1
|
Naffaa MM. Neurogenesis dynamics in the olfactory bulb: deciphering circuitry organization, function, and adaptive plasticity. Neural Regen Res 2025; 20:1565-1581. [PMID: 38934393 PMCID: PMC11688548 DOI: 10.4103/nrr.nrr-d-24-00312] [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: 03/19/2024] [Revised: 05/20/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Adult neurogenesis persists after birth in the subventricular zone, with new neurons migrating to the granule cell layer and glomerular layers of the olfactory bulb, where they integrate into existing circuitry as inhibitory interneurons. The generation of these new neurons in the olfactory bulb supports both structural and functional plasticity, aiding in circuit remodeling triggered by memory and learning processes. However, the presence of these neurons, coupled with the cellular diversity within the olfactory bulb, presents an ongoing challenge in understanding its network organization and function. Moreover, the continuous integration of new neurons in the olfactory bulb plays a pivotal role in regulating olfactory information processing. This adaptive process responds to changes in epithelial composition and contributes to the formation of olfactory memories by modulating cellular connectivity within the olfactory bulb and interacting intricately with higher-order brain regions. The role of adult neurogenesis in olfactory bulb functions remains a topic of debate. Nevertheless, the functionality of the olfactory bulb is intricately linked to the organization of granule cells around mitral and tufted cells. This organizational pattern significantly impacts output, network behavior, and synaptic plasticity, which are crucial for olfactory perception and memory. Additionally, this organization is further shaped by axon terminals originating from cortical and subcortical regions. Despite the crucial role of olfactory bulb in brain functions and behaviors related to olfaction, these complex and highly interconnected processes have not been comprehensively studied as a whole. Therefore, this manuscript aims to discuss our current understanding and explore how neural plasticity and olfactory neurogenesis contribute to enhancing the adaptability of the olfactory system. These mechanisms are thought to support olfactory learning and memory, potentially through increased complexity and restructuring of neural network structures, as well as the addition of new granule granule cells that aid in olfactory adaptation. Additionally, the manuscript underscores the importance of employing precise methodologies to elucidate the specific roles of adult neurogenesis amidst conflicting data and varying experimental paradigms. Understanding these processes is essential for gaining insights into the complexities of olfactory function and behavior.
Collapse
Affiliation(s)
- Moawiah M. Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
2
|
Zeppilli S, Gurrola AO, Demetci P, Brann DH, Pham TM, Attey R, Zilkha N, Kimchi T, Datta SR, Singh R, Tosches MA, Crombach A, Fleischmann A. Single-cell genomics of the mouse olfactory cortex reveals contrasts with neocortex and ancestral signatures of cell type evolution. Nat Neurosci 2025; 28:937-948. [PMID: 40200010 DOI: 10.1038/s41593-025-01924-3] [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: 09/10/2023] [Accepted: 02/19/2025] [Indexed: 04/10/2025]
Abstract
Understanding the molecular logic of cortical cell-type diversity can illuminate cortical circuit function and evolution. Here, we performed single-nucleus transcriptome and chromatin accessibility analyses to compare neurons across three- to six-layered cortical areas of adult mice and across tetrapod species. We found that, in contrast to the six-layered neocortex, glutamatergic neurons of the three-layered mouse olfactory (piriform) cortex displayed continuous rather than discrete variation in transcriptomic profiles. Subsets of piriform and neocortical glutamatergic cells with conserved transcriptomic profiles were distinguished by distinct, area-specific epigenetic states. Furthermore, we identified a prominent population of immature neurons in piriform cortex and observed that, in contrast to the neocortex, piriform cortex exhibited divergence between glutamatergic cells in laboratory versus wild-derived mice. Finally, we showed that piriform neurons displayed greater transcriptomic similarity to cortical neurons of turtles, lizards and salamanders than to those of the neocortex. In summary, despite over 200 million years of coevolution alongside the neocortex, olfactory cortex neurons retain molecular signatures of ancestral cortical identity.
Collapse
Affiliation(s)
- Sara Zeppilli
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Alonso O Gurrola
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Pinar Demetci
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - David H Brann
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Tuan M Pham
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Robin Attey
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Noga Zilkha
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Kimchi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Sandeep R Datta
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Ritambhara Singh
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Maria A Tosches
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Anton Crombach
- Inria Centre de Lyon, Villeurbanne, France.
- INSA-Lyon, CNRS, UCBL, LIRIS, UMR5205, Villeurbanne, France.
- INSA-Lyon, CITI, UR3720, Villeurbanne, France.
| | - Alexander Fleischmann
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Dong Q, Yang Y, Luo Z, Shen H, Shi X, Liu J. Robust Spatial Cell-Type Deconvolution with Qualitative Reference for Spatial Transcriptomics. SMALL METHODS 2025; 9:e2401145. [PMID: 40059456 PMCID: PMC12103236 DOI: 10.1002/smtd.202401145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/14/2025] [Indexed: 05/26/2025]
Abstract
Many spatially resolved transcriptomic technologies have been developed to provide gene expression profiles for spots that may contain heterogeneous mixtures of cells. To decompose cellular composition and expression levels, various deconvolution methods have been developed using single-cell RNA sequencing (scRNA-seq) data with known cell-type labels as a reference. However, in the absence of a reliable reference dataset or in the presence of heterogeneous batch effects, these methods may introduce bias. Here, a Qualitative-Reference-based Spatially-Informed Deconvolution method (QR-SIDE) is developed for multi-cellular spatial transcriptomic data. Uniquely, QR-SIDE provides a detailed map of spatial heterogeneity for individual marker genes and performs robust deconvolution by adaptively adjusting the contributions of each marker gene. Simultaneously, QR-SIDE unifies cell-type deconvolution with spatial clustering and incorporates spatial information via a Potts model to promote spatial continuity. The identified spatial domains represent a meaningful biological effect in potential tissue segments. Using simulated data and three real spatial transcriptomic datasets from the 10x Visium and ST platforms, QR-SIDE demonstrates improved accuracy and robustness in cell-type deconvolution and its superiority over established methods in recognizing and delineating spatial structures within a given context. These results can facilitate a range of downstream analyses and provide a refined understanding of cellular heterogeneity.
Collapse
Affiliation(s)
- Qishi Dong
- College of Big Data and InternetShenzhen Technology UniversityShenzhen518118China
| | - Yi Yang
- The Key Laboratory of Developmental Genes and Human DiseaseSchool of Life Science and TechnologySoutheast UniversityNanjing210018China
| | - Ziye Luo
- Department of BiometricsAstrazeneca Global R&D (China) CO. Ltd.Shanghai200085, China
| | - Haipeng Shen
- Faculty of Business and EconomicsHong Kong UniversityPokfulamHong Kong SARChina
| | - Xingjie Shi
- KLATASDS‐MOE, Academy of Statistics and Interdisciplinary SciencesSchool of StatisticsEast China Normal UniversityShanghai200062China
| | - Jin Liu
- School of Data ScienceThe Chinese University of Hong Kong‐ShenzhenShenzhen518172China
| |
Collapse
|
4
|
Klimpert N, Kollo M, Brann DH, Tan C, Barry D, Ma Y, Schaefer AT, Fleischmann A. 3D spatial transcriptomics reveals the molecular structure of input and output pathways in the mouse olfactory bulb. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639192. [PMID: 40060607 PMCID: PMC11888228 DOI: 10.1101/2025.02.19.639192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
A core organizing principle of the vertebrate brain is its symmetry along multiple axes. However, the precision with which neurons, circuit modules, and brain regions align to these axes remains poorly understood. Here, we used 3D spatial transcriptomics to reconstruct the anatomical and molecular organization of the mouse olfactory bulb. We mapped the positions of nearly one thousand molecularly distinct glomeruli, the structural and functional units of odor processing, revealing highly symmetric organization across hemispheres. Within each bulb, we delineated a curved axis of symmetry that divides pairs of sister glomeruli. Gene expression in the olfactory epithelium predicted glomerular position with near-glomerular resolution. However, glomerular symmetry did not extend to deeper layer mitral and granule cells, suggesting a reorganization from sensory input to cortical output pathways. Our findings provide the first comprehensive map of the olfactory bulb and reveal how its molecular structure is instructed by epithelial gene expression programs.
Collapse
|
5
|
Schubert C, Schulz K, Sonner JK, Hadjilaou A, Seemann AL, Gierke J, Vieira V, Meurs N, Woo MS, Lohr C, Morellini F, Hirnet D, Friese MA. Neuroinflammation causes mitral cell dysfunction and olfactory impairment in a multiple sclerosis model. J Neuroinflammation 2025; 22:71. [PMID: 40057769 PMCID: PMC11889885 DOI: 10.1186/s12974-025-03388-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 02/19/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Olfactory dysfunction is an underestimated symptom in multiple sclerosis (MS). Here, we examined the pathogenic mechanisms underlying inflammation-induced dysfunction of the olfactory bulb using the animal model of MS, experimental autoimmune encephalomyelitis (EAE). RESULTS Reduced olfactory function in EAE was associated with the degeneration of short-axon neurons, immature neurons, and both mitral and tufted cells, along with their synaptic interactions and axonal repertoire. To dissect the mechanisms underlying the susceptibility of mitral cells, the main projection neurons of the olfactory bulb, we profiled their responses to neuroinflammation by single-nucleus RNA sequencing followed by functional validation. Neuroinflammation resulted in the induction of potassium channel transcripts in mitral cells, which was reflected in increased halothane-induced outward currents of these cells, likely contributing to the impaired olfaction in EAE animals. CONCLUSION This study reveals the crucial role of mitral cells and their potassium channel activity in the olfactory bulb during EAE, thereby enhancing our understanding of neuroinflammation-induced neurodegeneration in MS.
Collapse
Affiliation(s)
- Charlotte Schubert
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Schulz
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexandros Hadjilaou
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Anna-Lena Seemann
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Janine Gierke
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Lohr
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Fabio Morellini
- Research Group Behavioral Biology, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniela Hirnet
- Institute of Cell and Systems Biology of Animals, University of Hamburg, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
6
|
Gadiwalla S, Guillaume C, Huang L, White SJB, Basha N, Petersen PH, Galliano E. Ex Vivo Functional Characterization of Mouse Olfactory Bulb Projection Neurons Reveals a Heterogeneous Continuum. eNeuro 2025; 12:ENEURO.0407-24.2025. [PMID: 39904626 PMCID: PMC11881907 DOI: 10.1523/eneuro.0407-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Mitral cells (MCs) and tufted cells (TCs) in the olfactory bulb (OB) act as an input convergence hub and transmit information to higher olfactory areas. Since first characterized, they have been classed as distinct projection neurons based on size and location: laminarly arranged MCs with a diameter larger than 20 µm in the mitral layer (ML) and smaller TCs spread across both the ML and external plexiform layers (EPL). Recent in vivo work has shown that these neurons encode complementary olfactory information, akin to parallel channels in other sensory systems. Yet, many ex vivo studies still collapse them into a single class, mitral/tufted, when describing their physiological properties and impact on circuit function. Using immunohistochemistry and whole-cell patch-clamp electrophysiology in fixed or acute slices from adult mice, we attempted to align in vivo and ex vivo data and test a soma size-based classifier of bulbar projection neurons using passive and intrinsic firing properties. We found that there is no clear separation between cell types based on passive or active properties. Rather, there is a heterogeneous continuum with three loosely clustered subgroups: TCs in the EPL, and putative tufted or putative MCs in the ML. These findings illustrate the large functional heterogeneity present within the OB projection neurons and complement existing literature highlighting how heterogeneity in sensory systems is preponderant and possibly used in the OB to decode complex olfactory information.
Collapse
Affiliation(s)
- Sana Gadiwalla
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EL, United Kingdom
- Department of Anatomy, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik 102, Iceland
| | - Chloé Guillaume
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EL, United Kingdom
| | - Li Huang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EL, United Kingdom
| | - Samuel J B White
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EL, United Kingdom
| | - Nihal Basha
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EL, United Kingdom
| | - Pétur Henry Petersen
- Department of Anatomy, Biomedical Center, Faculty of Medicine, University of Iceland, Reykjavik 102, Iceland
| | - Elisa Galliano
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB23EL, United Kingdom
| |
Collapse
|
7
|
Mi XQ, Liu BC, Qu L, Yuan Y, Li H, Xu AY, Zhang YL, Xie JX, Song N. Intranasal iron administration induces iron deposition, immunoactivation, and cell-specific vulnerability in the olfactory bulb of C57BL/6 mice. Zool Res 2025; 46:209-224. [PMID: 39846197 PMCID: PMC11891001 DOI: 10.24272/j.issn.2095-8137.2024.240] [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: 09/18/2024] [Accepted: 10/14/2024] [Indexed: 01/24/2025] Open
Abstract
Iron is the most abundant transition metal in the brain and is essential for brain development and neuronal function; however, its abnormal accumulation is also implicated in various neurological disorders. The olfactory bulb (OB), an early target in neurodegenerative diseases, acts as a gateway for environmental toxins and contains diverse neuronal populations with distinct roles. This study explored the cell-specific vulnerability to iron in the OB using a mouse model of intranasal administration of ferric ammonium citrate (FAC). Olfactory function was assessed through olfactory discrimination tests, while iron levels in OB tissues, cerebrospinal fluid (CSF), and serum were quantified using inductively coupled plasma mass spectrometry (ICP-MS), immunohistochemical staining, and iron assays. Transcriptomic changes and immune responses were assessed using RNA sequencing and immune cell infiltration analysis. Results showed that intranasal FAC administration impaired olfactory function, accompanied by iron deposition in the olfactory mucosa and OB, as well as damage to olfactory sensory neurons. Notably, these effects occurred without elevations in CSF or serum iron levels. OB iron accumulation activated multiple immune cells, including microglia and astrocytes, but did not trigger ferroptosis. Spatial transcriptomic sequencing of healthy adult mouse OBs revealed significant cellular heterogeneity, with an abundance of neuroglia and neurons. Among neurons, GABAergic neurons were the most prevalent, followed by glutamatergic and dopaminergic neurons, while cholinergic and serotonergic neurons were sparsely distributed. Under iron-stressed conditions, oligodendrocytes, dopaminergic neurons, and glutamatergic neurons exhibited significant damage, while GABAergic neurons remained unaffected. These findings highlight the selective vulnerability of neuronal and glial populations to iron-induced stress, offering novel insights into the loss of specific cell types in the OB during iron dysregulation.
Collapse
Affiliation(s)
- Xiao-Qing Mi
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
| | - Bao-Chen Liu
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
| | - Le Qu
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yu Yuan
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Hui Li
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
| | - Ao-Yang Xu
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
| | - Yu-Lin Zhang
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China
| | - Jun-Xia Xie
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China. E-mail:
| | - Ning Song
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Brain Diseases, Qingdao University, Qingdao, Shandong, 266071, China. E-mail:
| |
Collapse
|
8
|
Decoster L, Trova S, Zucca S, Bulk J, Gouveia A, Ternier G, Lhomme T, Legrand A, Gallet S, Boehm U, Wyatt A, Wahl V, Wartenberg P, Hrabovszky E, Rácz G, Luzzati F, Nato G, Fogli M, Peretto P, Schriever SC, Bernecker M, Pfluger PT, Steculorum SM, Bovetti S, Rasika S, Prevot V, Silva MSB, Giacobini P. A GnRH neuronal population in the olfactory bulb translates socially relevant odors into reproductive behavior in male mice. Nat Neurosci 2024; 27:1758-1773. [PMID: 39095587 DOI: 10.1038/s41593-024-01724-1] [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: 05/17/2023] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons regulate fertility and integrate hormonal status with environmental cues to ensure reproductive success. Here we show that GnRH neurons in the olfactory bulb (GnRHOB) of adult mice can mediate social recognition. Specifically, we show that GnRHOB neurons extend neurites into the vomeronasal organ and olfactory epithelium and project to the median eminence. GnRHOB neurons in males express vomeronasal and olfactory receptors, are activated by female odors and mediate gonadotropin release in response to female urine. Male preference for female odors required the presence and activation of GnRHOB neurons, was impaired after genetic inhibition or ablation of these cells and relied on GnRH signaling in the posterodorsal medial amygdala. GnRH receptor expression in amygdala kisspeptin neurons appear to be required for GnRHOB neurons' actions on male mounting behavior. Taken together, these results establish GnRHOB neurons as regulating fertility, sex recognition and mating in male mice.
Collapse
Affiliation(s)
- Laurine Decoster
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sara Trova
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Centro CMP3VdA, Istituto Italiano di Tecnologia (IIT), Aosta, Italy
| | - Stefano Zucca
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Janice Bulk
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Ayden Gouveia
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
| | - Gaetan Ternier
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Amandine Legrand
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Sarah Gallet
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Center for Gender-specific Biology and Medicine (CGBM), Saarland University School of Medicine, Homburg, Germany
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Hun-Ren Institute of Experimental Medicine, Budapest, Hungary
| | - Gergely Rácz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Federico Luzzati
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Giulia Nato
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
- Department of Neuroscience "Rita Levi Montalcini", University of Turin, Turin, Italy
| | - Marco Fogli
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sonja C Schriever
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Miriam Bernecker
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Paul T Pfluger
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of Neurobiology of Diabetes, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Sophie M Steculorum
- Max Planck Institute for Metabolism Research, Max Planck Research Group Neurocircuit Wiring and Function, Cologne, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano, Italy
| | - Sowmyalakshmi Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
| | - Mauro S B Silva
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paolo Giacobini
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, FHU 1000 Days for Health, School of Medicine, Lille, France.
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, Labex DistAlz, Lille, France.
| |
Collapse
|
9
|
Ma Y, Zhou X. Accurate and efficient integrative reference-informed spatial domain detection for spatial transcriptomics. Nat Methods 2024; 21:1231-1244. [PMID: 38844627 PMCID: PMC11831598 DOI: 10.1038/s41592-024-02284-9] [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: 05/10/2023] [Accepted: 04/18/2024] [Indexed: 06/23/2024]
Abstract
Spatially resolved transcriptomics (SRT) studies are becoming increasingly common and large, offering unprecedented opportunities in mapping complex tissue structures and functions. Here we present integrative and reference-informed tissue segmentation (IRIS), a computational method designed to characterize tissue spatial organization in SRT studies through accurately and efficiently detecting spatial domains. IRIS uniquely leverages single-cell RNA sequencing data for reference-informed detection of biologically interpretable spatial domains, integrating multiple SRT slices while explicitly considering correlations both within and across slices. We demonstrate the advantages of IRIS through in-depth analysis of six SRT datasets encompassing diverse technologies, tissues, species and resolutions. In these applications, IRIS achieves substantial accuracy gains (39-1,083%) and speed improvements (4.6-666.0) in moderate-sized datasets, while representing the only method applicable for large datasets including Stereo-seq and 10x Xenium. As a result, IRIS reveals intricate brain structures, uncovers tumor microenvironment heterogeneity and detects structural changes in diabetes-affected testis, all with exceptional speed and accuracy.
Collapse
Affiliation(s)
- Ying Ma
- Department of Biostatistics, Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
McKissick O, Klimpert N, Ritt JT, Fleischmann A. Odors in space. Front Neural Circuits 2024; 18:1414452. [PMID: 38978957 PMCID: PMC11228174 DOI: 10.3389/fncir.2024.1414452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/29/2024] [Indexed: 07/10/2024] Open
Abstract
As an evolutionarily ancient sense, olfaction is key to learning where to find food, shelter, mates, and important landmarks in an animal's environment. Brain circuitry linking odor and navigation appears to be a well conserved multi-region system among mammals; the anterior olfactory nucleus, piriform cortex, entorhinal cortex, and hippocampus each represent different aspects of olfactory and spatial information. We review recent advances in our understanding of the neural circuits underlying odor-place associations, highlighting key choices of behavioral task design and neural circuit manipulations for investigating learning and memory.
Collapse
Affiliation(s)
- Olivia McKissick
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Nell Klimpert
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Jason T Ritt
- Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Alexander Fleischmann
- Department of Neuroscience and Carney Institute for Brain Science, Brown University, Providence, RI, United States
| |
Collapse
|
11
|
Park SJ, Wang IH, Lee N, Jiang HC, Uemura T, Futai K, Kim D, Macosko E, Greer P. Combinatorial expression of neurexin genes regulates glomerular targeting by olfactory sensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587570. [PMID: 38617205 PMCID: PMC11014570 DOI: 10.1101/2024.04.01.587570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Precise connectivity between specific neurons is essential for the formation of the complex neural circuitry necessary for executing intricate motor behaviors and higher cognitive functions. While trans -interactions between synaptic membrane proteins have emerged as crucial elements in orchestrating the assembly of these neural circuits, the synaptic surface proteins involved in neuronal wiring remain largely unknown. Here, using unbiased single-cell transcriptomic and mouse genetic approaches, we uncover that the neurexin family of genes enables olfactory sensory neuron (OSNs) axons to form appropriate synaptic connections with their mitral and tufted (M/T) cell synaptic partners, within the mammalian olfactory system. Neurexin isoforms are differentially expressed within distinct populations of OSNs, resulting in unique pattern of neurexin expression that is specific to each OSN type, and synergistically cooperate to regulate axonal innervation, guiding OSN axons to their designated glomeruli. This process is facilitated through the interactions of neurexins with their postsynaptic partners, including neuroligins, which have distinct expression patterns in M/T cells. Our findings suggest a novel mechanism underpinning the precise assembly of olfactory neural circuits, driven by the trans -interaction between neurexins and their ligands.
Collapse
|
12
|
Hirata T. Olfactory information processing viewed through mitral and tufted cell-specific channels. Front Neural Circuits 2024; 18:1382626. [PMID: 38523698 PMCID: PMC10957668 DOI: 10.3389/fncir.2024.1382626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Parallel processing is a fundamental strategy of sensory coding. Through this processing, unique and distinct features of sensations are computed and projected to the central targets. This review proposes that mitral and tufted cells, which are the second-order projection neurons in the olfactory bulb, contribute to parallel processing within the olfactory system. Based on anatomical and functional evidence, I discuss potential features that could be conveyed through the unique channel formed by these neurons.
Collapse
Affiliation(s)
- Tatsumi Hirata
- Brain Function Laboratory, National Institute of Genetics, SOKENDAI, Mishima, Japan
| |
Collapse
|
13
|
Lindeman S, Fu X, Reinert JK, Fukunaga I. Value-related learning in the olfactory bulb occurs through pathway-dependent perisomatic inhibition of mitral cells. PLoS Biol 2024; 22:e3002536. [PMID: 38427708 PMCID: PMC10936853 DOI: 10.1371/journal.pbio.3002536] [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: 09/16/2023] [Revised: 03/13/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024] Open
Abstract
Associating values to environmental cues is a critical aspect of learning from experiences, allowing animals to predict and maximise future rewards. Value-related signals in the brain were once considered a property of higher sensory regions, but their wide distribution across many brain regions is increasingly recognised. Here, we investigate how reward-related signals begin to be incorporated, mechanistically, at the earliest stage of olfactory processing, namely, in the olfactory bulb. In head-fixed mice performing Go/No-Go discrimination of closely related olfactory mixtures, rewarded odours evoke widespread inhibition in one class of output neurons, that is, in mitral cells but not tufted cells. The temporal characteristics of this reward-related inhibition suggest it is odour-driven, but it is also context-dependent since it is absent during pseudo-conditioning and pharmacological silencing of the piriform cortex. Further, the reward-related modulation is present in the somata but not in the apical dendritic tuft of mitral cells, suggesting an involvement of circuit components located deep in the olfactory bulb. Depth-resolved imaging from granule cell dendritic gemmules suggests that granule cells that target mitral cells receive a reward-related extrinsic drive. Thus, our study supports the notion that value-related modulation of olfactory signals is a characteristic of olfactory processing in the primary olfactory area and narrows down the possible underlying mechanisms to deeper circuit components that contact mitral cells perisomatically.
Collapse
Affiliation(s)
- Sander Lindeman
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Xiaochen Fu
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Janine Kristin Reinert
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Izumi Fukunaga
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
14
|
Jammal Salameh L, Bitzenhofer SH, Hanganu-Opatz IL, Dutschmann M, Egger V. Blood pressure pulsations modulate central neuronal activity via mechanosensitive ion channels. Science 2024; 383:eadk8511. [PMID: 38301001 DOI: 10.1126/science.adk8511] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/11/2023] [Indexed: 02/03/2024]
Abstract
The transmission of the heartbeat through the cerebral vascular system causes intracranial pressure pulsations. We discovered that arterial pressure pulsations can directly modulate central neuronal activity. In a semi-intact rat brain preparation, vascular pressure pulsations elicited correlated local field oscillations in the olfactory bulb mitral cell layer. These oscillations did not require synaptic transmission but reflected baroreceptive transduction in mitral cells. This transduction was mediated by a fast excitatory mechanosensitive ion channel and modulated neuronal spiking activity. In awake animals, the heartbeat entrained the activity of a subset of olfactory bulb neurons within ~20 milliseconds. Thus, we propose that this fast, intrinsic interoceptive mechanism can modulate perception-for example, during arousal-within the olfactory bulb and possibly across various other brain areas.
Collapse
Affiliation(s)
- Luna Jammal Salameh
- Neurophysiology Group, Zoological Institute, Regensburg University, 93040 Regensburg, Germany
| | - Sebastian H Bitzenhofer
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ileana L Hanganu-Opatz
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, Hamburg Center of Neuroscience, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Mathias Dutschmann
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Veronica Egger
- Neurophysiology Group, Zoological Institute, Regensburg University, 93040 Regensburg, Germany
| |
Collapse
|
15
|
Prakash N, Matos HY, Sebaoui S, Tsai L, Tran T, Aromolaran A, Atrachji I, Campbell N, Goodrich M, Hernandez-Pineda D, Jesus Herrero M, Hirata T, Lischinsky J, Martinez W, Torii S, Yamashita S, Hosseini H, Sokolowski K, Esumi S, Kawasawa YI, Hashimoto-Torii K, Jones KS, Corbin JG. Connectivity and molecular profiles of Foxp2- and Dbx1-lineage neurons in the accessory olfactory bulb and medial amygdala. J Comp Neurol 2024; 532:e25545. [PMID: 37849047 PMCID: PMC10922300 DOI: 10.1002/cne.25545] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
In terrestrial vertebrates, the olfactory system is divided into main (MOS) and accessory (AOS) components that process both volatile and nonvolatile cues to generate appropriate behavioral responses. While much is known regarding the molecular diversity of neurons that comprise the MOS, less is known about the AOS. Here, focusing on the vomeronasal organ (VNO), the accessory olfactory bulb (AOB), and the medial amygdala (MeA), we reveal that populations of neurons in the AOS can be molecularly subdivided based on their ongoing or prior expression of the transcription factors Foxp2 or Dbx1, which delineate separate populations of GABAergic output neurons in the MeA. We show that a majority of AOB neurons that project directly to the MeA are of the Foxp2 lineage. Using single-neuron patch-clamp electrophysiology, we further reveal that in addition to sex-specific differences across lineage, the frequency of excitatory input to MeA Dbx1- and Foxp2-lineage neurons differs between sexes. Together, this work uncovers a novel molecular diversity of AOS neurons, and lineage and sex differences in patterns of connectivity.
Collapse
Affiliation(s)
- Nandkishore Prakash
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Heidi Y Matos
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Sonia Sebaoui
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Luke Tsai
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Tuyen Tran
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Adejimi Aromolaran
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Isabella Atrachji
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Nya Campbell
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Meredith Goodrich
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - David Hernandez-Pineda
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Maria Jesus Herrero
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Tsutomu Hirata
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Julieta Lischinsky
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Wendolin Martinez
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Shisui Torii
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Hassan Hosseini
- Department of Pharmacology, University of Michigan Medical
School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan
Medical School, Ann Arbor, MI 48109, USA
| | - Katie Sokolowski
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Shigeyuki Esumi
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, PA, USA
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| | - Kevin S Jones
- Department of Pharmacology, University of Michigan Medical
School, Ann Arbor, MI, USA; Neuroscience Graduate Program, University of Michigan
Medical School, Ann Arbor, MI 48109, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children’s
Research Institute, Children’s National Hospital, Washington DC, USA
| |
Collapse
|
16
|
Fang Z, Liu T, Zheng R, A J, Yin M, Li M. stAA: adversarial graph autoencoder for spatial clustering task of spatially resolved transcriptomics. Brief Bioinform 2023; 25:bbad500. [PMID: 38189544 PMCID: PMC10772985 DOI: 10.1093/bib/bbad500] [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: 10/11/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
With the development of spatially resolved transcriptomics technologies, it is now possible to explore the gene expression profiles of single cells while preserving their spatial context. Spatial clustering plays a key role in spatial transcriptome data analysis. In the past 2 years, several graph neural network-based methods have emerged, which significantly improved the accuracy of spatial clustering. However, accurately identifying the boundaries of spatial domains remains a challenging task. In this article, we propose stAA, an adversarial variational graph autoencoder, to identify spatial domain. stAA generates cell embedding by leveraging gene expression and spatial information using graph neural networks and enforces the distribution of cell embeddings to a prior distribution through Wasserstein distance. The adversarial training process can make cell embeddings better capture spatial domain information and more robust. Moreover, stAA incorporates global graph information into cell embeddings using labels generated by pre-clustering. Our experimental results show that stAA outperforms the state-of-the-art methods and achieves better clustering results across different profiling platforms and various resolutions. We also conducted numerous biological analyses and found that stAA can identify fine-grained structures in tissues, recognize different functional subtypes within tumors and accurately identify developmental trajectories.
Collapse
Affiliation(s)
- Zhaoyu Fang
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Teng Liu
- Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Chongqing 404031, China
- Translational Medicine Research Center (TMRC), School of Medicine, Chongqing University, Chongqing 401331, China
| | - Ruiqing Zheng
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Jin A
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Mingzhu Yin
- Clinical Research Center (CRC), Medical Pathology Center (MPC), Cancer Early Detection and Treatment Center (CEDTC), Chongqing University Three Gorges Hospital, Chongqing University, Chongqing 404031, China
- Translational Medicine Research Center (TMRC), School of Medicine, Chongqing University, Chongqing 401331, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| |
Collapse
|
17
|
Guo T, Yuan Z, Pan Y, Wang J, Chen F, Zhang MQ, Li X. SPIRAL: integrating and aligning spatially resolved transcriptomics data across different experiments, conditions, and technologies. Genome Biol 2023; 24:241. [PMID: 37864231 PMCID: PMC10590036 DOI: 10.1186/s13059-023-03078-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/29/2023] [Indexed: 10/22/2023] Open
Abstract
Properly integrating spatially resolved transcriptomics (SRT) generated from different batches into a unified gene-spatial coordinate system could enable the construction of a comprehensive spatial transcriptome atlas. Here, we propose SPIRAL, consisting of two consecutive modules: SPIRAL-integration, with graph domain adaptation-based data integration, and SPIRAL-alignment, with cluster-aware optimal transport-based coordination alignment. We verify SPIRAL with both synthetic and real SRT datasets. By encoding spatial correlations to gene expressions, SPIRAL-integration surpasses state-of-the-art methods in both batch effect removal and joint spatial domain identification. By aligning spots cluster-wise, SPIRAL-alignment achieves more accurate coordinate alignments than existing methods.
Collapse
Affiliation(s)
- Tiantian Guo
- School of Software Engineering, Beijing Jiaotong University, Beijing, 100044, China
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic & Systems Biology, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, 200433, China
| | - Yan Pan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jiakang Wang
- School of Software Engineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Fengling Chen
- Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Michael Q Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas, Richardson, TX, 75080-3021, USA.
| | - Xiangyu Li
- School of Software Engineering, Beijing Jiaotong University, Beijing, 100044, China.
| |
Collapse
|
18
|
Chitra U, Arnold BJ, Sarkar H, Ma C, Lopez-Darwin S, Sanno K, Raphael BJ. Mapping the topography of spatial gene expression with interpretable deep learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561757. [PMID: 37873258 PMCID: PMC10592770 DOI: 10.1101/2023.10.10.561757] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Spatially resolved transcriptomics technologies provide high-throughput measurements of gene expression in a tissue slice, but the sparsity of this data complicates the analysis of spatial gene expression patterns such as gene expression gradients. We address these issues by deriving a topographic map of a tissue slice-analogous to a map of elevation in a landscape-using a novel quantity called the isodepth. Contours of constant isodepth enclose spatial domains with distinct cell type composition, while gradients of the isodepth indicate spatial directions of maximum change in gene expression. We develop GASTON, an unsupervised and interpretable deep learning algorithm that simultaneously learns the isodepth, spatial gene expression gradients, and piecewise linear functions of the isodepth that model both continuous gradients and discontinuous spatial variation in the expression of individual genes. We validate GASTON by showing that it accurately identifies spatial domains and marker genes across several biological systems. In SRT data from the brain, GASTON reveals gradients of neuronal differentiation and firing, and in SRT data from a tumor sample, GASTON infers gradients of metabolic activity and epithelial-mesenchymal transition (EMT)-related gene expression in the tumor microenvironment.
Collapse
Affiliation(s)
- Uthsav Chitra
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Brian J. Arnold
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | | | - Kohei Sanno
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | | |
Collapse
|
19
|
Zeppilli S, Gurrola AO, Demetci P, Brann DH, Attey R, Zilkha N, Kimchi T, Datta SR, Singh R, Tosches MA, Crombach A, Fleischmann A. Mammalian olfactory cortex neurons retain molecular signatures of ancestral cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.13.553130. [PMID: 37645751 PMCID: PMC10461972 DOI: 10.1101/2023.08.13.553130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The cerebral cortex diversified extensively during vertebrate evolution. Intriguingly, the three-layered mammalian olfactory cortex resembles the cortical cytoarchitecture of non-mammals yet evolved alongside the six-layered neocortex, enabling unique comparisons for investigating cortical neuron diversification. We performed single-nucleus multiome sequencing across mouse three- to six-layered cortices and compared neuron types across mice, reptiles and salamander. We identified neurons that are olfactory cortex-specific or conserved across mouse cortical areas. However, transcriptomically similar neurons exhibited area-specific epigenetic states. Additionally, the olfactory cortex showed transcriptomic divergence between lab and wild-derived mice, suggesting enhanced circuit plasticity through adult immature neurons. Finally, olfactory cortex neurons displayed marked transcriptomic similarities to reptile and salamander neurons. Together, these data indicate that the mammalian olfactory cortex retains molecular signatures representative of ancestral cortical traits.
Collapse
|
20
|
Schoof M, Epplen GD, Walter C, Ballast A, Holdhof D, Göbel C, Neyazi S, Varghese J, Albert TK, Kerl K, Schüller U. The tumor suppressor CREBBP and the oncogene MYCN cooperate to induce malignant brain tumors in mice. Oncogenesis 2023; 12:36. [PMID: 37407554 DOI: 10.1038/s41389-023-00481-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
The tumor suppressor and chromatin modifier cAMP response element-binding protein binding protein (CREBBP) and v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog (MYCN), a member of the MYC oncogene family, are critically involved in brain development. Both genes are frequently mutated in the same tumor entities, including high-grade glioma and medulloblastoma. Therefore, we hypothesized that alterations in both genes cooperate to induce brain tumor formation. For further investigation, hGFAP-cre::CrebbpFl/Fl::lsl-MYCN mice were generated, which combine Crebbp deletion with overexpression of MYCN in neural stem cells (NSCs). Within eight months, these animals developed aggressive forebrain tumors. The first tumors were detectable in the olfactory bulbs of seven-day-old mice. This location raises the possibility that presumptive founder cells are derived from the ventricular-subventricular zone (V-SVZ). To examine the cellular biology of these tumors, single-cell RNA sequencing was performed, which revealed high intratumoral heterogeneity. Data comparison with reference CNS cell types indicated the highest similarity of tumor cells with transit-amplifying NSCs or activated NSCs of the V-SVZ. Consequently, we analyzed V-SVZ NSCs of our mouse model aiming to confirm that the tumors originate from this stem cell niche. Mutant V-SVZ NSCs showed significantly increased cell viability and proliferation as well as reduced glial and neural differentiation in vitro compared to control cells. In summary, we demonstrate the oncogenic potential of a combined loss of function of CREBBP and overexpression of MYCN in this cell population. hGFAP-cre::CrebbpFl/Fl::lsl-MYCN mice thus provide a valuable tool to study tumor-driving mechanisms in a key neural stem/ progenitor cell niche.
Collapse
Affiliation(s)
- Melanie Schoof
- Research Institute Children`s Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Carolin Walter
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Annika Ballast
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Dörthe Holdhof
- Research Institute Children`s Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carolin Göbel
- Research Institute Children`s Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina Neyazi
- Research Institute Children`s Cancer Center, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Thomas Karl Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Ulrich Schüller
- Research Institute Children`s Cancer Center, Hamburg, Germany.
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
21
|
Piwecka M, Rajewsky N, Rybak-Wolf A. Single-cell and spatial transcriptomics: deciphering brain complexity in health and disease. Nat Rev Neurol 2023; 19:346-362. [PMID: 37198436 PMCID: PMC10191412 DOI: 10.1038/s41582-023-00809-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 05/19/2023]
Abstract
In the past decade, single-cell technologies have proliferated and improved from their technically challenging beginnings to become common laboratory methods capable of determining the expression of thousands of genes in thousands of cells simultaneously. The field has progressed by taking the CNS as a primary research subject - the cellular complexity and multiplicity of neuronal cell types provide fertile ground for the increasing power of single-cell methods. Current single-cell RNA sequencing methods can quantify gene expression with sufficient accuracy to finely resolve even subtle differences between cell types and states, thus providing a great tool for studying the molecular and cellular repertoire of the CNS and its disorders. However, single-cell RNA sequencing requires the dissociation of tissue samples, which means that the interrelationships between cells are lost. Spatial transcriptomic methods bypass tissue dissociation and retain this spatial information, thereby allowing gene expression to be assessed across thousands of cells within the context of tissue structural organization. Here, we discuss how single-cell and spatially resolved transcriptomics have been contributing to unravelling the pathomechanisms underlying brain disorders. We focus on three areas where we feel these new technologies have provided particularly useful insights: selective neuronal vulnerability, neuroimmune dysfunction and cell-type-specific treatment response. We also discuss the limitations and future directions of single-cell and spatial RNA sequencing technologies.
Collapse
Affiliation(s)
- Monika Piwecka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Nikolaus Rajewsky
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Agnieszka Rybak-Wolf
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
22
|
Kraus A, Garcia B, Ma J, Herrera KJ, Zwaka H, Harpaz R, Wong RY, Engert F, Salinas I. Olfactory detection of viruses shapes brain immunity and behavior in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.17.533129. [PMID: 37034630 PMCID: PMC10081220 DOI: 10.1101/2023.03.17.533129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Olfactory sensory neurons (OSNs) are constantly exposed to pathogens, including viruses. However, serious brain infection via the olfactory route rarely occurs. When OSNs detect a virus, they coordinate local antiviral immune responses to stop virus progression to the brain. Despite effective immune control in the olfactory periphery, pathogen-triggered neuronal signals reach the CNS via the olfactory bulb (OB). We hypothesized that neuronal detection of a virus by OSNs initiates neuroimmune responses in the OB that prevent pathogen invasion. Using zebrafish ( Danio rerio ) as a model, we demonstrate viral-specific neuronal activation of OSNs projecting into the OB, indicating that OSNs are electrically activated by viruses. Further, behavioral changes are seen in both adult and larval zebrafish after viral exposure. By profiling the transcription of single cells in the OB after OSNs are exposed to virus, we found that both microglia and neurons enter a protective state. Microglia and macrophage populations in the OB respond within minutes of nasal viral delivery followed decreased expression of neuronal differentiation factors and enrichment of genes in the neuropeptide signaling pathway in neuronal clusters. Pituitary adenylate-cyclase-activating polypeptide ( pacap ), a known antimicrobial, was especially enriched in a neuronal cluster. We confirm that PACAP is antiviral in vitro and that PACAP expression increases in the OB 1 day post-viral treatment. Our work reveals how encounters with viruses in the olfactory periphery shape the vertebrate brain by inducing antimicrobial programs in neurons and by altering host behavior.
Collapse
|
23
|
Organizational Principles of the Centrifugal Projections to the Olfactory Bulb. Int J Mol Sci 2023; 24:ijms24054579. [PMID: 36902010 PMCID: PMC10002860 DOI: 10.3390/ijms24054579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Centrifugal projections in the olfactory system are critical to both olfactory processing and behavior. The olfactory bulb (OB), the first relay station in odor processing, receives a substantial number of centrifugal inputs from the central brain regions. However, the anatomical organization of these centrifugal connections has not been fully elucidated, especially for the excitatory projection neurons of the OB, the mitral/tufted cells (M/TCs). Using rabies virus-mediated retrograde monosynaptic tracing in Thy1-Cre mice, we identified that the three most prominent inputs of the M/TCs came from the anterior olfactory nucleus (AON), the piriform cortex (PC), and the basal forebrain (BF), similar to the granule cells (GCs), the most abundant population of inhibitory interneurons in the OB. However, M/TCs received proportionally less input from the primary olfactory cortical areas, including the AON and PC, but more input from the BF and contralateral brain regions than GCs. Unlike organizationally distinct inputs from the primary olfactory cortical areas to these two types of OB neurons, inputs from the BF were organized similarly. Furthermore, individual BF cholinergic neurons innervated multiple layers of the OB, forming synapses on both M/TCs and GCs. Taken together, our results indicate that the centrifugal projections to different types of OB neurons may provide complementary and coordinated strategies in olfactory processing and behavior.
Collapse
|
24
|
Chen Y, Chen X, Baserdem B, Zhan H, Li Y, Davis MB, Kebschull JM, Zador AM, Koulakov AA, Albeanu DF. High-throughput sequencing of single neuron projections reveals spatial organization in the olfactory cortex. Cell 2022; 185:4117-4134.e28. [PMID: 36306734 PMCID: PMC9681627 DOI: 10.1016/j.cell.2022.09.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 07/22/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022]
Abstract
In most sensory modalities, neuronal connectivity reflects behaviorally relevant stimulus features, such as spatial location, orientation, and sound frequency. By contrast, the prevailing view in the olfactory cortex, based on the reconstruction of dozens of neurons, is that connectivity is random. Here, we used high-throughput sequencing-based neuroanatomical techniques to analyze the projections of 5,309 mouse olfactory bulb and 30,433 piriform cortex output neurons at single-cell resolution. Surprisingly, statistical analysis of this much larger dataset revealed that the olfactory cortex connectivity is spatially structured. Single olfactory bulb neurons targeting a particular location along the anterior-posterior axis of piriform cortex also project to matched, functionally distinct, extra-piriform targets. Moreover, single neurons from the targeted piriform locus also project to the same matched extra-piriform targets, forming triadic circuit motifs. Thus, as in other sensory modalities, olfactory information is routed at early stages of processing to functionally diverse targets in a coordinated manner.
Collapse
Affiliation(s)
- Yushu Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xiaoyin Chen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Huiqing Zhan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Yan Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Martin B Davis
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Anthony M Zador
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| | | | - Dinu F Albeanu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
25
|
Kolling LJ, Tatti R, Lowry T, Loeven AM, Fadool JM, Fadool DA. Modulating the Excitability of Olfactory Output Neurons Affects Whole-Body Metabolism. J Neurosci 2022; 42:5966-5990. [PMID: 35710623 PMCID: PMC9337614 DOI: 10.1523/jneurosci.0190-22.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 01/29/2023] Open
Abstract
Metabolic state can alter olfactory sensitivity, but it is unknown whether the activity of the olfactory bulb (OB) may fine tune metabolic homeostasis. Our objective was to use CRISPR gene editing in male and female mice to enhance the excitability of mitral/tufted projection neurons (M/TCs) of the OB to test for improved metabolic health. Ex vivo slice recordings of MCs in CRISPR mice confirmed increased excitability due the targeted loss of Kv1.3 channels, which resulted in a less negative resting membrane potential (RMP), enhanced action potential (AP) firing, and insensitivity to the selective channel blocker margatoxin (MgTx). CRISPR mice exhibited enhanced odor discrimination using a habituation/dishabituation paradigm. CRISPR mice were challenged for 25 weeks with a moderately high-fat (MHF) diet, and compared with littermate controls, male mice were resistance to diet-induced obesity (DIO). Female mice did not exhibit DIO. CRISPR male mice gained less body weight, accumulated less white adipose tissue, cleared a glucose challenge more quickly, and had less serum leptin and liver triglycerides. CRISPR male mice consumed equivalent calories as control littermates, and had unaltered energy expenditure (EE) and locomotor activity, but used more fats for metabolic substrate over that of carbohydrates. Counter to CRISPR-engineered mice, by using chemogenetics to decrease M/TC excitability in male mice, activation of inhibitory designer receptors exclusively activated by designer drugs (DREADDs) caused a decrease in odor discrimination, and resulted in a metabolic profile that was obesogenic, mice had reduced EE and oxygen consumption (VO2). We conclude that the activity of M/TC projection neurons canonically carries olfactory information and simultaneously can regulate whole-body metabolism.SIGNIFICANCE STATEMENT The olfactory system drives food choice, and olfactory sensitivity is strongly correlated to hunger and fullness. Olfactory function thereby influences nutritional balance and obesity outcomes. Obesity has become a health and financial crisis in America, shortening life expectancy and increasing the severity of associated illnesses. It is expected that 51% of Americans will be obese by the year 2030. Using CRISPR gene editing and chemogenetic approaches, we discovered that changing the excitability of output neurons in the olfactory bulb (OB) affects metabolism and body weight stabilization in mice. Our results suggest that long-term therapeutic targeting of OB activity to higher processing centers may be a future clinical treatment of obesity or type II Diabetes.
Collapse
Affiliation(s)
- Louis John Kolling
- Institute of Molecular Biophysics, The Florida State University, Tallahassee, Florida 32306
| | - Roberta Tatti
- Department of Biological Science, The Florida State University, Tallahassee, Florida 32306
| | - Troy Lowry
- Department of Biological Science, The Florida State University, Tallahassee, Florida 32306
| | - Ashley M Loeven
- Department of Biological Science, The Florida State University, Tallahassee, Florida 32306
| | - James M Fadool
- Department of Biological Science, The Florida State University, Tallahassee, Florida 32306
- Program in Neuroscience, The Florida State University, Tallahassee, Florida 32306
| | - Debra Ann Fadool
- Institute of Molecular Biophysics, The Florida State University, Tallahassee, Florida 32306
- Department of Biological Science, The Florida State University, Tallahassee, Florida 32306
- Program in Neuroscience, The Florida State University, Tallahassee, Florida 32306
| |
Collapse
|
26
|
Aghvami SS, Kubota Y, Egger V. Anatomical and Functional Connectivity at the Dendrodendritic Reciprocal Mitral Cell-Granule Cell Synapse: Impact on Recurrent and Lateral Inhibition. Front Neural Circuits 2022; 16:933201. [PMID: 35937203 PMCID: PMC9355734 DOI: 10.3389/fncir.2022.933201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
In the vertebrate olfactory bulb, reciprocal dendrodendritic interactions between its principal neurons, the mitral and tufted cells, and inhibitory interneurons in the external plexiform layer mediate both recurrent and lateral inhibition, with the most numerous of these interneurons being granule cells. Here, we used recently established anatomical parameters and functional data on unitary synaptic transmission to simulate the strength of recurrent inhibition of mitral cells specifically from the reciprocal spines of rat olfactory bulb granule cells in a quantitative manner. Our functional data allowed us to derive a unitary synaptic conductance on the order of 0.2 nS. The simulations predicted that somatic voltage deflections by even proximal individual granule cell inputs are below the detection threshold and that attenuation with distance is roughly linear, with a passive length constant of 650 μm. However, since recurrent inhibition in the wake of a mitral cell action potential will originate from hundreds of reciprocal spines, the summated recurrent IPSP will be much larger, even though there will be substantial mutual shunting across the many inputs. Next, we updated and refined a preexisting model of connectivity within the entire rat olfactory bulb, first between pairs of mitral and granule cells, to estimate the likelihood and impact of recurrent inhibition depending on the distance between cells. Moreover, to characterize the substrate of lateral inhibition, we estimated the connectivity via granule cells between any two mitral cells or all the mitral cells that belong to a functional glomerular ensemble (i.e., which receive their input from the same glomerulus), again as a function of the distance between mitral cells and/or entire glomerular mitral cell ensembles. Our results predict the extent of the three regimes of anatomical connectivity between glomerular ensembles: high connectivity within a glomerular ensemble and across the first four rings of adjacent glomeruli, substantial connectivity to up to eleven glomeruli away, and negligible connectivity beyond. Finally, in a first attempt to estimate the functional strength of granule-cell mediated lateral inhibition, we combined this anatomical estimate with our above simulation results on attenuation with distance, resulting in slightly narrowed regimes of a functional impact compared to the anatomical connectivity.
Collapse
Affiliation(s)
- S. Sara Aghvami
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Yoshiyuki Kubota
- Division of Cerebral Circuitry, National Institute for Physiological Sciences (NIPS), Okazaki, Japan
| | - Veronica Egger
- Neurophysiology, Institute of Zoology, Regensburg University, Regensburg, Germany
| |
Collapse
|
27
|
Nagel M, Chesler AT. PIEZO2 ion channels in proprioception. Curr Opin Neurobiol 2022; 75:102572. [PMID: 35689908 DOI: 10.1016/j.conb.2022.102572] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/28/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022]
Abstract
PIEZO2 is a stretch-gated ion channel that is expressed at high levels in somatosensory neurons. Humans with rare mutations in the PIEZO2 gene have profound mechanosensory deficits that include a loss of the sense of proprioception. These striking phenotypes match those seen in conditional knockout mouse models demonstrating the highly conserved function for this gene. Here, we review the ramifications of loss of PIEZO2 function on normal daily activities and what studies like these have revealed about proprioception at the molecular and cellular level. Additionally, we highlight recent work that has uncovered the surprising functional and molecular diversity of proprioceptors. Together, these findings pioneer a path toward determining how the detection of mechanosensory input from muscles and tendons is used to control posture and refine motor performance.
Collapse
Affiliation(s)
- Maximilian Nagel
- Sensory Cells and Circuits Section, National Center for Complementary and Integrative Health, 35 Convent Drive, Bethesda, MD, 20892, USA
| | - Alexander T Chesler
- Sensory Cells and Circuits Section, National Center for Complementary and Integrative Health, 35 Convent Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
28
|
Dasgupta D, Warner TPA, Erskine A, Schaefer AT. Coupling of Mouse Olfactory Bulb Projection Neurons to Fluctuating Odor Pulses. J Neurosci 2022; 42:4278-4296. [PMID: 35440491 PMCID: PMC9145232 DOI: 10.1523/jneurosci.1422-21.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 11/28/2022] Open
Abstract
Odors are transported by turbulent air currents, creating complex temporal fluctuations in odor concentration that provide a potentially informative stimulus dimension. We have shown that mice are able to discriminate odor stimuli based on their temporal structure, indicating that information contained in the temporal structure of odor plumes can be extracted by the mouse olfactory system. Here, using in vivo extracellular and intracellular electrophysiological recordings, we show that mitral cells (MCs) and tufted cells (TCs) of the male C57BL/6 mouse olfactory bulb can encode the dominant temporal frequencies present in odor stimuli up to at least 20 Hz. A substantial population of cell-odor pairs showed significant coupling of their subthreshold membrane potential with the odor stimulus at both 2 Hz (29/70) and the suprasniff frequency 20 Hz (24/70). Furthermore, mitral/tufted cells (M/TCs) show differential coupling of their membrane potential to odor concentration fluctuations with tufted cells coupling more strongly for the 20 Hz stimulation. Frequency coupling was always observed to be invariant to odor identity, and M/TCs that coupled well to a mixture also coupled to at least one of the components of the mixture. Interestingly, pharmacological blocking of the inhibitory circuitry strongly modulated frequency coupling of cell-odor pairs at both 2 Hz (10/15) and 20 Hz (9/15). These results provide insight into how both cellular and circuit properties contribute to the encoding of temporal odor features in the mouse olfactory bulb.SIGNIFICANCE STATEMENT Odors in the natural environment have a strong temporal structure that can be extracted and used by mice in their behavior. Here, using in vivo extracellular and intracellular electrophysiological techniques, we show that the projection neurons in the olfactory bulb can encode and couple to the dominant frequency present in an odor stimulus. Furthermore, frequency coupling was observed to be differential between mitral and tufted cells and was odor invariant but strongly modulated by local inhibitory circuits. In summary, this study provides insight into how both cellular and circuit properties modulate encoding of odor temporal features in the mouse olfactory bulb.
Collapse
Affiliation(s)
- Debanjan Dasgupta
- Sensory Circuits and Neurotechnology Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Tom P A Warner
- Sensory Circuits and Neurotechnology Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Andrew Erskine
- Sensory Circuits and Neurotechnology Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Andreas T Schaefer
- Sensory Circuits and Neurotechnology Laboratory, Francis Crick Institute, London NW1 1AT, United Kingdom
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
29
|
Tufo C, Poopalasundaram S, Dorrego-Rivas A, Ford MC, Graham A, Grubb MS. Development of the mammalian main olfactory bulb. Development 2022; 149:274348. [PMID: 35147186 PMCID: PMC8918810 DOI: 10.1242/dev.200210] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The mammalian main olfactory bulb is a crucial processing centre for the sense of smell. The olfactory bulb forms early during development and is functional from birth. However, the olfactory system continues to mature and change throughout life as a target of constitutive adult neurogenesis. Our Review synthesises current knowledge of prenatal, postnatal and adult olfactory bulb development, focusing on the maturation, morphology, functions and interactions of its diverse constituent glutamatergic and GABAergic cell types. We highlight not only the great advances in the understanding of olfactory bulb development made in recent years, but also the gaps in our present knowledge that most urgently require addressing. Summary: This Review describes the morphological and functional maturation of cells in the mammalian main olfactory bulb, from embryonic development to adult neurogenesis.
Collapse
Affiliation(s)
- Candida Tufo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Subathra Poopalasundaram
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Ana Dorrego-Rivas
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Marc C Ford
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Anthony Graham
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Matthew S Grubb
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| |
Collapse
|
30
|
Koldaeva A, Zhang C, Huang YP, Reinert JK, Mizuno S, Sugiyama F, Takahashi S, Soliman T, Matsunami H, Fukunaga I. Generation and Characterization of a Cell Type-Specific, Inducible Cre-Driver Line to Study Olfactory Processing. J Neurosci 2021; 41:6449-6467. [PMID: 34099512 PMCID: PMC8318078 DOI: 10.1523/jneurosci.3076-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
In sensory systems of the brain, mechanisms exist to extract distinct features from stimuli to generate a variety of behavioral repertoires. These often correspond to different cell types at various stages in sensory processing. In the mammalian olfactory system, complex information processing starts in the olfactory bulb, whose output is conveyed by mitral cells (MCs) and tufted cells (TCs). Despite many differences between them, and despite the crucial position they occupy in the information hierarchy, Cre-driver lines that distinguish them do not yet exist. Here, we sought to identify genes that are differentially expressed between MCs and TCs of the mouse, with an ultimate goal to generate a cell type-specific Cre-driver line, starting from a transcriptome analysis using a large and publicly available single-cell RNA-seq dataset (Zeisel et al., 2018). Many genes were differentially expressed, but only a few showed consistent expressions in MCs and at the specificity required. After further validating these putative markers using ISH, two genes (i.e., Pkib and Lbdh2) remained as promising candidates. Using CRISPR/Cas9-mediated gene editing, we generated Cre-driver lines and analyzed the resulting recombination patterns. This indicated that our new inducible Cre-driver line, Lbhd2-CreERT2, can be used to genetically label MCs in a tamoxifen dose-dependent manner, both in male and female mice, as assessed by soma locations, projection patterns, and sensory-evoked responses in vivo Hence, this is a promising tool for investigating cell type-specific contributions to olfactory processing and demonstrates the power of publicly accessible data in accelerating science.SIGNIFICANCE STATEMENT In the brain, distinct cell types play unique roles. It is therefore important to have tools for studying unique cell types specifically. For the sense of smell in mammals, information is processed first by circuits of the olfactory bulb, where two types of cells, mitral cells and tufted cells, output different information. We generated a transgenic mouse line that enables mitral cells to be specifically labeled or manipulated. This was achieved by looking for genes that are specific to mitral cells using a large and public gene expression dataset, and creating a transgenic mouse using the gene editing technique, CRISPR/Cas9. This will allow scientists to better investigate parallel information processing underlying the sense of smell.
Collapse
Affiliation(s)
- Anzhelika Koldaeva
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan, 904-0495
| | - Cary Zhang
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan, 904-0495
| | - Yu-Pei Huang
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan, 904-0495
| | - Janine Kristin Reinert
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan, 904-0495
| | - Seiya Mizuno
- Laboratory Animal Resource Center, Tsukuba University, Ibaraki, Japan, 305-8577
| | - Fumihiro Sugiyama
- Laboratory Animal Resource Center, Tsukuba University, Ibaraki, Japan, 305-8577
| | - Satoru Takahashi
- Laboratory Animal Resource Center, Tsukuba University, Ibaraki, Japan, 305-8577
| | - Taha Soliman
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan, 904-0495
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology and Department of Neurobiology, Duke University, Durham, North Carolina, 27710
| | - Izumi Fukunaga
- Sensory and Behavioural Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan, 904-0495
| |
Collapse
|