51
|
Saunders RA, Allen WE, Pan X, Sandhu J, Lu J, Lau TK, Smolyar K, Sullivan ZA, Dulac C, Weissman JS, Zhuang X. A platform for multimodal in vivo pooled genetic screens reveals regulators of liver function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.624217. [PMID: 39605605 PMCID: PMC11601512 DOI: 10.1101/2024.11.18.624217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Organ function requires coordinated activities of thousands of genes in distinct, spatially organized cell types. Understanding the basis of emergent tissue function requires approaches to dissect the genetic control of diverse cellular and tissue phenotypes in vivo. Here, we develop paired imaging and sequencing methods to construct large-scale, multi-modal genotype-phenotypes maps in tissue with pooled genetic perturbations. Using imaging, we identify genetic perturbations in individual cells while simultaneously measuring their gene expression and subcellular morphology. Using single-cell sequencing, we measure transcriptomic responses to the same genetic perturbations. We apply this approach to study hundreds of genetic perturbations in the mouse liver. Our study reveals regulators of hepatocyte zonation and liver unfolded protein response, as well as distinct pathways that cause hepatocyte steatosis. Our approach enables new ways of interrogating the genetic basis of complex cellular and organismal physiology and provides crucial training data for emerging machine-learning models of cellular function.
Collapse
Affiliation(s)
- Reuben A. Saunders
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- University of California, San Francisco, San Francisco, CA 94158, USA
- Present address: Society of Fellows, Harvard University, MA 02138, USA
- These authors contributed equally
| | - William E. Allen
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Society of Fellows, Harvard University, Cambridge, MA 02138, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Present address: Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305; Arc Institute, Palo Alto, CA 94304
- These authors contributed equally
- Lead contact
| | - Xingjie Pan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Lead AI Scientist
| | - Jaspreet Sandhu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jiaqi Lu
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Thomas K. Lau
- Department of Statistics, Stanford University, Stanford, CA 94305
| | - Karina Smolyar
- Whitehead Institute, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Zuri A. Sullivan
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Catherine Dulac
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jonathan S. Weissman
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Whitehead Institute, Cambridge, MA 02139, USA
- Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Xiaowei Zhuang
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Physics, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
52
|
Qian J, Shao X, Bao H, Fang Y, Guo W, Li C, Li A, Hua H, Fan X. Identification and characterization of cell niches in tissue from spatial omics data at single-cell resolution. Nat Commun 2025; 16:1693. [PMID: 39956823 PMCID: PMC11830827 DOI: 10.1038/s41467-025-57029-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Deciphering the features, structure, and functions of the cell niche in tissues remains a major challenge. Here, we present scNiche, a computational framework to identify and characterize cell niches from spatial omics data at single-cell resolution. We benchmark scNiche with both simulated and biological datasets, and demonstrate that scNiche can effectively and robustly identify cell niches while outperforming other existing methods. In spatial proteomics data from human triple-negative breast cancer, scNiche reveals the influence of the microenvironment on cellular phenotypes, and further dissects patient-specific niches with distinct cellular compositions or phenotypic characteristics. By analyzing mouse liver spatial transcriptomics data across normal and early-onset liver failure donors, scNiche uncovers disease-specific liver injury niches, and further delineates the niche remodeling from normal liver to liver failure. Overall, scNiche enables decoding the cellular microenvironment in tissues from single-cell spatial omics data.
Collapse
Affiliation(s)
- Jingyang Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
| | - Hudong Bao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yin Fang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310013, China
| | - Wenbo Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Chengyu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Anyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, SiChuan Institute for Translational Chinese Medicine, Chengdu, 610041, China.
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China.
| |
Collapse
|
53
|
Posani L, Wang S, Muscinelli SP, Paninski L, Fusi S. Rarely categorical, always high-dimensional: how the neural code changes along the cortical hierarchy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.15.623878. [PMID: 39605683 PMCID: PMC11601379 DOI: 10.1101/2024.11.15.623878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
A long-standing debate in neuroscience concerns whether individual neurons are organized into functionally distinct populations that encode information differently ("categorical" representations [1-3]) and the implications for neural computation. Here, we systematically analyzed how cortical neurons encode cognitive, sensory, and movement variables across 43 cortical regions during a complex task (14,000+ units from the International Brain Laboratory public Brain-wide Map data set [4]) and studied how these properties change across the sensory-cognitive cortical hierarchy [5]. We found that the structure of the neural code was scale-dependent: on a whole-cortex scale, neural selectivity was categorical and organized across regions in a way that reflected their anatomical connectivity. However, within individual regions, categorical representations were rare and limited to primary sensory areas. Remarkably, the degree of categorical clustering of neural selectivity was inversely correlated to the dimensionality of neural representations, suggesting a link between single-neuron selectivity and computational properties of population codes that we explained in a mathematical model. Finally, we found that the fraction of linearly separable combinations of experimental conditions ("Shattering Dimensionality" [6]) was near maximal across all areas, indicating a robust and uniform ability for flexible information encoding throughout the cortex. In conclusion, our results provide systematic evidence for a non-categorical, high-dimensional neural code in all but the lower levels of the cortical hierarchy.
Collapse
Affiliation(s)
- Lorenzo Posani
- Zuckerman Institute, Columbia University, New York, NY, USA
- School of Computer and Communication Sciences, EPFL, Street, Lausanne, Switzerland
| | - Shuqi Wang
- School of Computer and Communication Sciences, EPFL, Street, Lausanne, Switzerland
- Department of Statistics, Columbia University, New York, NY, USA
| | | | - Liam Paninski
- Zuckerman Institute, Columbia University, New York, NY, USA
- Department of Statistics, Columbia University, New York, NY, USA
- Co-senior authors
| | - Stefano Fusi
- Zuckerman Institute, Columbia University, New York, NY, USA
- Co-senior authors
| |
Collapse
|
54
|
Kinman AI, Merryweather DN, Erwin SR, Campbell RE, Sullivan KE, Kraus L, Kapustina M, Bristow BN, Zhang MY, Elder MW, Wood SC, Tarik A, Kim E, Tindall J, Daniels W, Anwer M, Guo C, Cembrowski MS. Atypical hippocampal excitatory neurons express and govern object memory. Nat Commun 2025; 16:1195. [PMID: 39939601 PMCID: PMC11822006 DOI: 10.1038/s41467-025-56260-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/10/2025] [Indexed: 02/14/2025] Open
Abstract
Classically, pyramidal cells of the hippocampus are viewed as flexibly representing spatial and non-spatial information. Recent work has illustrated distinct types of hippocampal excitatory neurons, suggesting that hippocampal representations and functions may be constrained and interpreted by these underlying cell-type identities. In mice, here we reveal a non-pyramidal excitatory neuron type - the "ovoid" neuron - that is spatially adjacent to subiculum pyramidal cells but differs in gene expression, electrophysiology, morphology, and connectivity. Functionally, novel object encounters drive sustained ovoid neuron activity, whereas familiar objects fail to drive activity even months after single-trial learning. Silencing ovoid neurons prevents non-spatial object learning but leaves spatial learning intact, and activating ovoid neurons toggles novel-object seeking to familiar-object seeking. Such function is doubly dissociable from pyramidal neurons, wherein manipulation of pyramidal cells affects spatial assays but not non-spatial learning. Ovoid neurons of the subiculum thus illustrate selective cell-type-specific control of non-spatial memory and behavioral preference.
Collapse
Affiliation(s)
- Adrienne I Kinman
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Derek N Merryweather
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Sarah R Erwin
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Regan E Campbell
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Kaitlin E Sullivan
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Larissa Kraus
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Margarita Kapustina
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Brianna N Bristow
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Mingjia Y Zhang
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Madeline W Elder
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Sydney C Wood
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Ali Tarik
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Esther Kim
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Joshua Tindall
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - William Daniels
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Mehwish Anwer
- Dept. of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, V6T 1Z7, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Caiying Guo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, USA
| | - Mark S Cembrowski
- Dept. of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, V6T 1Z3, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, V6T 1Z3, Canada.
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, 20147, USA.
- School of Biomedical Engineering, University of British Columbia, Vancouver, V6T 1Z3, Canada.
- Department of Mathematics, University of British Columbia, Vancouver, V6T 1Z2, Canada.
| |
Collapse
|
55
|
Budoff SA, Poleg-Polsky A. A Complete Spatial Map of Mouse Retinal Ganglion Cells Reveals Density and Gene Expression Specializations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637538. [PMID: 39990332 PMCID: PMC11844403 DOI: 10.1101/2025.02.10.637538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Retinal ganglion cells (RGCs) transmit visual information from the eye to the brain. In mice, several RGC subtypes show nonuniform spatial distributions, potentially mediating specific visual functions. However, the full extent of RGC specialization remains unknown. Here, we used en-face cryosectioning, spatial transcriptomics, and machine learning to map the spatial distribution of all RGC subtypes identified in previous single-cell studies. While two-thirds of RGC subtypes were evenly distributed, others showed strong biases toward ventral or dorso-temporal regions associated with sky vision and the area retinae temporalis (ART), the predicted homolog of the area centralis. Additionally, we observed unexpected spatial variation in gene expression within several subtypes along the dorso-ventral axis or within vs. outside the ART, independent of RGC density profiles. Finally, we found limited correlations between the gene profiles of the ART and the primate macula, suggesting divergent specialization between the mouse and primate central vision.
Collapse
Affiliation(s)
- Samuel A. Budoff
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| | - Alon Poleg-Polsky
- University of Colorado Anschutz Medical Center, Department of Physiology and Biophysics, Aurora, 80045, USA
| |
Collapse
|
56
|
Miller JA, Travaglini KJ, Luquez T, Hostetler RE, Oster A, Daniel S, Tasic B, Menon V. Annotation Comparison Explorer (ACE): connecting brain cell types across studies of health and Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637559. [PMID: 39990500 PMCID: PMC11844562 DOI: 10.1101/2025.02.11.637559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Single-cell multiomic technologies have allowed unprecedented access to gene profiles of individual cells across species and organ systems, including >1000 papers focused on brain cell types alone. The Allen Institute has created foundational atlases characterizing mammalian brain cell types in the adult mouse brain and the neocortex of aged humans with and without Alzheimer's disease (AD). With so many public cell type classifications (or 'taxonomies') available and many groups choosing to define their own, linking cell types and associated knowledge between studies remains a major challenge. Here, we introduce Annotation Comparison Explorer (ACE), a web application for comparing cell type assignments and other cell-based annotations (e.g., donor demographics, anatomic locations, batch variables, and quality control metrics). ACE allows filtering of cells and includes an interactive set of tools for comparing two or more taxonomy annotations alongside collected knowledge (e.g., increased abundance in disease conditions, cell type aliases, or other information about a specific cell type). We present three primary use cases for ACE. First, we demonstrate how a user can assign cell type labels from the Seattle Alzheimer's Disease Brain Cell Atlas (SEA-AD) taxonomy to cells from their own study and compare these cell type mappings to existing cell type assignments and cell metadata. Second, we extend this approach to ten published human AD studies which we previously reprocessed through a common data analysis pipeline. This allowed us to compare brain taxonomies across otherwise incomparable studies and identify congruent cell type abundance changes in AD, including a decrease in abundance of subsets of somatostatin interneurons. Finally, ACE includes translation tables between different mouse and human brain cell type taxonomies publicly accessible on Allen Brain Map, from initial studies in individual neocortical areas to more recent studies spanning the whole brain. ACE can be freely and publicly accessed as a web application (https://sea-ad.shinyapps.io/ACEapp/) and on GitHub (github.com/AllenInstitute/ACE).
Collapse
|
57
|
Sun ED, Zhou OY, Hauptschein M, Rappoport N, Xu L, Navarro Negredo P, Liu L, Rando TA, Zou J, Brunet A. Spatial transcriptomic clocks reveal cell proximity effects in brain ageing. Nature 2025; 638:160-171. [PMID: 39695234 PMCID: PMC11798877 DOI: 10.1038/s41586-024-08334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Old age is associated with a decline in cognitive function and an increase in neurodegenerative disease risk1. Brain ageing is complex and is accompanied by many cellular changes2. Furthermore, the influence that aged cells have on neighbouring cells and how this contributes to tissue decline is unknown. More generally, the tools to systematically address this question in ageing tissues have not yet been developed. Here we generate a spatially resolved single-cell transcriptomics brain atlas of 4.2 million cells from 20 distinct ages across the adult lifespan and across two rejuvenating interventions-exercise and partial reprogramming. We build spatial ageing clocks, machine learning models trained on this spatial transcriptomics atlas, to identify spatial and cell-type-specific transcriptomic fingerprints of ageing, rejuvenation and disease, including for rare cell types. Using spatial ageing clocks and deep learning, we find that T cells, which increasingly infiltrate the brain with age, have a marked pro-ageing proximity effect on neighbouring cells. Surprisingly, neural stem cells have a strong pro-rejuvenating proximity effect on neighbouring cells. We also identify potential mediators of the pro-ageing effect of T cells and the pro-rejuvenating effect of neural stem cells on their neighbours. These results suggest that rare cell types can have a potent influence on their neighbours and could be targeted to counter tissue ageing. Spatial ageing clocks represent a useful tool for studying cell-cell interactions in spatial contexts and should allow scalable assessment of the efficacy of interventions for ageing and disease.
Collapse
Affiliation(s)
- Eric D Sun
- Biomedical Data Science Graduate Program, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Olivia Y Zhou
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biophysics Graduate Program, Stanford University, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Max Hauptschein
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Lucy Xu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Biology Graduate Program, Stanford University, Stanford, CA, USA
| | | | - Ling Liu
- Department of Neurology, Stanford University, Stanford, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - Thomas A Rando
- Department of Neurology, Stanford University, Stanford, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Center for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
58
|
Johnston KG, Berackey BT, Tran KM, Gelber A, Yu Z, MacGregor GR, Mukamel EA, Tan Z, Green KN, Xu X. Single-cell spatial transcriptomics reveals distinct patterns of dysregulation in non-neuronal and neuronal cells induced by the Trem2 R47H Alzheimer's risk gene mutation. Mol Psychiatry 2025; 30:461-477. [PMID: 39103533 PMCID: PMC11746152 DOI: 10.1038/s41380-024-02651-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 08/07/2024]
Abstract
The R47H missense mutation of the TREM2 gene is a known risk factor for development of Alzheimer's Disease. In this study, we analyze the impact of the Trem2R47H mutation on specific cell types in multiple cortical and subcortical brain regions in the context of wild-type and 5xFAD mouse background. We profile 19 mouse brain sections consisting of wild-type, Trem2R47H, 5xFAD and Trem2R47H; 5xFAD genotypes using MERFISH spatial transcriptomics, a technique that enables subcellular profiling of spatial gene expression. Spatial transcriptomics and neuropathology data are analyzed using our custom pipeline to identify plaque and Trem2R47H-induced transcriptomic dysregulation. We initially analyze cell type-specific transcriptomic alterations induced by plaque proximity. Next, we analyze spatial distributions of disease associated microglia and astrocytes, and how they vary between 5xFAD and Trem2R47H; 5xFAD mouse models. Finally, we analyze the impact of the Trem2R47H mutation on neuronal transcriptomes. The Trem2R47H mutation induces consistent upregulation of Bdnf and Ntrk2 across many cortical excitatory neuron types, independent of amyloid pathology. Spatial investigation of genotype enriched subclusters identified spatially localized neuronal subpopulations reduced in 5xFAD and Trem2R47H; 5xFAD mice. Overall, our MERFISH spatial transcriptomics analysis identifies glial and neuronal transcriptomic alterations induced independently by 5xFAD and Trem2R47H mutations, impacting inflammatory responses in microglia and astrocytes, and activity and BDNF signaling in neurons.
Collapse
Affiliation(s)
- Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Bereket T Berackey
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| | - Kristine M Tran
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA
| | - Alon Gelber
- Department of Cognitive Science, University of California, San Diego, CA, 92037, USA
| | - Zhaoxia Yu
- Department of Statistics, School of Computer and Information Science, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Grant R MacGregor
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA
| | - Eran A Mukamel
- Department of Cognitive Science, University of California, San Diego, CA, 92037, USA
| | - Zhiqun Tan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA
- Department of Molecular Biology and Biochemistry School of Biological Sciences, University of California, Irvine, CA, 92697, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, CA, 92697, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), Irvine, USA.
| |
Collapse
|
59
|
Jin K, Yao Z, van Velthoven CTJ, Kaplan ES, Glattfelder K, Barlow ST, Boyer G, Carey D, Casper T, Chakka AB, Chakrabarty R, Clark M, Departee M, Desierto M, Gary A, Gloe J, Goldy J, Guilford N, Guzman J, Hirschstein D, Lee C, Liang E, Pham T, Reding M, Ronellenfitch K, Ruiz A, Sevigny J, Shapovalova N, Shulga L, Sulc J, Torkelson A, Tung H, Levi B, Sunkin SM, Dee N, Esposito L, Smith KA, Tasic B, Zeng H. Brain-wide cell-type-specific transcriptomic signatures of healthy ageing in mice. Nature 2025; 638:182-196. [PMID: 39743592 PMCID: PMC11798837 DOI: 10.1038/s41586-024-08350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/06/2024] [Indexed: 01/04/2025]
Abstract
Biological ageing can be defined as a gradual loss of homeostasis across various aspects of molecular and cellular function1,2. Mammalian brains consist of thousands of cell types3, which may be differentially susceptible or resilient to ageing. Here we present a comprehensive single-cell RNA sequencing dataset containing roughly 1.2 million high-quality single-cell transcriptomes of brain cells from young adult and aged mice of both sexes, from regions spanning the forebrain, midbrain and hindbrain. High-resolution clustering of all cells results in 847 cell clusters and reveals at least 14 age-biased clusters that are mostly glial types. At the broader cell subclass and supertype levels, we find age-associated gene expression signatures and provide a list of 2,449 unique differentially expressed genes (age-DE genes) for many neuronal and non-neuronal cell types. Whereas most age-DE genes are unique to specific cell types, we observe common signatures with ageing across cell types, including a decrease in expression of genes related to neuronal structure and function in many neuron types, major astrocyte types and mature oligodendrocytes, and an increase in expression of genes related to immune function, antigen presentation, inflammation, and cell motility in immune cell types and some vascular cell types. Finally, we observe that some of the cell types that demonstrate the greatest sensitivity to ageing are concentrated around the third ventricle in the hypothalamus, including tanycytes, ependymal cells, and certain neuron types in the arcuate nucleus, dorsomedial nucleus and paraventricular nucleus that express genes canonically related to energy homeostasis. Many of these types demonstrate both a decrease in neuronal function and an increase in immune response. These findings suggest that the third ventricle in the hypothalamus may be a hub for ageing in the mouse brain. Overall, this study systematically delineates a dynamic landscape of cell-type-specific transcriptomic changes in the brain associated with normal ageing that will serve as a foundation for the investigation of functional changes in ageing and the interaction of ageing and disease.
Collapse
Affiliation(s)
- Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Max Departee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Josh Sevigny
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA.
| |
Collapse
|
60
|
Yan G, Hua SH, Li JJ. Categorization of 34 computational methods to detect spatially variable genes from spatially resolved transcriptomics data. Nat Commun 2025; 16:1141. [PMID: 39880807 PMCID: PMC11779979 DOI: 10.1038/s41467-025-56080-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
In the analysis of spatially resolved transcriptomics data, detecting spatially variable genes (SVGs) is crucial. Numerous computational methods exist, but varying SVG definitions and methodologies lead to incomparable results. We review 34 state-of-the-art methods, classifying SVGs into three categories: overall, cell-type-specific, and spatial-domain-marker SVGs. Our review explains the intuitions underlying these methods, summarizes their applications, and categorizes the hypothesis tests they use in the trade-off between generality and specificity for SVG detection. We discuss challenges in SVG detection and propose future directions for improvement. Our review offers insights for method developers and users, advocating for category-specific benchmarking.
Collapse
Affiliation(s)
- Guanao Yan
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095-1554, USA
| | - Shuo Harper Hua
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Jingyi Jessica Li
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095-1554, USA.
- Department of Human Genetics, University of California, Los Angeles, CA, 90095-7088, USA.
- Department of Computational Medicine, University of California, Los Angeles, CA, 90095-1766, USA.
- Department of Biostatistics, University of California, Los Angeles, CA, 90095-1772, USA.
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
61
|
Tetzlaff SK, Reyhan E, Layer N, Bengtson CP, Heuer A, Schroers J, Faymonville AJ, Langeroudi AP, Drewa N, Keifert E, Wagner J, Soyka SJ, Schubert MC, Sivapalan N, Pramatarov RL, Buchert V, Wageringel T, Grabis E, Wißmann N, Alhalabi OT, Botz M, Bojcevski J, Campos J, Boztepe B, Scheck JG, Conic SH, Puschhof MC, Villa G, Drexler R, Zghaibeh Y, Hausmann F, Hänzelmann S, Karreman MA, Kurz FT, Schröter M, Thier M, Suwala AK, Forsberg-Nilsson K, Acuna C, Saez-Rodriguez J, Abdollahi A, Sahm F, Breckwoldt MO, Suchorska B, Ricklefs FL, Heiland DH, Venkataramani V. Characterizing and targeting glioblastoma neuron-tumor networks with retrograde tracing. Cell 2025; 188:390-411.e36. [PMID: 39644898 DOI: 10.1016/j.cell.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/16/2024] [Accepted: 11/04/2024] [Indexed: 12/09/2024]
Abstract
Glioblastomas are invasive brain tumors with high therapeutic resistance. Neuron-to-glioma synapses have been shown to promote glioblastoma progression. However, a characterization of tumor-connected neurons has been hampered by a lack of technologies. Here, we adapted retrograde tracing using rabies viruses to investigate and manipulate neuron-tumor networks. Glioblastoma rapidly integrated into neural circuits across the brain, engaging in widespread functional communication, with cholinergic neurons driving glioblastoma invasion. We uncovered patient-specific and tumor-cell-state-dependent differences in synaptogenic gene expression associated with neuron-tumor connectivity and subsequent invasiveness. Importantly, radiotherapy enhanced neuron-tumor connectivity by increased neuronal activity. In turn, simultaneous neuronal activity inhibition and radiotherapy showed increased therapeutic effects, indicative of a role for neuron-to-glioma synapses in contributing to therapeutic resistance. Lastly, rabies-mediated genetic ablation of tumor-connected neurons halted glioblastoma progression, offering a viral strategy to tackle glioblastoma. Together, this study provides a framework to comprehensively characterize neuron-tumor networks and target glioblastoma.
Collapse
Affiliation(s)
- Svenja K Tetzlaff
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ekin Reyhan
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nikolas Layer
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, Heidelberg, Germany
| | - Alina Heuer
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Julian Schroers
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anton J Faymonville
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Nina Drewa
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Elijah Keifert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Julia Wagner
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Stella J Soyka
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Marc C Schubert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Nirosan Sivapalan
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Rangel L Pramatarov
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Verena Buchert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Tim Wageringel
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Elena Grabis
- Translational Neurosurgery, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany; Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
| | - Niklas Wißmann
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Obada T Alhalabi
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Botz
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Jovana Bojcevski
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joaquín Campos
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Berin Boztepe
- Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jonas G Scheck
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany
| | - Sascha Henry Conic
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Maria C Puschhof
- Faculty of Medicine, Heidelberg University, and Institute for Computational Biomedicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Giulia Villa
- Translational Neurosurgery, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Richard Drexler
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Yahya Zghaibeh
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fabian Hausmann
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sonja Hänzelmann
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthia A Karreman
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix T Kurz
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Neuroradiology, University Hospital Geneva, Geneva, Switzerland
| | - Manuel Schröter
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Marc Thier
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Abigail K Suwala
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology (B300), German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 75185 Uppsala, Sweden
| | - Claudio Acuna
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Faculty of Medicine, Heidelberg University, and Institute for Computational Biomedicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Sahm
- Department of Neuropathology, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuropathology (B300), German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael O Breckwoldt
- Neuroradiology Department, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bogdana Suchorska
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dieter Henrik Heiland
- Translational Neurosurgery, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany; Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany; Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany; Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; German Cancer Consortium (DKTK), partner site Freiburg, Freiburg, Germany
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
62
|
Hunker AC, Mich JK, Taskin N, Torkelson A, Pham T, Bertagnolli D, Chakka AB, Chakrabarty R, Donadio NP, Ferrer R, Gasperini M, Goldy J, Guzman JB, Jin K, Khem S, Kutsal R, Lalanne JB, Martinez RA, Newman D, Pena N, Shapovalova NV, Weed N, Zhou T, Yao S, Shendure J, Smith KA, Lein ES, Tasic B, Levi BP, Ting JT. Technical and biological sources of noise confound multiplexed enhancer AAV screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633018. [PMID: 39868122 PMCID: PMC11760716 DOI: 10.1101/2025.01.14.633018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Cis -acting regulatory enhancer elements are valuable tools for gaining cell type-specific genetic access. Leveraging large chromatin accessibility atlases, putative enhancer sequences can be identified and deployed in adeno-associated virus (AAV) delivery platforms. However, a significant bottleneck in enhancer AAV discovery is charting their detailed expression patterns in vivo , a process that currently requires gold-standard one-by-one testing. Here we present a barcoded multiplex strategy for screening enhancer AAVs at cell type resolution using single cell RNA sequencing and taxonomy mapping. We executed a proof-of-concept study using a small pool of validated enhancer AAVs expressing in a variety of neuronal and non-neuronal cell types across the mouse brain. Unexpectedly, we encountered substantial technical and biological noise including chimeric packaging products, necessitating development of novel techniques to accurately deconvolve enhancer expression patterns. These results underscore the need for improved methods to mitigate noise and highlight the complexity of enhancer AAV biology in vivo .
Collapse
|
63
|
Yu J, Liu H, Gao R, Wang TV, Li C, Liu Y, Yang L, Xu Y, Cui Y, Jia C, Huang J, Chen PR, Rao Y. Calcineurin: An essential regulator of sleep revealed by biochemical, chemical biological, and genetic approaches. Cell Chem Biol 2025; 32:157-173.e7. [PMID: 39740665 DOI: 10.1016/j.chembiol.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025]
Abstract
Research into mechanisms underlying sleep traditionally relies on electrophysiology and genetics. Because sleep can only be measured on whole animals by behavioral observations and physical means, no sleep research was initiated by biochemical and chemical biological approaches. We used phosphorylation sites of kinases important for sleep as targets for biochemical and chemical biological approaches. Sleep was increased in mice carrying a threonine-to-alanine substitution at residue T469 of salt-inducible kinase 3 (SIK3). Our biochemical purification and photo-crosslinking revealed calcineurin (CaN) dephosphorylation, both in vitro and in vivo, of SIK3 at T469 and S551, but not T221. Knocking down CaN regulatory subunit reduced daily sleep by more than 5 h, exceeding all known mouse mutants. Our work uncovered a critical physiological role for CaN in sleep and pioneered biochemical purification and chemical biology as effective approaches to study sleep.
Collapse
Affiliation(s)
- Jianjun Yu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Huijie Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Rui Gao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Tao V Wang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenggang Li
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Yuxiang Liu
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Lu Yang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Ying Xu
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Yunfeng Cui
- Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China
| | - Chenxi Jia
- National Center for Protein Sciences Phoenix, Beijing, China
| | - Juan Huang
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Peng R Chen
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yi Rao
- Laboratory of Neurochemical Biology, Peking-Tsinghua Center for Life Sciences, Peking-Tsinghua-NIBS (PTN) Graduate Program, School of Life Sciences, Peking University, Beijing, China; Chinese Institute for Brain Research (CIBR), Beijing, China; Department of Chemical Biology, College of Chemistry and Chemical Engineering; School of Pharmaceutical Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China; Chinese Institutes for Medical Research (CIMR), Beijing, China; Capital Medical University, Beijing, China.
| |
Collapse
|
64
|
Kim K, Han M, Lee D. InTiCAR: Network-based identification of significant inter-tissue communicators for autoimmune diseases. Comput Struct Biotechnol J 2025; 27:333-345. [PMID: 39897058 PMCID: PMC11782887 DOI: 10.1016/j.csbj.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 02/04/2025] Open
Abstract
Inter-tissue communicators (ITCs) are intricate and essential aspects of our body, as they are the keepers of homeostatic equilibrium. It is no surprise that the dysregulation of the exchange between tissues are at the core of various disorders. Among such conditions, autoimmune diseases (AIDs) refer to a collection of pathological conditions where the miscommunication drives the immune system to mistakenly attack one's own body. Due to their myriad and diverse pathophysiologies, AIDs cannot be easily diagnosed or treated, and continuous efforts are required to seek for potential diagnostic markers or therapeutic targets. The identification of ITCs with significant involvement in the disease states is therefore crucial. Here, we present InTiCAR, Inter-Tissue Communicators for Autoimmune diseases by Random walk with restart, which is a network exploration-based analysis method that suggests disease-specific ITCs based on prior knowledge of disease genes, without the need for the external expression data. We first show that distinct ITC profile s can be acquired for various diseases by InTiCAR. We further illustrate that, for autoimmune diseases (AIDs) specifically, the disease-specific ITCs outperform disease genes in diagnosing patients using the UK Biobank plasma proteome dataset. Also, through CMap LINCS dataset, we find that high perturbation on the AIDs genes can be observed by the disease-specific ITCs. Our results provide and highlight unique perspectives on biological network analysis by focusing on the entities of extracellular communications.
Collapse
Affiliation(s)
- Kwansoo Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Manyoung Han
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| |
Collapse
|
65
|
Labarta-Bajo L, Allen NJ. Astrocytes in aging. Neuron 2025; 113:109-126. [PMID: 39788083 PMCID: PMC11735045 DOI: 10.1016/j.neuron.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The mammalian nervous system is impacted by aging. Aging alters brain architecture, is associated with molecular damage, and can manifest with cognitive and motor deficits that diminish the quality of life. Astrocytes are glial cells of the CNS that regulate the development, function, and repair of neural circuits during development and adulthood; however, their functions in aging are less understood. Astrocytes change their transcriptome during aging, with astrocytes in areas such as the cerebellum, the hypothalamus, and white matter-rich regions being the most affected. While numerous studies describe astrocyte transcriptional changes in aging, many questions still remain. For example, how is astrocyte function altered by transcriptional changes that occur during aging? What are the mechanisms promoting astrocyte aged states? How do aged astrocytes impact brain function? This review discusses features of aged astrocytes and their potential triggers and proposes ways in which they may impact brain function and health span.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Nicola J Allen
- Salk Institute for Biological Studies, Molecular Neurobiology Laboratory, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
66
|
Yuan Y, Liu H, Dai Z, He C, Qin S, Su Z. From Physiology to Pathology of Astrocytes: Highlighting Their Potential as Therapeutic Targets for CNS Injury. Neurosci Bull 2025; 41:131-154. [PMID: 39080102 PMCID: PMC11748647 DOI: 10.1007/s12264-024-01258-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 01/19/2025] Open
Abstract
In the mammalian central nervous system (CNS), astrocytes are the ubiquitous glial cells that have complex morphological and molecular characteristics. These fascinating cells play essential neurosupportive and homeostatic roles in the healthy CNS and undergo morphological, molecular, and functional changes to adopt so-called 'reactive' states in response to CNS injury or disease. In recent years, interest in astrocyte research has increased dramatically and some new biological features and roles of astrocytes in physiological and pathological conditions have been discovered thanks to technological advances. Here, we will review and discuss the well-established and emerging astroglial biology and functions, with emphasis on their potential as therapeutic targets for CNS injury, including traumatic and ischemic injury. This review article will highlight the importance of astrocytes in the neuropathological process and repair of CNS injury.
Collapse
Affiliation(s)
- Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
- Department of Pain Medicine, School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Ziwei Dai
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
67
|
Kaplan HS, Horvath PM, Rahman MM, Dulac C. The neurobiology of parenting and infant-evoked aggression. Physiol Rev 2025; 105:315-381. [PMID: 39146250 DOI: 10.1152/physrev.00036.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Parenting behavior comprises a variety of adult-infant and adult-adult interactions across multiple timescales. The state transition from nonparent to parent requires an extensive reorganization of individual priorities and physiology and is facilitated by combinatorial hormone action on specific cell types that are integrated throughout interconnected and brainwide neuronal circuits. In this review, we take a comprehensive approach to integrate historical and current literature on each of these topics across multiple species, with a focus on rodents. New and emerging molecular, circuit-based, and computational technologies have recently been used to address outstanding gaps in our current framework of knowledge on infant-directed behavior. This work is raising fundamental questions about the interplay between instinctive and learned components of parenting and the mutual regulation of affiliative versus agonistic infant-directed behaviors in health and disease. Whenever possible, we point to how these technologies have helped gain novel insights and opened new avenues of research into the neurobiology of parenting. We hope this review will serve as an introduction for those new to the field, a comprehensive resource for those already studying parenting, and a guidepost for designing future studies.
Collapse
Affiliation(s)
- Harris S Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Patricia M Horvath
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Mohammed Mostafizur Rahman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| |
Collapse
|
68
|
Ma J, Qi R, Wang J, Berto S, Wang GZ. Human-unique brain cell clusters are associated with learning disorders and human episodic memory activity. Mol Psychiatry 2025; 30:353-359. [PMID: 39227435 DOI: 10.1038/s41380-024-02722-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
The advanced evolution of the human cerebral cortex forms the basis for our high-level cognitive functions. Through a comparative analysis of single-nucleus transcriptome data from the human neocortex and that of chimpanzees, macaques, and marmosets, we discovered 20 subgroups of cell types unique to the human brain, which include 11 types of excitatory neurons. Many of these human-unique cell clusters exhibit significant overexpression of genes regulated by human-specific enhancers. Notably, these specific cell clusters also express genes associated with disease risk, particularly those related to brain dysfunctions like learning disorders. Furthermore, genes linked to cortical thickness and human episodic memory encoding activities show heightened expression within these cell subgroups. These findings underscore the critical role of human brain-unique cell clusters in the evolution of human brain functions.
Collapse
Affiliation(s)
- Junjie 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
| | - Ruicheng Qi
- 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
| | - Jing Wang
- 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
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Guang-Zhong Wang
- 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.
| |
Collapse
|
69
|
Hrovatin K, Sikkema L, Shitov VA, Heimberg G, Shulman M, Oliver AJ, Mueller MF, Ibarra IL, Wang H, Ramírez-Suástegui C, He P, Schaar AC, Teichmann SA, Theis FJ, Luecken MD. Considerations for building and using integrated single-cell atlases. Nat Methods 2025; 22:41-57. [PMID: 39672979 DOI: 10.1038/s41592-024-02532-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/22/2024] [Indexed: 12/15/2024]
Abstract
The rapid adoption of single-cell technologies has created an opportunity to build single-cell 'atlases' integrating diverse datasets across many laboratories. Such atlases can serve as a reference for analyzing and interpreting current and future data. However, it has become apparent that atlasing approaches differ, and the impact of these differences are often unclear. Here we review the current atlasing literature and present considerations for building and using atlases. Importantly, we find that no one-size-fits-all protocol for atlas building exists, but rather we discuss context-specific considerations and workflows, including atlas conceptualization, data collection, curation and integration, atlas evaluation and atlas sharing. We further highlight the benefits of integrated atlases for analyses of new datasets and deriving biological insights beyond what is possible from individual datasets. Our overview of current practices and associated recommendations will improve the quality of atlases to come, facilitating the shift to a unified, reference-based understanding of single-cell biology.
Collapse
Affiliation(s)
- Karin Hrovatin
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lisa Sikkema
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Vladimir A Shitov
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Graham Heimberg
- Department of OMNI Bioinformatics, Genentech, South San Francisco, CA, USA
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
| | - Maiia Shulman
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Michaela F Mueller
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Ignacio L Ibarra
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Hanchen Wang
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
- Department of Computer Science, Stanford University, Palo Alto, CA, USA
| | - Ciro Ramírez-Suástegui
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Anna C Schaar
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
- CIFAR MacMillan Multiscale Human Programme, Toronto, Ontario, Canada
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Garching, Germany.
| | - Malte D Luecken
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
70
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
71
|
Calabrese RL, Marder E. Degenerate neuronal and circuit mechanisms important for generating rhythmic motor patterns. Physiol Rev 2025; 105:95-135. [PMID: 39453990 DOI: 10.1152/physrev.00003.2024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 10/27/2024] Open
Abstract
In 1996, we published a review article (Marder E, Calabrese RL. Physiol Rev 76: 687-717, 1996) describing the state of knowledge about the structure and function of the central pattern-generating circuits important for producing rhythmic behaviors. Although many of the core questions persist, much has changed since 1996. Here, we focus on newer studies that reveal ambiguities that complicate understanding circuit dynamics, despite the enormous technical advances of the recent past. In particular, we highlight recent studies of animal-to-animal variability and our understanding that circuit rhythmicity may be supported by multiple state-dependent mechanisms within the same animal and that robustness and resilience in the face of perturbation may depend critically on the presence of modulators and degenerate circuit mechanisms. Additionally, we highlight the use of computational models to ask whether there are generalizable principles about circuit motifs that can be found across rhythmic motor systems in different animal species.
Collapse
Affiliation(s)
| | - Eve Marder
- Brandeis University, Waltham, Massachusetts, United States
| |
Collapse
|
72
|
Schaub DP, Yousefi B, Kaiser N, Khatri R, Puelles VG, Krebs CF, Panzer U, Bonn S. PCA-based spatial domain identification with state-of-the-art performance. Bioinformatics 2024; 41:btaf005. [PMID: 39775801 PMCID: PMC11761416 DOI: 10.1093/bioinformatics/btaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
MOTIVATION The identification of biologically meaningful domains is a central step in the analysis of spatial transcriptomic data. RESULTS Following Occam's razor, we show that a simple PCA-based algorithm for unsupervised spatial domain identification rivals the performance of ten competing state-of-the-art methods across six single-cell spatial transcriptomic datasets. Our reductionist approach, NichePCA, provides researchers with intuitive domain interpretation and excels in execution speed, robustness, and scalability. AVAILABILITY AND IMPLEMENTATION The code is available at https://github.com/imsb-uke/nichepca.
Collapse
Affiliation(s)
- Darius P Schaub
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Behnam Yousefi
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Nico Kaiser
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Robin Khatri
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Victor G Puelles
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Christian F Krebs
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Ulf Panzer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Stefan Bonn
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
73
|
Qiu X, Zhu DY, Lu Y, Yao J, Jing Z, Min KH, Cheng M, Pan H, Zuo L, King S, Fang Q, Zheng H, Wang M, Wang S, Zhang Q, Yu S, Liao S, Liu C, Wu X, Lai Y, Hao S, Zhang Z, Wu L, Zhang Y, Li M, Tu Z, Lin J, Yang Z, Li Y, Gu Y, Ellison D, Chen A, Liu L, Weissman JS, Ma J, Xu X, Liu S, Bai Y. Spatiotemporal modeling of molecular holograms. Cell 2024; 187:7351-7373.e61. [PMID: 39532097 DOI: 10.1016/j.cell.2024.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Quantifying spatiotemporal dynamics during embryogenesis is crucial for understanding congenital diseases. We developed Spateo (https://github.com/aristoteleo/spateo-release), a 3D spatiotemporal modeling framework, and applied it to a 3D mouse embryogenesis atlas at E9.5 and E11.5, capturing eight million cells. Spateo enables scalable, partial, non-rigid alignment, multi-slice refinement, and mesh correction to create molecular holograms of whole embryos. It introduces digitization methods to uncover multi-level biology from subcellular to whole organ, identifying expression gradients along orthogonal axes of emergent 3D structures, e.g., secondary organizers such as midbrain-hindbrain boundary (MHB). Spateo further jointly models intercellular and intracellular interaction to dissect signaling landscapes in 3D structures, including the zona limitans intrathalamica (ZLI). Lastly, Spateo introduces "morphometric vector fields" of cell migration and integrates spatial differential geometry to unveil molecular programs underlying asymmetrical murine heart organogenesis and others, bridging macroscopic changes with molecular dynamics. Thus, Spateo enables the study of organ ecology at a molecular level in 3D space over time.
Collapse
Affiliation(s)
- Xiaojie Qiu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| | - Daniel Y Zhu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifan Lu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Electronic Information School, Wuhan University, Wuhan 430072, China
| | - Jiajun Yao
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kyung Hoi Min
- Ginkgo Bioworks, The Innovation and Design Building, Boston, MA 02210, USA
| | - Mengnan Cheng
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | | | - Lulu Zuo
- BGI Research, Shenzhen 518083, China
| | - Samuel King
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Fang
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingyue Wang
- BGI Research, Hangzhou 310030, China; Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shuai Wang
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingquan Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Sichao Yu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Chao Liu
- BGI Research, Wuhan 430074, China
| | - Xinchao Wu
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiwei Lai
- BGI Research, Shenzhen 518083, China
| | | | - Zhewei Zhang
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liang Wu
- BGI Research, Chongqing 401329, China
| | | | - Mei Li
- STOmics Tech Co., Ltd, Shenzhen 518083, China
| | - Zhencheng Tu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinpei Lin
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China
| | - Zhuoxuan Yang
- BGI Research, Hangzhou 310030, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Ying Gu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ao Chen
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA
| | - Jiayi Ma
- Electronic Information School, Wuhan University, Wuhan 430072, China.
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China.
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China; The Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, Guangdong, China.
| | - Yinqi Bai
- BGI Research, Sanya 572025, China; Hainan Technology Innovation Center for Marine Biological Resources Utilization (Preparatory Period), BGI Research, Sanya 572025, China.
| |
Collapse
|
74
|
Gaertner Z, Oram C, Schneeweis A, Schonfeld E, Bolduc C, Chen C, Dombeck D, Parisiadou L, Poulin JF, Awatramani R. Molecular and spatial transcriptomic classification of midbrain dopamine neurons and their alterations in a LRRK2 G2019S model of Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597807. [PMID: 38895448 PMCID: PMC11185743 DOI: 10.1101/2024.06.06.597807] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Several studies have revealed that midbrain dopamine (DA) neurons, even within a single neuroanatomical area, display heterogeneous properties. In parallel, studies using single cell profiling techniques have begun to cluster DA neurons into subtypes based on their molecular signatures. Recent work has shown that molecularly defined DA subtypes within the substantia nigra (SNc) display distinctive anatomic and functional properties, and differential vulnerability in Parkinson's disease (PD). Based on these provocative results, a granular understanding of these putative subtypes and their alterations in PD models, is imperative. We developed an optimized pipeline for single-nuclear RNA sequencing (snRNA-seq) and generated a high-resolution hierarchically organized map revealing 20 molecularly distinct DA neuron subtypes belonging to three main families. We integrated this data with spatial MERFISH technology to map, with high definition, the location of these subtypes in the mouse midbrain, revealing heterogeneity even within neuroanatomical sub-structures. Finally, we demonstrate that in the preclinical LRRK2G2019S knock-in mouse model of PD, subtype organization and proportions are preserved. Transcriptional alterations occur in many subtypes including those localized to the ventral tier SNc, where differential expression is observed in synaptic pathways, which might account for previously described DA release deficits in this model. Our work provides an advancement of current taxonomic schemes of the mouse midbrain DA neuron subtypes, a high-resolution view of their spatial locations, and their alterations in a prodromal mouse model of PD.
Collapse
Affiliation(s)
- Zachary Gaertner
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Cameron Oram
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Amanda Schneeweis
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Elan Schonfeld
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
| | - Cyril Bolduc
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Chuyu Chen
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Dombeck
- Northwestern University, Dept of Neurobiology, Evanston, IL 60201
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Loukia Parisiadou
- Northwestern University Feinberg School of Medicine, Dept of Pharmacology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jean-Francois Poulin
- McGill University (Montreal Neurological Institute), Faculty of Medicine and Health Sciences, Dept of Neurology and Neurosurgery, Montreal (QC), Canada
| | - Rajeshwar Awatramani
- Northwestern University Feinberg School of Medicine, Dept of Neurology, Chicago, IL 60611
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
75
|
El-Khatib SM, Vagadia AR, Le ACD, Baulch JE, Ng DQ, Du M, Johnston KG, Tan Z, Xu X, Chan A, Acharya MM. BDNF augmentation reverses cranial radiation therapy-induced cognitive decline and neurodegenerative consequences. Acta Neuropathol Commun 2024; 12:190. [PMID: 39696694 DOI: 10.1186/s40478-024-01906-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Cranial radiation therapy (RT) for brain cancers is often associated with the development of radiation-induced cognitive dysfunction (RICD). RICD significantly impacts the quality of life for cancer survivors, highlighting an unmet medical need. Previous human studies revealed a marked reduction in plasma brain-derived neurotrophic factor (BDNF) post-chronic chemotherapy, linking this decline to a substantial cognitive dysfunction among cancer survivors. Moreover, riluzole (RZ)-mediated increased BDNF in vivo in the chemotherapy-exposed mice reversed cognitive decline. RZ is an FDA-approved medication for ALS known to increase BDNF in vivo. In an effort to mitigate the detrimental effects of RT-induced BDNF decline in RICD, we tested the efficacy of RZ in a cranially irradiated (9 Gy) adult mouse model. Notably, RT-exposed mice exhibited significantly reduced hippocampal BDNF, accompanied by increased neuroinflammation, loss of neuronal plasticity-related immediate early gene product, cFos, and synaptic density. Spatial transcriptomic profiling comparing the RT + Vehicle with the RT + RZ group showed gene expression signatures of neuroprotection of hippocampal excitatory neurons post-RZ. RT-exposed mice performed poorly on learning and memory, and memory consolidation tasks. However, irradiated mice receiving RZ (13 mg/kg, drinking water) for 6-7 weeks showed a significant improvement in cognitive function compared to RT-exposed mice receiving vehicle. Dual-immunofluorescence staining, spatial transcriptomics, and biochemical assessment of RZ-treated irradiated brains demonstrated preservation of synaptic integrity and mature neuronal plasticity but not neurogenesis and reduced neuroinflammation concurrent with elevated BDNF levels and transcripts compared to vehicle-treated irradiated brains. In summary, oral administration of RZ represents a viable and translationally feasible neuroprotective approach against RICD.
Collapse
Affiliation(s)
- Sanad M El-Khatib
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Arya R Vagadia
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Anh C D Le
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
| | - Janet E Baulch
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, USA
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA
| | - Mingyu Du
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Kevin G Johnston
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Zhiqun Tan
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA
- Center for Neural Circuit Mapping, School of Medicine, University of California, Irvine, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, USA.
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, USA.
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, USA.
| |
Collapse
|
76
|
Boldrini M, Xiao Y, Singh T, Zhu C, Jabbi M, Pantazopoulos H, Gürsoy G, Martinowich K, Punzi G, Vallender EJ, Zody M, Berretta S, Hyde TM, Kleinman JE, Marenco S, Roussos P, Lewis DA, Turecki G, Lehner T, Mann JJ. Omics Approaches to Investigate the Pathogenesis of Suicide. Biol Psychiatry 2024; 96:919-928. [PMID: 38821194 PMCID: PMC11563882 DOI: 10.1016/j.biopsych.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Suicide is the second leading cause of death in U.S. adolescents and young adults and is generally associated with a psychiatric disorder. Suicidal behavior has a complex etiology and pathogenesis. Moderate heritability suggests genetic causes. Associations between childhood and recent life adversity indicate contributions from epigenetic factors. Genomic contributions to suicide pathogenesis remain largely unknown. This article is based on a workshop held to design strategies to identify molecular drivers of suicide neurobiology that would be putative new treatment targets. The panel determined that while bulk tissue studies provide comprehensive information, single-nucleus approaches that identify cell type-specific changes are needed. While single-nuclei techniques lack information on cytoplasm, processes, spines, and synapses, spatial multiomic technologies on intact tissue detect cell alterations specific to brain tissue layers and subregions. Because suicide has genetic and environmental drivers, multiomic approaches that combine cell type-specific epigenome, transcriptome, and proteome provide a more complete picture of pathogenesis. To determine the direction of effect of suicide risk gene variants on RNA and protein expression and how these interact with epigenetic marks, single-nuclei and spatial multiomics quantitative trait loci maps should be integrated with whole-genome sequencing and genome-wide association databases. The workshop concluded with a recommendation for the formation of an international suicide biology consortium that will bring together brain banks and investigators with expertise in cutting-edge omics technologies to delineate the biology of suicide and identify novel potential treatment targets to be tested in cellular and animal models for drug and biomarker discovery to guide suicide prevention.
Collapse
Affiliation(s)
- Maura Boldrini
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York.
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Tarjinder Singh
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York; New York Genome Center, New York, New York
| | - Chenxu Zhu
- New York Genome Center, New York, New York; Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York
| | - Mbemba Jabbi
- Department of Psychiatry and Behavioral Sciences, Mulva Clinics for the Neurosciences, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | - Gamze Gürsoy
- New York Genome Center, New York, New York; Departments of Biomedical Informatics and Computer Science, Columbia University, New York, New York
| | - Keri Martinowich
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Giovanna Punzi
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Eric J Vallender
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Sabina Berretta
- Department of Psychiatry, Harvard Brain Tissue Resource Center, Harvard Medical School, McLean Hospital, Belmont, Massachusetts
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Department of Psychiatry and Behavioral Sciences, Baltimore, Maryland
| | - Stefano Marenco
- Human Brain Collection Core, National Institute of Mental Health's (NIMH) Division of Intramural Research Programs, Bethesda, Maryland
| | - Panagiotis Roussos
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, Bronx, New York
| | - David A Lewis
- Departments of Psychiatry and Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Institute, McGill University, Montréal, Québec, Canada
| | | | - J John Mann
- Department of Psychiatry, Columbia University, New York, New York; Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, New York
| |
Collapse
|
77
|
Yang L, Fang LZ, Lynch MR, Xu CS, Hahm HJ, Zhang Y, Heitmeier MR, Costa VD, Samineni VK, Creed MC. Transcriptomic landscape of mammalian ventral pallidum at single-cell resolution. SCIENCE ADVANCES 2024; 10:eadq6017. [PMID: 39661664 PMCID: PMC11633743 DOI: 10.1126/sciadv.adq6017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
The ventral pallidum (VP) is critical for motivated behaviors. While contemporary work has begun to elucidate the functional diversity of VP neurons, the molecular heterogeneity underlying this functional diversity remains incompletely understood. We used single-nucleus RNA sequencing and in situ hybridization to define the transcriptional taxonomy of VP cell types in mice, macaques, and baboons. We found transcriptional conservation between all three species, within the broader neurochemical cell types. Unique dopaminoceptive and cholinergic subclusters were identified and conserved across both primate species but had no homolog in mice. This harmonized consensus VP cellular atlas will pave the way for understanding the structure and function of the VP and identified key neuropeptides, neurotransmitters, and neurotransmitter receptors that could be targeted within specific VP cell types for functional investigations.
Collapse
Affiliation(s)
- Lite Yang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa Z. Fang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Michelle R. Lynch
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- NINDS Neuroscience Postbaccalaureate Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Chang S. Xu
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah J. Hahm
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yufen Zhang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Monique R. Heitmeier
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Vincent D. Costa
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
| | - Vijay K. Samineni
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Meaghan C. Creed
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
78
|
Thompson JR, Nelson ED, Tippani M, Ramnauth AD, Divecha HR, Miller RA, Eagles NJ, Pattie EA, Kwon SH, Bach SV, Kaipa UM, Yao J, Hou C, Kleinman JE, Collado-Torres L, Han S, Maynard KR, Hyde TM, Martinowich K, Page SC, Hicks SC. An integrated single-nucleus and spatial transcriptomics atlas reveals the molecular landscape of the human hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590643. [PMID: 38712198 PMCID: PMC11071618 DOI: 10.1101/2024.04.26.590643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The hippocampus contains many unique cell types, which serve the structure's specialized functions, including learning, memory and cognition. These cells have distinct spatial organization, morphology, physiology, and connectivity, highlighting the importance of transcriptome-wide profiling strategies that retain cytoarchitectural organization. Here, we generated spatially-resolved transcriptomics (SRT) and single-nucleus RNA-sequencing (snRNA-seq) data from adjacent tissue sections of the anterior human hippocampus in ten adult neurotypical donors to define molecular profiles for hippocampal cell types and spatial domains. Using non-negative matrix factorization (NMF) and label transfer, we integrated these data by defining gene expression patterns within the snRNA-seq data and inferring their expression in the SRT data. We identified NMF patterns that captured transcriptional variation across neuronal cell types and indicated that the response of excitatory and inhibitory postsynaptic specializations were prioritized in different SRT spatial domains. We used the NMF and label transfer approach to leverage existing rodent datasets, identifying patterns of activity-dependent transcription and subpopulations of dentate gyrus granule cells in our SRT dataset that may be predisposed to participate in learning and memory ensembles. Finally, we characterized the spatial organization of NMF patterns corresponding to non-cornu ammonis pyramidal neurons and identified snRNA-seq clusters mapping to distinct regions of the retrohippocampus, to three subiculum layers, and to a population of presubiculum neurons. To make this comprehensive molecular atlas accessible to the scientific community, both raw and processed data are freely available, including through interactive web applications.
Collapse
Affiliation(s)
- Jacqueline R. Thompson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Erik D. Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Anthony D. Ramnauth
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Ryan A. Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Nicholas J. Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Elizabeth A. Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Svitlana V. Bach
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Uma M. Kaipa
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Jianing Yao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Hou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Shizhong Han
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, MD, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Stephanie C. Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
79
|
Martin N, Olsen P, Quon J, Campos J, Cuevas NV, Nagra J, VanNess M, Maltzer Z, Gelfand EC, Oyama A, Gary A, Wang Y, Alaya A, Ruiz A, Reynoldson C, Bielstein C, Pom CA, Huang C, Slaughterbeck C, Liang E, Alexander J, Ariza J, Malone J, Melchor J, Colbert K, Brouner K, Shulga L, Reding M, Latimer P, Sanchez R, Barta S, Egdorf T, Madigan Z, Pagan CM, Close JL, Long B, Kunst M, Lein ES, Zeng H, McMillen D, Waters J. MerQuaCo: a computational tool for quality control in image-based spatial transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626766. [PMID: 39677693 PMCID: PMC11643037 DOI: 10.1101/2024.12.04.626766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Image-based spatial transcriptomics platforms are powerful tools often used to identify cell populations and describe gene expression in intact tissue. Spatial experiments return large, high-dimension datasets and several open-source software packages are available to facilitate analysis and visualization. Spatial results are typically imperfect. For example, local variations in transcript detection probability are common. Software tools to characterize imperfections and their impact on downstream analyses are lacking so the data quality is assessed manually, a laborious and often a subjective process. Here we describe imperfections in a dataset of 641 fresh-frozen adult mouse brain sections collected using the Vizgen MERSCOPE. Common imperfections included the local loss of tissue from the section, tissue outside the imaging volume due to detachment from the coverslip, transcripts missing due to dropped images, varying detection probability through space, and differences in transcript detection probability between experiments. We describe the incidence of each imperfection and the likely impact on the accuracy of cell type labels. We develop MerQuaCo, open-source code that detects and quantifies imperfections without user input, facilitating the selection of sections for further analysis with existing packages. Together, our results and MerQuaCo facilitate rigorous, objective assessment of the quality of spatial transcriptomics results.
Collapse
Affiliation(s)
| | | | - Jacob Quon
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jazmin Campos
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | - Josh Nagra
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Marshall VanNess
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Zoe Maltzer
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Emily C Gelfand
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Alana Oyama
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Amanda Gary
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Yimin Wang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Angela Alaya
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Augustin Ruiz
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Cade Reynoldson
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | | | - Cindy Huang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | | | - Elizabeth Liang
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jason Alexander
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jeanelle Ariza
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jocelin Malone
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jose Melchor
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Kaity Colbert
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Krissy Brouner
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Lyudmila Shulga
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Melissa Reding
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Patrick Latimer
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Raymond Sanchez
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Stuard Barta
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Tom Egdorf
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Zachary Madigan
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Chelsea M Pagan
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jennie L Close
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Brian Long
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Michael Kunst
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Ed S Lein
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Hongkui Zeng
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Delissa McMillen
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| | - Jack Waters
- Allen Institute for Brain Science, 615 Westlake Ave N, Seattle WA
| |
Collapse
|
80
|
Ament SA, Campbell RR, Lobo MK, Receveur JP, Agrawal K, Borjabad A, Byrareddy SN, Chang L, Clarke D, Emani P, Gabuzda D, Gaulton KJ, Giglio M, Giorgi FM, Gok B, Guda C, Hadas E, Herb BR, Hu W, Huttner A, Ishmam MR, Jacobs MM, Kelschenbach J, Kim DW, Lee C, Liu S, Liu X, Madras BK, Mahurkar AA, Mash DC, Mukamel EA, Niu M, O'Connor RM, Pagan CM, Pang APS, Pillai P, Repunte-Canonigo V, Ruzicka WB, Stanley J, Tickle T, Tsai SYA, Wang A, Wills L, Wilson AM, Wright SN, Xu S, Yang J, Zand M, Zhang L, Zhang J, Akbarian S, Buch S, Cheng CS, Corley MJ, Fox HS, Gerstein M, Gummuluru S, Heiman M, Ho YC, Kellis M, Kenny PJ, Kluger Y, Milner TA, Moore DJ, Morgello S, Ndhlovu LC, Rana TM, Sanna PP, Satterlee JS, Sestan N, Spector SA, Spudich S, Tilgner HU, Volsky DJ, White OR, Williams DW, Zeng H. The single-cell opioid responses in the context of HIV (SCORCH) consortium. Mol Psychiatry 2024; 29:3950-3961. [PMID: 38879719 PMCID: PMC11609103 DOI: 10.1038/s41380-024-02620-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/19/2024]
Abstract
Substance use disorders (SUD) and drug addiction are major threats to public health, impacting not only the millions of individuals struggling with SUD, but also surrounding families and communities. One of the seminal challenges in treating and studying addiction in human populations is the high prevalence of co-morbid conditions, including an increased risk of contracting a human immunodeficiency virus (HIV) infection. Of the ~15 million people who inject drugs globally, 17% are persons with HIV. Conversely, HIV is a risk factor for SUD because chronic pain syndromes, often encountered in persons with HIV, can lead to an increased use of opioid pain medications that in turn can increase the risk for opioid addiction. We hypothesize that SUD and HIV exert shared effects on brain cell types, including adaptations related to neuroplasticity, neurodegeneration, and neuroinflammation. Basic research is needed to refine our understanding of these affected cell types and adaptations. Studying the effects of SUD in the context of HIV at the single-cell level represents a compelling strategy to understand the reciprocal interactions among both conditions, made feasible by the availability of large, extensively-phenotyped human brain tissue collections that have been amassed by the Neuro-HIV research community. In addition, sophisticated animal models that have been developed for both conditions provide a means to precisely evaluate specific exposures and stages of disease. We propose that single-cell genomics is a uniquely powerful technology to characterize the effects of SUD and HIV in the brain, integrating data from human cohorts and animal models. We have formed the Single-Cell Opioid Responses in the Context of HIV (SCORCH) consortium to carry out this strategy.
Collapse
Affiliation(s)
- Seth A Ament
- University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | - Mary Kay Lobo
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | - Linda Chang
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Dana Gabuzda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Michelle Giglio
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Eran Hadas
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian R Herb
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wen Hu
- Weill Cornell Medicine, New York, NY, USA
| | | | | | | | | | | | - Cheyu Lee
- University of California Irvine, Irvine, CA, USA
| | - Shuhui Liu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiaokun Liu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anup A Mahurkar
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Meng Niu
- University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | - Piya Pillai
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - W Brad Ruzicka
- McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | | | | | | | - Allen Wang
- University of California San Diego, La Jolla, CA, USA
| | - Lauren Wills
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Siwei Xu
- University of California Irvine, Irvine, CA, USA
| | | | - Maryam Zand
- University of California San Diego, La Jolla, CA, USA
| | - Le Zhang
- Yale School of Medicine, New Haven, CT, USA
| | - Jing Zhang
- University of California Irvine, Irvine, CA, USA
| | | | - Shilpa Buch
- University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | - Howard S Fox
- University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | - Myriam Heiman
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ya-Chi Ho
- Yale School of Medicine, New Haven, CT, USA
| | - Manolis Kellis
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paul J Kenny
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - David J Moore
- University of California San Diego, La Jolla, CA, USA
| | - Susan Morgello
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Tariq M Rana
- University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | - David J Volsky
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Owen R White
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
81
|
Bonev B, Castelo-Branco G, Chen F, Codeluppi S, Corces MR, Fan J, Heiman M, Harris K, Inoue F, Kellis M, Levine A, Lotfollahi M, Luo C, Maynard KR, Nitzan M, Ramani V, Satijia R, Schirmer L, Shen Y, Sun N, Green GS, Theis F, Wang X, Welch JD, Gokce O, Konopka G, Liddelow S, Macosko E, Ali Bayraktar O, Habib N, Nowakowski TJ. Opportunities and challenges of single-cell and spatially resolved genomics methods for neuroscience discovery. Nat Neurosci 2024; 27:2292-2309. [PMID: 39627587 PMCID: PMC11999325 DOI: 10.1038/s41593-024-01806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/23/2024] [Indexed: 12/13/2024]
Abstract
Over the past decade, single-cell genomics technologies have allowed scalable profiling of cell-type-specific features, which has substantially increased our ability to study cellular diversity and transcriptional programs in heterogeneous tissues. Yet our understanding of mechanisms of gene regulation or the rules that govern interactions between cell types is still limited. The advent of new computational pipelines and technologies, such as single-cell epigenomics and spatially resolved transcriptomics, has created opportunities to explore two new axes of biological variation: cell-intrinsic regulation of cell states and expression programs and interactions between cells. Here, we summarize the most promising and robust technologies in these areas, discuss their strengths and limitations and discuss key computational approaches for analysis of these complex datasets. We highlight how data sharing and integration, documentation, visualization and benchmarking of results contribute to transparency, reproducibility, collaboration and democratization in neuroscience, and discuss needs and opportunities for future technology development and analysis.
Collapse
Affiliation(s)
- Boyan Bonev
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fei Chen
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Myriam Heiman
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | - Kenneth Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Fumitaka Inoue
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Manolis Kellis
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ariel Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Mo Lotfollahi
- Institute of Computational Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Chongyuan Luo
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mor Nitzan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vijay Ramani
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, San Francisco, CA, USA
| | - Rahul Satijia
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Lucas Schirmer
- Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yin Shen
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Na Sun
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gilad S Green
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Fabian Theis
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xiao Wang
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Ozgun Gokce
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Shane Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Evan Macosko
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | | | - Naomi Habib
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Tomasz J Nowakowski
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
82
|
Tahmasian N, Feng MY, Arbabi K, Rusu B, Cao W, Kukreja B, Lubotzky A, Wainberg M, Tripathy SJ, Kalish BT. Neonatal Brain Injury Triggers Niche-Specific Changes to Cellular Biogeography. eNeuro 2024; 11:ENEURO.0224-24.2024. [PMID: 39681473 DOI: 10.1523/eneuro.0224-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Preterm infants are at risk for brain injury and neurodevelopmental impairment due, in part, to white matter injury following chronic hypoxia exposure. However, the precise molecular mechanisms by which neonatal hypoxia disrupts early neurodevelopment are poorly understood. Here, we constructed a brain-wide map of the regenerative response to newborn brain injury using high-resolution imaging-based spatial transcriptomics to analyze over 800,000 cells in a mouse model of chronic neonatal hypoxia. Additionally, we developed a new method for inferring condition-associated differences in cell type spatial proximity, enabling the identification of niche-specific changes in cellular architecture. We observed hypoxia-associated changes in region-specific cell states, cell type composition, and spatial organization. Importantly, our analysis revealed mechanisms underlying reparative neurogenesis and gliogenesis, while also nominating pathways that may impede circuit rewiring following neonatal hypoxia. Altogether, our work provides a comprehensive description of the molecular response to newborn brain injury.
Collapse
Affiliation(s)
- Nareh Tahmasian
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Min Yi Feng
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Keon Arbabi
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
| | - Bianca Rusu
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Wuxinhao Cao
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Bharti Kukreja
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Asael Lubotzky
- Division of Neurology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| | - Michael Wainberg
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario M5G 1X5, Canada
| | - Shreejoy J Tripathy
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, Ontario M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Brian T Kalish
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
83
|
Yen A, Sarafinovska S, Chen X, Skinner DD, Leti F, Crosby M, Hoisington-Lopez J, Wu Y, Chen J, Li ZA, Noguchi KK, Mitra RD, Dougherty JD. MYT1L deficiency impairs excitatory neuron trajectory during cortical development. Nat Commun 2024; 15:10308. [PMID: 39604385 PMCID: PMC11603064 DOI: 10.1038/s41467-024-54371-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Mutations reducing the function of MYT1L, a neuron-specific transcription factor, are associated with a syndromic neurodevelopmental disorder. MYT1L is used as a pro-neural factor in fibroblast-to-neuron transdifferentiation and is hypothesized to influence neuronal specification and maturation, but it is not clear which neuron types are most impacted by MYT1L loss. In this study, we profile 412,132 nuclei from the forebrains of wild-type and MYT1L-deficient mice at three developmental stages: E14 at the peak of neurogenesis, P1 when cortical neurons have been born, and P21 when neurons are maturing, to examine the role of MYT1L levels on neuronal development. MYT1L deficiency disrupts cortical neuron proportions and gene expression, primarily affecting neuronal maturation programs. Effects are mostly cell autonomous and persistent through development. While MYT1L can both activate and repress gene expression, the repressive effects are most sensitive to haploinsufficiency, likely mediating MYT1L syndrome. These findings illuminate MYT1L's role in orchestrating gene expression during neuronal development, providing insights into the molecular underpinnings of MYT1L syndrome.
Collapse
Affiliation(s)
- Allen Yen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Simona Sarafinovska
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Xuhua Chen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | | | | | - MariaLynn Crosby
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
- DNA Sequencing and Innovation Lab, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jessica Hoisington-Lopez
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
- DNA Sequencing and Innovation Lab, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yizhe Wu
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jiayang Chen
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Zipeng A Li
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kevin K Noguchi
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Robi D Mitra
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
84
|
Iyer L, Johnson K, Collier S, Koretsky AP, Petrus E. Post-Critical Period Transcriptional and Physiological Adaptations of Thalamocortical Connections after Sensory Loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624130. [PMID: 39876977 PMCID: PMC11774545 DOI: 10.1101/2024.11.19.624130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Unilateral whisker denervation activates plasticity mechanisms and circuit adaptations in adults. Single nucleus RNA sequencing and multiplex fluorescence in situ hybridization revealed differentially expressed genes related to altered glutamate receptor distributions and synaptogenesis in thalamocortical (TC) recipient layer 4 (L4) neurons of the sensory cortex, specifically those receiving input from the intact whiskers after whisker denervation. Electrophysiology detected increased spontaneous excitatory events at L4 neurons, confirming an increase in synaptic connections. Elevated expression levels of Gria2 mRNA and functional GluA2 subunit of AMPA receptors at the TC synapse indicate the presence of stabilized and potentiated TC synapses to L4 excitatory neurons along the intact pathway after unilateral whisker denervation. These adaptations likely underlie the increased cortical activity observed in rodents during intact whisker sensation after unilateral whisker denervation. Our findings provide new insights into the mechanisms by which the adult brain supports recovery after unilateral sensory loss.
Collapse
|
85
|
Chen CH, Yao Z, Wu S, Regehr WG. Characterization of direct Purkinje cell outputs to the brainstem. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608221. [PMID: 39605653 PMCID: PMC11601412 DOI: 10.1101/2024.08.16.608221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Purkinje cells (PCs) primarily project to cerebellar nuclei but also directly innervate the brainstem. Some PC-brainstem projections have been described previously, but most have not been thoroughly characterized. Here we use a PC-specific cre line to anatomically and electrophysiologically characterize PC projections to the brainstem. PC synapses are surprisingly widespread, with the highest densities found in the vestibular and parabrachial nuclei. However, there are pronounced regional differences in synaptic densities within both the vestibular and parabrachial nuclei. Large optogenetically-evoked PC-IPSCs are preferentially observed in subregions with the highest densities of PC synapses, suggesting that PCs selectively influence these areas and the behaviors they regulate. Unexpectedly, the pontine central gray and nearby subnuclei also contained a low density of PC synapses, and large PC-IPSCs are observed in a small fraction of cells. We combined electrophysiological recordings with immunohistochemistry to assess the molecular identities of two putative PC targets: PC synapses onto mesencephalic trigeminal neurons were not observed even though these cells are in close proximity to PC boutons. PC synapses onto locus coeruleus neurons are exceedingly rare or absent, even though previous studies concluded that PCs are a major input to these neurons. The availability of a highly selective cre line for PCs allowed us to study functional synapses, while avoiding complications that can accompany the use of viral approaches. We conclude that PCs directly innervate numerous brainstem nuclei, but only inhibit a small fraction of cells in many nuclei. This suggests that PCs target cell types with specific behavioral roles in brainstem regions.
Collapse
Affiliation(s)
- Christopher H. Chen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, Pennsylvania
- These authors contributed equally
| | - Zhiyi Yao
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Neural and Behavioral Sciences, The Pennsylvania State University, Hershey, Pennsylvania
- These authors contributed equally
| | - Shuting Wu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Wade G. Regehr
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
86
|
Ceballos CC, Ma L, Qin M, Zhong H. Widespread co-release of glutamate and GABA throughout the mouse brain. Commun Biol 2024; 7:1502. [PMID: 39537846 PMCID: PMC11560972 DOI: 10.1038/s42003-024-07198-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: 06/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Several brain neuronal populations transmit both the excitatory and inhibitory neurotransmitters, glutamate, and GABA. However, it remains largely unknown whether these opposing neurotransmitters are co-released simultaneously or are independently transmitted at different times and locations. By recording from acute mouse brain slices, we observed biphasic miniature postsynaptic currents, i.e., minis with time-locked excitatory and inhibitory currents, in striatal spiny projection neurons. This observation cannot be explained by accidental coincidence of monophasic excitatory and inhibitory minis. Interestingly, these biphasic minis could either be an excitatory current leading an inhibitory current or vice versa. Deletion of dopaminergic neurons did not eliminate biphasic minis, indicating that they originate from another source. Importantly, we found that both types of biphasic minis were present in multiple striatal neuronal types and in nine out of ten other brain regions. Overall, co-release of glutamate and GABA appears to be a widespread mode of neurotransmission in the brain.
Collapse
Affiliation(s)
- Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
87
|
Wang MG, Chen L, Zhang XF. Dual decoding of cell types and gene expression in spatial transcriptomics with PANDA. Nucleic Acids Res 2024; 52:12173-12190. [PMID: 39404057 PMCID: PMC11551751 DOI: 10.1093/nar/gkae876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/24/2024] [Accepted: 09/24/2024] [Indexed: 11/12/2024] Open
Abstract
Sequencing-based spatial transcriptomics technologies have revolutionized our understanding of complex biological systems by enabling transcriptome profiling while preserving spatial context. However, spot-level expression measurements often amalgamate signals from diverse cells, obscuring potential heterogeneity. Existing methods aim to deconvolute spatial transcriptomics data into cell type proportions for each spot using single-cell RNA sequencing references but overlook cell-type-specific gene expression, essential for uncovering intra-type heterogeneity. We present PANDA (ProbAbilistic-based decoNvolution with spot-aDaptive cell type signAtures), a novel method that concurrently deciphers spot-level gene expression into both cell type proportions and cell-type-specific gene expression. PANDA integrates archetypal analysis to capture within-cell-type heterogeneity and dynamically learns cell type signatures for each spot during deconvolution. Simulations demonstrate PANDA's superior performance. Applied to real spatial transcriptomics data from diverse tissues, including tumor, brain, and developing heart, PANDA reconstructs spatial structures and reveals subtle transcriptional variations within specific cell types, offering a comprehensive understanding of tissue dynamics.
Collapse
Affiliation(s)
- Meng-Guo Wang
- School of Mathematics and Statistics, and Hubei Key Lab–Math. Sci., Central China Normal University, Wuhan 430079, Hubei, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, Zhejiang, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai 519031, Guangdong, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics, and Hubei Key Lab–Math. Sci., Central China Normal University, Wuhan 430079, Hubei, China
- Key Laboratory of Nonlinear Analysis & Applications (Ministry of Education), Central China Normal University, Wuhan 430079, Hubei, China
| |
Collapse
|
88
|
Bartels T, Rowitch DH, Bayraktar OA. Generation of Mammalian Astrocyte Functional Heterogeneity. Cold Spring Harb Perspect Biol 2024; 16:a041351. [PMID: 38692833 PMCID: PMC11529848 DOI: 10.1101/cshperspect.a041351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Mammalian astrocytes have regional roles within the brain parenchyma. Indeed, the notion that astrocytes are molecularly heterogeneous could help explain how the central nervous system (CNS) retains embryonic positional information through development into specialized regions into adulthood. A growing body of evidence supports the concept of morphological and molecular differences between astrocytes in different brain regions, which might relate to their derivation from regionally patterned radial glia and/or local neuron inductive cues. Here, we review evidence for regionally encoded functions of astrocytes to provide an integrated concept on lineage origins and heterogeneity to understand regional brain organization, as well as emerging technologies to identify and further investigate novel roles for astrocytes.
Collapse
Affiliation(s)
- Theresa Bartels
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - David H Rowitch
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
89
|
Chen X. Reimagining Cortical Connectivity by Deconstructing Its Molecular Logic into Building Blocks. Cold Spring Harb Perspect Biol 2024; 16:a041509. [PMID: 38621822 PMCID: PMC11529856 DOI: 10.1101/cshperspect.a041509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Comprehensive maps of neuronal connectivity provide a foundation for understanding the structure of neural circuits. In a circuit, neurons are diverse in morphology, electrophysiology, gene expression, activity, and other neuronal properties. Thus, constructing a comprehensive connectivity map requires associating various properties of neurons, including their connectivity, at cellular resolution. A commonly used approach is to use the gene expression profiles as an anchor to which all other neuronal properties are associated. Recent advances in genomics and anatomical techniques dramatically improved the ability to determine and associate the long-range projections of neurons with their gene expression profiles. These studies revealed unprecedented details of the gene-projection relationship, but also highlighted conceptual challenges in understanding this relationship. In this article, I delve into the findings and the challenges revealed by recent studies using state-of-the-art neuroanatomical and transcriptomic techniques. Building upon these insights, I propose an approach that focuses on understanding the gene-projection relationship through basic features in gene expression profiles and projections, respectively, that associate with underlying cellular processes. I then discuss how the developmental trajectories of projections and gene expression profiles create additional challenges and necessitate interrogating the gene-projection relationship across time. Finally, I explore complementary strategies that, together, can provide a comprehensive view of the gene-projection relationship.
Collapse
Affiliation(s)
- Xiaoyin Chen
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| |
Collapse
|
90
|
Cao J, Zheng Z, Sun D, Chen X, Cheng R, Lv T, An Y, Zheng J, Song J, Wu L, Yang C. Decoder-seq enhances mRNA capture efficiency in spatial RNA sequencing. Nat Biotechnol 2024; 42:1735-1746. [PMID: 38228777 DOI: 10.1038/s41587-023-02086-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024]
Abstract
Spatial transcriptomics technologies with high resolution often lack high sensitivity in mRNA detection. Here we report a dendrimeric DNA coordinate barcoding design for spatial RNA sequencing (Decoder-seq), which offers both high sensitivity and high resolution. Decoder-seq combines dendrimeric nanosubstrates with microfluidic coordinate barcoding to generate spatial arrays with a DNA density approximately ten times higher than previously reported methods while maintaining flexibility in resolution. We show that the high RNA capture efficiency of Decoder-seq improved the detection of lowly expressed olfactory receptor (Olfr) genes in mouse olfactory bulbs and contributed to the discovery of a unique layer enrichment pattern for two Olfr genes. The near-cellular resolution provided by Decoder-seq has enabled the construction of a spatial single-cell atlas of the mouse hippocampus, revealing dendrite-enriched mRNAs in neurons. When applying Decoder-seq to human renal cell carcinomas, we dissected the heterogeneous tumor microenvironment across different cancer subtypes and identified spatial gradient-expressed genes related to epithelial-mesenchymal transition with the potential to predict tumor prognosis and progression.
Collapse
Affiliation(s)
- Jiao Cao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhong Zheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Sun
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Cheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianpeng Lv
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu An
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junhua Zheng
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jia Song
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lingling Wu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chaoyong Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, State Key Laboratory of Physical Chemical of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, China.
| |
Collapse
|
91
|
Sheng J, Wang D. VAPOR: Variational autoencoder with transport operators decouples co-occurring biological processes in development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620534. [PMID: 39554007 PMCID: PMC11565819 DOI: 10.1101/2024.10.27.620534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Background Emerging single-cell and spatial transcriptomic data enable the investigation of gene expression dynamics of various biological processes, especially for development. To this end, existing computational methods typically infer trajectories that sequentially order cells for revealing gene expression changes in development, e.g., to assign a pseudotime to each cell indicating the ordering. However, these trajectories can aggregate different biological processes that cells undergo simultaneously-such as maturation for specialized function and differentiation into specific cell types-which do not occur on the same timescale. Therefore, a single pseudotime axis may not distinguish gene expression dynamics from co-occurring processes. Methods We introduce a method, VAPOR (variational autoencoder with transport operators), to decouple dynamic patterns from developmental gene expression data. Particularly, VAPOR learns a latent space for gene expression dynamics and decomposes the space into multiple subspaces. The dynamics on each subspace are governed by an ordinary differential equation model, attempting to recapitulate specific biological processes. Furthermore, we can infer the process-specific pseudotimes, revealing multifaceted timescales of distinct processes in which cells may simultaneously be involved during development. Results Initially tested on simulated datasets, VAPOR effectively recovered the topology and decoupled distinct dynamic patterns in the data. We then applied VAPOR to a developmental human brain scRNA-seq dataset across postconceptional weeks and identified gene expression dynamics for several key processes, such as differentiation and maturation. Moreover, our benchmarking analyses also demonstrated the outperformance of VAPOR over other methods. Additionally, we applied VAPOR to spatial transcriptomics data in the human dorsolateral prefrontal cortex. VAPOR captured the 'inside-out' pattern across cortical layers, potentially revealing how layers were formed, characterized by their gene expression dynamics. Conclusion VAPOR is open source for general use ( https://github.com/daifengwanglab/VAPOR ) to parameterize and infer developmental gene expression dynamics. It can be further extended for other single-cell and spatial omics such as chromatin accessibility to reveal developmental epigenomic dynamics.
Collapse
|
92
|
Howe JR, Chan CL, Lee D, Blanquart M, Lee JH, Romero HK, Zadina AN, Lemieux ME, Mills F, Desplats PA, Tye KM, Root CM. Control of innate olfactory valence by segregated cortical amygdala circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600895. [PMID: 38979308 PMCID: PMC11230396 DOI: 10.1101/2024.06.26.600895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Animals exhibit innate behaviors that are stereotyped responses to specific evolutionarily relevant stimuli in the absence of prior learning or experience. These behaviors can be reduced to an axis of valence, whereby specific odors evoke approach or avoidance responses. The posterolateral cortical amygdala (plCoA) mediates innate attraction and aversion to odor. However, little is known about how this brain area gives rise to behaviors of opposing motivational valence. Here, we sought to define the circuit features of plCoA that give rise to innate attraction and aversion to odor. We characterized the physiology, gene expression, and projections of this structure, identifying a divergent, topographic organization that selectively controls innate attraction and avoidance to odor. First, we examined odor-evoked responses in these areas and found sparse encoding of odor identity, but not valence. We next considered a topographic organization and found that optogenetic stimulation of the anterior and posterior domains of plCoA elicits attraction and avoidance, respectively, suggesting a functional axis for valence. Using single cell and spatial RNA sequencing, we identified the molecular cell types in plCoA, revealing an anteroposterior gradient in cell types, whereby anterior glutamatergic neurons preferentially express VGluT2 and posterior neurons express VGluT1. Activation of these respective cell types recapitulates appetitive and aversive behaviors, and chemogenetic inhibition reveals partial necessity for responses to innate appetitive or aversive odors. Finally, we identified topographically organized circuits defined by projections, whereby anterior neurons preferentially project to medial amygdala, and posterior neurons preferentially project to nucleus accumbens, which are respectively sufficient and necessary for innate attraction and aversion. Together, these data advance our understanding of how the olfactory system generates stereotypic, hardwired attraction and avoidance, and supports a model whereby distinct, topographically distributed plCoA populations direct innate olfactory responses by signaling to divergent valence-specific targets, linking upstream olfactory identity to downstream valence behaviors, through a population code. This suggests a novel amygdala circuit motif in which valence encoding is represented not by the firing properties of individual neurons, but by population level identity encoding that is routed through divergent targets to mediate distinct behaviors of opposing appetitive and aversive responses.
Collapse
Affiliation(s)
- James R. Howe
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Chung-Lung Chan
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
- These authors contributed equally
| | - Donghyung Lee
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marlon Blanquart
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - James H. Lee
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Haylie K. Romero
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Abigail N. Zadina
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
| | | | - Fergil Mills
- Salk Institute for Biological Sciences, La Jolla, CA 92037, USA
| | - Paula A. Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- Center for Circadian Biology, University of California, San Diego, La Jolla, CA 92093, USA
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kay M. Tye
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
- Salk Institute for Biological Sciences, La Jolla, CA 92037, USA
- Howard Hughes Medical Institute, La Jolla, CA 92037, USA
| | - Cory M. Root
- Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
93
|
Vatsa N, Brynildsen JK, Goralski TM, Kurgat K, Meyerdirk L, Breton L, DeWeerd D, Brasseur L, Turner L, Becker K, Gallik KL, Bassett DS, Henderson MX. Network analysis of α-synuclein pathology progression reveals p21-activated kinases as regulators of vulnerability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619411. [PMID: 39484617 PMCID: PMC11526907 DOI: 10.1101/2024.10.22.619411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
α-Synuclein misfolding and progressive accumulation drives a pathogenic process in Parkinson's disease. To understand cellular and network vulnerability to α-synuclein pathology, we developed a framework to quantify network-level vulnerability and identify new therapeutic targets at the cellular level. Full brain α-synuclein pathology was mapped in mice over 9 months. Empirical pathology data was compared to theoretical pathology estimates from a diffusion model of pathology progression along anatomical connections. Unexplained variance in the model enabled us to derive regional vulnerability that we compared to regional gene expression. We identified gene expression patterns that relate to regional vulnerability, including 12 kinases that were enriched in vulnerable regions. Among these, an inhibitor of group II PAKs demonstrated protection from neuron death and α-synuclein pathology, even after delayed compound treatment. This study provides a framework for the derivation of cellular vulnerability from network-based studies and identifies a promising therapeutic pathway for Parkinson's disease.
Collapse
Affiliation(s)
- Naman Vatsa
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Julia K. Brynildsen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas M. Goralski
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Kevin Kurgat
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Lindsay Meyerdirk
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Libby Breton
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniella DeWeerd
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Brasseur
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | | | | | - Dani S. Bassett
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Electrical & Systems Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Santa Fe Institute, Santa Fe, NM, USA
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Michael X. Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Lead Contact
| |
Collapse
|
94
|
Kronman FN, Liwang JK, Betty R, Vanselow DJ, Wu YT, Tustison NJ, Bhandiwad A, Manjila SB, Minteer JA, Shin D, Lee CH, Patil R, Duda JT, Xue J, Lin Y, Cheng KC, Puelles L, Gee JC, Zhang J, Ng L, Kim Y. Developmental mouse brain common coordinate framework. Nat Commun 2024; 15:9072. [PMID: 39433760 PMCID: PMC11494176 DOI: 10.1038/s41467-024-53254-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
3D brain atlases are key resources to understand the brain's spatial organization and promote interoperability across different studies. However, unlike the adult mouse brain, the lack of developing mouse brain 3D reference atlases hinders advancements in understanding brain development. Here, we present a 3D developmental common coordinate framework (DevCCF) spanning embryonic day (E)11.5, E13.5, E15.5, E18.5, and postnatal day (P)4, P14, and P56, featuring undistorted morphologically averaged atlas templates created from magnetic resonance imaging and co-registered high-resolution light sheet fluorescence microscopy templates. The DevCCF with 3D anatomical segmentations can be downloaded or explored via an interactive 3D web-visualizer. As a use case, we utilize the DevCCF to unveil GABAergic neuron emergence in embryonic brains. Moreover, we map the Allen CCFv3 and spatial transcriptome cell-type data to our stereotaxic P56 atlas. In summary, the DevCCF is an openly accessible resource for multi-study data integration to advance our understanding of brain development.
Collapse
Affiliation(s)
- Fae N Kronman
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Josephine K Liwang
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Rebecca Betty
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Daniel J Vanselow
- Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Nicholas J Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | | | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jennifer A Minteer
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Choong Heon Lee
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Rohan Patil
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jeffrey T Duda
- Department of Radiology, Penn Image Computing and Science Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Xue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yingxi Lin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Keith C Cheng
- Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Universidad de Murcia, and Murcia Arrixaca Institute for Biomedical Research (IMIB), Murcia, Spain
| | - James C Gee
- Department of Radiology, Penn Image Computing and Science Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA.
| |
Collapse
|
95
|
De Filippo R, Schmitz D. Transcriptomic mapping of the 5-HT receptor landscape. PATTERNS (NEW YORK, N.Y.) 2024; 5:101048. [PMID: 39569210 PMCID: PMC11574285 DOI: 10.1016/j.patter.2024.101048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/19/2024] [Accepted: 07/31/2024] [Indexed: 11/22/2024]
Abstract
Serotonin (5-HT) is crucial for regulating brain functions such as mood, sleep, and cognition. This study presents a comprehensive transcriptomic analysis of 5-HT receptors (Htrs) across ≈4 million cells in the adult mouse brain using single-cell RNA sequencing (scRNA-seq) data from the Allen Institute. We observed differential transcription patterns of all 14 Htr subtypes, revealing diverse prevalence and distribution across cell classes. Remarkably, we found that 65.84% of cells transcribe RNA of at least one Htr, with frequent co-transcription of multiple Htrs, underscoring the complexity of the 5-HT system even at the single-cell dimension. Leveraging a multiplexed error-robust fluorescence in situ hybridization (MERFISH) dataset provided by Harvard University of ≈10 million cells, we analyzed the spatial distribution of each Htr, confirming previous findings and uncovering novel transcription patterns. To aid in exploring Htr transcription, we provide an online interactive visualizer.
Collapse
Affiliation(s)
- Roberto De Filippo
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
| | - Dietmar Schmitz
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117 Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, Einstein Center for Neuroscience, 10117 Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, 10117 Berlin, Germany
- Humboldt-Universität zu Berlin, Bernstein Center for Computational Neuroscience, Philippstr. 13, 10115 Berlin, Germany
| |
Collapse
|
96
|
Liu Y, Bech P, Tamura K, Délez LT, Crochet S, Petersen CCH. Cell class-specific long-range axonal projections of neurons in mouse whisker-related somatosensory cortices. eLife 2024; 13:RP97602. [PMID: 39392390 PMCID: PMC11469677 DOI: 10.7554/elife.97602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024] Open
Abstract
Long-range axonal projections of diverse classes of neocortical excitatory neurons likely contribute to brain-wide interactions processing sensory, cognitive and motor signals. Here, we performed light-sheet imaging of fluorescently labeled axons from genetically defined neurons located in posterior primary somatosensory barrel cortex and supplemental somatosensory cortex. We used convolutional networks to segment axon-containing voxels and quantified their distribution within the Allen Mouse Brain Atlas Common Coordinate Framework. Axonal density was analyzed for different classes of glutamatergic neurons using transgenic mouse lines selectively expressing Cre recombinase in layer 2/3 intratelencephalic projection neurons (Rasgrf2-dCre), layer 4 intratelencephalic projection neurons (Scnn1a-Cre), layer 5 intratelencephalic projection neurons (Tlx3-Cre), layer 5 pyramidal tract projection neurons (Sim1-Cre), layer 5 projection neurons (Rbp4-Cre), and layer 6 corticothalamic neurons (Ntsr1-Cre). We found distinct axonal projections from the different neuronal classes to many downstream brain areas, which were largely similar for primary and supplementary somatosensory cortices. Functional connectivity maps obtained from optogenetic activation of sensory cortex and wide-field imaging revealed topographically organized evoked activity in frontal cortex with neurons located more laterally in somatosensory cortex signaling to more anteriorly located regions in motor cortex, consistent with the anatomical projections. The current methodology therefore appears to quantify brain-wide axonal innervation patterns supporting brain-wide signaling.
Collapse
Affiliation(s)
- Yanqi Liu
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Pol Bech
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Keita Tamura
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridgeUnited Kingdom
- International Research Center for Medical Sciences, Kumamoto UniversityKumamotoJapan
| | - Lucas T Délez
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Sylvain Crochet
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| | - Carl CH Petersen
- Laboratory of Sensory Processing, Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL)LausanneSwitzerland
| |
Collapse
|
97
|
Manoharan VT, Abdelkareem A, Gill G, Brown S, Gillmor A, Hall C, Seo H, Narta K, Grewal S, Dang NH, Ahn BY, Osz K, Lun X, Mah L, Zemp F, Mahoney D, Senger DL, Chan JA, Morrissy AS. Spatiotemporal modeling reveals high-resolution invasion states in glioblastoma. Genome Biol 2024; 25:264. [PMID: 39390467 PMCID: PMC11465563 DOI: 10.1186/s13059-024-03407-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 09/29/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Diffuse invasion of glioblastoma cells through normal brain tissue is a key contributor to tumor aggressiveness, resistance to conventional therapies, and dismal prognosis in patients. A deeper understanding of how components of the tumor microenvironment (TME) contribute to overall tumor organization and to programs of invasion may reveal opportunities for improved therapeutic strategies. RESULTS Towards this goal, we apply a novel computational workflow to a spatiotemporally profiled GBM xenograft cohort, leveraging the ability to distinguish human tumor from mouse TME to overcome previous limitations in the analysis of diffuse invasion. Our analytic approach, based on unsupervised deconvolution, performs reference-free discovery of cell types and cell activities within the complete GBM ecosystem. We present a comprehensive catalogue of 15 tumor cell programs set within the spatiotemporal context of 90 mouse brain and TME cell types, cell activities, and anatomic structures. Distinct tumor programs related to invasion align with routes of perivascular, white matter, and parenchymal invasion. Furthermore, sub-modules of genes serving as program network hubs are highly prognostic in GBM patients. CONCLUSION The compendium of programs presented here provides a basis for rational targeting of tumor and/or TME components. We anticipate that our approach will facilitate an ecosystem-level understanding of the immediate and long-term consequences of such perturbations, including the identification of compensatory programs that will inform improved combinatorial therapies.
Collapse
Affiliation(s)
- Varsha Thoppey Manoharan
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Aly Abdelkareem
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Gurveer Gill
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Samuel Brown
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Aaron Gillmor
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Courtney Hall
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Heewon Seo
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Kiran Narta
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Sean Grewal
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Ngoc Ha Dang
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Bo Young Ahn
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Kata Osz
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Xueqing Lun
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Laura Mah
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Franz Zemp
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Douglas Mahoney
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Microbiology, Immunology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Donna L Senger
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.
| | - Jennifer A Chan
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| | - A Sorana Morrissy
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada.
- Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
98
|
Hemminger Z, Sanchez-Tam G, Ocampo HD, Wang A, Underwood T, Xie F, Zhao Q, Song D, Li JJ, Dong H, Wollman R. Spatial Single-Cell Mapping of Transcriptional Differences Across Genetic Backgrounds in Mouse Brains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617260. [PMID: 39416191 PMCID: PMC11483037 DOI: 10.1101/2024.10.08.617260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Genetic variation can alter brain structure and, consequently, function. Comparative statistical analysis of mouse brains across genetic backgrounds requires spatial, single-cell, atlas-scale data, in replicates-a challenge for current technologies. We introduce Atlas-scale Transcriptome Localization using Aggregate Signatures (ATLAS), a scalable tissue mapping method. ATLAS learns transcriptional signatures from scRNAseq data, encodes them in situ with tens of thousands of oligonucleotide probes, and decodes them to infer cell types and imputed transcriptomes. We validated ATLAS by comparing its cell type inferences with direct MERFISH measurements of marker genes and quantitative comparisons to four other technologies. Using ATLAS, we mapped the central brains of five male and five female C57BL/6J (B6) mice and five male BTBR T+ tf/J (BTBR) mice, an idiopathic model of autism, collectively profiling over 40 million cells across over 400 coronal sections. Our analysis revealed over 40 significant differences in cell type distributions and identified 16 regional composition changes across male-female and B6-BTBR comparisons. ATLAS thus enables systematic comparative studies, facilitating organ-level structure-function analysis of disease models.
Collapse
Affiliation(s)
| | | | | | - Aihui Wang
- Department of Chemistry and Biochemistry, UCLA
| | | | - Fangming Xie
- Department of Chemical Biology, David Geffen School of Medicine at UCLA
| | - Qiuying Zhao
- Department of Neurobiology, David Geffen School of Medicine at UCLA
| | | | - Jingyi Jessica Li
- Department of Statistics and Data Science, UCLA
- Institute of Quantitative Biosciences, UCLA
| | - Hongwei Dong
- Department of Neurobiology, David Geffen School of Medicine at UCLA
| | - Roy Wollman
- Department of Chemistry and Biochemistry, UCLA
- Institute of Quantitative Biosciences, UCLA
- Department of Integrative Biology and Physiology, UCLA
| |
Collapse
|
99
|
Butrus S, Monday HR, Yoo CJ, Feldman DE, Shekhar K. Molecular states underlying neuronal cell type development and plasticity in the whisker cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617106. [PMID: 39416021 PMCID: PMC11482765 DOI: 10.1101/2024.10.07.617106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Mouse whisker somatosensory cortex (wS1) is a major model system to study the experience-dependent plasticity of cortical neuron physiology, morphology, and sensory coding. However, the role of sensory experience in regulating neuronal cell type development and gene expression in wS1 remains poorly understood. We assembled and annotated a transcriptomic atlas of wS1 during postnatal development comprising 45 molecularly distinct neuronal types that can be grouped into eight excitatory and four inhibitory neuron subclasses. Using this atlas, we examined the influence of whisker experience from postnatal day (P) 12, the onset of active whisking, to P22, on the maturation of molecularly distinct cell types. During this developmental period, when whisker experience was normal, ~250 genes were regulated in a neuronal subclass-specific fashion. At the resolution of neuronal types, we found that only the composition of layer (L) 2/3 glutamatergic neuronal types, but not other neuronal types, changed substantially between P12 and P22. These compositional changes resemble those observed previously in the primary visual cortex (V1), and the temporal gene expression changes were also highly conserved between the two regions. In contrast to V1, however, cell type maturation in wS1 is not substantially dependent on sensory experience, as 10-day full-face whisker deprivation did not influence the transcriptomic identity and composition of L2/3 neuronal types. A one-day competitive whisker deprivation protocol also did not affect cell type identity but induced moderate changes in plasticity-related gene expression. Thus, developmental maturation of cell types is similar in V1 and wS1, but sensory deprivation minimally affects cell type development in wS1.
Collapse
Affiliation(s)
- Salwan Butrus
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hannah R. Monday
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher J. Yoo
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Daniel E. Feldman
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Center for Computational Biology; Vision Sciences and Optometry; University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
100
|
Kuralay A, McDonough MC, Resch JM. Control of sodium appetite by hindbrain aldosterone-sensitive neurons. Mol Cell Endocrinol 2024; 592:112323. [PMID: 38936597 PMCID: PMC11381173 DOI: 10.1016/j.mce.2024.112323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/25/2024] [Indexed: 06/29/2024]
Abstract
Mineralocorticoids play a key role in hydromineral balance by regulating sodium retention and potassium wasting. Through favoring sodium, mineralocorticoids can cause hypertension from fluid overload under conditions of hyperaldosteronism, such as aldosterone-secreting tumors. An often-overlooked mechanism by which aldosterone functions to increase sodium is through stimulation of salt appetite. To drive sodium intake, aldosterone targets neurons in the hindbrain which uniquely express 11β-hydroxysteroid dehydrogenase type 2 (HSD2). This enzyme is a necessary precondition for aldosterone-sensing cells as it metabolizes glucocorticoids - preventing their activation of the mineralocorticoid receptor. In this review, we will consider the role of hindbrain HSD2 neurons in regulating sodium appetite by discussing HSD2 expression in the brain, regulation of hindbrain HSD2 neuron activity, and the circuitry mediating the effects of these aldosterone-sensitive neurons. Reducing the activity of hindbrain HSD2 neurons may be a viable strategy to reduce sodium intake and cardiovascular risk, particularly for conditions of hyperaldosteronism.
Collapse
Affiliation(s)
- Ahmet Kuralay
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA
| | - Miriam C McDonough
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Jon M Resch
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA; Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA, USA; Molecular Medicine Graduate Program, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|