101
|
Lee AJ, Yao S, Lusk N, Ng L, Kunst M, Zeng H, Tasic B, Abbasi-Asl R. Data-driven fine-grained region discovery in the mouse brain with transformers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592608. [PMID: 38766132 PMCID: PMC11100623 DOI: 10.1101/2024.05.05.592608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Technologies such as spatial transcriptomics offer unique opportunities to define the spatial organization of the mouse brain. We developed an unsupervised training scheme and novel transformer-based deep learning architecture to detect spatial domains across the whole mouse brain using spatial transcriptomics data. Our model learns local representations of molecular and cellular statistical patterns which can be clustered to identify spatial domains within the brain from coarse to fine-grained. Discovered domains are spatially regular, even with several hundreds of spatial clusters. They are also consistent with existing anatomical ontologies such as the Allen Mouse Brain Common Coordinate Framework version 3 (CCFv3) and can be visually interpreted at the cell type or transcript level. We demonstrate our method can be used to identify previously uncatalogued subregions, such as in the midbrain, where we uncover gradients of inhibitory neuron complexity and abundance. Notably, these subregions cannot be discovered using other methods. We apply our method to a separate multi-animal whole-brain spatial transcriptomic dataset and show that our method can also robustly integrate spatial domains across animals.
Collapse
Affiliation(s)
- Alex J. Lee
- University of California, San Francisco
- Weill Institute for Neurosciences
| | | | | | - Lydia Ng
- Allen Institute for Brain Science
| | | | | | | | - Reza Abbasi-Asl
- University of California, San Francisco
- Weill Institute for Neurosciences
| |
Collapse
|
102
|
Atta L, Clifton K, Anant M, Aihara G, Fan J. Gene count normalization in single-cell imaging-based spatially resolved transcriptomics. Genome Biol 2024; 25:153. [PMID: 38867267 PMCID: PMC11167774 DOI: 10.1186/s13059-024-03303-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: 08/18/2023] [Accepted: 06/06/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Recent advances in imaging-based spatially resolved transcriptomics (im-SRT) technologies now enable high-throughput profiling of targeted genes and their locations in fixed tissues. Normalization of gene expression data is often needed to account for technical factors that may confound underlying biological signals. RESULTS Here, we investigate the potential impact of different gene count normalization methods with different targeted gene panels in the analysis and interpretation of im-SRT data. Using different simulated gene panels that overrepresent genes expressed in specific tissue regions or cell types, we demonstrate how normalization methods based on detected gene counts per cell differentially impact normalized gene expression magnitudes in a region- or cell type-specific manner. We show that these normalization-induced effects may reduce the reliability of downstream analyses including differential gene expression, gene fold change, and spatially variable gene analysis, introducing false positive and false negative results when compared to results obtained from gene panels that are more representative of the gene expression of the tissue's component cell types. These effects are not observed with normalization approaches that do not use detected gene counts for gene expression magnitude adjustment, such as with cell volume or cell area normalization. CONCLUSIONS We recommend using non-gene count-based normalization approaches when feasible and evaluating gene panel representativeness before using gene count-based normalization methods if necessary. Overall, we caution that the choice of normalization method and gene panel may impact the biological interpretation of the im-SRT data.
Collapse
Affiliation(s)
- Lyla Atta
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
| | - Kalen Clifton
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
| | - Manjari Anant
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Gohta Aihara
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Center for Computational Biology, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21211, USA.
| |
Collapse
|
103
|
Brown ST, Medina-Pizarro M, Holla M, Vaaga CE, Raman IM. Simple spike patterns and synaptic mechanisms encoding sensory and motor signals in Purkinje cells and the cerebellar nuclei. Neuron 2024; 112:1848-1861.e4. [PMID: 38492575 PMCID: PMC11156563 DOI: 10.1016/j.neuron.2024.02.014] [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: 03/28/2022] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 03/18/2024]
Abstract
Whisker stimulation in awake mice evokes transient suppression of simple spike probability in crus I/II Purkinje cells. Here, we investigated how simple spike suppression arises synaptically, what it encodes, and how it affects cerebellar output. In vitro, monosynaptic parallel fiber (PF)-excitatory postsynaptic currents (EPSCs) facilitated strongly, whereas disynaptic inhibitory postsynaptic currents (IPSCs) remained stable, maximizing relative inhibitory strength at the onset of PF activity. Short-term plasticity thus favors the inhibition of Purkinje spikes before PFs facilitate. In vivo, whisker stimulation evoked a 2-6 ms synchronous spike suppression, just 6-8 ms (∼4 synaptic delays) after sensory onset, whereas active whisker movements elicited broadly timed spike rate increases that did not modulate sensory-evoked suppression. Firing in the cerebellar nuclei (CbN) inversely correlated with disinhibition from sensory-evoked simple spike suppressions but was decoupled from slow, non-synchronous movement-associated elevations of Purkinje firing rates. Synchrony thus allows the CbN to high-pass filter Purkinje inputs, facilitating sensory-evoked cerebellar outputs that can drive movements.
Collapse
Affiliation(s)
- Spencer T Brown
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Mauricio Medina-Pizarro
- Department of Neurobiology, Northwestern University, Evanston, IL, USA; Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | - Meghana Holla
- Department of Neurobiology, Northwestern University, Evanston, IL, USA; Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA
| | | | - Indira M Raman
- Department of Neurobiology, Northwestern University, Evanston, IL, USA; Northwestern University Interdepartmental Neuroscience Program, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
104
|
Choi YK, Feng L, Jeong WK, Kim J. Connecto-informatics at the mesoscale: current advances in image processing and analysis for mapping the brain connectivity. Brain Inform 2024; 11:15. [PMID: 38833195 DOI: 10.1186/s40708-024-00228-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024] Open
Abstract
Mapping neural connections within the brain has been a fundamental goal in neuroscience to understand better its functions and changes that follow aging and diseases. Developments in imaging technology, such as microscopy and labeling tools, have allowed researchers to visualize this connectivity through high-resolution brain-wide imaging. With this, image processing and analysis have become more crucial. However, despite the wealth of neural images generated, access to an integrated image processing and analysis pipeline to process these data is challenging due to scattered information on available tools and methods. To map the neural connections, registration to atlases and feature extraction through segmentation and signal detection are necessary. In this review, our goal is to provide an updated overview of recent advances in these image-processing methods, with a particular focus on fluorescent images of the mouse brain. Our goal is to outline a pathway toward an integrated image-processing pipeline tailored for connecto-informatics. An integrated workflow of these image processing will facilitate researchers' approach to mapping brain connectivity to better understand complex brain networks and their underlying brain functions. By highlighting the image-processing tools available for fluroscent imaging of the mouse brain, this review will contribute to a deeper grasp of connecto-informatics, paving the way for better comprehension of brain connectivity and its implications.
Collapse
Affiliation(s)
- Yoon Kyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Department of Computer Science and Engineering, Korea University, Seoul, South Korea
| | | | - Won-Ki Jeong
- Department of Computer Science and Engineering, Korea University, Seoul, South Korea
| | - Jinhyun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea.
- Department of Computer Science and Engineering, Korea University, Seoul, South Korea.
- KIST-SKKU Brain Research Center, SKKU Institute for Convergence, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
105
|
Liu Z, Li A, Gong H, Yang X, Luo Q, Feng Z, Li X. The cytoarchitectonic landscape revealed by deep learning method facilitated precise positioning in mouse neocortex. Cereb Cortex 2024; 34:bhae229. [PMID: 38836835 DOI: 10.1093/cercor/bhae229] [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: 03/19/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/06/2024] Open
Abstract
Neocortex is a complex structure with different cortical sublayers and regions. However, the precise positioning of cortical regions can be challenging due to the absence of distinct landmarks without special preparation. To address this challenge, we developed a cytoarchitectonic landmark identification pipeline. The fluorescence micro-optical sectioning tomography method was employed to image the whole mouse brain stained by general fluorescent nucleotide dye. A fast 3D convolution network was subsequently utilized to segment neuronal somas in entire neocortex. By approach, the cortical cytoarchitectonic profile and the neuronal morphology were analyzed in 3D, eliminating the influence of section angle. And the distribution maps were generated that visualized the number of neurons across diverse morphological types, revealing the cytoarchitectonic landscape which characterizes the landmarks of cortical regions, especially the typical signal pattern of barrel cortex. Furthermore, the cortical regions of various ages were aligned using the generated cytoarchitectonic landmarks suggesting the structural changes of barrel cortex during the aging process. Moreover, we observed the spatiotemporally gradient distributions of spindly neurons, concentrated in the deep layer of primary visual area, with their proportion decreased over time. These findings could improve structural understanding of neocortex, paving the way for further exploration with this method.
Collapse
Affiliation(s)
- Zhixiang Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, No. 1037 Luoyu Road, Wuhan 430070, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, No. 1037 Luoyu Road, Wuhan 430070, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, No. 58 Renmin Road, Haikou 570228, China
- HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, No. 388 Ruoshui Road, Suzhou 215000, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, No. 1037 Luoyu Road, Wuhan 430070, China
- HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, No. 388 Ruoshui Road, Suzhou 215000, China
| | - Xiaoquan Yang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, No. 58 Renmin Road, Haikou 570228, China
- HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, No. 388 Ruoshui Road, Suzhou 215000, China
| | - Qingming Luo
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, No. 58 Renmin Road, Haikou 570228, China
| | - Zhao Feng
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, No. 58 Renmin Road, Haikou 570228, China
- HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, No. 388 Ruoshui Road, Suzhou 215000, China
| | - Xiangning Li
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, No. 58 Renmin Road, Haikou 570228, China
- HUST-Suzhou Institute for Brainsmatics, Jiangsu Industrial Technology Research Institute, No. 388 Ruoshui Road, Suzhou 215000, China
| |
Collapse
|
106
|
Kurz A, Müller H, Kather JN, Schneider L, Bucher TC, Brinker TJ. 3-Dimensional Reconstruction From Histopathological Sections: A Systematic Review. J Transl Med 2024; 104:102049. [PMID: 38513977 DOI: 10.1016/j.labinv.2024.102049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/18/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
Although pathological tissue analysis is typically performed on single 2-dimensional (2D) histologic reference slides, 3-dimensional (3D) reconstruction from a sequence of histologic sections could provide novel opportunities for spatial analysis of the extracted tissue. In this review, we analyze recent works published after 2018 and report information on the extracted tissue types, the section thickness, and the number of sections used for reconstruction. By analyzing the technological requirements for 3D reconstruction, we observe that software tools exist, both free and commercial, which include the functionality to perform 3D reconstruction from a sequence of histologic images. Through the analysis of the most recent works, we provide an overview of the workflows and tools that are currently used for 3D reconstruction from histologic sections and address points for future work, such as a missing common file format or computer-aided analysis of the reconstructed model.
Collapse
Affiliation(s)
- Alexander Kurz
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heimo Müller
- Diagnostics and Research Institute for Pathology, Medical University of Graz, Graz, Austria
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tabea C Bucher
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
107
|
Wan T, Fu C, Peng J, Lu J, Li P, Zhuo J. Repairing the in situ hybridization missing data in the hippocampus region by using a 3D residual U-Net model. BIOMEDICAL OPTICS EXPRESS 2024; 15:3541-3554. [PMID: 38867784 PMCID: PMC11166418 DOI: 10.1364/boe.522078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/31/2024] [Accepted: 04/22/2024] [Indexed: 06/14/2024]
Abstract
The hippocampus is a critical brain region. Transcriptome data provides valuable insights into the structure and function of the hippocampus at the gene level. However, transcriptome data is often incomplete. To address this issue, we use the convolutional neural network model to repair the missing voxels in the hippocampus region, based on Allen institute coronal slices in situ hybridization (ISH) dataset. Moreover, we analyze the gene expression correlation between coronal and sagittal dataset in the hippocampus region. The results demonstrated that the trend of gene expression correlation between the coronal and sagittal datasets remained consistent following the repair of missing data in the coronal ISH dataset. In the last, we use repaired ISH dataset to identify novel genes specific to hippocampal subregions. Our findings demonstrate the accuracy and effectiveness of using deep learning method to repair ISH missing data. After being repaired, ISH has the potential to improve our comprehension of the hippocampus's structure and function.
Collapse
Affiliation(s)
- Tong Wan
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Changping Fu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Jiinbo Peng
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Jinling Lu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215100, China
| | - Pengcheng Li
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
- Britton Chance Center for Biomedical Photonics and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Science, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou 215100, China
| | - JunJie Zhuo
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| |
Collapse
|
108
|
Zhang R, Huang D, Gasparini S, Geerling JC. Efferent projections of Nps-expressing neurons in the parabrachial region. J Comp Neurol 2024; 532:e25629. [PMID: 39031887 DOI: 10.1002/cne.25629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 07/22/2024]
Abstract
In the brain, connectivity determines function. Neurons in the parabrachial nucleus (PB) relay diverse information to widespread brain regions, but the connections and functions of PB neurons that express Nps (neuropeptide S, NPS) remain mysterious. Here, we use Cre-dependent anterograde tracing and whole-brain analysis to map their output connections. While many other PB neurons project ascending axons through the central tegmental tract, NPS axons reach the forebrain via distinct periventricular and ventral pathways. Along the periventricular pathway, NPS axons target the tectal longitudinal column and periaqueductal gray, then continue rostrally to target the paraventricular nucleus of the thalamus. Along the ventral pathway, NPS axons blanket much of the hypothalamus but avoid the ventromedial and mammillary nuclei. They also project prominently to the ventral bed nucleus of the stria terminalis, A13 cell group, and magnocellular subparafasciular nucleus. In the hindbrain, NPS axons have fewer descending projections, targeting primarily the superior salivatory nucleus, nucleus of the lateral lemniscus, and periolivary region. Combined with what is known already about NPS and its receptor, the output pattern of Nps-expressing neurons in the PB region predicts roles in threat response and circadian behavior.
Collapse
Affiliation(s)
- Richie Zhang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Dake Huang
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Silvia Gasparini
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Joel C Geerling
- Department of Neurology and Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
109
|
Zhang Y, Yuan L, Zhu Q, Wu J, Nöbauer T, Zhang R, Xiao G, Wang M, Xie H, Guo Z, Dai Q, Vaziri A. A miniaturized mesoscope for the large-scale single-neuron-resolved imaging of neuronal activity in freely behaving mice. Nat Biomed Eng 2024; 8:754-774. [PMID: 38902522 DOI: 10.1038/s41551-024-01226-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/03/2024] [Indexed: 06/22/2024]
Abstract
Exploring the relationship between neuronal dynamics and ethologically relevant behaviour involves recording neuronal-population activity using technologies that are compatible with unrestricted animal behaviour. However, head-mounted microscopes that accommodate weight limits to allow for free animal behaviour typically compromise field of view, resolution or depth range, and are susceptible to movement-induced artefacts. Here we report a miniaturized head-mounted fluorescent mesoscope that we systematically optimized for calcium imaging at single-neuron resolution, for increased fields of view and depth of field, and for robustness against motion-generated artefacts. Weighing less than 2.5 g, the mesoscope enabled recordings of neuronal-population activity at up to 16 Hz, with 4 μm resolution over 300 μm depth-of-field across a field of view of 3.6 × 3.6 mm2 in the cortex of freely moving mice. We used the mesoscope to record large-scale neuronal-population activity in socially interacting mice during free exploration and during fear-conditioning experiments, and to investigate neurovascular coupling across multiple cortical regions.
Collapse
Affiliation(s)
- Yuanlong Zhang
- Department of Automation, Tsinghua University, Beijing, China
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Lekang Yuan
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Qiyu Zhu
- School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Jiamin Wu
- Department of Automation, Tsinghua University, Beijing, China
| | - Tobias Nöbauer
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Rujin Zhang
- Department of Anesthesiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guihua Xiao
- Department of Automation, Tsinghua University, Beijing, China
| | - Mingrui Wang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Hao Xie
- Department of Automation, Tsinghua University, Beijing, China
| | - Zengcai Guo
- School of Medicine, Tsinghua University, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Qionghai Dai
- Department of Automation, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Alipasha Vaziri
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA.
- The Kavli Neural Systems Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
110
|
Vickers ED, McCormick DA. Pan-cortical 2-photon mesoscopic imaging and neurobehavioral alignment in awake, behaving mice. eLife 2024; 13:RP94167. [PMID: 38808733 PMCID: PMC11136495 DOI: 10.7554/elife.94167] [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: 05/30/2024] Open
Abstract
The flow of neural activity across the neocortex during active sensory discrimination is constrained by task-specific cognitive demands, movements, and internal states. During behavior, the brain appears to sample from a broad repertoire of activation motifs. Understanding how these patterns of local and global activity are selected in relation to both spontaneous and task-dependent behavior requires in-depth study of densely sampled activity at single neuron resolution across large regions of cortex. In a significant advance toward this goal, we developed procedures to record mesoscale 2-photon Ca2+ imaging data from two novel in vivo preparations that, between them, allow for simultaneous access to nearly all 0f the mouse dorsal and lateral neocortex. As a proof of principle, we aligned neural activity with both behavioral primitives and high-level motifs to reveal the existence of large populations of neurons that coordinated their activity across cortical areas with spontaneous changes in movement and/or arousal. The methods we detail here facilitate the identification and exploration of widespread, spatially heterogeneous neural ensembles whose activity is related to diverse aspects of behavior.
Collapse
Affiliation(s)
- Evan D Vickers
- Institute of Neuroscience, University of OregonEugeneUnited States
| | - David A McCormick
- Institute of Neuroscience, University of OregonEugeneUnited States
- Department of Biology, University of OregonEugeneUnited States
| |
Collapse
|
111
|
Lazari A, Tachrount M, Valverde JM, Papp D, Beauchamp A, McCarthy P, Ellegood J, Grandjean J, Johansen-Berg H, Zerbi V, Lerch JP, Mars RB. The mouse motor system contains multiple premotor areas and partially follows human organizational principles. Cell Rep 2024; 43:114191. [PMID: 38717901 DOI: 10.1016/j.celrep.2024.114191] [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: 01/12/2023] [Revised: 12/10/2023] [Accepted: 04/17/2024] [Indexed: 06/01/2024] Open
Abstract
While humans are known to have several premotor cortical areas, secondary motor cortex (M2) is often considered to be the only higher-order motor area of the mouse brain and is thought to combine properties of various human premotor cortices. Here, we show that axonal tracer, functional connectivity, myelin mapping, gene expression, and optogenetics data contradict this notion. Our analyses reveal three premotor areas in the mouse, anterior-lateral motor cortex (ALM), anterior-lateral M2 (aM2), and posterior-medial M2 (pM2), with distinct structural, functional, and behavioral properties. By using the same techniques across mice and humans, we show that ALM has strikingly similar functional and microstructural properties to human anterior ventral premotor areas and that aM2 and pM2 amalgamate properties of human pre-SMA and cingulate cortex. These results provide evidence for the existence of multiple premotor areas in the mouse and chart a comparative map between the motor systems of humans and mice.
Collapse
Affiliation(s)
- Alberto Lazari
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK.
| | - Mohamed Tachrount
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Juan Miguel Valverde
- DTU Compute, Technical University of Denmark, Kongens Lyngby, Denmark; A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70150 Kuopio, Finland
| | - Daniel Papp
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Antoine Beauchamp
- Mouse Imaging Centre, The Hospital for Sick Children, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Paul McCarthy
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacob Ellegood
- Mouse Imaging Centre, The Hospital for Sick Children, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Joanes Grandjean
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Valerio Zerbi
- Neuro-X Institute, School of Engineering (STI), EPFL, 1015 Lausanne, Switzerland; CIBM Center for Biomedical Imaging, 1015 Lausanne, Switzerland
| | - Jason P Lerch
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Mouse Imaging Centre, The Hospital for Sick Children, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Rogier B Mars
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
112
|
Liu Y, Zhang J, Jiang Z, Qin M, Xu M, Zhang S, Ma G. Organization of corticocortical and thalamocortical top-down inputs in the primary visual cortex. Nat Commun 2024; 15:4495. [PMID: 38802410 PMCID: PMC11130321 DOI: 10.1038/s41467-024-48924-8] [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: 10/16/2023] [Accepted: 05/16/2024] [Indexed: 05/29/2024] Open
Abstract
Unified visual perception requires integration of bottom-up and top-down inputs in the primary visual cortex (V1), yet the organization of top-down inputs in V1 remains unclear. Here, we used optogenetics-assisted circuit mapping to identify how multiple top-down inputs from higher-order cortical and thalamic areas engage V1 excitatory and inhibitory neurons. Top-down inputs overlap in superficial layers yet segregate in deep layers. Inputs from the medial secondary visual cortex (V2M) and anterior cingulate cortex (ACA) converge on L6 Pyrs, whereas ventrolateral orbitofrontal cortex (ORBvl) and lateral posterior thalamic nucleus (LP) inputs are processed in parallel in Pyr-type-specific subnetworks (Pyr←ORBvl and Pyr←LP) and drive mutual inhibition between them via local interneurons. Our study deepens understanding of the top-down modulation mechanisms of visual processing and establishes that V2M and ACA inputs in L6 employ integrated processing distinct from the parallel processing of LP and ORBvl inputs in L5.
Collapse
Affiliation(s)
- Yanmei Liu
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiahe Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhishan Jiang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Meiling Qin
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Min Xu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Siyu Zhang
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Guofen Ma
- Songjiang Hospital and Songjiang Research Institute, Shanghai Key Laboratory of Emotions and Affective Disorders, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
113
|
Markicevic M, Mandino F, Toyonaga T, Cai Z, Fesharaki-Zadeh A, Shen X, Strittmatter SM, Lake E. Repetitive mild closed-head injury induced synapse loss and increased local BOLD-fMRI signal homogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595651. [PMID: 38826468 PMCID: PMC11142233 DOI: 10.1101/2024.05.24.595651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Repeated mild head injuries due to sports, or domestic violence and military service are increasingly linked to debilitating symptoms in the long term. Although symptoms may take decades to manifest, potentially treatable neurobiological alterations must begin shortly after injury. Better means to diagnose and treat traumatic brain injuries, requires an improved understanding of the mechanisms underlying progression and means through which they can be measured. Here, we employ a repetitive mild closed-head injury (rmTBI) and chronic variable stress (CVS) mouse model to investigate emergent structural and functional brain abnormalities. Brain imaging is achieved with [ 18 F]SynVesT-1 positron emission tomography, with the synaptic vesicle glycoprotein 2A ligand marking synapse density and BOLD (blood-oxygen-level-dependent) functional magnetic resonance imaging (fMRI). Animals were scanned six weeks after concluding rmTBI/Stress procedures. Injured mice showed widespread decreases in synaptic density coupled with an i ncrease in local BOLD-fMRI synchrony detected as regional homogeneity. Injury-affected regions with higher synapse density showed a greater increase in fMRI regional homogeneity. Taken together, these observations may reflect compensatory mechanisms following injury. Multimodal studies are needed to provide deeper insights into these observations.
Collapse
|
114
|
Aboharb F, Davoudian PA, Shao LX, Liao C, Rzepka GN, Wojtasiewicz C, Dibbs M, Rondeau J, Sherwood AM, Kaye AP, Kwan AC. Classification of psychedelic drugs based on brain-wide imaging of cellular c-Fos expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.23.590306. [PMID: 38826215 PMCID: PMC11142187 DOI: 10.1101/2024.05.23.590306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Psilocybin, ketamine, and MDMA are psychoactive compounds that exert behavioral effects with distinguishable but also overlapping features. The growing interest in using these compounds as therapeutics necessitates preclinical assays that can accurately screen psychedelics and related analogs. We posit that a promising approach may be to measure drug action on markers of neural plasticity in native brain tissues. We therefore developed a pipeline for drug classification using light sheet fluorescence microscopy of immediate early gene expression at cellular resolution followed by machine learning. We tested male and female mice with a panel of drugs, including psilocybin, ketamine, 5-MeO-DMT, 6-fluoro-DET, MDMA, acute fluoxetine, chronic fluoxetine, and vehicle. In one-versus-rest classification, the exact drug was identified with 67% accuracy, significantly above the chance level of 12.5%. In one-versus-one classifications, psilocybin was discriminated from 5-MeO-DMT, ketamine, MDMA, or acute fluoxetine with >95% accuracy. We used Shapley additive explanation to pinpoint the brain regions driving the machine learning predictions. Our results support a novel approach for screening psychoactive drugs with psychedelic properties.
Collapse
Affiliation(s)
- Farid Aboharb
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Weill Cornell Medicine/Rockefeller/Sloan-Kettering Tri-Institutional MD/PhD Program, New York, NY, 10021, USA
| | - Pasha A. Davoudian
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, 06511, USA
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Ling-Xiao Shao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Gillian N. Rzepka
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
| | | | - Mark Dibbs
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Jocelyne Rondeau
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | | | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, 06477, USA
- Wu Tsai Institute, Yale University, New Haven, CT, 06511, USA
| | - Alex C. Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, 10065, USA
| |
Collapse
|
115
|
Swain AK, Pandit V, Sharma J, Yadav P. SpatialPrompt: spatially aware scalable and accurate tool for spot deconvolution and domain identification in spatial transcriptomics. Commun Biol 2024; 7:639. [PMID: 38796505 PMCID: PMC11127982 DOI: 10.1038/s42003-024-06349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Efficiently mapping of cell types in situ remains a major challenge in spatial transcriptomics. Most spot deconvolution tools ignore spatial coordinate information and perform extremely slow on large datasets. Here, we introduce SpatialPrompt, a spatially aware and scalable tool for spot deconvolution and domain identification. SpatialPrompt integrates gene expression, spatial location, and single-cell RNA sequencing (scRNA-seq) dataset as reference to accurately infer cell-type proportions of spatial spots. SpatialPrompt uses non-negative ridge regression and graph neural network to efficiently capture local microenvironment information. Our extensive benchmarking analysis on Visium, Slide-seq, and MERFISH datasets demonstrated superior performance of SpatialPrompt over 15 existing tools. On mouse hippocampus dataset, SpatialPrompt achieves spot deconvolution and domain identification within 2 minutes for 50,000 spots. Overall, domain identification using SpatialPrompt was 44 to 150 times faster than existing methods. We build a database housing 40 plus curated scRNA-seq datasets for seamless integration with SpatialPrompt for spot deconvolution.
Collapse
Affiliation(s)
- Asish Kumar Swain
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Vrushali Pandit
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Jyoti Sharma
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India
| | - Pankaj Yadav
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India.
- School of Artificial Intelligence and Data Science, Indian Institute of Technology, Jodhpur, Rajasthan, 342030, India.
| |
Collapse
|
116
|
Liu Z, Feng Z, Liu G, Li A, Gong H, Yang X, Li X. A complementary approach for neocortical cytoarchitecture inspection with cellular resolution imaging at whole brain scale. Front Neuroanat 2024; 18:1388084. [PMID: 38846539 PMCID: PMC11153794 DOI: 10.3389/fnana.2024.1388084] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/26/2024] [Indexed: 06/09/2024] Open
Abstract
Cytoarchitecture, the organization of cells within organs and tissues, serves as a crucial anatomical foundation for the delineation of various regions. It enables the segmentation of the cortex into distinct areas with unique structural and functional characteristics. While traditional 2D atlases have focused on cytoarchitectonic mapping of cortical regions through individual sections, the intricate cortical gyri and sulci demands a 3D perspective for unambiguous interpretation. In this study, we employed fluorescent micro-optical sectioning tomography to acquire architectural datasets of the entire macaque brain at a resolution of 0.65 μm × 0.65 μm × 3 μm. With these volumetric data, the cortical laminar textures were remarkably presented in appropriate view planes. Additionally, we established a stereo coordinate system to represent the cytoarchitectonic information as surface-based tomograms. Utilizing these cytoarchitectonic features, we were able to three-dimensionally parcel the macaque cortex into multiple regions exhibiting contrasting architectural patterns. The whole-brain analysis was also conducted on mice that clearly revealed the presence of barrel cortex and reflected biological reasonability of this method. Leveraging these high-resolution continuous datasets, our method offers a robust tool for exploring the organizational logic and pathological mechanisms of the brain's 3D anatomical structure.
Collapse
Affiliation(s)
- Zhixiang Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao Feng
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Guangcai Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Xiangning Li
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| |
Collapse
|
117
|
Si Z, Li H, Shang W, Zhao Y, Kong L, Long C, Zuo Y, Feng Z. SpaNCMG: improving spatial domains identification of spatial transcriptomics using neighborhood-complementary mixed-view graph convolutional network. Brief Bioinform 2024; 25:bbae259. [PMID: 38811360 PMCID: PMC11136618 DOI: 10.1093/bib/bbae259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
The advancement of spatial transcriptomics (ST) technology contributes to a more profound comprehension of the spatial properties of gene expression within tissues. However, due to challenges of high dimensionality, pronounced noise and dynamic limitations in ST data, the integration of gene expression and spatial information to accurately identify spatial domains remains challenging. This paper proposes a SpaNCMG algorithm for the purpose of achieving precise spatial domain description and localization based on a neighborhood-complementary mixed-view graph convolutional network. The algorithm enables better adaptation to ST data at different resolutions by integrating the local information from KNN and the global structure from r-radius into a complementary neighborhood graph. It also introduces an attention mechanism to achieve adaptive fusion of different reconstructed expressions, and utilizes KPCA method for dimensionality reduction. The application of SpaNCMG on five datasets from four sequencing platforms demonstrates superior performance to eight existing advanced methods. Specifically, the algorithm achieved highest ARI accuracies of 0.63 and 0.52 on the datasets of the human dorsolateral prefrontal cortex and mouse somatosensory cortex, respectively. It accurately identified the spatial locations of marker genes in the mouse olfactory bulb tissue and inferred the biological functions of different regions. When handling larger datasets such as mouse embryos, the SpaNCMG not only identified the main tissue structures but also explored unlabeled domains. Overall, the good generalization ability and scalability of SpaNCMG make it an outstanding tool for understanding tissue structure and disease mechanisms. Our codes are available at https://github.com/ZhihaoSi/SpaNCMG.
Collapse
Affiliation(s)
- Zhihao Si
- College of Sciences, Inner Mongolia University of Technology, Hohhot 010051, China
| | - Hanshuang Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Wenjing Shang
- College of Sciences, Inner Mongolia University of Technology, Hohhot 010051, China
| | - Yanan Zhao
- College of Sciences, Inner Mongolia University of Technology, Hohhot 010051, China
| | - Lingjiao Kong
- College of Sciences, Inner Mongolia University of Technology, Hohhot 010051, China
| | - Chunshen Long
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yongchun Zuo
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Zhenxing Feng
- College of Sciences, Inner Mongolia University of Technology, Hohhot 010051, China
| |
Collapse
|
118
|
Mezias C, Huo B, Bota M, Jayakumar J, Mitra PP. Establishing neuroanatomical correspondences across mouse and marmoset brain structures. RESEARCH SQUARE 2024:rs.3.rs-4373678. [PMID: 38826382 PMCID: PMC11142350 DOI: 10.21203/rs.3.rs-4373678/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Interest in the common marmoset is growing due to evolutionarily proximity to humans compared to laboratory mice, necessitating a comparison of mouse and marmoset brain architectures, including connectivity and cell type distributions. Creating an actionable comparative platform is challenging since these brains have distinct spatial organizations and expert neuroanatomists disagree. We propose a general theoretical framework to relate named atlas compartments across taxa and use it to establish a detailed correspondence between marmoset and mice brains. Contrary to conventional wisdom that brain structures may be easier to relate at higher levels of the atlas hierarchy, we find that finer parcellations at the leaf levels offer greater reconcilability despite naming discrepancies. Utilizing existing atlases and associated literature, we created a list of leaf-level structures for both species and establish five types of correspondence between them. One-to-one relations were found between 43% of the structures in mouse and 47% in marmoset, whereas 25% of mouse and 10% of marmoset structures were not relatable. The remaining structures show a set of more complex mappings which we quantify. Implementing this correspondence with volumetric atlases of the two species, we make available a computational tool for querying and visualizing relationships between the corresponding brains. Our findings provide a foundation for computational comparative analyses of mesoscale connectivity and cell type distributions in the laboratory mouse and the common marmoset.
Collapse
Affiliation(s)
- Christopher Mezias
- Cold Spring Harbor Laboratory, Department of Neuroscience, 1 Bungtown Rd, Cold Spring Harbor, NY
| | - Bingxing Huo
- Broad Institute of MIT and Harvard, Data Sciences Platform Division, 105 Broadway, Cambridge, MA
| | - Mihail Bota
- 15 Cismelei, 15 Bl. Constanta, Romania, 900842
| | - Jaikishan Jayakumar
- Indian Institute of Technology-Madras, Center for Computational Brain Research, Chennai, TM, India
| | - Partha P. Mitra
- Cold Spring Harbor Laboratory, Department of Neuroscience, 1 Bungtown Rd, Cold Spring Harbor, NY
| |
Collapse
|
119
|
Gurdon B, Yates SC, Csucs G, Groeneboom NE, Hadad N, Telpoukhovskaia M, Ouellette A, Ouellette T, O'Connell KMS, Singh S, Murdy TJ, Merchant E, Bjerke I, Kleven H, Schlegel U, Leergaard TB, Puchades MA, Bjaalie JG, Kaczorowski CC. Detecting the effect of genetic diversity on brain composition in an Alzheimer's disease mouse model. Commun Biol 2024; 7:605. [PMID: 38769398 PMCID: PMC11106287 DOI: 10.1038/s42003-024-06242-1] [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: 08/16/2023] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
Alzheimer's disease (AD) is broadly characterized by neurodegeneration, pathology accumulation, and cognitive decline. There is considerable variation in the progression of clinical symptoms and pathology in humans, highlighting the importance of genetic diversity in the study of AD. To address this, we analyze cell composition and amyloid-beta deposition of 6- and 14-month-old AD-BXD mouse brains. We utilize the analytical QUINT workflow- a suite of software designed to support atlas-based quantification, which we expand to deliver a highly effective method for registering and quantifying cell and pathology changes in diverse disease models. In applying the expanded QUINT workflow, we quantify near-global age-related increases in microglia, astrocytes, and amyloid-beta, and we identify strain-specific regional variation in neuron load. To understand how individual differences in cell composition affect the interpretation of bulk gene expression in AD, we combine hippocampal immunohistochemistry analyses with bulk RNA-sequencing data. This approach allows us to categorize genes whose expression changes in response to AD in a cell and/or pathology load-dependent manner. Ultimately, our study demonstrates the use of the QUINT workflow to standardize the quantification of immunohistochemistry data in diverse mice, - providing valuable insights into regional variation in cellular load and amyloid deposition in the AD-BXD model.
Collapse
Affiliation(s)
- Brianna Gurdon
- The Jackson Laboratory, Bar Harbor, ME, USA
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA
| | - Sharon C Yates
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Gergely Csucs
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nicolaas E Groeneboom
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Niran Hadad
- The Jackson Laboratory, Bar Harbor, ME, USA
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | - Andrew Ouellette
- The Jackson Laboratory, Bar Harbor, ME, USA
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA
| | - Tionna Ouellette
- The Jackson Laboratory, Bar Harbor, ME, USA
- Tufts University Graduate School of Biomedical Sciences, Medford, MA, USA
| | - Kristen M S O'Connell
- The Jackson Laboratory, Bar Harbor, ME, USA
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA
- Tufts University Graduate School of Biomedical Sciences, Medford, MA, USA
| | - Surjeet Singh
- The Jackson Laboratory, Bar Harbor, ME, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Ingvild Bjerke
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Heidi Kleven
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ulrike Schlegel
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Trygve B Leergaard
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Maja A Puchades
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jan G Bjaalie
- Neural Systems Laboratory, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Catherine C Kaczorowski
- The University of Maine Graduate School of Biomedical Sciences and Engineering, Orono, ME, USA.
- Tufts University Graduate School of Biomedical Sciences, Medford, MA, USA.
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
120
|
Barraclough BN, Stubbs WT, Bohic M, Upadhyay A, Abraira VE, Ramer MS. Direct comparison of Hoxb8-driven reporter distribution in the brains of four transgenic mouse lines: towards a spinofugal projection atlas. Front Neuroanat 2024; 18:1400015. [PMID: 38817241 PMCID: PMC11137224 DOI: 10.3389/fnana.2024.1400015] [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: 03/12/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Hox genes govern rostro-caudal identity along the developing spinal cord, which has a well-defined division of function between dorsal (sensory) and ventral (motor) halves. Here we exploit developmental Hoxb8 expression, normally restricted to the dorsal cord below the obex, to genetically label spinal cord-to-brain ("spinofugal") axons. Methods We crossed two targeted (knock-in) and two non-targeted recombinase-expressing lines (Hoxb8-IRES-Cre and Hoxb8-T2AFlpO; Hoxb8-Cre and Hoxb8-FlpO, respectively) with appropriate tdtomato-expressing reporter strains. Serial sectioning, confocal and superresolution microscopy, as well as light-sheet imaging was used to reveal robust labeling of ascending axons and their terminals in expected and unexpected regions. Results This strategy provides unprecedented anatomical detail of ascending spinal tracts anterior to the brainstem, and reveals a previously undescribed decussating tract in the ventral hypothalamus (the spinofugal hypothalamic decussating tract, or shxt). The absence of Hoxb8-suppressing elements led to multiple instances of ectopic reporter expression in Hoxb8-Cre mice (retinal ganglion and vomeronasal axons, anterior thalamic nuclei and their projections to the anterior cingulate and retrosplenial cortices and subiculum, and a population of astrocytes at the cephalic flexure) and Hoxb8-FlpO mice (Cajal-Retzius cells of the dentate gyrus, and mesenchymal cells of the choroid plexus). While targeted transgenic lines were similar in terms of known spinofugal projections, Hoxb8-IRES-Cre reporters had an additional projection to the core of the facial motor nucleus, and more abundant Hoxb8-lineage microglia scattered throughout the brain than Hoxb8-T2A-FlpO (or any other) mice, suggesting dysregulated Hoxb8-driven reporter expression in one or both lines. Discussion This work complements structural and connectivity atlases of the mouse central nervous system, and provides a platform upon which their reactions to injury or disease can be studied. Ectopic Hoxb8-driven recombinase expression may also be a useful tool to study structure and function of other cell populations in non-targeted lines.
Collapse
Affiliation(s)
- Bridget N. Barraclough
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - W. Terrence Stubbs
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
| | - Manon Bohic
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Aman Upadhyay
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Victoria E. Abraira
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Matt S. Ramer
- International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
121
|
Scaglione A, Resta F, Goretti F, Pavone FS. Group ICA of wide-field calcium imaging data reveals the retrosplenial cortex as a major contributor to cortical activity during anesthesia. Front Cell Neurosci 2024; 18:1258793. [PMID: 38799987 PMCID: PMC11116703 DOI: 10.3389/fncel.2024.1258793] [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: 07/14/2023] [Accepted: 03/14/2024] [Indexed: 05/29/2024] Open
Abstract
Large-scale cortical dynamics play a crucial role in many cognitive functions such as goal-directed behaviors, motor learning and sensory processing. It is well established that brain states including wakefulness, sleep, and anesthesia modulate neuronal firing and synchronization both within and across different brain regions. However, how the brain state affects cortical activity at the mesoscale level is less understood. This work aimed to identify the cortical regions engaged in different brain states. To this end, we employed group ICA (Independent Component Analysis) to wide-field imaging recordings of cortical activity in mice during different anesthesia levels and the awake state. Thanks to this approach we identified independent components (ICs) representing elements of the cortical networks that are common across subjects under decreasing levels of anesthesia toward the awake state. We found that ICs related to the retrosplenial cortices exhibited a pronounced dependence on brain state, being most prevalent in deeper anesthesia levels and diminishing during the transition to the awake state. Analyzing the occurrence of the ICs we found that activity in deeper anesthesia states was characterized by a strong correlation between the retrosplenial components and this correlation decreases when transitioning toward wakefulness. Overall these results indicate that during deeper anesthesia states coactivation of the posterior-medial cortices is predominant over other connectivity patterns, whereas a richer repertoire of dynamics is expressed in lighter anesthesia levels and the awake state.
Collapse
Affiliation(s)
- Alessandro Scaglione
- Department of Physics and Astronomy, University of Florence, Florence, Italy
- European Laboratory for Non-Linear Spectroscopy (LENS), Florence, Italy
| | - Francesco Resta
- European Laboratory for Non-Linear Spectroscopy (LENS), Florence, Italy
- National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy
| | - Francesco Goretti
- European Laboratory for Non-Linear Spectroscopy (LENS), Florence, Italy
| | - Francesco S. Pavone
- Department of Physics and Astronomy, University of Florence, Florence, Italy
- European Laboratory for Non-Linear Spectroscopy (LENS), Florence, Italy
- National Institute of Optics, National Research Council (INO-CNR), Sesto Fiorentino, Italy
| |
Collapse
|
122
|
Noel JP, Balzani E, Acerbi L, Benson J, Savin C, Angelaki DE. A common computational and neural anomaly across mouse models of autism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593232. [PMID: 38766250 PMCID: PMC11100696 DOI: 10.1101/2024.05.08.593232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Computational psychiatry has suggested that humans within the autism spectrum disorder (ASD) inflexibly update their expectations (i.e., Bayesian priors). Here, we leveraged high-yield rodent psychophysics (n = 75 mice), extensive behavioral modeling (including principled and heuristics), and (near) brain-wide single cell extracellular recordings (over 53k units in 150 brain areas) to ask (1) whether mice with different genetic perturbations associated with ASD show this same computational anomaly, and if so, (2) what neurophysiological features are shared across genotypes in subserving this deficit. We demonstrate that mice harboring mutations in Fmr1 , Cntnap2 , and Shank3B show a blunted update of priors during decision-making. Neurally, the differentiating factor between animals flexibly and inflexibly updating their priors was a shift in the weighting of prior encoding from sensory to frontal cortices. Further, in mouse models of ASD frontal areas showed a preponderance of units coding for deviations from the animals' long-run prior, and sensory responses did not differentiate between expected and unexpected observations. These findings demonstrate that distinct genetic instantiations of ASD may yield common neurophysiological and behavioral phenotypes.
Collapse
|
123
|
Zheng W, Mu H, Chen Z, Liu J, Xia D, Cheng Y, Jing Q, Lau PM, Tang J, Bi GQ, Wu F, Wang H. NEATmap: a high-efficiency deep learning approach for whole mouse brain neuronal activity trace mapping. Natl Sci Rev 2024; 11:nwae109. [PMID: 38831937 PMCID: PMC11145917 DOI: 10.1093/nsr/nwae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/26/2024] [Accepted: 02/25/2024] [Indexed: 06/05/2024] Open
Abstract
Quantitative analysis of activated neurons in mouse brains by a specific stimulation is usually a primary step to locate the responsive neurons throughout the brain. However, it is challenging to comprehensively and consistently analyze the neuronal activity trace in whole brains of a large cohort of mice from many terabytes of volumetric imaging data. Here, we introduce NEATmap, a deep learning-based high-efficiency, high-precision and user-friendly software for whole-brain neuronal activity trace mapping by automated segmentation and quantitative analysis of immunofluorescence labeled c-Fos+ neurons. We applied NEATmap to study the brain-wide differentiated neuronal activation in response to physical and psychological stressors in cohorts of mice.
Collapse
Affiliation(s)
- Weijie Zheng
- AHU-IAI AI Joint Laboratory, Anhui University, Hefei 230039, China
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Huawei Mu
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zhiyi Chen
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Jiajun Liu
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Debin Xia
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Yuxiao Cheng
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Qi Jing
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Pak-Ming Lau
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jin Tang
- AHU-IAI AI Joint Laboratory, Anhui University, Hefei 230039, China
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
| | - Guo-Qiang Bi
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Feng Wu
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| | - Hao Wang
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- National Engineering Laboratory for Brain-inspired Intelligence Technology and Application, School of Information Science and Technology, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
124
|
Jamiolkowski RM, Nguyen QA, Farrell JS, McGinn RJ, Hartmann DA, Nirschl JJ, Sanchez MI, Buch VP, Soltesz I. The fasciola cinereum of the hippocampal tail as an interventional target in epilepsy. Nat Med 2024; 30:1292-1299. [PMID: 38632391 PMCID: PMC11108783 DOI: 10.1038/s41591-024-02924-9] [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: 10/24/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
Targeted tissue ablation involving the anterior hippocampus is the standard of care for patients with drug-resistant mesial temporal lobe epilepsy. However, a substantial proportion continues to suffer from seizures even after surgery. We identified the fasciola cinereum (FC) neurons of the posterior hippocampal tail as an important seizure node in both mice and humans with epilepsy. Genetically defined FC neurons were highly active during spontaneous seizures in epileptic mice, and closed-loop optogenetic inhibition of these neurons potently reduced seizure duration. Furthermore, we specifically targeted and found the prominent involvement of FC during seizures in a cohort of six patients with epilepsy. In particular, targeted lesioning of the FC in a patient reduced the seizure burden present after ablation of anterior mesial temporal structures. Thus, the FC may be a promising interventional target in epilepsy.
Collapse
Affiliation(s)
| | - Quynh-Anh Nguyen
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- Department of Pharmacology and the Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| | - Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- F.M. Kirby Neurobiology Center and Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan J McGinn
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - David A Hartmann
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Jeff J Nirschl
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Mateo I Sanchez
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Vivek P Buch
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
125
|
Hoops D, Yee Y, Hammill C, Wong S, Manitt C, Bedell BJ, Cahill L, Lerch JP, Flores C, Sled JG. Disproportionate neuroanatomical effects of DCC haploinsufficiency in adolescence compared with adulthood: links to dopamine, connectivity, covariance, and gene expression brain maps in mice. J Psychiatry Neurosci 2024; 49:E157-E171. [PMID: 38692693 PMCID: PMC11068426 DOI: 10.1503/jpn.230106] [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: 07/31/2023] [Revised: 01/23/2024] [Accepted: 03/06/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Critical adolescent neural refinement is controlled by the DCC (deleted in colorectal cancer) protein, a receptor for the netrin-1 guidance cue. We sought to describe the effects of reduced DCC on neuroanatomy in the adolescent and adult mouse brain. METHODS We examined neuronal connectivity, structural covariance, and molecular processes in a DCC-haploinsufficient mouse model, compared with wild-type mice, using new, custom analytical tools designed to leverage publicly available databases from the Allen Institute. RESULTS We included 11 DCC-haploinsufficient mice and 16 wild-type littermates. Neuroanatomical effects of DCC haploinsufficiency were more severe in adolescence than adulthood and were largely restricted to the mesocorticolimbic dopamine system. The latter finding was consistent whether we identified the regions of the mesocorticolimbic dopamine system a priori or used connectivity data from the Allen Brain Atlas to determine de novo where these dopamine axons terminated. Covariance analyses found that DCC haploinsufficiency disrupted the coordinated development of the brain regions that make up the mesocorticolimbic dopamine system. Gene expression maps pointed to molecular processes involving the expression of DCC, UNC5C (encoding DCC's co-receptor), and NTN1 (encoding its ligand, netrin-1) as underlying our structural findings. LIMITATIONS Our study involved a single sex (males) at only 2 ages. CONCLUSION The neuroanatomical phenotype of DCC haploinsufficiency described in mice parallels that observed in DCC-haploinsufficient humans. It is critical to understand the DCC-haploinsufficient mouse as a clinically relevant model system.
Collapse
Affiliation(s)
- Daniel Hoops
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Yohan Yee
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Christopher Hammill
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Sammi Wong
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Colleen Manitt
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Barry J Bedell
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Lindsay Cahill
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Jason P Lerch
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - Cecilia Flores
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| | - John G Sled
- From the Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ont. (Hoops, Yee, Hammill, Wong, Lerch, Sled); the Department of Medical Biophysics, University of Toronto, Ont. (Hoops, Yee, Lerch, Sled); the Department of Psychiatry, McGill University, Montréal, Que. (Hoops, Flores); the Douglas Mental Health University Institute, Montréal, Que. (Hoops, Manitt, Flores); the Department of Chemistry, Memorial University, St. John's, N.L. (Hoops, Cahill); the Department of Neurology and Neurosurgery, McGill University, Montréal, Que. (Bedell, Flores); the Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain, Nuffield Department of Clinical Neuroscience, University of Oxford, U.K. (Lerch); the Ludmer Centre for Neuroinformatics & Mental Health, McGill University, Montréal, Que. (Flores)
| |
Collapse
|
126
|
Yang L, Liu F, Hahm H, Okuda T, Li X, Zhang Y, Kalyanaraman V, Heitmeier MR, Samineni VK. Projection-TAGs enable multiplex projection tracing and multi-modal profiling of projection neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590975. [PMID: 38712231 PMCID: PMC11071495 DOI: 10.1101/2024.04.24.590975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Single-cell multiomic techniques have sparked immense interest in developing a comprehensive multi-modal map of diverse neuronal cell types and their brain wide projections. However, investigating the spatial organization, transcriptional and epigenetic landscapes of brain wide projection neurons is hampered by the lack of efficient and easily adoptable tools. Here we introduce Projection-TAGs, a retrograde AAV platform that allows multiplex tagging of projection neurons using RNA barcodes. By using Projection-TAGs, we performed multiplex projection tracing of the mouse cortex and high-throughput single-cell profiling of the transcriptional and epigenetic landscapes of the cortical projection neurons. Projection-TAGs can be leveraged to obtain a snapshot of activity-dependent recruitment of distinct projection neurons and their molecular features in the context of a specific stimulus. Given its flexibility, usability, and compatibility, we envision that Projection-TAGs can be readily applied to build a comprehensive multi-modal map of brain neuronal cell types and their projections.
Collapse
Affiliation(s)
- Lite Yang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Fang Liu
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Hannah Hahm
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Takao Okuda
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Xiaoyue Li
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Yufen Zhang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Vani Kalyanaraman
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Monique R. Heitmeier
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Vijay K. Samineni
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
127
|
Cai HR, Chen SQ, Xiang XJ, Zhang XQ, Ma RZ, Zhu G, Ding SL. Comparison of the connectivity of the posterior intralaminar thalamic nucleus and peripeduncular nucleus in rats and mice. Front Neural Circuits 2024; 18:1384621. [PMID: 38736977 PMCID: PMC11082296 DOI: 10.3389/fncir.2024.1384621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
The posterior intralaminar thalamic nucleus (PIL) and peripeduncular nucleus (PP) are two adjoining structures located medioventral to the medial geniculate nucleus. The PIL-PP region plays important roles in auditory fear conditioning and in social, maternal and sexual behaviors. Previous studies often lumped the PIL and PP into single entity, and therefore it is not known if they have common and/or different brain-wide connections. In this study, we investigate brain-wide efferent and afferent projections of the PIL and PP using reliable anterograde and retrograde tracing methods. Both PIL and PP project strongly to lateral, medial and anterior basomedial amygdaloid nuclei, posteroventral striatum (putamen and external globus pallidus), amygdalostriatal transition area, zona incerta, superior and inferior colliculi, and the ectorhinal cortex. However, the PP rather than the PIL send stronger projections to the hypothalamic regions such as preoptic area/nucleus, anterior hypothalamic nucleus, and ventromedial nucleus of hypothalamus. As for the afferent projections, both PIL and PP receive multimodal information from auditory (inferior colliculus, superior olivary nucleus, nucleus of lateral lemniscus, and association auditory cortex), visual (superior colliculus and ectorhinal cortex), somatosensory (gracile and cuneate nuclei), motor (external globus pallidus), and limbic (central amygdaloid nucleus, hypothalamus, and insular cortex) structures. However, the PP rather than PIL receives strong projections from the visual related structures parabigeminal nucleus and ventral lateral geniculate nucleus. Additional results from Cre-dependent viral tracing in mice have also confirmed the main results in rats. Together, the findings in this study would provide new insights into the neural circuits and functional correlation of the PIL and PP.
Collapse
Affiliation(s)
- Hui-Ru Cai
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Department of Psychology, School of Health Management, Guangzhou Medical University, Guangzhou, China
| | - Sheng-Qiang Chen
- Department of Psychology, School of Health Management, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Jun Xiang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xue-Qin Zhang
- Department of Psychology, School of Health Management, Guangzhou Medical University, Guangzhou, China
| | - Run-Zhe Ma
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ge Zhu
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Song-Lin Ding
- Department of Psychology, School of Health Management, Guangzhou Medical University, Guangzhou, China
- Allen Institute for Brain Science, Seattle, WA, United States
| |
Collapse
|
128
|
Stouffer KM, Trouvé A, Younes L, Kunst M, Ng L, Zeng H, Anant M, Fan J, Kim Y, Chen X, Rue M, Miller MI. Cross-modality mapping using image varifolds to align tissue-scale atlases to molecular-scale measures with application to 2D brain sections. Nat Commun 2024; 15:3530. [PMID: 38664422 PMCID: PMC11045777 DOI: 10.1038/s41467-024-47883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
This paper explicates a solution to building correspondences between molecular-scale transcriptomics and tissue-scale atlases. This problem arises in atlas construction and cross-specimen/technology alignment where specimens per emerging technology remain sparse and conventional image representations cannot efficiently model the high dimensions from subcellular detection of thousands of genes. We address these challenges by representing spatial transcriptomics data as generalized functions encoding position and high-dimensional feature (gene, cell type) identity. We map onto low-dimensional atlas ontologies by modeling regions as homogeneous random fields with unknown transcriptomic feature distribution. We solve simultaneously for the minimizing geodesic diffeomorphism of coordinates through LDDMM and for these latent feature densities. We map tissue-scale mouse brain atlases to gene-based and cell-based transcriptomics data from MERFISH and BARseq technologies and to histopathology and cross-species atlases to illustrate integration of diverse molecular and cellular datasets into a single coordinate system as a means of comparison and further atlas construction.
Collapse
Affiliation(s)
- Kaitlin M Stouffer
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
- Centre Borelli, ENS Paris-Saclay, Gif-sur-yvette, France.
| | - Alain Trouvé
- Centre Borelli, ENS Paris-Saclay, Gif-sur-yvette, France
| | - Laurent Younes
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Manjari Anant
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, Penn State University, College of Medicine, State College, PA, USA
| | - Xiaoyin Chen
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Mara Rue
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
129
|
Chen X, Fischer S, Rue MCP, Zhang A, Mukherjee D, Kanold PO, Gillis J, Zador AM. Whole-cortex in situ sequencing reveals input-dependent area identity. Nature 2024:10.1038/s41586-024-07221-6. [PMID: 38658747 DOI: 10.1038/s41586-024-07221-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/22/2024] [Indexed: 04/26/2024]
Abstract
The cerebral cortex is composed of neuronal types with diverse gene expression that are organized into specialized cortical areas. These areas, each with characteristic cytoarchitecture1,2, connectivity3,4 and neuronal activity5,6, are wired into modular networks3,4,7. However, it remains unclear whether these spatial organizations are reflected in neuronal transcriptomic signatures and how such signatures are established in development. Here we used BARseq, a high-throughput in situ sequencing technique, to interrogate the expression of 104 cell-type marker genes in 10.3 million cells, including 4,194,658 cortical neurons over nine mouse forebrain hemispheres, at cellular resolution. De novo clustering of gene expression in single neurons revealed transcriptomic types consistent with previous single-cell RNA sequencing studies8,9. The composition of transcriptomic types is highly predictive of cortical area identity. Moreover, areas with similar compositions of transcriptomic types, which we defined as cortical modules, overlap with areas that are highly connected, suggesting that the same modular organization is reflected in both transcriptomic signatures and connectivity. To explore how the transcriptomic profiles of cortical neurons depend on development, we assessed cell-type distributions after neonatal binocular enucleation. Notably, binocular enucleation caused the shifting of the cell-type compositional profiles of visual areas towards neighbouring cortical areas within the same module, suggesting that peripheral inputs sharpen the distinct transcriptomic identities of areas within cortical modules. Enabled by the high throughput, low cost and reproducibility of BARseq, our study provides a proof of principle for the use of large-scale in situ sequencing to both reveal brain-wide molecular architecture and understand its development.
Collapse
Affiliation(s)
- Xiaoyin Chen
- Allen Institute for Brain Science, Seattle, WA, USA.
| | - Stephan Fischer
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Mara C P Rue
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Aixin Zhang
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Didhiti Mukherjee
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Patrick O Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
130
|
Han RW, Zhang ZY, Jiao C, Hu ZY, Pan BX. Synergism between two BLA-to-BNST pathways for appropriate expression of anxiety-like behaviors in male mice. Nat Commun 2024; 15:3455. [PMID: 38658548 PMCID: PMC11043328 DOI: 10.1038/s41467-024-47966-2] [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: 08/22/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
Understanding how distinct functional circuits are coordinated to fine-tune mood and behavior is of fundamental importance. Here, we observe that within the dense projections from basolateral amygdala (BLA) to bed nucleus of stria terminalis (BNST), there are two functionally opposing pathways orchestrated to enable contextually appropriate expression of anxiety-like behaviors in male mice. Specifically, the anterior BLA neurons predominantly innervate the anterodorsal BNST (adBNST), while their posterior counterparts send massive fibers to oval BNST (ovBNST) with moderate to adBNST. Optogenetic activation of the anterior and posterior BLA inputs oppositely regulated the activity of adBNST neurons and anxiety-like behaviors, via disengaging and engaging the inhibitory ovBNST-to-adBNST microcircuit, respectively. Importantly, the two pathways exhibited synchronized but opposite responses to both anxiolytic and anxiogenic stimuli, partially due to their mutual inhibition within BLA and the different inputs they receive. These findings reveal synergistic interactions between two BLA-to-BNST pathways for appropriate anxiety expression with ongoing environmental demands.
Collapse
Affiliation(s)
- Ren-Wen Han
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| | - Zi-Yi Zhang
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Chen Jiao
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Ze-Yu Hu
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- College of Life Science, Nanchang University, Nanchang, 330031, China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
131
|
Ocklenburg S, Guo ZV. Cross-hemispheric communication: Insights on lateralized brain functions. Neuron 2024; 112:1222-1234. [PMID: 38458199 DOI: 10.1016/j.neuron.2024.02.010] [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: 07/31/2023] [Revised: 12/13/2023] [Accepted: 02/12/2024] [Indexed: 03/10/2024]
Abstract
On the surface, the two hemispheres of vertebrate brains look almost perfectly symmetrical, but several motor, sensory, and cognitive systems show a deeply lateralized organization. Importantly, the two hemispheres are connected by various commissures, white matter tracts that cross the brain's midline and enable cross-hemispheric communication. Cross-hemispheric communication has been suggested to play an important role in the emergence of lateralized brain functions. Here, we review current advances in understanding cross-hemispheric communication that have been made using modern neuroscientific tools in rodents and other model species, such as genetic labeling, large-scale recordings of neuronal activity, spatiotemporally precise perturbation, and quantitative behavior analyses. These findings suggest that the emergence of lateralized brain functions cannot be fully explained by largely static factors such as genetic variation and differences in structural brain asymmetries. In addition, learning-dependent asymmetric interactions between the left and right hemispheres shape lateralized brain functions.
Collapse
Affiliation(s)
- Sebastian Ocklenburg
- Department of Psychology, MSH Medical School Hamburg, Hamburg, Germany; ICAN Institute for Cognitive and Affective Neuroscience, MSH Medical School Hamburg, Hamburg, Germany; Biopsychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.
| | - Zengcai V Guo
- School of Medicine, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
132
|
Joshi J, Yao M, Kakazu A, Ouyang Y, Duan W, Aggarwal M. Distinguishing microgliosis and tau deposition in the mouse brain using paramagnetic and diamagnetic susceptibility source separation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.588962. [PMID: 38659855 PMCID: PMC11042227 DOI: 10.1101/2024.04.11.588962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Tauopathies, including Alzheimer's disease (AD), are neurodegenerative disorders characterized by hyperphosphorylated tau protein aggregates in the brain. In addition to protein aggregates, microglia-mediated inflammation and iron dyshomeostasis are other pathological features observed in AD and other tauopathies. It is known that these alterations at the subcellular level occur much before the onset of macroscopic tissue atrophy or cognitive deficits. The ability to detect these microstructural changes with MRI therefore has substantive importance for improved characterization of disease pathogenesis. In this study, we demonstrate that quantitative susceptibility mapping (QSM) with paramagnetic and diamagnetic susceptibility source separation has the potential to distinguish neuropathological alterations in a transgenic mouse model of tauopathy. 3D multi-echo gradient echo data were acquired from fixed brains of PS19 (Tau) transgenic mice and age-matched wild-type (WT) mice (n = 5 each) at 11.7 T. The multi-echo data were fit to a 3-pool complex signal model to derive maps of paramagnetic component susceptibility (PCS) and diamagnetic component susceptibility (DCS). Group-averaged signal fraction and composite susceptibility maps showed significant region-specific differences between the WT and Tau mouse brains. Significant bilateral increases in PCS and |DCS| were observed in specific hippocampal and cortical sub-regions of the Tau mice relative to WT controls. Comparison with immunohistological staining for microglia (Iba1) and phosphorylated-tau (AT8) further indicated that the PCS and DCS differences corresponded to regional microgliosis and tau deposition in the PS19 mouse brains, respectively. The results demonstrate that quantitative susceptibility source separation may provide sensitive imaging markers to detect distinct pathological alterations in tauopathies.
Collapse
Affiliation(s)
- Jayvik Joshi
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Minmin Yao
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aaron Kakazu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuxiao Ouyang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manisha Aggarwal
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
133
|
Martínez-Tazo P, Santos A, Selim MK, Espinós-Soler E, De Santis S. Sex matters: The MouseX DW-ALLEN Atlas for mice diffusion-weighted MR imaging. Neuroimage 2024; 292:120573. [PMID: 38521211 DOI: 10.1016/j.neuroimage.2024.120573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
Overcoming sex bias in preclinical research requires not only including animals of both sexes in the experiments, but also developing proper tools to handle such data. Recent work revealed sensitivity of diffusion-weighted MRI to glia morphological changes in response to inflammatory stimuli, opening up exciting possibilities to characterize inflammation in a variety of preclinical models of pathologies, the great majority of them available in mice. However, there are limited resources dedicated to mouse imaging, like those required for the data processing and analysis. To fill this gap, we build a mouse MRI template of both structural and diffusion contrasts, with anatomical annotation according to the Allen Mouse Brain Atlas, the most detailed public resource for mouse brain investigation. To achieve a standardized resource, we use a large cohort of animals in vivo, and include animals of both sexes. To prove the utility of this resource to integrate imaging and molecular data, we demonstrate significant association between the mean diffusivity from MRI and gene expression-based glia density. To demonstrate the need of equitable sex representation, we compared across sexes the warp fields needed to match a male-based template, and our template built with both sexes. Then, we use both templates for analysing mice imaging data obtained in animals of different ages, demonstrating that using a male-based template creates spurious significant sex effects, not present otherwise. All in all, our MouseX DW-ALLEN Atlas will be a widely useful resource getting us one step closer to equitable healthcare.
Collapse
Affiliation(s)
| | - Alexandra Santos
- Instituto de Neurociencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain
| | - Mohamed Kotb Selim
- Instituto de Neurociencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain
| | - Elena Espinós-Soler
- Instituto de Neurociencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain
| | - Silvia De Santis
- Instituto de Neurociencias de Alicante, CSIC-UMH, San Juan de Alicante, Spain.
| |
Collapse
|
134
|
Ding SL. Lamination, Borders, and Thalamic Projections of the Primary Visual Cortex in Human, Non-Human Primate, and Rodent Brains. Brain Sci 2024; 14:372. [PMID: 38672021 PMCID: PMC11048015 DOI: 10.3390/brainsci14040372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The primary visual cortex (V1) is one of the most studied regions of the brain and is characterized by its specialized and laminated layer 4 in human and non-human primates. However, studies aiming to harmonize the definition of the cortical layers and borders of V1 across rodents and primates are very limited. This article attempts to identify and harmonize the molecular markers and connectional patterns that can consistently link corresponding cortical layers of V1 and borders across mammalian species and ages. V1 in primates has at least two additional and unique layers (L3b2 and L3c) and two sublayers of layer 4 (L4a and L4b) compared to rodent V1. In all species examined, layers 4 and 3b of V1 receive strong inputs from the (dorsal) lateral geniculate nucleus, and V1 is mostly surrounded by the secondary visual cortex except for one location where V1 directly abuts area prostriata. The borders of primate V1 can also be clearly identified at mid-gestational ages using gene markers. In rodents, a novel posteromedial extension of V1 is identified, which expresses V1 marker genes and receives strong inputs from the lateral geniculate nucleus. This V1 extension was labeled as the posterior retrosplenial cortex and medial secondary visual cortex in the literature and brain atlases. Layer 6 of the rodent and primate V1 originates corticothalamic projections to the lateral geniculate, lateral dorsal, and reticular thalamic nuclei and the lateroposterior-pulvinar complex with topographic organization. Finally, the direct geniculo-extrastriate (particularly the strong geniculo-prostriata) projections are probably major contributors to blindsight after V1 lesions. Taken together, compared to rodents, primates, and humans, V1 has at least two unique middle layers, while other layers are comparable across species and display conserved molecular markers and similar connections with the visual thalamus with only subtle differences.
Collapse
Affiliation(s)
- Song-Lin Ding
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| |
Collapse
|
135
|
Weiler S, Rahmati V, Isstas M, Wutke J, Stark AW, Franke C, Graf J, Geis C, Witte OW, Hübener M, Bolz J, Margrie TW, Holthoff K, Teichert M. A primary sensory cortical interareal feedforward inhibitory circuit for tacto-visual integration. Nat Commun 2024; 15:3081. [PMID: 38594279 PMCID: PMC11003985 DOI: 10.1038/s41467-024-47459-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
Tactile sensation and vision are often both utilized for the exploration of objects that are within reach though it is not known whether or how these two distinct sensory systems combine such information. Here in mice, we used a combination of stereo photogrammetry for 3D reconstruction of the whisker array, brain-wide anatomical tracing and functional connectivity analysis to explore the possibility of tacto-visual convergence in sensory space and within the circuitry of the primary visual cortex (VISp). Strikingly, we find that stimulation of the contralateral whisker array suppresses visually evoked activity in a tacto-visual sub-region of VISp whose visual space representation closely overlaps with the whisker search space. This suppression is mediated by local fast-spiking interneurons that receive a direct cortico-cortical input predominantly from layer 6 neurons located in the posterior primary somatosensory barrel cortex (SSp-bfd). These data demonstrate functional convergence within and between two primary sensory cortical areas for multisensory object detection and recognition.
Collapse
Affiliation(s)
- Simon Weiler
- Sainsbury Wellcome Centre for Neuronal Circuits and Behaviour, University College London, 25 Howland Street, London, W1T 4JG, UK
| | - Vahid Rahmati
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany
| | - Marcel Isstas
- Friedrich Schiller University Jena, Institute of General Zoology and Animal Physiology, Erbertstraße 1, 07743, Jena, Germany
| | - Johann Wutke
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany
| | - Andreas Walter Stark
- Friedrich Schiller University Jena, Institute of Applied Optics and Biophysics, Fröbelstieg 1, 07743, Jena, Germany
| | - Christian Franke
- Friedrich Schiller University Jena, Institute of Applied Optics and Biophysics, Fröbelstieg 1, 07743, Jena, Germany
- Friedrich Schiller University Jena, Jena Center for Soft Matter, Philosophenweg 7, 07743, Jena, Germany
- Friedrich Schiller University Jena, Abbe Center of Photonics, Albert-Einstein-Straße 6, 07745, Jena, Germany
| | - Jürgen Graf
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany
| | - Christian Geis
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany
| | - Otto W Witte
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany
| | - Mark Hübener
- Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Jürgen Bolz
- Friedrich Schiller University Jena, Institute of General Zoology and Animal Physiology, Erbertstraße 1, 07743, Jena, Germany
| | - Troy W Margrie
- Sainsbury Wellcome Centre for Neuronal Circuits and Behaviour, University College London, 25 Howland Street, London, W1T 4JG, UK
| | - Knut Holthoff
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany
| | - Manuel Teichert
- Jena University Hospital, Department of Neurology, Am Klinikum 1, 07747, Jena, Germany.
| |
Collapse
|
136
|
Takahashi-Yamashiro K, Miyazono K. Tissue clearing method in visualization of cancer progression and metastasis. Ups J Med Sci 2024; 129:10634. [PMID: 38716075 PMCID: PMC11075440 DOI: 10.48101/ujms.v129.10634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 03/07/2024] [Indexed: 05/24/2024] Open
Abstract
Since various imaging modalities have been developed, cancer metastasis can be detected from an early stage. However, limitations still exist, especially in terms of spatial resolution. Tissue-clearing technology has emerged as a new imaging modality in cancer research, which has been developed and utilized for a long time mainly in neuroscience field. This method enables us to detect cancer metastatic foci with single-cell resolution at whole mouse body/organ level. On top of that, 3D images of cancer metastasis of whole mouse organs make it easy to understand their characteristics. Recently, further applications of tissue clearing methods were reported in combination with reporter systems, labeling, and machine learning. In this review, we would like to provide an overview of this technique and current applications in cancer research and discuss their potentials and limitations.
Collapse
Affiliation(s)
- Kei Takahashi-Yamashiro
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta, Canada
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Cancer Invasion and Metastasis, Institute for Medical Sciences, RIKEN, Yokohama City, Kanagawa, Japan
| |
Collapse
|
137
|
Fang W, Yin B, Fang Z, Tian M, Ke L, Ma X, Di Q. Heat stroke-induced cerebral cortex nerve injury by mitochondrial dysfunction: A comprehensive multi-omics profiling analysis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170869. [PMID: 38342446 DOI: 10.1016/j.scitotenv.2024.170869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/20/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
In recent years, global warming has led to frequent instances of extremely high temperatures during summer, arousing significant concern about the adverse effects of high temperature. Among these, heat stroke is the most serious, which has detrimental effects on the all organs of human body, especially on brain. However, the comprehensive pathogenesis leading to brain damage remains unclear. In this study, we constructed a mouse model of heat stroke and conducted multi-omics profiling to identify relevant pathogenesis induced by heat stroke. The mice were placed in a constant temperature chamber at 42 °C with a humidity of 50 %, and the criteria for success in modeling were that the rectal temperature reached 42 °C and that the mice were trembling. Then the mice were immediately taken out for further experiments. Firstly, we conducted cFos protein localization and identified the cerebral cortex, especially the anterior cingulate cortex as the region exhibiting the most pronounced damage. Secondly, we performed metabolomics, transcriptomics, and proteomics analysis on cerebral cortex. This multi-omics investigation unveiled noteworthy alterations in proteins and metabolites within pathways associated with neurotransmitter systems, heatstroke-induced mitochondrial dysfunction, encompassing histidine and pentose phosphate metabolic pathways, as well as oxidative stress. In addition, the cerebral cortex exhibited pronounced Reactive Oxygen Species (ROS) production, alongside significant downregulation of the mitochondrial outer membrane protein Tomm40 and mitochondrial permeability transition pore, implicating cerebral cortex mitochondrial dysfunction as the primary instigator of neural impairment. This study marks a significant milestone as the first to employ multi-omics analysis in exploring the molecular mechanisms underlying heat stroke-induced damage in cerebral cortex neurons. It comprehensively identifies all potentially impacted pathways by heat stroke, laying a solid foundation for ensuing research endeavors. Consequently, this study introduces a fresh angle to clinical approaches in heatstroke prevention and treatment, as well as establishes an innovative groundwork for shaping future-forward environmental policies.
Collapse
Affiliation(s)
- Wen Fang
- Division of Sports Science& Physical Education, Tsinghua University, Beijing, China; Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Bo Yin
- School of Medicine, Tsinghua University, Beijing, China
| | - Zijian Fang
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Mengyi Tian
- School of Medicine, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Limei Ke
- School of Medicine, Tsinghua University, Beijing, China
| | - Xindong Ma
- Division of Sports Science& Physical Education, Tsinghua University, Beijing, China; IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| | - Qian Di
- Vanke School of Public Health, Tsinghua University, Beijing, China; Institute for Healthy China, Tsinghua University, Beijing, China.
| |
Collapse
|
138
|
Kuroda KO, Fukumitsu K, Kurachi T, Ohmura N, Shiraishi Y, Yoshihara C. Parental brain through time: The origin and development of the neural circuit of mammalian parenting. Ann N Y Acad Sci 2024; 1534:24-44. [PMID: 38426943 DOI: 10.1111/nyas.15111] [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: 03/02/2024]
Abstract
This review consolidates current knowledge on mammalian parental care, focusing on its neural mechanisms, evolutionary origins, and derivatives. Neurobiological studies have identified specific neurons in the medial preoptic area as crucial for parental care. Unexpectedly, these neurons are characterized by the expression of molecules signaling satiety, such as calcitonin receptor and BRS3, and overlap with neurons involved in the reproductive behaviors of males but not females. A synthesis of comparative ecology and paleontology suggests an evolutionary scenario for mammalian parental care, possibly stemming from male-biased guarding of offspring in basal vertebrates. The terrestrial transition of tetrapods led to prolonged egg retention in females and the emergence of amniotes, skewing care toward females. The nocturnal adaptation of Mesozoic mammalian ancestors reinforced maternal care for lactation and thermal regulation via endothermy, potentially introducing metabolic gate control in parenting neurons. The established maternal care may have served as the precursor for paternal and cooperative care in mammals and also fostered the development of group living, which may have further contributed to the emergence of empathy and altruism. These evolution-informed working hypotheses require empirical validation, yet they offer promising avenues to investigate the neural underpinnings of mammalian social behaviors.
Collapse
Affiliation(s)
- Kumi O Kuroda
- RIKEN Center for Brain Science, Saitama, Japan
- School of Life Sciences and Technologies, Tokyo Institute of Technology, Kanagawa, Japan
| | - Kansai Fukumitsu
- RIKEN Center for Brain Science, Saitama, Japan
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takuma Kurachi
- RIKEN Center for Brain Science, Saitama, Japan
- Department of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Nami Ohmura
- RIKEN Center for Brain Science, Saitama, Japan
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| | - Yuko Shiraishi
- RIKEN Center for Brain Science, Saitama, Japan
- Kawamura Gakuen Woman's University, Chiba, Japan
| | - Chihiro Yoshihara
- RIKEN Center for Brain Science, Saitama, Japan
- School of Life Sciences and Technologies, Tokyo Institute of Technology, Kanagawa, Japan
| |
Collapse
|
139
|
Kibbe RR, Muddiman DC. Quantitative mass spectrometry imaging (qMSI): A tutorial. JOURNAL OF MASS SPECTROMETRY : JMS 2024; 59:e5009. [PMID: 38488849 DOI: 10.1002/jms.5009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
Mass spectrometry imaging (MSI) is an analytical technique that enables the simultaneous detection of hundreds to thousands of chemical species while retaining their spatial information; usually, MSI is applied to biological tissues. Combining these elements can create ion images, which allows for the identification and localization of multiple chemical species within the sample. Being able to produce molecular images of biological tissues has already impacted the study of health and disease; however, the next logical step is being able to combine MSI with quantitative mass spectrometry methods to both quantify and determine the localization of disease progression or drug action. In this tutorial, we will detail the main factors to consider when designing a qMSI experiment and highlight the methods that have been developed to overcome these added complexities, specifically for those newer to the field of MSI.
Collapse
Affiliation(s)
- Russell R Kibbe
- Department of Chemistry, FTMS Laboratory for Human Health Research, North Carolina State University, Raleigh, North Carolina, USA
| | - David C Muddiman
- Department of Chemistry, FTMS Laboratory for Human Health Research, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
140
|
Fitzpatrick Z, Ghabdan Zanluqui N, Rosenblum JS, Tuong ZK, Lee CYC, Chandrashekhar V, Negro-Demontel ML, Stewart AP, Posner DA, Buckley M, Allinson KSJ, Mastorakos P, Chittiboina P, Maric D, Donahue D, Helmy A, Tajsic T, Ferdinand JR, Portet A, Peñalver A, Gillman E, Zhuang Z, Clatworthy MR, McGavern DB. Venous-plexus-associated lymphoid hubs support meningeal humoral immunity. Nature 2024; 628:612-619. [PMID: 38509366 PMCID: PMC11482273 DOI: 10.1038/s41586-024-07202-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 02/16/2024] [Indexed: 03/22/2024]
Abstract
There is increasing interest in how immune cells in the meninges-the membranes that surround the brain and spinal cord-contribute to homeostasis and disease in the central nervous system1,2. The outer layer of the meninges, the dura mater, has recently been described to contain both innate and adaptive immune cells, and functions as a site for B cell development3-6. Here we identify organized lymphoid structures that protect fenestrated vasculature in the dura mater. The most elaborate of these dural-associated lymphoid tissues (DALT) surrounded the rostral-rhinal confluence of the sinuses and included lymphatic vessels. We termed this structure, which interfaces with the skull bone marrow and a comparable venous plexus at the skull base, the rostral-rhinal venolymphatic hub. Immune aggregates were present in DALT during homeostasis and expanded with age or after challenge with systemic or nasal antigens. DALT contain germinal centre B cells and support the generation of somatically mutated, antibody-producing cells in response to a nasal pathogen challenge. Inhibition of lymphocyte entry into the rostral-rhinal hub at the time of nasal viral challenge abrogated the generation of germinal centre B cells and class-switched plasma cells, as did perturbation of B-T cell interactions. These data demonstrate a lymphoid structure around vasculature in the dura mater that can sample antigens and rapidly support humoral immune responses after local pathogen challenge.
Collapse
Affiliation(s)
- Zachary Fitzpatrick
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nagela Ghabdan Zanluqui
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
| | | | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | - Colin Y C Lee
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | | | - Maria Luciana Negro-Demontel
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
| | - Andrew P Stewart
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | - David A Posner
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| | - Monica Buckley
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
| | - Kieren S J Allinson
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Panagiotis Mastorakos
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA
- Department of Surgical Neurology, NINDS, NIH, Bethesda, MD, USA
| | | | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, NINDS, NIH, Bethesda, MD, USA
| | | | - Adel Helmy
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Tamara Tajsic
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - John R Ferdinand
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Anais Portet
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ana Peñalver
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Eleanor Gillman
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK.
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke (NINDS), National Institute of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
141
|
Deng F, Wan J, Li G, Dong H, Xia X, Wang Y, Li X, Zhuang C, Zheng Y, Liu L, Yan Y, Feng J, Zhao Y, Xie H, Li Y. Improved green and red GRAB sensors for monitoring spatiotemporal serotonin release in vivo. Nat Methods 2024; 21:692-702. [PMID: 38443508 PMCID: PMC11377854 DOI: 10.1038/s41592-024-02188-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 01/19/2024] [Indexed: 03/07/2024]
Abstract
The serotonergic system plays important roles in both physiological and pathological processes, and is a therapeutic target for many psychiatric disorders. Although several genetically encoded GFP-based serotonin (5-HT) sensors were recently developed, their sensitivities and spectral profiles are relatively limited. To overcome these limitations, we optimized green fluorescent G-protein-coupled receptor (GPCR)-activation-based 5-HT (GRAB5-HT) sensors and developed a red fluorescent GRAB5-HT sensor. These sensors exhibit excellent cell surface trafficking and high specificity, sensitivity and spatiotemporal resolution, making them suitable for monitoring 5-HT dynamics in vivo. Besides recording subcortical 5-HT release in freely moving mice, we observed both uniform and gradient 5-HT release in the mouse dorsal cortex with mesoscopic imaging. Finally, we performed dual-color imaging and observed seizure-induced waves of 5-HT release throughout the cortex following calcium and endocannabinoid waves. In summary, these 5-HT sensors can offer valuable insights regarding the serotonergic system in both health and disease.
Collapse
Affiliation(s)
- Fei Deng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jinxia Wan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Guochuan Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Hui Dong
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Xiju Xia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yipan Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Xuelin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Chaowei Zhuang
- Department of Automation, Tsinghua University, Beijing, China
| | - Yu Zheng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Laixin Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yuqi Yan
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Jiesi Feng
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Yulin Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
| | - Hao Xie
- Department of Automation, Tsinghua University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China.
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China.
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, New Cornerstone Science Laboratory, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
142
|
Miller MI, Trouvé A, Younes L. Space-feature measures on meshes for mapping spatial transcriptomics. Med Image Anal 2024; 93:103068. [PMID: 38176357 DOI: 10.1016/j.media.2023.103068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 09/18/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024]
Abstract
Advances in the development of largely automated microscopy methods such as MERFISH for imaging cellular structures in mouse brains are providing spatial detection of micron resolution gene expression. While there has been tremendous progress made in the field of Computational Anatomy (CA) to perform diffeomorphic mapping technologies at the tissue scales for advanced neuroinformatic studies in common coordinates, integration of molecular- and cellular-scale populations through statistical averaging via common coordinates remains yet unattained. This paper describes the first set of algorithms for calculating geodesics in the space of diffeomorphisms, what we term space-feature-measure LDDMM, extending the family of large deformation diffeomorphic metric mapping (LDDMM) algorithms to accommodate a space-feature action on marked particles which extends consistently to the tissue scales. It leads to the derivation of a cross-modality alignment algorithm of transcriptomic data to common coordinate systems attached to standard atlases. We represent the brain data as geometric measures, termed as space-feature measures supported by a large number of unstructured points, each point representing a small volume in space and carrying a list of densities of features elements of a high-dimensional feature space. The shape of space-feature measure brain spaces is measured by transforming them by diffeomorphisms. The metric between these measures is obtained after embedding these objects in a linear space equipped with the norm, yielding a so-called "chordal metric".
Collapse
Affiliation(s)
- Michael I Miller
- Center of Imaging Science and Department of Biomedical Engineering, Johns Hopkins University, United States of America.
| | - Alain Trouvé
- Centre Giovanni Borelli (UMR 9010), Ecole Normale Supérieure Paris-Saclay, Université Paris-Saclay, France.
| | - Laurent Younes
- Center of imaging Science and Department of Applied Mathematics and Statistics, Johns Hopkins University, United States of America.
| |
Collapse
|
143
|
Farrell JS, Hwaun E, Dudok B, Soltesz I. Neural and behavioural state switching during hippocampal dentate spikes. Nature 2024; 628:590-595. [PMID: 38480889 PMCID: PMC11023929 DOI: 10.1038/s41586-024-07192-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 02/01/2024] [Indexed: 04/06/2024]
Abstract
Distinct brain and behavioural states are associated with organized neural population dynamics that are thought to serve specific cognitive functions1-3. Memory replay events, for example, occur during synchronous population events called sharp-wave ripples in the hippocampus while mice are in an 'offline' behavioural state, enabling cognitive mechanisms such as memory consolidation and planning4-11. But how does the brain re-engage with the external world during this behavioural state and permit access to current sensory information or promote new memory formation? Here we found that the hippocampal dentate spike, an understudied population event that frequently occurs between sharp-wave ripples12, may underlie such a mechanism. We show that dentate spikes are associated with distinctly elevated brain-wide firing rates, primarily observed in higher order networks, and couple to brief periods of arousal. Hippocampal place coding during dentate spikes aligns to the mouse's current spatial location, unlike the memory replay accompanying sharp-wave ripples. Furthermore, inhibiting neural activity during dentate spikes disrupts associative memory formation. Thus, dentate spikes represent a distinct brain state and support memory during non-locomotor behaviour, extending the repertoire of cognitive processes beyond the classical offline functions.
Collapse
Affiliation(s)
- Jordan S Farrell
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- F.M. Kirby Neurobiology Center and Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
| | - Ernie Hwaun
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Barna Dudok
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
- Departments of Neurology and Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
144
|
Mansour H, Azrak R, Cook JJ, Hornburg KJ, Qi Y, Tian Y, Williams RW, Yeh FC, White LE, Johnson GA. An Open Resource: MR and light sheet microscopy stereotaxic atlas of the mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587246. [PMID: 38586051 PMCID: PMC10996689 DOI: 10.1101/2024.03.28.587246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
We have combined MR histology and light sheet microscopy (LSM) of five postmortem C57BL/6J mouse brains in a stereotaxic space based on micro-CT yielding a multimodal 3D atlas with the highest spatial and contrast resolution yet reported. Brains were imaged in situ with multi gradient echo (mGRE) and diffusion tensor imaging (DTI) at 15 μm resolution (∼ 2.4 million times that of clinical MRI). Scalar images derived from the average DTI and mGRE provide unprecedented contrast in 14 complementary 3D volumes, each highlighting distinct histologic features. The same tissues scanned with LSM and registered into the stereotaxic space provide 17 different molecular cell type stains. The common coordinate framework labels (CCFv3) complete the multimodal atlas. The atlas has been used to correct distortions in the Allen Brain Atlas and harmonize it with Franklin Paxinos. It provides a unique resource for stereotaxic labeling of mouse brain images from many sources.
Collapse
|
145
|
Ma X, Xing Y, Zhai R, Du Y, Yan H. Development and advancements in rodent MRI-based brain atlases. Heliyon 2024; 10:e27421. [PMID: 38510053 PMCID: PMC10950579 DOI: 10.1016/j.heliyon.2024.e27421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Rodents, particularly mice and rats, are extensively utilized in fundamental neuroscience research. Brain atlases have played a pivotal role in this field, evolving from traditional printed histology atlases to digital atlases incorporating diverse imaging datasets. Magnetic resonance imaging (MRI)-based brain atlases, also known as brain maps, have been employed in specific studies. However, the existence of numerous versions of MRI-based brain atlases has impeded their standardized application and widespread use, despite the consensus within the academic community regarding their significance in mice and rats. Furthermore, there is a dearth of comprehensive and systematic reviews on MRI-based brain atlases for rodents. This review aims to bridge this gap by providing a comprehensive overview of the advancements in MRI-based brain atlases for rodents, with a specific focus on mice and rats. It seeks to explore the advantages and disadvantages of histologically printed brain atlases in comparison to MRI brain atlases, delineate the standardized methods for creating MRI brain atlases, and summarize their primary applications in neuroscience research. Additionally, this review aims to assist researchers in selecting appropriate versions of MRI brain atlases for their studies or refining existing MRI brain atlas resources, thereby facilitating the development and widespread adoption of standardized MRI-based brain atlases in rodents.
Collapse
Affiliation(s)
- Xiaoyi Ma
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yao Xing
- School of Information Science and Technology, Fudan University, Shanghai, 200433, China
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Renkuan Zhai
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Yingying Du
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Huanhuan Yan
- Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, 518048, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
146
|
Willekens SMA, Morini F, Mediavilla T, Nilsson E, Orädd G, Hahn M, Chotiwan N, Visa M, Berggren PO, Ilegems E, Överby AK, Ahlgren U, Marcellino D. An MR-based brain template and atlas for optical projection tomography and light sheet fluorescence microscopy in neuroscience. Front Neurosci 2024; 18:1328815. [PMID: 38601090 PMCID: PMC11004350 DOI: 10.3389/fnins.2024.1328815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction Optical Projection Tomography (OPT) and light sheet fluorescence microscopy (LSFM) are high resolution optical imaging techniques, ideally suited for ex vivo 3D whole mouse brain imaging. Although they exhibit high specificity for their targets, the anatomical detail provided by tissue autofluorescence remains limited. Methods T1-weighted images were acquired from 19 BABB or DBE cleared brains to create an MR template using serial longitudinal registration. Afterwards, fluorescent OPT and LSFM images were coregistered/normalized to the MR template to create fusion images. Results Volumetric calculations revealed a significant difference between BABB and DBE cleared brains, leading to develop two optimized templates, with associated tissue priors and brain atlas, for BABB (OCUM) and DBE (iOCUM). By creating fusion images, we identified virus infected brain regions, mapped dopamine transporter and translocator protein expression, and traced innervation from the eye along the optic tract to the thalamus and superior colliculus using cholera toxin B. Fusion images allowed for precise anatomical identification of fluorescent signal in the detailed anatomical context provided by MR. Discussion The possibility to anatomically map fluorescent signals on magnetic resonance (MR) images, widely used in clinical and preclinical neuroscience, would greatly benefit applications of optical imaging of mouse brain. These specific MR templates for cleared brains enable a broad range of neuroscientific applications integrating 3D optical brain imaging.
Collapse
Affiliation(s)
- Stefanie M. A. Willekens
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Federico Morini
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Tomas Mediavilla
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Emma Nilsson
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Greger Orädd
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Max Hahn
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Nunya Chotiwan
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Montse Visa
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Erwin Ilegems
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Anna K. Överby
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Ulf Ahlgren
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Daniel Marcellino
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
147
|
Qian P, Manubens-Gil L, Jiang S, Peng H. Non-homogenous axonal bouton distribution in whole-brain single-cell neuronal networks. Cell Rep 2024; 43:113871. [PMID: 38451816 DOI: 10.1016/j.celrep.2024.113871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/08/2024] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
We examined the distribution of pre-synaptic contacts in axons of mouse neurons and constructed whole-brain single-cell neuronal networks using an extensive dataset of 1,891 fully reconstructed neurons. We found that bouton locations were not homogeneous throughout the axon and among brain regions. As our algorithm was able to generate whole-brain single-cell connectivity matrices from full morphology reconstruction datasets, we further found that non-homogeneous bouton locations have a significant impact on network wiring, including degree distribution, triad census, and community structure. By perturbing neuronal morphology, we further explored the link between anatomical details and network topology. In our in silico exploration, we found that dendritic and axonal tree span would have the greatest impact on network wiring, followed by synaptic contact deletion. Our results suggest that neuroanatomical details must be carefully addressed in studies of whole-brain networks at the single-cell level.
Collapse
Affiliation(s)
- Penghao Qian
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China; School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Linus Manubens-Gil
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Shengdian Jiang
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China; School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hanchuan Peng
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China.
| |
Collapse
|
148
|
Guo X, Ning J, Chen Y, Liu G, Zhao L, Fan Y, Sun S. Recent advances in differential expression analysis for single-cell RNA-seq and spatially resolved transcriptomic studies. Brief Funct Genomics 2024; 23:95-109. [PMID: 37022699 DOI: 10.1093/bfgp/elad011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/09/2022] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Differential expression (DE) analysis is a necessary step in the analysis of single-cell RNA sequencing (scRNA-seq) and spatially resolved transcriptomics (SRT) data. Unlike traditional bulk RNA-seq, DE analysis for scRNA-seq or SRT data has unique characteristics that may contribute to the difficulty of detecting DE genes. However, the plethora of DE tools that work with various assumptions makes it difficult to choose an appropriate one. Furthermore, a comprehensive review on detecting DE genes for scRNA-seq data or SRT data from multi-condition, multi-sample experimental designs is lacking. To bridge such a gap, here, we first focus on the challenges of DE detection, then highlight potential opportunities that facilitate further progress in scRNA-seq or SRT analysis, and finally provide insights and guidance in selecting appropriate DE tools or developing new computational DE methods.
Collapse
Affiliation(s)
- Xiya Guo
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Ning
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yuanze Chen
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Guoliang Liu
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Liyan Zhao
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yue Fan
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shiquan Sun
- School of Public Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
- Key Laboratory of Trace Elements and Endemic Diseases, Center for Single Cell Omics and Health, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
149
|
Vu MAT, Brown EH, Wen MJ, Noggle CA, Zhang Z, Monk KJ, Bouabid S, Mroz L, Graham BM, Zhuo Y, Li Y, Otchy TM, Tian L, Davison IG, Boas DA, Howe MW. Targeted micro-fiber arrays for measuring and manipulating localized multi-scale neural dynamics over large, deep brain volumes during behavior. Neuron 2024; 112:909-923.e9. [PMID: 38242115 PMCID: PMC10957316 DOI: 10.1016/j.neuron.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/11/2023] [Accepted: 12/15/2023] [Indexed: 01/21/2024]
Abstract
Neural population dynamics relevant to behavior vary over multiple spatial and temporal scales across three-dimensional volumes. Current optical approaches lack the spatial coverage and resolution necessary to measure and manipulate naturally occurring patterns of large-scale, distributed dynamics within and across deep brain regions such as the striatum. We designed a new micro-fiber array approach capable of chronically measuring and optogenetically manipulating local dynamics across over 100 targeted locations simultaneously in head-fixed and freely moving mice, enabling the investigation of cell-type- and neurotransmitter-specific signals over arbitrary 3D volumes at a spatial resolution and coverage previously inaccessible. We applied this method to resolve rapid dopamine release dynamics across the striatum, revealing distinct, modality-specific spatiotemporal patterns in response to salient sensory stimuli extending over millimeters of tissue. Targeted optogenetics enabled flexible control of neural signaling on multiple spatial scales, better matching endogenous signaling patterns, and the spatial localization of behavioral function across large circuits.
Collapse
Affiliation(s)
- Mai-Anh T Vu
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Eleanor H Brown
- Graduate Program for Neuroscience, Boston University, Boston, MA, USA
| | - Michelle J Wen
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA; Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Christian A Noggle
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zicheng Zhang
- Department of Biology, Boston University, Boston, MA, USA
| | - Kevin J Monk
- Department of Biology, Boston University, Boston, MA, USA
| | - Safa Bouabid
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Lydia Mroz
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA; Northeastern University, Boston, MA, USA
| | - Benjamin M Graham
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA
| | - Yizhou Zhuo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Yulong Li
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| | | | - Lin Tian
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; Max Planck Florida Institute of Neuroscience, Jupiter, FL, USA
| | - Ian G Davison
- Department of Biology, Boston University, Boston, MA, USA
| | - David A Boas
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Mark W Howe
- Department of Psychological & Brain Sciences, Boston University, Boston, MA, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
150
|
Pan MT, Zhang H, Li XJ, Guo XY. Genetically modified non-human primate models for research on neurodegenerative diseases. Zool Res 2024; 45:263-274. [PMID: 38287907 PMCID: PMC11017080 DOI: 10.24272/j.issn.2095-8137.2023.197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/25/2024] [Indexed: 01/31/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a group of debilitating neurological disorders that primarily affect elderly populations and include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). Currently, there are no therapies available that can delay, stop, or reverse the pathological progression of NDs in clinical settings. As the population ages, NDs are imposing a huge burden on public health systems and affected families. Animal models are important tools for preclinical investigations to understand disease pathogenesis and test potential treatments. While numerous rodent models of NDs have been developed to enhance our understanding of disease mechanisms, the limited success of translating findings from animal models to clinical practice suggests that there is still a need to bridge this translation gap. Old World non-human primates (NHPs), such as rhesus, cynomolgus, and vervet monkeys, are phylogenetically, physiologically, biochemically, and behaviorally most relevant to humans. This is particularly evident in the similarity of the structure and function of their central nervous systems, rendering such species uniquely valuable for neuroscience research. Recently, the development of several genetically modified NHP models of NDs has successfully recapitulated key pathologies and revealed novel mechanisms. This review focuses on the efficacy of NHPs in modeling NDs and the novel pathological insights gained, as well as the challenges associated with the generation of such models and the complexities involved in their subsequent analysis.
Collapse
Affiliation(s)
- Ming-Tian Pan
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
| | - Han Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China
| | - Xiang-Yu Guo
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong 510632, China. E-mail:
| |
Collapse
|