with limited molecular understanding. Collectively, this comprehensive map will facilitate decoding of discrete molecular, cellular and circuit mechanisms underlying specific PFC functions in health and disease.
Collapse
Affiliation(s)
- Aritra Bhattacherjee
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Chao Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Brianna R Watson
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Mohamed Nadhir Djekidel
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
| |
Collapse
|
103
|
Cerneckis J, Shi Y. Myelin organoids for the study of Alzheimer's disease. Front Neurosci 2023; 17:1283742. [PMID: 37942133 PMCID: PMC10628225 DOI: 10.3389/fnins.2023.1283742] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, United States
| |
Collapse
|
104
|
Sharma H, Chang KA, Hulme J, An SSA. Mammalian Models in Alzheimer's Research: An Update. Cells 2023; 12:2459. [PMID: 37887303 PMCID: PMC10605533 DOI: 10.3390/cells12202459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
A form of dementia distinct from healthy cognitive aging, Alzheimer's disease (AD) is a complex multi-stage disease that currently afflicts over 50 million people worldwide. Unfortunately, previous therapeutic strategies developed from murine models emulating different aspects of AD pathogenesis were limited. Consequently, researchers are now developing models that express several aspects of pathogenesis that better reflect the clinical situation in humans. As such, this review seeks to provide insight regarding current applications of mammalian models in AD research by addressing recent developments and characterizations of prominent transgenic models and their contributions to pathogenesis as well as discuss the advantages, limitations, and application of emerging models that better capture genetic heterogeneity and mixed pathologies observed in the clinical situation.
Collapse
Affiliation(s)
- Himadri Sharma
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| | - Keun-A Chang
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea
| | - John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| |
Collapse
|
105
|
Wilbers R, Galakhova AA, Driessens SL, Heistek TS, Metodieva VD, Hagemann J, Heyer DB, Mertens EJ, Deng S, Idema S, de Witt Hamer PC, Noske DP, van Schie P, Kommers I, Luan G, Li T, Shu Y, de Kock CP, Mansvelder HD, Goriounova NA. Structural and functional specializations of human fast-spiking neurons support fast cortical signaling. SCIENCE ADVANCES 2023; 9:eadf0708. [PMID: 37824618 PMCID: PMC10569701 DOI: 10.1126/sciadv.adf0708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 05/17/2023] [Indexed: 10/14/2023]
Abstract
Fast-spiking interneurons (FSINs) provide fast inhibition that synchronizes neuronal activity and is critical for cognitive function. Fast synchronization frequencies are evolutionary conserved in the expanded human neocortex despite larger neuron-to-neuron distances that challenge fast input-output transfer functions of FSINs. Here, we test in human neurons from neurosurgery tissue, which mechanistic specializations of human FSINs explain their fast-signaling properties in human cortex. With morphological reconstructions, multipatch recordings, and biophysical modeling, we find that despite threefold longer dendritic path, human FSINs maintain fast inhibition between connected pyramidal neurons through several mechanisms: stronger synapse strength of excitatory inputs, larger dendrite diameter with reduced complexity, faster AP initiation, and faster and larger inhibitory output, while Na+ current activation/inactivation properties are similar. These adaptations underlie short input-output delays in fast inhibition of human pyramidal neurons through FSINs, explaining how cortical synchronization frequencies are conserved despite expanded and sparse network topology of human cortex.
Collapse
Affiliation(s)
- René Wilbers
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Anna A. Galakhova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Stan L.W. Driessens
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Tim S. Heistek
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Verjinia D. Metodieva
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Jim Hagemann
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Djai B. Heyer
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Eline J. Mertens
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Suixin Deng
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, 19 Xinjiekou Wai Street, Beijing 100875, China
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 201508, China
| | - Sander Idema
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, VUmc Cancer Center, Amsterdam Brain Tumor Center, de Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Philip C. de Witt Hamer
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, VUmc Cancer Center, Amsterdam Brain Tumor Center, de Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - David P. Noske
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, VUmc Cancer Center, Amsterdam Brain Tumor Center, de Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Paul van Schie
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, VUmc Cancer Center, Amsterdam Brain Tumor Center, de Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Ivar Kommers
- Department of Neurosurgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, VUmc Cancer Center, Amsterdam Brain Tumor Center, de Boelelaan 1117, 1081 HV Amsterdam, Netherlands
| | - Guoming Luan
- Department of Neurosurgery, Epilepsy Center, Sanbo Brain Hospital, Capital Medical University, Xiangshan Yikesong 50, Beijing 100093, China
| | - Tianfu Li
- Department of Neurosurgery, Epilepsy Center, Sanbo Brain Hospital, Capital Medical University, Xiangshan Yikesong 50, Beijing 100093, China
| | - Yousheng Shu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, 19 Xinjiekou Wai Street, Beijing 100875, China
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 201508, China
| | - Christiaan P.J. de Kock
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Huibert D. Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | - Natalia A. Goriounova
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Vrije Universiteit Amsterdam, Amsterdam Neuroscience, de Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| |
Collapse
|
106
|
Jorstad NL, Close J, Johansen N, Yanny AM, Barkan ER, Travaglini KJ, Bertagnolli D, Campos J, Casper T, Crichton K, Dee N, Ding SL, Gelfand E, Goldy J, Hirschstein D, Kroll M, Kunst M, Lathia K, Long B, Martin N, McMillen D, Pham T, Rimorin C, Ruiz A, Shapovalova N, Shehata S, Siletti K, Somasundaram S, Sulc J, Tieu M, Torkelson A, Tung H, Ward K, Callaway EM, Hof PR, Keene CD, Levi BP, Linnarsson S, Mitra PP, Smith K, Hodge RD, Bakken TE, Lein ES. Transcriptomic cytoarchitecture reveals principles of human neocortex organization. Science 2023; 382:eadf6812. [PMID: 37824655 PMCID: PMC11687949 DOI: 10.1126/science.adf6812] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 09/08/2023] [Indexed: 10/14/2023]
Abstract
Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.
Collapse
Affiliation(s)
| | - Jennie Close
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | | | | | | | | | - Jazmin Campos
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Tamara Casper
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | - Nick Dee
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Song-Lin Ding
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Emily Gelfand
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Jeff Goldy
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | - Matthew Kroll
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Michael Kunst
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Kanan Lathia
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Brian Long
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Naomi Martin
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | | | | | - Augustin Ruiz
- Allen Institute for Brain Science; Seattle, WA, 98109
| | | | | | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet; Stockholm, Sweden, 171 77
| | | | - Josef Sulc
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Michael Tieu
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Amy Torkelson
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Herman Tung
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Katelyn Ward
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Edward M. Callaway
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA, 92037
| | - Patrick R. Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, WA, 98195
| | - Boaz P. Levi
- Allen Institute for Brain Science; Seattle, WA, 98109
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet; Stockholm, Sweden, 171 77
| | - Partha P. Mitra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724
| | | | | | | | - Ed S. Lein
- Allen Institute for Brain Science; Seattle, WA, 98109
| |
Collapse
|
107
|
Jorstad NL, Song JH, Exposito-Alonso D, Suresh H, Castro-Pacheco N, Krienen FM, Yanny AM, Close J, Gelfand E, Long B, Seeman SC, Travaglini KJ, Basu S, Beaudin M, Bertagnolli D, Crow M, Ding SL, Eggermont J, Glandon A, Goldy J, Kiick K, Kroes T, McMillen D, Pham T, Rimorin C, Siletti K, Somasundaram S, Tieu M, Torkelson A, Feng G, Hopkins WD, Höllt T, Keene CD, Linnarsson S, McCarroll SA, Lelieveldt BP, Sherwood CC, Smith K, Walsh CA, Dobin A, Gillis J, Lein ES, Hodge RD, Bakken TE. Comparative transcriptomics reveals human-specific cortical features. Science 2023; 382:eade9516. [PMID: 37824638 PMCID: PMC10659116 DOI: 10.1126/science.ade9516] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 09/13/2023] [Indexed: 10/14/2023]
Abstract
The cognitive abilities of humans are distinctive among primates, but their molecular and cellular substrates are poorly understood. We used comparative single-nucleus transcriptomics to analyze samples of the middle temporal gyrus (MTG) from adult humans, chimpanzees, gorillas, rhesus macaques, and common marmosets to understand human-specific features of the neocortex. Human, chimpanzee, and gorilla MTG showed highly similar cell-type composition and laminar organization as well as a large shift in proportions of deep-layer intratelencephalic-projecting neurons compared with macaque and marmoset MTG. Microglia, astrocytes, and oligodendrocytes had more-divergent expression across species compared with neurons or oligodendrocyte precursor cells, and neuronal expression diverged more rapidly on the human lineage. Only a few hundred genes showed human-specific patterning, suggesting that relatively few cellular and molecular changes distinctively define adult human cortical structure.
Collapse
Affiliation(s)
| | - Janet H.T. Song
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - David Exposito-Alonso
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Hamsini Suresh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Fenna M. Krienen
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jennie Close
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Emily Gelfand
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Brian Long
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | | | | | - Soumyadeep Basu
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
- Computer Graphics and Visualization Group, Delft University of Technology, Delft, Netherlands
| | - Marc Beaudin
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Megan Crow
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Song-Lin Ding
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Jeroen Eggermont
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
| | | | - Jeff Goldy
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Katelyn Kiick
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Thomas Kroes
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
| | | | | | | | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael Tieu
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Amy Torkelson
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - William D. Hopkins
- Keeling Center for Comparative Medicine and Research, University of Texas, MD Anderson Cancer Center, Houston, TX 78602, USA
| | - Thomas Höllt
- Computer Graphics and Visualization Group, Delft University of Technology, Delft, Netherlands
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 981915, USA
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Steven A. McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Boudewijn P. Lelieveldt
- LKEB, Dept of Radiology, Leiden University Medical Center; Leiden, The Netherlands
- Pattern Recognition and Bioinformatics group, Delft University of Technology, Delft, Netherlands
| | - Chet C. Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20037, USA
| | - Kimberly Smith
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | - Christopher A. Walsh
- Allen Discovery Center for Human Brain Evolution, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Alexander Dobin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Ed S. Lein
- Allen Institute for Brain Science; Seattle, WA, 98109, USA
| | | | | |
Collapse
|
108
|
Li YE, Preissl S, Miller M, Johnson ND, Wang Z, Jiao H, Zhu C, Wang Z, Xie Y, Poirion O, Kern C, Pinto-Duarte A, Tian W, Siletti K, Emerson N, Osteen J, Lucero J, Lin L, Yang Q, Zhu Q, Zemke N, Espinoza S, Yanny AM, Nyhus J, Dee N, Casper T, Shapovalova N, Hirschstein D, Hodge RD, Linnarsson S, Bakken T, Levi B, Keene CD, Shang J, Lein E, Wang A, Behrens MM, Ecker JR, Ren B. A comparative atlas of single-cell chromatin accessibility in the human brain. Science 2023; 382:eadf7044. [PMID: 37824643 PMCID: PMC10852054 DOI: 10.1126/science.adf7044] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 09/14/2023] [Indexed: 10/14/2023]
Abstract
Recent advances in single-cell transcriptomics have illuminated the diverse neuronal and glial cell types within the human brain. However, the regulatory programs governing cell identity and function remain unclear. Using a single-nucleus assay for transposase-accessible chromatin using sequencing (snATAC-seq), we explored open chromatin landscapes across 1.1 million cells in 42 brain regions from three adults. Integrating this data unveiled 107 distinct cell types and their specific utilization of 544,735 candidate cis-regulatory DNA elements (cCREs) in the human genome. Nearly a third of the cCREs demonstrated conservation and chromatin accessibility in the mouse brain cells. We reveal strong links between specific brain cell types and neuropsychiatric disorders including schizophrenia, bipolar disorder, Alzheimer's disease (AD), and major depression, and have developed deep learning models to predict the regulatory roles of noncoding risk variants in these disorders.
Collapse
Affiliation(s)
- Yang Eric Li
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Michael Miller
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | - Zihan Wang
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Henry Jiao
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Chenxu Zhu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhaoning Wang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yang Xie
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Olivier Poirion
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Colin Kern
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | - Wei Tian
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kimberly Siletti
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Nora Emerson
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Julia Osteen
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Jacinta Lucero
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Lin Lin
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Qian Yang
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Quan Zhu
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Nathan Zemke
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Sarah Espinoza
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Sten Linnarsson
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Trygve Bakken
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104, USA
| | - Jingbo Shang
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Allen Wang
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| | | | - Joseph R Ecker
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Epigenomics, University of California San Diego, School of Medicine, La Jolla, CA 92093, USA
| |
Collapse
|
109
|
Chartrand T, Dalley R, Close J, Goriounova NA, Lee BR, Mann R, Miller JA, Molnar G, Mukora A, Alfiler L, Baker K, Bakken TE, Berg J, Bertagnolli D, Braun T, Brouner K, Casper T, Csajbok EA, Dee N, Egdorf T, Enstrom R, Galakhova AA, Gary A, Gelfand E, Goldy J, Hadley K, Heistek TS, Hill D, Jorstad N, Kim L, Kocsis AK, Kruse L, Kunst M, Leon G, Long B, Mallory M, McGraw M, McMillen D, Melief EJ, Mihut N, Ng L, Nyhus J, Oláh G, Ozsvár A, Omstead V, Peterfi Z, Pom A, Potekhina L, Rajanbabu R, Rozsa M, Ruiz A, Sandle J, Sunkin SM, Szots I, Tieu M, Toth M, Trinh J, Vargas S, Vumbaco D, Williams G, Wilson J, Yao Z, Barzo P, Cobbs C, Ellenbogen RG, Esposito L, Ferreira M, Gouwens NW, Grannan B, Gwinn RP, Hauptman JS, Jarsky T, Keene CD, Ko AL, Koch C, Ojemann JG, Patel A, Ruzevick J, Silberberg DL, Smith K, Sorensen SA, Tasic B, Ting JT, Waters J, de Kock CP, Mansvelder HD, Tamas G, Zeng H, Kalmbach B, Lein ES. Morphoelectric and transcriptomic divergence of the layer 1 interneuron repertoire in human versus mouse neocortex. Science 2023; 382:eadf0805. [PMID: 37824667 PMCID: PMC11864503 DOI: 10.1126/science.adf0805] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 09/09/2023] [Indexed: 10/14/2023]
Abstract
Neocortical layer 1 (L1) is a site of convergence between pyramidal-neuron dendrites and feedback axons where local inhibitory signaling can profoundly shape cortical processing. Evolutionary expansion of human neocortex is marked by distinctive pyramidal neurons with extensive L1 branching, but whether L1 interneurons are similarly diverse is underexplored. Using Patch-seq recordings from human neurosurgical tissue, we identified four transcriptomic subclasses with mouse L1 homologs, along with distinct subtypes and types unmatched in mouse L1. Subclass and subtype comparisons showed stronger transcriptomic differences in human L1 and were correlated with strong morphoelectric variability along dimensions distinct from mouse L1 variability. Accompanied by greater layer thickness and other cytoarchitecture changes, these findings suggest that L1 has diverged in evolution, reflecting the demands of regulating the expanded human neocortical circuit.
Collapse
Affiliation(s)
| | | | | | - Natalia A. Goriounova
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | | | - Rusty Mann
- Allen Institute for Brain Science; Seattle, USA
| | | | - Gabor Molnar
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | | | | | | | - Jim Berg
- Allen Institute for Brain Science; Seattle, USA
| | | | | | | | | | - Eva Adrienn Csajbok
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Nick Dee
- Allen Institute for Brain Science; Seattle, USA
| | - Tom Egdorf
- Allen Institute for Brain Science; Seattle, USA
| | | | - Anna A. Galakhova
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - Amanda Gary
- Allen Institute for Brain Science; Seattle, USA
| | | | - Jeff Goldy
- Allen Institute for Brain Science; Seattle, USA
| | | | - Tim S. Heistek
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - DiJon Hill
- Allen Institute for Brain Science; Seattle, USA
| | - Nik Jorstad
- Allen Institute for Brain Science; Seattle, USA
| | - Lisa Kim
- Allen Institute for Brain Science; Seattle, USA
| | - Agnes Katalin Kocsis
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | | | | | - Brian Long
- Allen Institute for Brain Science; Seattle, USA
| | | | | | | | - Erica J. Melief
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, USA
| | - Norbert Mihut
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Lindsay Ng
- Allen Institute for Brain Science; Seattle, USA
| | - Julie Nyhus
- Allen Institute for Brain Science; Seattle, USA
| | - Gáspár Oláh
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Attila Ozsvár
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Zoltan Peterfi
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | - Alice Pom
- Allen Institute for Brain Science; Seattle, USA
| | | | | | - Marton Rozsa
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Joanna Sandle
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Ildiko Szots
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Martin Toth
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Sara Vargas
- Allen Institute for Brain Science; Seattle, USA
| | | | | | | | - Zizhen Yao
- Allen Institute for Brain Science; Seattle, USA
| | - Pal Barzo
- Department of Neurosurgery, University of Szeged; Szeged, Hungary
| | | | | | | | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | - Benjamin Grannan
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | - Jason S. Hauptman
- Department of Neurological Surgery, University of Washington; Seattle USA
| | - Tim Jarsky
- Allen Institute for Brain Science; Seattle, USA
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington; Seattle, USA
| | - Andrew L. Ko
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | - Jeffrey G. Ojemann
- Department of Neurological Surgery, University of Washington; Seattle USA
| | - Anoop Patel
- Department of Neurological Surgery, University of Washington; Seattle USA
| | - Jacob Ruzevick
- Department of Neurological Surgery, University of Washington; Seattle USA
| | | | | | | | | | - Jonathan T. Ting
- Allen Institute for Brain Science; Seattle, USA
- Department of Physiology and Biophysics, University of Washington; Seattle, USA
- Washington National Primate Research Center, University of Washington; Seattle, USA
| | - Jack Waters
- Allen Institute for Brain Science; Seattle, USA
| | - Christiaan P.J. de Kock
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - Huib D. Mansvelder
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit; Amsterdam, The Netherlands
| | - Gabor Tamas
- Research Group for Cortical Microcircuits of the Hungarian Academy of Science, University of Szeged; Szeged, Hungary
| | | | - Brian Kalmbach
- Allen Institute for Brain Science; Seattle, USA
- Department of Physiology and Biophysics, University of Washington; Seattle, USA
| | - Ed S. Lein
- Allen Institute for Brain Science; Seattle, USA
- Department of Neurological Surgery, University of Washington; Seattle USA
| |
Collapse
|
110
|
Han X, Guo S, Ji N, Li T, Liu J, Ye X, Wang Y, Yun Z, Xiong F, Rong J, Liu D, Ma H, Wang Y, Huang Y, Zhang P, Wu W, Ding L, Hawrylycz M, Lein E, Ascoli GA, Xie W, Liu L, Zhang L, Peng H. Whole human-brain mapping of single cortical neurons for profiling morphological diversity and stereotypy. SCIENCE ADVANCES 2023; 9:eadf3771. [PMID: 37824619 PMCID: PMC10569712 DOI: 10.1126/sciadv.adf3771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 04/18/2023] [Indexed: 10/14/2023]
Abstract
Quantifying neuron morphology and distribution at the whole-brain scale is essential to understand the structure and diversity of cell types. It is exceedingly challenging to reuse recent technologies of single-cell labeling and whole-brain imaging to study human brains. We propose adaptive cell tomography (ACTomography), a low-cost, high-throughput, and high-efficacy tomography approach, based on adaptive targeting of individual cells. We established a platform to inject dyes into cortical neurons in surgical tissues of 18 patients with brain tumors or other conditions and one donated fresh postmortem brain. We collected three-dimensional images of 1746 cortical neurons, of which 852 neurons were reconstructed to quantify local dendritic morphology, and mapped to standard atlases. In our data, human neurons are more diverse across brain regions than by subject age or gender. The strong stereotypy within cohorts of brain regions allows generating a statistical tensor field of neuron morphology to characterize anatomical modularity of a human brain.
Collapse
Affiliation(s)
- Xiaofeng Han
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Shuxia Guo
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Tian Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Xiangqiao Ye
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Yi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhixi Yun
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Feng Xiong
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Jing Rong
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Di Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Hui Ma
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Yujin Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yue Huang
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenhao Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liya Ding
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | | | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Giorgio A. Ascoli
- Center for Neural Informatics, Krasnow Institute for Advanced Studies and Bioengineering Department, College of Engineering and Computing, George Mason University, Fairfax, VA, USA
| | - Wei Xie
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Lijuan Liu
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Key Laboratory of Brain Tumor, Beijing, China
| | - Hanchuan Peng
- Institute for Brain and Intelligence, Southeast University, Nanjing, China
| |
Collapse
|
111
|
Jung N, Kim TK. Spatial transcriptomics in neuroscience. Exp Mol Med 2023; 55:2105-2115. [PMID: 37779145 PMCID: PMC10618223 DOI: 10.1038/s12276-023-01093-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 10/03/2023] Open
Abstract
The brain is one of the most complex living tissue types and is composed of an exceptional diversity of cell types displaying unique functional connectivity. Single-cell RNA sequencing (scRNA-seq) can be used to efficiently map the molecular identities of the various cell types in the brain by providing the transcriptomic profiles of individual cells isolated from the tissue. However, the lack of spatial context in scRNA-seq prevents a comprehensive understanding of how different configurations of cell types give rise to specific functions in individual brain regions and how each distinct cell is connected to form a functional unit. To understand how the various cell types contribute to specific brain functions, it is crucial to correlate the identities of individual cells obtained through scRNA-seq with their spatial information in intact tissue. Spatial transcriptomics (ST) can resolve the complex spatial organization of cell types in the brain and their connectivity. Various ST tools developed during the past decade based on imaging and sequencing technology have permitted the creation of functional atlases of the brain and have pulled the properties of neural circuits into ever-sharper focus. In this review, we present a summary of several ST tools and their applications in neuroscience and discuss the unprecedented insights these tools have made possible.
Collapse
Affiliation(s)
- Namyoung Jung
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
112
|
Chen X, Deng R, Su D, Ma X, Han X, Wang S, Xia Y, Yang Z, Gong N, Jia Y, Gao X, Ren X. Visual genetic typing of glioma using proximity-anchored in situ spectral coding amplification. EXPLORATION (BEIJING, CHINA) 2023; 3:20220175. [PMID: 37933281 PMCID: PMC10582607 DOI: 10.1002/exp.20220175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/21/2023] [Indexed: 11/08/2023]
Abstract
Gliomas are histologically and genetically heterogeneous tumors. However, classical histopathological typing often ignores the high heterogeneity of tumors and thus cannot meet the requirements of precise pathological diagnosis. Here, proximity-anchored in situ spectral coding amplification (ProxISCA) is proposed for multiplexed imaging of RNA mutations, enabling visual typing of brain gliomas with different pathological grades at the single-cell and tissue levels. The ligation-based padlock probe can discriminate one-nucleotide variations, and the design of proximity primers enables the anchoring of amplicons on target RNA, thus improving localization accuracy. The DNA module-based spectral coding strategy can dramatically improve the multiplexing capacity for imaging RNA mutations through one-time labelling, with low cost and simple operation. One-target-one-amplicon amplification confers ProxISCA the ability to quantify RNA mutation copy number with single-molecule resolution. Based on this approach, it is found that gliomas with higher malignant grades express more genes with high correlation at the cellular and tissue levels and show greater cellular heterogeneity. ProxISCA provides a tool for glioma research and precise diagnosis, which can reveal the relationship between cellular heterogeneity and glioma occurrence or development and assist in pathological prognosis.
Collapse
Affiliation(s)
- Xiaolei Chen
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Ruijie Deng
- College of Biomass Science and EngineeringHealthy Food Evaluation Research CenterSichuan UniversityChengduChina
| | - Dongdong Su
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Xiaochen Ma
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Xu Han
- Institute of High Energy PhysicsChinese Academy of SciencesBeijingChina
| | - Shizheng Wang
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Yuqing Xia
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Zifu Yang
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Ningqiang Gong
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaUSA
| | - Yanwei Jia
- State‐Key Laboratory of Analog and Mixed‐Signal VLSIInstitute of MicroelectronicsUniversity of MacauMacauChina
| | - Xueyun Gao
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| | - Xiaojun Ren
- Department of Chemistry and BiologyFaculty of Environment and Life ScienceBeijing University of TechnologyBeijingChina
| |
Collapse
|
113
|
Tang Z, Liu X, Li Z, Zhang T, Yang B, Su J, Song Q. SpaRx: elucidate single-cell spatial heterogeneity of drug responses for personalized treatment. Brief Bioinform 2023; 24:bbad338. [PMID: 37798249 PMCID: PMC10555713 DOI: 10.1093/bib/bbad338] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/08/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
Spatial cellular authors heterogeneity contributes to differential drug responses in a tumor lesion and potential therapeutic resistance. Recent emerging spatial technologies such as CosMx, MERSCOPE and Xenium delineate the spatial gene expression patterns at the single cell resolution. This provides unprecedented opportunities to identify spatially localized cellular resistance and to optimize the treatment for individual patients. In this work, we present a graph-based domain adaptation model, SpaRx, to reveal the heterogeneity of spatial cellular response to drugs. SpaRx transfers the knowledge from pharmacogenomics profiles to single-cell spatial transcriptomics data, through hybrid learning with dynamic adversarial adaption. Comprehensive benchmarking demonstrates the superior and robust performance of SpaRx at different dropout rates, noise levels and transcriptomics coverage. Further application of SpaRx to the state-of-the-art single-cell spatial transcriptomics data reveals that tumor cells in different locations of a tumor lesion present heterogenous sensitivity or resistance to drugs. Moreover, resistant tumor cells interact with themselves or the surrounding constituents to form an ecosystem for drug resistance. Collectively, SpaRx characterizes the spatial therapeutic variability, unveils the molecular mechanisms underpinning drug resistance and identifies personalized drug targets and effective drug combinations.
Collapse
Affiliation(s)
- Ziyang Tang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Xiang Liu
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Zuotian Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
- Department of Computer Graphics Technology, Purdue University, Indiana, USA
| | - Tonglin Zhang
- Department of Statistics, Purdue University, Indiana, USA
| | - Baijian Yang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest University School of Medicine, North Carolina, USA
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Florida, USA
| |
Collapse
|
114
|
Luo X, Liu Z, Xu R. Adult tissue-specific stem cell interaction: novel technologies and research advances. Front Cell Dev Biol 2023; 11:1220694. [PMID: 37808078 PMCID: PMC10551553 DOI: 10.3389/fcell.2023.1220694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Adult tissue-specific stem cells play a dominant role in tissue homeostasis and regeneration. Various in vivo markers of adult tissue-specific stem cells have been increasingly reported by lineage tracing in genetic mouse models, indicating that marked cells differentiation is crucial during homeostasis and regeneration. How adult tissue-specific stem cells with indicated markers contact the adjacent lineage with indicated markers is of significance to be studied. Novel methods bring future findings. Recent advances in lineage tracing, synthetic receptor systems, proximity labeling, and transcriptomics have enabled easier and more accurate cell behavior visualization and qualitative and quantitative analysis of cell-cell interactions than ever before. These technological innovations have prompted researchers to re-evaluate previous experimental results, providing increasingly compelling experimental results for understanding the mechanisms of cell-cell interactions. This review aimed to describe the recent methodological advances of dual enzyme lineage tracing system, the synthetic receptor system, proximity labeling, single-cell RNA sequencing and spatial transcriptomics in the study of adult tissue-specific stem cells interactions. An enhanced understanding of the mechanisms of adult tissue-specific stem cells interaction is important for tissue regeneration and maintenance of homeostasis in organisms.
Collapse
Affiliation(s)
| | | | - Ruoshi Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
115
|
Li J, Jaiswal MK, Chien JF, Kozlenkov A, Jung J, Zhou P, Gardashli M, Pregent LJ, Engelberg-Cook E, Dickson DW, Belzil VV, Mukamel EA, Dracheva S. Divergent single cell transcriptome and epigenome alterations in ALS and FTD patients with C9orf72 mutation. Nat Commun 2023; 14:5714. [PMID: 37714849 PMCID: PMC10504300 DOI: 10.1038/s41467-023-41033-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
A repeat expansion in the C9orf72 (C9) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we investigate single nucleus transcriptomics (snRNA-seq) and epigenomics (snATAC-seq) in postmortem motor and frontal cortices from C9-ALS, C9-FTD, and control donors. C9-ALS donors present pervasive alterations of gene expression with concordant changes in chromatin accessibility and histone modifications. The greatest alterations occur in upper and deep layer excitatory neurons, as well as in astrocytes. In neurons, the changes imply an increase in proteostasis, metabolism, and protein expression pathways, alongside a decrease in neuronal function. In astrocytes, the alterations suggest activation and structural remodeling. Conversely, C9-FTD donors have fewer high-quality neuronal nuclei in the frontal cortex and numerous gene expression changes in glial cells. These findings highlight a context-dependent molecular disruption in C9-ALS and C9-FTD, indicating unique effects across cell types, brain regions, and diseases.
Collapse
Affiliation(s)
- Junhao Li
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, 92037, US
| | - Manoj K Jaiswal
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Jo-Fan Chien
- Department of Physics, University of California San Diego, La Jolla, CA, 92037, US
| | - Alexey Kozlenkov
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Jinyoung Jung
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | - Ping Zhou
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US
| | | | - Luc J Pregent
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, US
| | | | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, US
| | | | - Eran A Mukamel
- Department of Cognitive Science, University of California San Diego, La Jolla, CA, 92037, US.
| | - Stella Dracheva
- Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, US.
- Research & Development and VISN2 MIREC, James J, Peters VA Medical Center, Bronx, NY, 10468, US.
| |
Collapse
|
116
|
Tang Z, Li Z, Hou T, Zhang T, Yang B, Su J, Song Q. SiGra: single-cell spatial elucidation through an image-augmented graph transformer. Nat Commun 2023; 14:5618. [PMID: 37699885 PMCID: PMC10497630 DOI: 10.1038/s41467-023-41437-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/01/2023] [Indexed: 09/14/2023] Open
Abstract
Recent advances in high-throughput molecular imaging have pushed spatial transcriptomics technologies to subcellular resolution, which surpasses the limitations of both single-cell RNA-seq and array-based spatial profiling. The multichannel immunohistochemistry images in such data provide rich information on the cell types, functions, and morphologies of cellular compartments. In this work, we developed a method, single-cell spatial elucidation through image-augmented Graph transformer (SiGra), to leverage such imaging information for revealing spatial domains and enhancing substantially sparse and noisy transcriptomics data. SiGra applies hybrid graph transformers over a single-cell spatial graph. SiGra outperforms state-of-the-art methods on both single-cell and spot-level spatial transcriptomics data from complex tissues. The inclusion of immunohistochemistry images improves the model performance by 37% (95% CI: 27-50%). SiGra improves the characterization of intratumor heterogeneity and intercellular communication and recovers the known microscopic anatomy. Overall, SiGra effectively integrates different spatial modality data to gain deep insights into spatial cellular ecosystems.
Collapse
Affiliation(s)
- Ziyang Tang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Zuotian Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
- Department of Computer Graphics Technology, Purdue University, Indiana, USA
| | - Tieying Hou
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indiana, USA
| | - Tonglin Zhang
- Department of Statistics, Purdue University, Indiana, USA
| | - Baijian Yang
- Department of Computer and Information Technology, Purdue University, Indiana, USA.
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA.
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest University School of Medicine, North Carolina, USA.
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Florida, USA.
| |
Collapse
|
117
|
Lin X, Sun Y, Dong X, Liu Z, Sugimura R, Xie G. IPSC-derived CAR-NK cells for cancer immunotherapy. Biomed Pharmacother 2023; 165:115123. [PMID: 37406511 DOI: 10.1016/j.biopha.2023.115123] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023] Open
Abstract
Adoptive cell therapies (ACT) based on chimeric antigen receptor (CAR)-modified immune cells have made great progress with six CAR-T cell products approved by the U.S. FDA for hematological malignancies. Compared with CAR-T cells, CAR-NK cells have attracted increasing attention owing to their multiple killing mechanisms, higher safety profile, and broad sources. Induced pluripotent stem cell (iPSC)-derived NK (iPSC-NK) cells possess a mature phenotype and potent cytolytic activity, and can provide a homogeneous population of CAR-NK cells that can be expanded to clinical scale. Thus, iPSC-derived CAR-NK (CAR-iNK) cells could be used as a standardized and "off-the-shelf" product for cancer immunotherapy. In this review, we summarize the current status of the manufacturing techniques, genetic modification strategies, preclinical and clinical evidence of CAR-iNK cells, and discuss the challenges and future prospects of CAR-iNK cell therapy as a novel cellular immunotherapy in cancer.
Collapse
Affiliation(s)
- Xiaotong Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yao Sun
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xin Dong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zishen Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ryohichi Sugimura
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region of China.
| | - Guozhu Xie
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
118
|
Wang Y, Liu B, Zhao G, Lee Y, Buzdin A, Mu X, Zhao J, Chen H, Li X. Spatial transcriptomics: Technologies, applications and experimental considerations. Genomics 2023; 115:110671. [PMID: 37353093 PMCID: PMC10571167 DOI: 10.1016/j.ygeno.2023.110671] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
The diverse cell types of an organ have a highly structured organization to enable their efficient and correct function. To fully appreciate gene functions in a given cell type, one needs to understand how much, when and where the gene is expressed. Classic bulk RNA sequencing and popular single cell sequencing destroy cell structural organization and fail to provide spatial information. However, the spatial location of gene expression or of the cell in a complex tissue provides key clues to comprehend how the neighboring genes or cells cross talk, transduce signals and work together as a team to complete the job. The functional requirement for the spatial content has been a driving force for rapid development of the spatial transcriptomics technologies in the past few years. Here, we present an overview of current spatial technologies with a special focus on the commercially available or currently being commercialized technologies, highlight their applications by category and discuss experimental considerations for a first spatial experiment.
Collapse
Affiliation(s)
- Ye Wang
- Clinical Laboratory, The Affiliated Qingdao Central Hospital of Medical College of Qingdao University, Qingdao 266042, China.
| | - Bin Liu
- Departments of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China
| | - Gexin Zhao
- UCLA Technology Center for Genomics & Bioinformatics, Department of Pathology & Laboratory Medicine, 650 Charles E Young Dr., Los Angeles, CA 90095, USA
| | - YooJin Lee
- UCLA Technology Center for Genomics & Bioinformatics, Department of Pathology & Laboratory Medicine, 650 Charles E Young Dr., Los Angeles, CA 90095, USA
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia; Moscow Institute of Physics and Technology, Moscow Region, 141701, Russia; World-Class Research Center "Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Xiaofeng Mu
- Clinical Laboratory, The Affiliated Qingdao Central Hospital of Medical College of Qingdao University, Qingdao 266042, China
| | - Joseph Zhao
- UCLA Technology Center for Genomics & Bioinformatics, Department of Pathology & Laboratory Medicine, 650 Charles E Young Dr., Los Angeles, CA 90095, USA
| | - Hong Chen
- Heilongjiang Academy of Traditional Chinese Medicine, No. 142, Sanfu Street, Xiangfang District, Harbin City, Heilongjiang Province 150036, China
| | - Xinmin Li
- UCLA Technology Center for Genomics & Bioinformatics, Department of Pathology & Laboratory Medicine, 650 Charles E Young Dr., Los Angeles, CA 90095, USA.
| |
Collapse
|
119
|
徐 晨, 王 寅, 魏 东, 李 文, 钱 晔, 潘 新, 雷 大. [Advances of spatial omics in the individualized diagnosis and treatment of head and neck cancer]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2023; 37:729-733;739. [PMID: 37830120 PMCID: PMC10722126 DOI: 10.13201/j.issn.2096-7993.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Indexed: 10/14/2023]
Abstract
Spatialomics is another research hotspot of biotechnology after single-cell sequencing technology, which can make up for the defect that single-cell sequencing technology can not obtain cell spatial distribution information. Spatialomics mainly studies the relative position of cells in tissue samples to reveal the effect of cell spatial distribution on diseases. In recent years, spatialomics has made new progress in the pathogenesis, target exploration, drug development and many other aspects of head and neck tumors. This paper summarizes the latest progress of spatialomics in the diagnosis and treatment of head and neck cancer.
Collapse
Affiliation(s)
- 晨阳 徐
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| | - 寅 王
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| | - 东敏 魏
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| | - 文明 李
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| | - 晔 钱
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| | - 新良 潘
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| | - 大鹏 雷
- 山东大学齐鲁医院耳鼻咽喉科,国家卫生健康委员会耳鼻喉科学重点实验室(山东大学)(济南,250012)Department of Otorhinolaryngology, Qilu Hospital of Shandong University, National Health Commission Key Laboratory of Otorhinolaryngology[Shandong University], Jinan, 250012, China
| |
Collapse
|
120
|
Johansen N, Hu H, Quon G. Projecting RNA measurements onto single cell atlases to extract cell type-specific expression profiles using scProjection. Nat Commun 2023; 14:5192. [PMID: 37626024 PMCID: PMC10457395 DOI: 10.1038/s41467-023-40744-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Multi-modal single cell RNA assays capture RNA content as well as other data modalities, such as spatial cell position or the electrophysiological properties of cells. Compared to dedicated scRNA-seq assays however, they may unintentionally capture RNA from multiple adjacent cells, exhibit lower RNA sequencing depth compared to scRNA-seq, or lack genome-wide RNA measurements. We present scProjection, a method for mapping individual multi-modal RNA measurements to deeply sequenced scRNA-seq atlases to extract cell type-specific, single cell gene expression profiles. We demonstrate several use cases of scProjection, including identifying spatial motifs from spatial transcriptome assays, distinguishing RNA contributions from neighboring cells in both spatial and multi-modal single cell assays, and imputing expression measurements of un-measured genes from gene markers. scProjection therefore combines the advantages of both multi-modal and scRNA-seq assays to yield precise multi-modal measurements of single cells.
Collapse
Affiliation(s)
- Nelson Johansen
- Graduate Group in Computer Science, University of California, Davis, Davis, CA, USA.
| | - Hongru Hu
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, Davis, CA, USA
| | - Gerald Quon
- Graduate Group in Computer Science, University of California, Davis, Davis, CA, USA.
- Integrative Genetics and Genomics Graduate Group, University of California, Davis, Davis, CA, USA.
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
121
|
Guma E, Beauchamp A, Liu S, Levitis E, Ellegood J, Pham L, Mars RB, Raznahan A, Lerch JP. Comparative neuroimaging of sex differences in human and mouse brain anatomy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.23.554334. [PMID: 37662398 PMCID: PMC10473765 DOI: 10.1101/2023.08.23.554334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
In vivo neuroimaging studies have established several reproducible volumetric sex differences in the human brain, but the causes of such differences are hard to parse. While mouse models are useful for understanding the cellular and mechanistic bases of sex-biased brain development in mammals, there have been no attempts to formally compare mouse and human sex differences across the whole brain to ascertain how well they translate. Addressing this question would shed critical light on use of the mouse as a translational model for sex differences in the human brain and provide insights into the degree to which sex differences in brain volume are conserved across mammals. Here, we use cross-species structural magnetic resonance imaging to carry out the first comparative neuroimaging study of sex-biased neuroanatomical organization of the human and mouse brain. In line with previous findings, we observe that in humans, males have significantly larger and more variable total brain volume; these sex differences are not mirrored in mice. After controlling for total brain volume, we observe modest cross-species congruence in the volumetric effect size of sex across 60 homologous brain regions (r=0.30; e.g.: M>F amygdala, hippocampus, bed nucleus of the stria terminalis, and hypothalamus and F>M anterior cingulate, somatosensory, and primary auditory cortices). This cross-species congruence is greater in the cortex (r=0.33) than non-cortex (r=0.16). By incorporating regional measures of gene expression in both species, we reveal that cortical regions with greater cross-species congruence in volumetric sex differences also show greater cross-species congruence in the expression profile of 2835 homologous genes. This phenomenon differentiates primary sensory regions with high congruence of sex effects and gene expression from limbic cortices where congruence in both these features was weaker between species. These findings help identify aspects of sex-biased brain anatomy present in mice that are retained, lost, or inverted in humans. More broadly, our work provides an empirical basis for targeting mechanistic studies of sex-biased brain development in mice to brain regions that best echo sex-biased brain development in humans.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Antoine Beauchamp
- Mouse Imaging Centre, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Siyuan Liu
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Elizabeth Levitis
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jacob Ellegood
- Mouse Imaging Centre, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Linh Pham
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Rogier B Mars
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Jason P Lerch
- Mouse Imaging Centre, Toronto, Ontario, Canada
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Wellcome Centre for Integrative Neuroimaging, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
122
|
Kalhor K, Chen CJ, Lee HS, Cai M, Nafisi M, Que R, Palmer C, Yuan Y, Zhang Y, Song J, Knoten A, Lake BB, Gaut JP, Keene D, Lein E, Kharchenko PV, Chun J, Jain S, Fan JB, Zhang K. Mapping Human Tissues with Highly Multiplexed RNA in situ Hybridization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.16.553610. [PMID: 37645998 PMCID: PMC10462101 DOI: 10.1101/2023.08.16.553610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
In situ transcriptomic techniques promise a holistic view of tissue organization and cell-cell interactions. Recently there has been a surge of multiplexed RNA in situ techniques but their application to human tissues and clinical biopsies has been limited due to their large size, general lower tissue quality and high background autofluorescence. Here we report DART-FISH, a versatile padlock probe-based technology capable of profiling hundreds to thousands of genes in centimeter-sized human tissue sections at cellular resolution. We introduced an omni-cell type cytoplasmic stain, dubbed RiboSoma that substantially improves the segmentation of cell bodies. We developed a computational decoding-by-deconvolution workflow to extract gene spots even in the presence of optical crowding. Our enzyme-free isothermal decoding procedure allowed us to image 121 genes in a large section from the human neocortex in less than 10 hours, where we successfully recapitulated the cytoarchitecture of 20 neuronal and non-neuronal subclasses. Additionally, we demonstrated the detection of transcripts as short as 461 nucleotides, including neuropeptides and discovered new cortical layer markers. We further performed in situ mapping of 300 genes on a diseased human kidney, profiled >20 healthy and pathological cell states, and identified diseased niches enriched in transcriptionally altered epithelial cells and myofibroblasts.
Collapse
Affiliation(s)
- Kian Kalhor
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Chien-Ju Chen
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Program in Bioinformatics and Systems Biology, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally
| | - Ho Suk Lee
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Electrical Engineering, University of California San Diego, La Jolla, CA, USA
| | - Matthew Cai
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Mahsa Nafisi
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Richard Que
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Carter Palmer
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
- Program in Biomedical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Yixu Yuan
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Yida Zhang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jinghui Song
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Amanda Knoten
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Blue B. Lake
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Joseph P. Gaut
- Department of Pathology and Immunology, Washington University School of Medicine, St
| | - Dirk Keene
- University of Washington School of Medicine, Seattle, WA, USA
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA, USA Louis, MO, USA
| | - Peter V. Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Sanjay Jain
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St
| | | | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
123
|
Chen A, Sun Y, Lei Y, Li C, Liao S, Meng J, Bai Y, Liu Z, Liang Z, Zhu Z, Yuan N, Yang H, Wu Z, Lin F, Wang K, Li M, Zhang S, Yang M, Fei T, Zhuang Z, Huang Y, Zhang Y, Xu Y, Cui L, Zhang R, Han L, Sun X, Chen B, Li W, Huangfu B, Ma K, Ma J, Li Z, Lin Y, Wang H, Zhong Y, Zhang H, Yu Q, Wang Y, Liu X, Peng J, Liu C, Chen W, Pan W, An Y, Xia S, Lu Y, Wang M, Song X, Liu S, Wang Z, Gong C, Huang X, Yuan Y, Zhao Y, Chai Q, Tan X, Liu J, Zheng M, Li S, Huang Y, Hong Y, Huang Z, Li M, Jin M, Li Y, Zhang H, Sun S, Gao L, Bai Y, Cheng M, Hu G, Liu S, Wang B, Xiang B, Li S, Li H, Chen M, Wang S, Li M, Liu W, Liu X, Zhao Q, Lisby M, Wang J, Fang J, Lin Y, Xie Q, Liu Z, He J, Xu H, Huang W, Mulder J, Yang H, Sun Y, Uhlen M, Poo M, Wang J, Yao J, Wei W, et alChen A, Sun Y, Lei Y, Li C, Liao S, Meng J, Bai Y, Liu Z, Liang Z, Zhu Z, Yuan N, Yang H, Wu Z, Lin F, Wang K, Li M, Zhang S, Yang M, Fei T, Zhuang Z, Huang Y, Zhang Y, Xu Y, Cui L, Zhang R, Han L, Sun X, Chen B, Li W, Huangfu B, Ma K, Ma J, Li Z, Lin Y, Wang H, Zhong Y, Zhang H, Yu Q, Wang Y, Liu X, Peng J, Liu C, Chen W, Pan W, An Y, Xia S, Lu Y, Wang M, Song X, Liu S, Wang Z, Gong C, Huang X, Yuan Y, Zhao Y, Chai Q, Tan X, Liu J, Zheng M, Li S, Huang Y, Hong Y, Huang Z, Li M, Jin M, Li Y, Zhang H, Sun S, Gao L, Bai Y, Cheng M, Hu G, Liu S, Wang B, Xiang B, Li S, Li H, Chen M, Wang S, Li M, Liu W, Liu X, Zhao Q, Lisby M, Wang J, Fang J, Lin Y, Xie Q, Liu Z, He J, Xu H, Huang W, Mulder J, Yang H, Sun Y, Uhlen M, Poo M, Wang J, Yao J, Wei W, Li Y, Shen Z, Liu L, Liu Z, Xu X, Li C. Single-cell spatial transcriptome reveals cell-type organization in the macaque cortex. Cell 2023; 186:3726-3743.e24. [PMID: 37442136 DOI: 10.1016/j.cell.2023.06.009] [Show More Authors] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/24/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023]
Abstract
Elucidating the cellular organization of the cerebral cortex is critical for understanding brain structure and function. Using large-scale single-nucleus RNA sequencing and spatial transcriptomic analysis of 143 macaque cortical regions, we obtained a comprehensive atlas of 264 transcriptome-defined cortical cell types and mapped their spatial distribution across the entire cortex. We characterized the cortical layer and region preferences of glutamatergic, GABAergic, and non-neuronal cell types, as well as regional differences in cell-type composition and neighborhood complexity. Notably, we discovered a relationship between the regional distribution of various cell types and the region's hierarchical level in the visual and somatosensory systems. Cross-species comparison of transcriptomic data from human, macaque, and mouse cortices further revealed primate-specific cell types that are enriched in layer 4, with their marker genes expressed in a region-dependent manner. Our data provide a cellular and molecular basis for understanding the evolution, development, aging, and pathogenesis of the primate brain.
Collapse
Affiliation(s)
- Ao Chen
- BGI-Shenzhen, Shenzhen 518103, China; Department of Biology, University of Copenhagen, Copenhagen 2200, Denmark; BGI Research-Southwest, BGI, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Yidi Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Ying Lei
- BGI-Shenzhen, Shenzhen 518103, China; BGI-Hangzhou, Hangzhou 310012, China
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sha Liao
- BGI-Shenzhen, Shenzhen 518103, China; BGI Research-Southwest, BGI, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Juan Meng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yiqin Bai
- Lingang Laboratory, Shanghai 200031, China
| | - Zhen 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
| | - Zhifeng Liang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Nini Yuan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zihan Wu
- Tencent AI Lab, Shenzhen 518057, China
| | - Feng Lin
- BGI-Shenzhen, Shenzhen 518103, China
| | - Kexin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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-Shenzhen, Shenzhen 518103, China
| | - Shuzhen Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Tianyi Fei
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenkun Zhuang
- BGI-Shenzhen, Shenzhen 518103, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yiming Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yong Zhang
- BGI-Shenzhen, Shenzhen 518103, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yuanfang Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luman Cui
- BGI-Shenzhen, Shenzhen 518103, China
| | - Ruiyi Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lei Han
- BGI-Shenzhen, Shenzhen 518103, China
| | - Xing Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | - Baoqian Huangfu
- BGI-Shenzhen, Shenzhen 518103, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | | | - Jianyun Ma
- 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
| | - Zhao Li
- BGI-Shenzhen, Shenzhen 518103, China
| | - Yikun Lin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanqing Zhong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huifang Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yaqian Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xing Liu
- BGI-Shenzhen, Shenzhen 518103, China
| | - Jian Peng
- BGI-Shenzhen, Shenzhen 518103, China
| | | | - Wei Chen
- BGI-Shenzhen, Shenzhen 518103, China
| | | | - Yingjie An
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shihui Xia
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingli Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuai Liu
- BGI-Shenzhen, Shenzhen 518103, China
| | | | - Chun Gong
- BGI-Shenzhen, Shenzhen 518103, China; China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Xin Huang
- BGI-Shenzhen, Shenzhen 518103, China
| | - Yue Yuan
- BGI-Shenzhen, Shenzhen 518103, China
| | - Yun Zhao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qinwen Chai
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, 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 Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shengkang Li
- BGI-Shenzhen, Shenzhen 518103, China; Guangdong Bigdata Engineering Technology Research Center for Life Sciences, Shenzhen 518083, China
| | | | - Yan Hong
- BGI-Shenzhen, Shenzhen 518103, China
| | | | - Min Li
- BGI-Shenzhen, Shenzhen 518103, China
| | - Mengmeng Jin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Gao
- 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
| | - Yinqi Bai
- BGI-Shenzhen, Shenzhen 518103, China
| | | | - Guohai Hu
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Shiping Liu
- BGI-Shenzhen, Shenzhen 518103, China; BGI-Hangzhou, Hangzhou 310012, China
| | - Bo Wang
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Bin Xiang
- 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
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huanhuan Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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 Neuroscience, 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 Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Minglong Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xin Liu
- BGI-Shenzhen, Shenzhen 518103, China
| | - Qian Zhao
- BGI-Shenzhen, Shenzhen 518103, China
| | - Michael Lisby
- Department of Biology, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jing Wang
- BGI-Shenzhen, Shenzhen 518103, China
| | - Jiao Fang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun Lin
- BGI-Shenzhen, Shenzhen 518103, China
| | - Qing Xie
- BGI-Shenzhen, Shenzhen 518103, China
| | - Zhen Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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
| | - Jie He
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huatai Xu
- Lingang Laboratory, Shanghai 200031, China
| | - Wei Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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
| | | | - Yangang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mathias Uhlen
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
| | - Muming Poo
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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
| | - Jian Wang
- BGI-Shenzhen, Shenzhen 518103, China; China National GeneBank, BGI-Shenzhen, Shenzhen 518120, 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.
| | - Yuxiang Li
- BGI-Shenzhen, Shenzhen 518103, China; BGI Research-Wuhan, BGI, Wuhan 430074, China.
| | - Zhiming Shen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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.
| | - Longqi Liu
- BGI-Shenzhen, Shenzhen 518103, China; BGI-Hangzhou, Hangzhou 310012, China.
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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.
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518103, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen 518120, China.
| | - Chengyu Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, 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; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
124
|
Tang Z, Liu X, Li Z, Zhang T, Yang B, Su J, Song Q. SpaRx: Elucidate single-cell spatial heterogeneity of drug responses for personalized treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551911. [PMID: 37577665 PMCID: PMC10418183 DOI: 10.1101/2023.08.03.551911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Spatial cellular heterogeneity contributes to differential drug responses in a tumor lesion and potential therapeutic resistance. Recent emerging spatial technologies such as CosMx SMI, MERSCOPE, and Xenium delineate the spatial gene expression patterns at the single cell resolution. This provides unprecedented opportunities to identify spatially localized cellular resistance and to optimize the treatment for individual patients. In this work, we present a graph-based domain adaptation model, SpaRx, to reveal the heterogeneity of spatial cellular response to drugs. SpaRx transfers the knowledge from pharmacogenomics profiles to single-cell spatial transcriptomics data, through hybrid learning with dynamic adversarial adaption. Comprehensive benchmarking demonstrates the superior and robust performance of SpaRx at different dropout rates, noise levels, and transcriptomics coverage. Further application of SpaRx to the state-of-art single-cell spatial transcriptomics data reveals that tumor cells in different locations of a tumor lesion present heterogenous sensitivity or resistance to drugs. Moreover, resistant tumor cells interact with themselves or the surrounding constituents to form an ecosystem for drug resistance. Collectively, SpaRx characterizes the spatial therapeutic variability, unveils the molecular mechanisms underpinning drug resistance, and identifies personalized drug targets and effective drug combinations. Key Points We have developed a novel graph-based domain adaption model named SpaRx, to reveal the heterogeneity of spatial cellular response to different types of drugs, which bridges the gap between pharmacogenomics knowledgebase and single-cell spatial transcriptomics data.SpaRx is developed tailored for single-cell spatial transcriptomics data and is provided available as a ready-to-use open-source software, which demonstrates high accuracy and robust performance.SpaRx uncovers that tumor cells located in different areas within tumor lesion exhibit varying levels of sensitivity or resistance to drugs. Moreover, SpaRx reveals that tumor cells interact with themselves and the surrounding microenvironment to form an ecosystem capable of drug resistance.
Collapse
Affiliation(s)
- Ziyang Tang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Xiang Liu
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Zuotian Li
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
- Department of Computer Graphics Technology, Purdue University, Indiana, USA
| | - Tonglin Zhang
- Department of Statistics, Purdue University, Indiana, USA
| | - Baijian Yang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Qianqian Song
- Center for Cancer Genomics and Precision Oncology, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, North Carolina, USA
- Department of Cancer Biology, Wake Forest University School of Medicine, North Carolina, USA
| |
Collapse
|
125
|
Huilgol D, Russ JB, Srivas S, Huang ZJ. The progenitor basis of cortical projection neuron diversity. Curr Opin Neurobiol 2023; 81:102726. [PMID: 37148649 PMCID: PMC10557529 DOI: 10.1016/j.conb.2023.102726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/04/2023] [Accepted: 04/09/2023] [Indexed: 05/08/2023]
Abstract
Diverse glutamatergic projection neurons (PNs) mediate myriad processing streams and output channels of the cerebral cortex. Yet, how different types of neural progenitors, such as radial glia (RGs) and intermediate progenitors (IPs), produce PN diversity, and hierarchical organization remains unclear. A fundamental issue is whether RGs constitute a homogeneous, multipotent lineage capable of generating all major PN types through a temporally regulated developmental program, or whether RGs comprise multiple transcriptionally heterogenous pools, each fated to generate a subset of PNs. Beyond RGs, the role of IPs in PN diversification remains underexplored. Addressing these questions requires tracking PN developmental trajectories with cell-type resolution - from transcription factor-defined RGs and IPs to their PN progeny, which are defined not only by laminar location but also by projection patterns and gene expression. Advances in cell-type resolution genetic fate mapping, axon tracing, and spatial transcriptomics may provide the technical capability for answering these fundamental questions.
Collapse
Affiliation(s)
- Dhananjay Huilgol
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeffrey B Russ
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Division of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sweta Srivas
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Z Josh Huang
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC 27708, USA.
| |
Collapse
|
126
|
Chen H, Li D, Bar-Joseph Z. SCS: cell segmentation for high-resolution spatial transcriptomics. Nat Methods 2023; 20:1237-1243. [PMID: 37429992 DOI: 10.1038/s41592-023-01939-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/02/2023] [Indexed: 07/12/2023]
Abstract
Spatial transcriptomics promises to greatly improve our understanding of tissue organization and cell-cell interactions. While most current platforms for spatial transcriptomics only offer multi-cellular resolution, with 10-15 cells per spot, recent technologies provide a much denser spot placement leading to subcellular resolution. A key challenge for these newer methods is cell segmentation and the assignment of spots to cells. Traditional image-based segmentation methods are limited and do not make full use of the information profiled by spatial transcriptomics. Here we present subcellular spatial transcriptomics cell segmentation (SCS), which combines imaging data with sequencing data to improve cell segmentation accuracy. SCS assigns spots to cells by adaptively learning the position of each spot relative to the center of its cell using a transformer neural network. SCS was tested on two new subcellular spatial transcriptomics technologies and outperformed traditional image-based segmentation methods. SCS achieved better accuracy, identified more cells and provided more realistic cell size estimation. Subcellular analysis of RNAs using SCS spot assignments provides information on RNA localization and further supports the segmentation results.
Collapse
Affiliation(s)
- Hao Chen
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Dongshunyi Li
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ziv Bar-Joseph
- Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA.
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
127
|
Cerneckis J, Bu G, Shi Y. Pushing the boundaries of brain organoids to study Alzheimer's disease. Trends Mol Med 2023; 29:659-672. [PMID: 37353408 PMCID: PMC10374393 DOI: 10.1016/j.molmed.2023.05.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 06/25/2023]
Abstract
Progression of Alzheimer's disease (AD) entails deterioration or aberrant function of multiple brain cell types, eventually leading to neurodegeneration and cognitive decline. Defining how complex cell-cell interactions become dysregulated in AD requires novel human cell-based in vitro platforms that could recapitulate the intricate cytoarchitecture and cell diversity of the human brain. Brain organoids (BOs) are 3D self-organizing tissues that partially resemble the human brain architecture and can recapitulate AD-relevant pathology. In this review, we highlight the versatile applications of different types of BOs to model AD pathogenesis, including amyloid-β and tau aggregation, neuroinflammation, myelin breakdown, vascular dysfunction, and other phenotypes, as well as to accelerate therapeutic development for AD.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Guojun Bu
- SciNeuro Pharmaceuticals, Rockville, MD 20850, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA; Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA.
| |
Collapse
|
128
|
|
129
|
Hong Y, Song K, Zhang Z, Deng Y, Zhang X, Zhao J, Jiang J, Zhang Q, Guo C, Peng C. The spatiotemporal dynamics of spatially variable genes in developing mouse brain revealed by a novel computational scheme. Cell Death Discov 2023; 9:264. [PMID: 37500639 PMCID: PMC10374563 DOI: 10.1038/s41420-023-01569-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
To understand how brain regions form and work, it is important to explore the spatially variable genes (SVGs) enriched in specific brain regions during development. Spatial transcriptomics techniques provide opportunity to select SVGs in the high-throughput way. However, previous methods neglected the ranking order and combinatorial effect of SVGs, making them difficult to automatically select the high-priority SVGs from spatial transcriptomics data. Here, we proposed a novel computational pipeline, called SVGbit, to rank the individual and combinatorial SVGs for marker selection in various brain regions, which was tested in different kinds of public datasets for both human and mouse brains. We then generated the spatial transcriptomics and immunohistochemistry data from mouse brain at critical embryonic and neonatal stages. The results show that our ranking and clustering scheme captures the key SVGs which coincide with known anatomic regions in the developing mouse brain. More importantly, SVGbit can facilitate the identification of multiple gene combination sets in different brain regions. We identified three dynamical sub-regions which can be segregated by the staining of Sox2 and Calb2 in thalamus, and we also found that Nr4a2 expression gradually segregates the neocortex and hippocampus during the development. In summary, our work not only reveals the spatiotemporal dynamics of individual and combinatorial SVGs in developing mouse brain, but also provides a novel computational pipeline to facilitate the selection of marker genes from spatial transcriptomics data.
Collapse
Affiliation(s)
- Yingzhou Hong
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Kai Song
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Zongbo Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Yuxia Deng
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Xue Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Jinqian Zhao
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Jun Jiang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Qing Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Chunming Guo
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| | - Cheng Peng
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
130
|
Fu XD, Mobley WC. Therapeutic Potential of PTB Inhibition Through Converting Glial Cells to Neurons in the Brain. Annu Rev Neurosci 2023; 46:145-165. [PMID: 37428606 DOI: 10.1146/annurev-neuro-083022-113120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Cell replacement therapy represents a promising approach for treating neurodegenerative diseases. Contrary to the common addition strategy to generate new neurons from glia by overexpressing a lineage-specific transcription factor(s), a recent study introduced a subtraction strategy by depleting a single RNA-binding protein, Ptbp1, to convert astroglia to neurons not only in vitro but also in the brain. Given its simplicity, multiple groups have attempted to validate and extend this attractive approach but have met with difficulty in lineage tracing newly induced neurons from mature astrocytes, raising the possibility of neuronal leakage as an alternative explanation for apparent astrocyte-to-neuron conversion. This review focuses on the debate over this critical issue. Importantly, multiple lines of evidence suggest that Ptbp1 depletion can convert a selective subpopulation of glial cells into neurons and, via this and other mechanisms, reverse deficits in a Parkinson's disease model, emphasizing the importance of future efforts in exploring this therapeutic strategy.
Collapse
Affiliation(s)
- Xiang-Dong Fu
- Westlake Laboratory of Life Sciences and Biomedicine, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China;
| | - William C Mobley
- Department of Neuroscience, University of California, San Diego, La Jolla, California, USA;
| |
Collapse
|
131
|
Fangma Y, Liu M, Liao J, Chen Z, Zheng Y. Dissecting the brain with spatially resolved multi-omics. J Pharm Anal 2023; 13:694-710. [PMID: 37577383 PMCID: PMC10422112 DOI: 10.1016/j.jpha.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 08/15/2023] Open
Abstract
Recent studies have highlighted spatially resolved multi-omics technologies, including spatial genomics, transcriptomics, proteomics, and metabolomics, as powerful tools to decipher the spatial heterogeneity of the brain. Here, we focus on two major approaches in spatial transcriptomics (next-generation sequencing-based technologies and image-based technologies), and mass spectrometry imaging technologies used in spatial proteomics and spatial metabolomics. Furthermore, we discuss their applications in neuroscience, including building the brain atlas, uncovering gene expression patterns of neurons for special behaviors, deciphering the molecular basis of neuronal communication, and providing a more comprehensive explanation of the molecular mechanisms underlying central nervous system disorders. However, further efforts are still needed toward the integrative application of multi-omics technologies, including the real-time spatial multi-omics analysis in living cells, the detailed gene profile in a whole-brain view, and the combination of functional verification.
Collapse
Affiliation(s)
- Yijia Fangma
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengting Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jie Liao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
132
|
Zhang Z, Wiencke JK, Kelsey KT, Koestler DC, Molinaro AM, Pike SC, Karra P, Christensen BC, Salas LA. Hierarchical deconvolution for extensive cell type resolution in the human brain using DNA methylation. Front Neurosci 2023; 17:1198243. [PMID: 37404460 PMCID: PMC10315586 DOI: 10.3389/fnins.2023.1198243] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction The human brain comprises heterogeneous cell types whose composition can be altered with physiological and pathological conditions. New approaches to discern the diversity and distribution of brain cells associated with neurological conditions would significantly advance the study of brain-related pathophysiology and neuroscience. Unlike single-nuclei approaches, DNA methylation-based deconvolution does not require special sample handling or processing, is cost-effective, and easily scales to large study designs. Existing DNA methylation-based methods for brain cell deconvolution are limited in the number of cell types deconvolved. Methods Using DNA methylation profiles of the top cell-type-specific differentially methylated CpGs, we employed a hierarchical modeling approach to deconvolve GABAergic neurons, glutamatergic neurons, astrocytes, microglial cells, oligodendrocytes, endothelial cells, and stromal cells. Results We demonstrate the utility of our method by applying it to data on normal tissues from various brain regions and in aging and diseased tissues, including Alzheimer's disease, autism, Huntington's disease, epilepsy, and schizophrenia. Discussion We expect that the ability to determine the cellular composition in the brain using only DNA from bulk samples will accelerate understanding brain cell type composition and cell-type-specific epigenetic states in normal and diseased brain tissues.
Collapse
Affiliation(s)
- Ze Zhang
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - John K. Wiencke
- Department of Neurological Surgery, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States
| | - Karl T. Kelsey
- Department of Epidemiology, Department of Pathology and Laboratory Medicine, Brown University School of Public Health, Providence, RI, United States
| | - Devin C. Koestler
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, United States
| | - Annette M. Molinaro
- Department of Neurological Surgery, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, United States
| | - Steven C. Pike
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
- Department of Neurology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Prasoona Karra
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Brock C. Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| | - Lucas A. Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, United States
| |
Collapse
|
133
|
Chen H, Li D, Bar-Joseph Z. SCS: cell segmentation for high-resolution spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523658. [PMID: 37398213 PMCID: PMC10312435 DOI: 10.1101/2023.01.11.523658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Spatial transcriptomics promises to greatly improve our understanding of tissue organization and cell-cell interactions. While most current platforms for spatial transcriptomics only offer multi-cellular resolution, with 10-15 cells per spot, recent technologies provide a much denser spot placement leading to sub-cellular resolution. A key challenge for these newer methods is cell segmentation and the assignment of spots to cells. Traditional image-based segmentation methods are limited and do not make full use of the information profiled by spatial transcrip-tomics. Here we present SCS, which combines imaging data with sequencing data to improve cell segmentation accuracy. SCS assigns spots to cells by adaptively learning the position of each spot relative to the center of its cell using a transformer neural network. SCS was tested on two new sub-cellular spatial transcriptomics technologies and outperformed traditional image-based segmentation methods. SCS achieved better accuracy, identified more cells, and provided more realistic cell size estimation. Sub-cellular analysis of RNAs using SCS spots assignments provides information on RNA localization and further supports the segmentation results.
Collapse
|
134
|
Zhang Y, Miller JA, Park J, Lelieveldt BP, Long B, Abdelaal T, Aevermann BD, Biancalani T, Comiter C, Dzyubachyk O, Eggermont J, Langseth CM, Petukhov V, Scalia G, Vaishnav ED, Zhao Y, Lein ES, Scheuermann RH. Reference-based cell type matching of in situ image-based spatial transcriptomics data on primary visual cortex of mouse brain. Sci Rep 2023; 13:9567. [PMID: 37311768 DOI: 10.1038/s41598-023-36638-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
With the advent of multiplex fluorescence in situ hybridization (FISH) and in situ RNA sequencing technologies, spatial transcriptomics analysis is advancing rapidly, providing spatial location and gene expression information about cells in tissue sections at single cell resolution. Cell type classification of these spatially-resolved cells can be inferred by matching the spatial transcriptomics data to reference atlases derived from single cell RNA-sequencing (scRNA-seq) in which cell types are defined by differences in their gene expression profiles. However, robust cell type matching of the spatially-resolved cells to reference scRNA-seq atlases is challenging due to the intrinsic differences in resolution between the spatial and scRNA-seq data. In this study, we systematically evaluated six computational algorithms for cell type matching across four image-based spatial transcriptomics experimental protocols (MERFISH, smFISH, BaristaSeq, and ExSeq) conducted on the same mouse primary visual cortex (VISp) brain region. We find that many cells are assigned as the same type by multiple cell type matching algorithms and are present in spatial patterns previously reported from scRNA-seq studies in VISp. Furthermore, by combining the results of individual matching strategies into consensus cell type assignments, we see even greater alignment with biological expectations. We present two ensemble meta-analysis strategies used in this study and share the consensus cell type matching results in the Cytosplore Viewer ( https://viewer.cytosplore.org ) for interactive visualization and data exploration. The consensus matching can also guide spatial data analysis using SSAM, allowing segmentation-free cell type assignment.
Collapse
Affiliation(s)
- Yun Zhang
- J. Craig Venter Institute, La Jolla, CA, USA
| | | | - Jeongbin Park
- School of Biomedical Convergence Engineering, Pusan National University, Busan, Korea
| | - Boudewijn P Lelieveldt
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, The Netherlands
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tamim Abdelaal
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Pattern Recognition and Bioinformatics Group, Delft University of Technology, Delft, The Netherlands
| | - Brian D Aevermann
- J. Craig Venter Institute, La Jolla, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Tommaso Biancalani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | | | - Oleh Dzyubachyk
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen Eggermont
- LKEB, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Viktor Petukhov
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Gabriele Scalia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | | | - Yilin Zhao
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Richard H Scheuermann
- J. Craig Venter Institute, La Jolla, CA, USA.
- Department of Pathology, University of California, San Diego, CA, USA.
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA.
| |
Collapse
|
135
|
Arioka Y, Okumura H, Sakaguchi H, Ozaki N. Shedding light on latent pathogenesis and pathophysiology of mental disorders: The potential of iPS cell technology. Psychiatry Clin Neurosci 2023; 77:308-314. [PMID: 36929185 PMCID: PMC11488641 DOI: 10.1111/pcn.13545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
Mental disorders are considered as one of the major healthcare issues worldwide owing to their significant impact on the quality of life of patients, causing serious social burdens. However, it is hard to examine the living brain-a source of psychiatric symptoms-at the cellular, subcellular, and molecular levels, which poses difficulty in determining the pathogenesis and pathophysiology of mental disorders. Recently, induced pluripotent stem cell (iPSC) technology has been used as a novel tool for research on mental disorders. We believe that the iPSC-based studies will address the limitations of other research approaches, such as human genome, postmortem brain study, brain imaging, and animal model analysis. Notably, studies using integrated iPSC technology with genetic information have provided significant novel findings to date. This review aimed to discuss the history, current trends, potential, and future of iPSC technology in the field of mental disorders. Although iPSC technology has several limitations, this technology can be used in combination with the other approaches to facilitate studies on mental disorders.
Collapse
Affiliation(s)
- Yuko Arioka
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Center for Advanced Medicine and Clinical ResearchNagoya University HospitalNagoyaJapan
| | - Hiroki Okumura
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Hospital PharmacyNagoya University HospitalNagoyaJapan
| | - Hideya Sakaguchi
- BDR‐Otsuka Pharmaceutical Collaboration Center, RIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Norio Ozaki
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityNagoyaJapan
| |
Collapse
|
136
|
Gabitto MI, Travaglini KJ, Rachleff VM, Kaplan ES, Long B, Ariza J, Ding Y, Mahoney JT, Dee N, Goldy J, Melief EJ, Brouner K, Campos J, Carr AJ, Casper T, Chakrabarty R, Clark M, Compos J, Cool J, Valera Cuevas NJ, Dalley R, Darvas M, Ding SL, Dolbeare T, Mac Donald CL, Egdorf T, Esposito L, Ferrer R, Gala R, Gary A, Gloe J, Guilford N, Guzman J, Ho W, Jarksy T, Johansen N, Kalmbach BE, Keene LM, Khawand S, Kilgore M, Kirkland A, Kunst M, Lee BR, Malone J, Maltzer Z, Martin N, McCue R, McMillen D, Meyerdierks E, Meyers KP, Mollenkopf T, Montine M, Nolan AL, Nyhus J, Olsen PA, Pacleb M, Pham T, Pom CA, Postupna N, Ruiz A, Schantz AM, Sorensen SA, Staats B, Sullivan M, Sunkin SM, Thompson C, Tieu M, Ting J, Torkelson A, Tran T, Wang MQ, Waters J, Wilson AM, Haynor D, Gatto N, Jayadev S, Mufti S, Ng L, Mukherjee S, Crane PK, Latimer CS, Levi BP, Smith K, Close JL, Miller JA, Hodge RD, Larson EB, Grabowski TJ, Hawrylycz M, Keene CD, Lein ES. Integrated multimodal cell atlas of Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-2921860. [PMID: 37292694 PMCID: PMC10246227 DOI: 10.21203/rs.3.rs-2921860/v1] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in older adults. Neuropathological and imaging studies have demonstrated a progressive and stereotyped accumulation of protein aggregates, but the underlying molecular and cellular mechanisms driving AD progression and vulnerable cell populations affected by disease remain coarsely understood. The current study harnesses single cell and spatial genomics tools and knowledge from the BRAIN Initiative Cell Census Network to understand the impact of disease progression on middle temporal gyrus cell types. We used image-based quantitative neuropathology to place 84 donors spanning the spectrum of AD pathology along a continuous disease pseudoprogression score and multiomic technologies to profile single nuclei from each donor, mapping their transcriptomes, epigenomes, and spatial coordinates to a common cell type reference with unprecedented resolution. Temporal analysis of cell-type proportions indicated an early reduction of Somatostatin-expressing neuronal subtypes and a late decrease of supragranular intratelencephalic-projecting excitatory and Parvalbumin-expressing neurons, with increases in disease-associated microglial and astrocytic states. We found complex gene expression differences, ranging from global to cell type-specific effects. These effects showed different temporal patterns indicating diverse cellular perturbations as a function of disease progression. A subset of donors showed a particularly severe cellular and molecular phenotype, which correlated with steeper cognitive decline. We have created a freely available public resource to explore these data and to accelerate progress in AD research at SEA-AD.org.
Collapse
Affiliation(s)
| | | | - Victoria M. Rachleff
- Allen Institute for Brain Science, Seattle, WA, 98109
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | | | - Brian Long
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Jeanelle Ariza
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Yi Ding
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Erica J. Melief
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | | | - John Campos
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | | | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Michael Clark
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Jazmin Compos
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Jonah Cool
- Chan Zuckerberg Initiative, Redwood City, CA 94063
| | | | - Rachel Dalley
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Martin Darvas
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Song-Lin Ding
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Tim Dolbeare
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Tom Egdorf
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Luke Esposito
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Rohan Gala
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | | | - Windy Ho
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Tim Jarksy
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | | | - Lisa M. Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Sarah Khawand
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Mitch Kilgore
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Amanda Kirkland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Michael Kunst
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Brian R. Lee
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Zoe Maltzer
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Rachel McCue
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | | | - Kelly P. Meyers
- Kaiser Permanente Washington Research Institute, Seattle, WA, 98101
| | | | - Mark Montine
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Amber L. Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Paul A. Olsen
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Maiya Pacleb
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Thanh Pham
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Nadia Postupna
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Augustin Ruiz
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Aimee M. Schantz
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | | | - Brian Staats
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Matt Sullivan
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | | | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Jonathan Ting
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Amy Torkelson
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Tracy Tran
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Angela M. Wilson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - David Haynor
- Department of Radiology, University of Washington, Seattle, WA 98014
| | - Nicole Gatto
- Kaiser Permanente Washington Research Institute, Seattle, WA, 98101
| | - Suman Jayadev
- Department of Neurology, University of Washington, Seattle, WA 98104
| | - Shoaib Mufti
- Allen Institute for Brain Science, Seattle, WA, 98109
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | - Paul K. Crane
- Department of Medicine, University of Washington, Seattle, WA 98104
| | - Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Boaz P. Levi
- Allen Institute for Brain Science, Seattle, WA, 98109
| | | | | | | | | | - Eric B. Larson
- Department of Medicine, University of Washington, Seattle, WA 98104
| | | | | | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98104
| | - Ed S. Lein
- Allen Institute for Brain Science, Seattle, WA, 98109
| |
Collapse
|
137
|
Cadinu P, Sivanathan KN, Misra A, Xu RJ, Mangani D, Yang E, Rone JM, Tooley K, Kye YC, Bod L, Geistlinger L, Lee T, Ono N, Wang G, Sanmarco L, Quintana FJ, Anderson AC, Kuchroo VK, Moffitt JR, Nowarski R. Charting the cellular biogeography in colitis reveals fibroblast trajectories and coordinated spatial remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539701. [PMID: 37214800 PMCID: PMC10197602 DOI: 10.1101/2023.05.08.539701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Gut inflammation involves contributions from immune and non-immune cells, whose interactions are shaped by the spatial organization of the healthy gut and its remodeling during inflammation. The crosstalk between fibroblasts and immune cells is an important axis in this process, but our understanding has been challenged by incomplete cell-type definition and biogeography. To address this challenge, we used MERFISH to profile the expression of 940 genes in 1.35 million cells imaged across the onset and recovery from a mouse colitis model. We identified diverse cell populations; charted their spatial organization; and revealed their polarization or recruitment in inflammation. We found a staged progression of inflammation-associated tissue neighborhoods defined, in part, by multiple inflammation-associated fibroblasts, with unique expression profiles, spatial localization, cell-cell interactions, and healthy fibroblast origins. Similar signatures in ulcerative colitis suggest conserved human processes. Broadly, we provide a framework for understanding inflammation-induced remodeling in the gut and other tissues.
Collapse
Affiliation(s)
- Paolo Cadinu
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
- These authors contributed equally
| | - Kisha N. Sivanathan
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
- These authors contributed equally
| | - Aditya Misra
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Rosalind J. Xu
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138 USA
| | - Davide Mangani
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Evan Yang
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Joseph M. Rone
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Katherine Tooley
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Yoon-Chul Kye
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Lloyd Bod
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Ludwig Geistlinger
- Center for Computational Biomedicine, Harvard Medical School, Boston, MA 02115, USA
| | - Tyrone Lee
- Center for Computational Biomedicine, Harvard Medical School, Boston, MA 02115, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77030 USA
| | - Gang Wang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
| | - Liliana Sanmarco
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Francisco J. Quintana
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Ana C. Anderson
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Jeffrey R. Moffitt
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115 USA
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
| | - Roni Nowarski
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
- Lead contact
| |
Collapse
|
138
|
Andersen J, Thom N, Shadrach JL, Chen X, Onesto MM, Amin ND, Yoon SJ, Li L, Greenleaf WJ, Müller F, Pașca AM, Kaltschmidt JA, Pașca SP. Single-cell transcriptomic landscape of the developing human spinal cord. Nat Neurosci 2023; 26:902-914. [PMID: 37095394 DOI: 10.1038/s41593-023-01311-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Understanding spinal cord assembly is essential to elucidate how motor behavior is controlled and how disorders arise. The human spinal cord is exquisitely organized, and this complex organization contributes to the diversity and intricacy of motor behavior and sensory processing. But how this complexity arises at the cellular level in the human spinal cord remains unknown. Here we transcriptomically profiled the midgestation human spinal cord with single-cell resolution and discovered remarkable heterogeneity across and within cell types. Glia displayed diversity related to positional identity along the dorso-ventral and rostro-caudal axes, while astrocytes with specialized transcriptional programs mapped into white and gray matter subtypes. Motor neurons clustered at this stage into groups suggestive of alpha and gamma neurons. We also integrated our data with multiple existing datasets of the developing human spinal cord spanning 22 weeks of gestation to investigate the cell diversity over time. Together with mapping of disease-related genes, this transcriptomic mapping of the developing human spinal cord opens new avenues for interrogating the cellular basis of motor control in humans and guides human stem cell-based models of disease.
Collapse
Affiliation(s)
- Jimena Andersen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Nicholas Thom
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | | | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Massimo Mario Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Se-Jin Yoon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Li Li
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Fabian Müller
- Department of Genetics, Stanford University, Stanford, CA, USA
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Anca M Pașca
- Department of Pediatrics, Division of Neonatology, Stanford University, Stanford, CA, USA
| | | | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA.
| |
Collapse
|
139
|
Covert I, Gala R, Wang T, Svoboda K, Sümbül U, Lee SI. Predictive and robust gene selection for spatial transcriptomics. Nat Commun 2023; 14:2091. [PMID: 37045821 PMCID: PMC10097645 DOI: 10.1038/s41467-023-37392-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
A prominent trend in single-cell transcriptomics is providing spatial context alongside a characterization of each cell's molecular state. This typically requires targeting an a priori selection of genes, often covering less than 1% of the genome, and a key question is how to optimally determine the small gene panel. We address this challenge by introducing a flexible deep learning framework, PERSIST, to identify informative gene targets for spatial transcriptomics studies by leveraging reference scRNA-seq data. Using datasets spanning different brain regions, species, and scRNA-seq technologies, we show that PERSIST reliably identifies panels that provide more accurate prediction of the genome-wide expression profile, thereby capturing more information with fewer genes. PERSIST can be adapted to specific biological goals, and we demonstrate that PERSIST's binarization of gene expression levels enables models trained on scRNA-seq data to generalize with to spatial transcriptomics data, despite the complex shift between these technologies.
Collapse
Affiliation(s)
- Ian Covert
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Rohan Gala
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Wang
- HHMI Janelia Research Campus, Ashburn, VA, USA
| | - Karel Svoboda
- HHMI Janelia Research Campus, Ashburn, VA, USA
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Uygar Sümbül
- Allen Institute for Brain Science, Seattle, WA, USA.
| | - Su-In Lee
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
140
|
Zemke NR, Armand EJ, Wang W, Lee S, Zhou J, Li YE, Liu H, Tian W, Nery JR, Castanon RG, Bartlett A, Osteen JK, Li D, Zhuo X, Xu V, Miller M, Krienen FM, Zhang Q, Taskin N, Ting J, Feng G, McCarroll SA, Callaway EM, Wang T, Behrens MM, Lein ES, Ecker JR, Ren B. Comparative single cell epigenomic analysis of gene regulatory programs in the rodent and primate neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.08.536119. [PMID: 37066152 PMCID: PMC10104177 DOI: 10.1101/2023.04.08.536119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Sequence divergence of cis- regulatory elements drives species-specific traits, but how this manifests in the evolution of the neocortex at the molecular and cellular level remains to be elucidated. We investigated the gene regulatory programs in the primary motor cortex of human, macaque, marmoset, and mouse with single-cell multiomics assays, generating gene expression, chromatin accessibility, DNA methylome, and chromosomal conformation profiles from a total of over 180,000 cells. For each modality, we determined species-specific, divergent, and conserved gene expression and epigenetic features at multiple levels. We find that cell type-specific gene expression evolves more rapidly than broadly expressed genes and that epigenetic status at distal candidate cis -regulatory elements (cCREs) evolves faster than promoters. Strikingly, transposable elements (TEs) contribute to nearly 80% of the human-specific cCREs in cortical cells. Through machine learning, we develop sequence-based predictors of cCREs in different species and demonstrate that the genomic regulatory syntax is highly preserved from rodents to primates. Lastly, we show that epigenetic conservation combined with sequence similarity helps uncover functional cis -regulatory elements and enhances our ability to interpret genetic variants contributing to neurological disease and traits.
Collapse
|
141
|
Monné Rodríguez JM, Frisk AL, Kreutzer R, Lemarchand T, Lezmi S, Saravanan C, Stierstorfer B, Thuilliez C, Vezzali E, Wieczorek G, Yun SW, Schaudien D. European Society of Toxicologic Pathology (Pathology 2.0 Molecular Pathology Special Interest Group): Review of In Situ Hybridization Techniques for Drug Research and Development. Toxicol Pathol 2023; 51:92-111. [PMID: 37449403 PMCID: PMC10467011 DOI: 10.1177/01926233231178282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
In situ hybridization (ISH) is used for the localization of specific nucleic acid sequences in cells or tissues by complementary binding of a nucleotide probe to a specific target nucleic acid sequence. In the last years, the specificity and sensitivity of ISH assays were improved by innovative techniques like synthetic nucleic acids and tandem oligonucleotide probes combined with signal amplification methods like branched DNA, hybridization chain reaction and tyramide signal amplification. These improvements increased the application spectrum for ISH on formalin-fixed paraffin-embedded tissues. ISH is a powerful tool to investigate DNA, mRNA transcripts, regulatory noncoding RNA, and therapeutic oligonucleotides. ISH can be used to obtain spatial information of a cell type, subcellular localization, or expression levels of targets. Since immunohistochemistry and ISH share similar workflows, their combination can address simultaneous transcriptomics and proteomics questions. The goal of this review paper is to revisit the current state of the scientific approaches in ISH and its application in drug research and development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Seong-Wook Yun
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| |
Collapse
|
142
|
Zhang D, Deng Y, Kukanja P, Agirre E, Bartosovic M, Dong M, Ma C, Ma S, Su G, Bao S, Liu Y, Xiao Y, Rosoklija GB, Dwork AJ, Mann JJ, Leong KW, Boldrini M, Wang L, Haeussler M, Raphael BJ, Kluger Y, Castelo-Branco G, Fan R. Spatial epigenome-transcriptome co-profiling of mammalian tissues. Nature 2023; 616:113-122. [PMID: 36922587 PMCID: PMC10076218 DOI: 10.1038/s41586-023-05795-1] [Citation(s) in RCA: 171] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 02/03/2023] [Indexed: 03/17/2023]
Abstract
Emerging spatial technologies, including spatial transcriptomics and spatial epigenomics, are becoming powerful tools for profiling of cellular states in the tissue context1-5. However, current methods capture only one layer of omics information at a time, precluding the possibility of examining the mechanistic relationship across the central dogma of molecular biology. Here, we present two technologies for spatially resolved, genome-wide, joint profiling of the epigenome and transcriptome by cosequencing chromatin accessibility and gene expression, or histone modifications (H3K27me3, H3K27ac or H3K4me3) and gene expression on the same tissue section at near-single-cell resolution. These were applied to embryonic and juvenile mouse brain, as well as adult human brain, to map how epigenetic mechanisms control transcriptional phenotype and cell dynamics in tissue. Although highly concordant tissue features were identified by either spatial epigenome or spatial transcriptome we also observed distinct patterns, suggesting their differential roles in defining cell states. Linking epigenome to transcriptome pixel by pixel allows the uncovering of new insights in spatial epigenetic priming, differentiation and gene regulation within the tissue architecture. These technologies are of great interest in life science and biomedical research.
Collapse
Affiliation(s)
- Di Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yanxiang Deng
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Department of Pathology and Laboratory Medicine, Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Marek Bartosovic
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mingze Dong
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Sai Ma
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Shuozhen Bao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yang Liu
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Gorazd B Rosoklija
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, USA
- Macedonian Academy of Sciences & Arts, Skopje, Republic of Macedonia
| | - Andrew J Dwork
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, USA
- Macedonian Academy of Sciences & Arts, Skopje, Republic of Macedonia
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, USA
- Department of Radiology, Columbia University, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maura Boldrini
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, USA
| | - Liya Wang
- AtlasXomics, Inc., New Haven, CT, USA
| | | | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Yuval Kluger
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Applied Mathematics Program, Yale University, New Haven, CT, USA
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
143
|
Hippenmeyer S. Principles of neural stem cell lineage progression: Insights from developing cerebral cortex. Curr Opin Neurobiol 2023; 79:102695. [PMID: 36842274 DOI: 10.1016/j.conb.2023.102695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/18/2023] [Accepted: 01/29/2023] [Indexed: 02/28/2023]
Abstract
How to generate a brain of correct size and with appropriate cell-type diversity during development is a major question in Neuroscience. In the developing neocortex, radial glial progenitor (RGP) cells are the main neural stem cells that produce cortical excitatory projection neurons, glial cells, and establish the prospective postnatal stem cell niche in the lateral ventricles. RGPs follow a tightly orchestrated developmental program that when disrupted can result in severe cortical malformations such as microcephaly and megalencephaly. The precise cellular and molecular mechanisms instructing faithful RGP lineage progression are however not well understood. This review will summarize recent conceptual advances that contribute to our understanding of the general principles of RGP lineage progression.
Collapse
Affiliation(s)
- Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
144
|
Fair T, Pollen AA. Genetic architecture of human brain evolution. Curr Opin Neurobiol 2023; 80:102710. [PMID: 37003107 DOI: 10.1016/j.conb.2023.102710] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/20/2023] [Accepted: 02/26/2023] [Indexed: 04/03/2023]
Abstract
Comparative studies of hominids have long sought to identify mutational events that shaped the evolution of the human nervous system. However, functional genetic differences are outnumbered by millions of nearly neutral mutations, and the developmental mechanisms underlying human nervous system specializations are difficult to model and incompletely understood. Candidate-gene studies have attempted to map select human-specific genetic differences to neurodevelopmental functions, but it remains unclear how to contextualize the relative effects of genes that are investigated independently. Considering these limitations, we discuss scalable approaches for probing the functional contributions of human-specific genetic differences. We propose that a systems-level view will enable a more quantitative and integrative understanding of the genetic, molecular and cellular underpinnings of human nervous system evolution.
Collapse
Affiliation(s)
- Tyler Fair
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA. https://twitter.com/@TylerFair_
| | - Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
145
|
Luria V, Ma S, Shibata M, Pattabiraman K, Sestan N. Molecular and cellular mechanisms of human cortical connectivity. Curr Opin Neurobiol 2023; 80:102699. [PMID: 36921362 DOI: 10.1016/j.conb.2023.102699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 02/05/2023] [Indexed: 03/18/2023]
Abstract
Comparative studies of the cerebral cortex have identified various human and primate-specific changes in both local and long-range connectivity, which are thought to underlie our advanced cognitive capabilities. These changes are likely mediated by the divergence of spatiotemporal regulation of gene expression, which is particularly prominent in the prenatal and early postnatal human and non-human primate cerebral cortex. In this review, we describe recent advances in characterizing human and primate genetic and cellular innovations including identification of novel species-specific, especially human-specific, genes, gene expression patterns, and cell types. Finally, we highlight three recent studies linking these molecular changes to reorganization of cortical connectivity.
Collapse
Affiliation(s)
- Victor Luria
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Mikihito Shibata
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Kartik Pattabiraman
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA.
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA; Departments of Psychiatry, Genetics and Comparative Medicine, Program in Cellular Neuroscience, Neurodegeneration and Repair, and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
146
|
Zhang M, Pan X, Jung W, Halpern A, Eichhorn SW, Lei Z, Cohen L, Smith KA, Tasic B, Yao Z, Zeng H, Zhuang X. A molecularly defined and spatially resolved cell atlas of the whole mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531348. [PMID: 36945367 PMCID: PMC10028822 DOI: 10.1101/2023.03.06.531348] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
In mammalian brains, tens of millions to billions of cells form complex interaction networks to enable a wide range of functions. The enormous diversity and intricate organization of cells in the brain have so far hindered our understanding of the molecular and cellular basis of its functions. Recent advances in spatially resolved single-cell transcriptomics have allowed systematic mapping of the spatial organization of molecularly defined cell types in complex tissues1-3. However, these approaches have only been applied to a few brain regions1-11 and a comprehensive cell atlas of the whole brain is still missing. Here, we imaged a panel of >1,100 genes in ~8 million cells across the entire adult mouse brain using multiplexed error-robust fluorescence in situ hybridization (MERFISH)12 and performed spatially resolved, single-cell expression profiling at the whole-transcriptome scale by integrating MERFISH and single-cell RNA-sequencing (scRNA-seq) data. Using this approach, we generated a comprehensive cell atlas of >5,000 transcriptionally distinct cell clusters, belonging to ~300 major cell types, in the whole mouse brain with high molecular and spatial resolution. Registration of the MERFISH images to the common coordinate framework (CCF) of the mouse brain further allowed systematic quantifications of the cell composition and organization in individual brain regions defined in the CCF. We further identified spatial modules characterized by distinct cell-type compositions and spatial gradients featuring gradual changes in the gene-expression profiles of cells. Finally, this high-resolution spatial map of cells, with a transcriptome-wide expression profile associated with each cell, allowed us to infer cell-type-specific interactions between several hundred pairs of molecularly defined cell types and predict potential molecular (ligand-receptor) basis and functional implications of these cell-cell interactions. These results provide rich insights into the molecular and cellular architecture of the brain and a valuable resource for future functional investigations of neural circuits and their dysfunction in diseases.
Collapse
Affiliation(s)
- Meng Zhang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- These authors contributed equally
| | - Xingjie Pan
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- These authors contributed equally
| | - Won Jung
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
- These authors contributed equally
| | - Aaron Halpern
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Stephen W. Eichhorn
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Zhiyun Lei
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | - Limor Cohen
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| | | | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
147
|
Yuan Z, Pan W, Zhao X, Zhao F, Xu Z, Li X, Zhao Y, Zhang MQ, Yao J. SODB facilitates comprehensive exploration of spatial omics data. Nat Methods 2023; 20:387-399. [PMID: 36797409 DOI: 10.1038/s41592-023-01773-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/06/2023] [Indexed: 02/18/2023]
Abstract
Spatial omics technologies generate wealthy but highly complex datasets. Here we present Spatial Omics DataBase (SODB), a web-based platform providing both rich data resources and a suite of interactive data analytical modules. SODB currently maintains >2,400 experiments from >25 spatial omics technologies, which are freely accessible as a unified data format compatible with various computational packages. SODB also provides multiple interactive data analytical modules, especially a unique module, Spatial Omics View (SOView). We conduct comprehensive statistical analyses and illustrate the utility of both basic and advanced analytical modules using multiple spatial omics datasets. We demonstrate SOView utility with brain spatial transcriptomics data and recover known anatomical structures. We further delineate functional tissue domains with associated marker genes that were obscured when analyzed using previous methods. We finally show how SODB may efficiently facilitate computational method development. The SODB website is https://gene.ai.tencent.com/SpatialOmics/ . The command-line package is available at https://pysodb.readthedocs.io/en/latest/ .
Collapse
Affiliation(s)
- Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Tencent AI Lab, Shenzhen, China.
| | - Wentao Pan
- Tencent AI Lab, Shenzhen, China
- Shenzhen International Graduate School, Tsinghua University, Shenzen, China
| | | | - Fangyuan Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Xiu Li
- Shenzhen International Graduate School, Tsinghua University, Shenzen, China
| | - Yi Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Michael Q Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas, Richardson, TX, USA.
| | | |
Collapse
|
148
|
Huuki-Myers L, Spangler A, Eagles N, Montgomery KD, Kwon SH, Guo B, Grant-Peters M, Divecha HR, Tippani M, Sriworarat C, Nguyen AB, Ravichandran P, Tran MN, Seyedian A, Hyde TM, Kleinman JE, Battle A, Page SC, Ryten M, Hicks SC, Martinowich K, Collado-Torres L, Maynard KR. Integrated single cell and unsupervised spatial transcriptomic analysis defines molecular anatomy of the human dorsolateral prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528722. [PMID: 36824961 PMCID: PMC9949126 DOI: 10.1101/2023.02.15.528722] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Generation of a molecular neuroanatomical map of the human prefrontal cortex reveals novel spatial domains and cell-cell interactions relevant for psychiatric disease. The molecular organization of the human neocortex has been historically studied in the context of its histological layers. However, emerging spatial transcriptomic technologies have enabled unbiased identification of transcriptionally-defined spatial domains that move beyond classic cytoarchitecture. Here we used the Visium spatial gene expression platform to generate a data-driven molecular neuroanatomical atlas across the anterior-posterior axis of the human dorsolateral prefrontal cortex (DLPFC). Integration with paired single nucleus RNA-sequencing data revealed distinct cell type compositions and cell-cell interactions across spatial domains. Using PsychENCODE and publicly available data, we map the enrichment of cell types and genes associated with neuropsychiatric disorders to discrete spatial domains. Finally, we provide resources for the scientific community to explore these integrated spatial and single cell datasets at research.libd.org/spatialDLPFC/.
Collapse
Affiliation(s)
- Louise Huuki-Myers
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Abby Spangler
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Nick Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Kelsey D Montgomery
- 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
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Melissa Grant-Peters
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Chaichontat Sriworarat
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Annie B Nguyen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | | | - Matthew N Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Arta Seyedian
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 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
| | - 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
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The 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
| | | | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- The 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
| |
Collapse
|
149
|
Szegedi V, Bakos E, Furdan S, Kovács BH, Varga D, Erdélyi M, Barzó P, Szücs A, Tamás G, Lamsa K. HCN channels at the cell soma ensure the rapid electrical reactivity of fast-spiking interneurons in human neocortex. PLoS Biol 2023; 21:e3002001. [PMID: 36745683 PMCID: PMC9934405 DOI: 10.1371/journal.pbio.3002001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 02/16/2023] [Accepted: 01/17/2023] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence indicates that there are substantial species differences in the properties of mammalian neurons, yet theories on circuit activity and information processing in the human brain are based heavily on results obtained from rodents and other experimental animals. This knowledge gap may be particularly important for understanding the neocortex, the brain area responsible for the most complex neuronal operations and showing the greatest evolutionary divergence. Here, we examined differences in the electrophysiological properties of human and mouse fast-spiking GABAergic basket cells, among the most abundant inhibitory interneurons in cortex. Analyses of membrane potential responses to current input, pharmacologically isolated somatic leak currents, isolated soma outside-out patch recordings, and immunohistochemical staining revealed that human neocortical basket cells abundantly express hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel isoforms HCN1 and HCN2 at the cell soma membrane, whereas these channels are sparse at the rodent basket cell soma membrane. Antagonist experiments showed that HCN channels in human neurons contribute to the resting membrane potential and cell excitability at the cell soma, accelerate somatic membrane potential kinetics, and shorten the lag between excitatory postsynaptic potentials and action potential generation. These effects are important because the soma of human fast-spiking neurons without HCN channels exhibit low persistent ion leak and slow membrane potential kinetics, compared with mouse fast-spiking neurons. HCN channels speed up human cell membrane potential kinetics and help attain an input-output rate close to that of rodent cells. Computational modeling demonstrated that HCN channel activity at the human fast-spiking cell soma membrane is sufficient to accelerate the input-output function as observed in cell recordings. Thus, human and mouse fast-spiking neurons exhibit functionally significant differences in ion channel composition at the cell soma membrane to set the speed and fidelity of their input-output function. These HCN channels ensure fast electrical reactivity of fast-spiking cells in human neocortex.
Collapse
Affiliation(s)
- Viktor Szegedi
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Emőke Bakos
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Szabina Furdan
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
| | - Bálint H. Kovács
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Dániel Varga
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Pál Barzó
- Department of Neurosurgery, University of Szeged, Szeged, Hungary
| | - Attila Szücs
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
- Neuronal Cell Biology Research Group, Eötvös Loránd University, Budapest, Budapest, Hungary
| | - Gábor Tamás
- MTA-SZTE Research Group for Cortical Microcircuits, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Karri Lamsa
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine Research Group for Human neuron physiology and therapy, Szeged, Hungary
- * E-mail: ,
| |
Collapse
|
150
|
Tao Y, Zhou X, Sun L, Lin D, Cai H, Chen X, Zhou W, Yang B, Hu Z, Yu J, Zhang J, Yang X, Yang F, Shen B, Qi W, Fu Z, Dai J, Cao G. Highly efficient and robust π-FISH rainbow for multiplexed in situ detection of diverse biomolecules. Nat Commun 2023; 14:443. [PMID: 36707540 PMCID: PMC9883232 DOI: 10.1038/s41467-023-36137-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
In the unprecedented single-cell sequencing and spatial multiomics era of biology, fluorescence in situ hybridization (FISH) technologies with higher sensitivity and robustness, especially for detecting short RNAs and other biomolecules, are greatly desired. Here, we develop the robust multiplex π-FISH rainbow method to detect diverse biomolecules (DNA, RNA, proteins, and neurotransmitters) individually or simultaneously with high efficiency. This versatile method is successfully applied to detect gene expression in different species, from microorganisms to plants and animals. Furthermore, we delineate the landscape of diverse neuron subclusters by decoding the spatial distribution of 21 marker genes via only two rounds of hybridization. Significantly, we combine π-FISH rainbow with hybridization chain reaction to develop π-FISH+ technology for short nucleic acid fragments, such as microRNA and prostate cancer anti-androgen therapy-resistant marker ARV7 splicing variant in circulating tumour cells from patients. Our study provides a robust biomolecule in situ detection technology for spatial multiomics investigation and clinical diagnosis.
Collapse
Affiliation(s)
- Yingfeng Tao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Xiaoliu Zhou
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Leqiang Sun
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Da Lin
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Huaiyuan Cai
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Xi Chen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Wei Zhou
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China
| | - Zhe Hu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China
| | - Jing Yu
- Department of Blood Transfusion, Wuhan hospital of Traditional Chinese and Western Medicine, Huazhong University of Science and Technology, 430070, Wuhan, China
| | - Jing Zhang
- Department of the 1st Thoracic Medical Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430070, Wuhan, China
| | - Xiaoqing Yang
- Hospital of Huazhong Agricultural University, 430070, Wuhan, China
| | - Fang Yang
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, 430070, Wuhan, China
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China.,Key Laboratory of Preventive Medicine in Hubei Province, 430070, Wuhan, Hubei Province, China
| | - Wenbao Qi
- College of Veterinary Medicine, South China Agricultural University, 510642, Guangzhou, China.,African Swine Fever Regional Laboratory of China, Guangzhou, China
| | - Zhenfang Fu
- Departments of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Jinxia Dai
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China. .,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China.
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070, Wuhan, China. .,College of Veterinary Medicine, Huazhong Agricultural University, 430070, Wuhan, China. .,College of Biomedicine and Health, Huazhong Agricultural University, 430070, Wuhan, China.
| |
Collapse
|