1
|
Gamlin CR, Schneider-Mizell CM, Mallory M, Elabbady L, Gouwens N, Williams G, Mukora A, Dalley R, Bodor AL, Brittain D, Buchanan J, Bumbarger DJ, Joyce E, Kapner D, Kinn S, Mahalingam G, Seshamani S, Takeno M, Torres R, Yin W, Nicovich PR, Bae JA, Castro MA, Dorkenwald S, Halageri A, Jia Z, Jordan C, Kemnitz N, Lee K, Li K, Lu R, Macrina T, Mitchell E, Mondal SS, Mu S, Nehoran B, Popovych S, Silversmith W, Turner NL, Wong W, Wu J, Yu SC, Berg J, Jarsky T, Lee B, Seung HS, Zeng H, Reid RC, Collman F, da Costa NM, Sorensen SA. Connectomics of predicted Sst transcriptomic types in mouse visual cortex. Nature 2025; 640:497-505. [PMID: 40205210 PMCID: PMC11981948 DOI: 10.1038/s41586-025-08805-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 02/18/2025] [Indexed: 04/11/2025]
Abstract
Neural circuit function is shaped both by the cell types that comprise the circuit and the connections between them1. Neural cell types have previously been defined by morphology2,3, electrophysiology4, transcriptomic expression5,6, connectivity7-9 or a combination of such modalities10-12. The Patch-seq technique enables the characterization of morphology, electrophysiology and transcriptomic properties from individual cells13-15. These properties were integrated to define 28 inhibitory, morpho-electric-transcriptomic (MET) cell types in mouse visual cortex16, which do not include synaptic connectivity. Conversely, large-scale electron microscopy (EM) enables morphological reconstruction and a near-complete description of a neuron's local synaptic connectivity, but does not include transcriptomic or electrophysiological information. Here, we leveraged morphological information from Patch-seq to predict the transcriptomically defined cell subclass and/or MET-type of inhibitory neurons within a large-scale EM dataset. We further analysed Martinotti cells-a somatostatin (Sst)-positive17 morphological cell type18,19-which were classified successfully into Sst MET-types with distinct axon myelination and synaptic output connectivity patterns. We demonstrate that morphological features can be used to link cell types across experimental modalities, enabling further comparison of connectivity to gene expression and electrophysiology. We observe unique connectivity rules for predicted Sst cell types.
Collapse
Affiliation(s)
| | | | | | - Leila Elabbady
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | | | | | - Alice Mukora
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Emily Joyce
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Sam Kinn
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Marc Takeno
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Wenjing Yin
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - J Alexander Bae
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Electrical and Computer Engineering Department, Princeton University, Princeton, NJ, USA
| | - Manuel A Castro
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Sven Dorkenwald
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Akhilesh Halageri
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Zhen Jia
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Chris Jordan
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Nico Kemnitz
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Kisuk Lee
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Brain and Cognitive Sciences Department, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kai Li
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Ran Lu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Thomas Macrina
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Eric Mitchell
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Shanka Subhra Mondal
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Electrical and Computer Engineering Department, Princeton University, Princeton, NJ, USA
| | - Shang Mu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Barak Nehoran
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Sergiy Popovych
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | | | - Nicholas L Turner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - William Wong
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Jingpeng Wu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Szi-Chieh Yu
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Jim Berg
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Tim Jarsky
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Brian Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | - H Sebastian Seung
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
- Computer Science Department, Princeton University, Princeton, NJ, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - R Clay Reid
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | |
Collapse
|
2
|
Luo W, Egger M, Cruz-Ochoa N, Tse A, Maloveczky G, Tamás B, Lukacsovich D, Seng C, Amrein I, Lukacsovich T, Wolfer D, Földy C. Activation of feedforward wiring in adult hippocampal neurons by the basic-helix-loop-helix transcription factor Ascl4. PNAS NEXUS 2024; 3:pgae174. [PMID: 38711810 PMCID: PMC11071515 DOI: 10.1093/pnasnexus/pgae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024]
Abstract
Although evidence indicates that the adult brain retains a considerable capacity for circuit formation, adult wiring has not been broadly considered and remains poorly understood. In this study, we investigate wiring activation in adult neurons. We show that the basic-helix-loop-helix transcription factor Ascl4 can induce wiring in different types of hippocampal neurons of adult mice. The new axons are mainly feedforward and reconfigure synaptic weights in the circuit. Mice with the Ascl4-induced circuits do not display signs of pathology and solve spatial problems equally well as controls. Our results demonstrate reprogrammed connectivity by a single transcriptional factor and provide insights into the regulation of brain wiring in adults.
Collapse
Affiliation(s)
- Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Matteo Egger
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| | - Natalia Cruz-Ochoa
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| | - Alice Tse
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Gyula Maloveczky
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Bálint Tamás
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Charlotte Seng
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - Irmgard Amrein
- Institute of Anatomy, Faculty of Medicine, University of Zürich, Zürich 8057, Switzerland
| | - Tamás Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
| | - David Wolfer
- Institute of Anatomy, Faculty of Medicine, University of Zürich, Zürich 8057, Switzerland
- Institute of Human Movement Sciences and Sport, D-HEST, ETH Zürich, Zürich 8057, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich 8057, Switzerland
- Adaptive Brain Circuits in Development and Learning (AdaBD), University Research Priority Program (URPP), University of Zürich, Zürich 8057, Switzerland
| |
Collapse
|
3
|
Liu Z, Xue J, Liu C, Tang J, Wu S, Lin J, Han J, Zhang Q, Wu C, Huang H, Zhao L, Zhuo Y, Li Y. Selective deletion of zinc transporter 3 in amacrine cells promotes retinal ganglion cell survival and optic nerve regeneration after injury. Neural Regen Res 2023; 18:2773-2780. [PMID: 37449644 DOI: 10.4103/1673-5374.373660] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Vision depends on accurate signal conduction from the retina to the brain through the optic nerve, an important part of the central nervous system that consists of bundles of axons originating from retinal ganglion cells. The mammalian optic nerve, an important part of the central nervous system, cannot regenerate once it is injured, leading to permanent vision loss. To date, there is no clinical treatment that can regenerate the optic nerve and restore vision. Our previous study found that the mobile zinc (Zn2+) level increased rapidly after optic nerve injury in the retina, specifically in the vesicles of the inner plexiform layer. Furthermore, chelating Zn2+ significantly promoted axonal regeneration with a long-term effect. In this study, we conditionally knocked out zinc transporter 3 (ZnT3) in amacrine cells or retinal ganglion cells to construct two transgenic mouse lines (VGATCreZnT3fl/fl and VGLUT2CreZnT3fl/fl, respectively). We obtained direct evidence that the rapidly increased mobile Zn2+ in response to injury was from amacrine cells. We also found that selective deletion of ZnT3 in amacrine cells promoted retinal ganglion cell survival and axonal regeneration after optic nerve crush injury, improved retinal ganglion cell function, and promoted vision recovery. Sequencing analysis of reginal ganglion cells revealed that inhibiting the release of presynaptic Zn2+ affected the transcription of key genes related to the survival of retinal ganglion cells in postsynaptic neurons, regulated the synaptic connection between amacrine cells and retinal ganglion cells, and affected the fate of retinal ganglion cells. These results suggest that amacrine cells release Zn2+ to trigger transcriptomic changes related to neuronal growth and survival in reginal ganglion cells, thereby influencing the synaptic plasticity of retinal networks. These results make the theory of zinc-dependent retinal ganglion cell death more accurate and complete and provide new insights into the complex interactions between retinal cell networks.
Collapse
Affiliation(s)
- Zhe Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Jingfei Xue
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Canying Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Siting Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Jicheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Jiaxu Han
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Caiqing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Haishun Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, Guangdong Province, China, Guangzhou
| |
Collapse
|
4
|
Abashkin DA, Karpov DS, Kurishev AO, Marilovtseva EV, Golimbet VE. ASCL1 Is Involved in the Pathogenesis of Schizophrenia by Regulation of Genes Related to Cell Proliferation, Neuronal Signature Formation, and Neuroplasticity. Int J Mol Sci 2023; 24:15746. [PMID: 37958729 PMCID: PMC10648210 DOI: 10.3390/ijms242115746] [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: 09/08/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Schizophrenia (SZ) is a common psychiatric neurodevelopmental disorder with a complex genetic architecture. Genome-wide association studies indicate the involvement of several transcription factors, including ASCL1, in the pathogenesis of SZ. We aimed to identify ASCL1-dependent cellular and molecular mechanisms associated with SZ. We used Capture-C, CRISPR/Cas9 systems and RNA-seq analysis to confirm the involvement of ASCL1 in SZ-associated pathogenesis, establish a mutant SH-SY5Y line with a functional ASCL1 knockout (ASCL1-del) and elucidate differentially expressed genes that may underlie ASCL1-dependent pathogenic mechanisms. Capture-C confirmed the spatial interaction of the ASCL1 promoter with SZ-associated loci. Transcriptome analysis showed that ASCL1 regulation may be through a negative feedback mechanism. ASCL1 dysfunction affects the expression of genes associated with the pathogenesis of SZ, as well as bipolar and depressive disorders. Genes differentially expressed in ASCL1-del are involved in cell mitosis, neuronal projection, neuropeptide signaling, and the formation of intercellular contacts, including the synapse. After retinoic acid (RA)-induced differentiation, ASCL1 activity is restricted to a small subset of genes involved in neuroplasticity. These data suggest that ASCL1 dysfunction promotes SZ development predominantly before the onset of neuronal differentiation by slowing cell proliferation and impeding the formation of neuronal signatures.
Collapse
Affiliation(s)
| | - Dmitry S. Karpov
- Mental Health Research Center, Kashirskoe Sh., 34, Moscow 115522, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str. 32, Moscow 119991, Russia
| | | | | | - Vera E. Golimbet
- Mental Health Research Center, Kashirskoe Sh., 34, Moscow 115522, Russia
| |
Collapse
|
5
|
Danielli S, Ma Z, Pantazi E, Kumar A, Demarco B, Fischer FA, Paudel U, Weissenrieder J, Lee RJ, Joyce S, Foskett JK, Bezbradica JS. The ion channel CALHM6 controls bacterial infection-induced cellular cross-talk at the immunological synapse. EMBO J 2023; 42:e111450. [PMID: 36861806 PMCID: PMC10068325 DOI: 10.15252/embj.2022111450] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 03/03/2023] Open
Abstract
Membrane ion channels of the calcium homeostasis modulator (CALHM) family promote cell-cell crosstalk at neuronal synapses via ATP release, where ATP acts as a neurotransmitter. CALHM6, the only CALHM highly expressed in immune cells, has been linked to the induction of natural killer (NK) cell anti-tumour activity. However, its mechanism of action and broader functions in the immune system remain unclear. Here, we generated Calhm6-/- mice and report that CALHM6 is important for the regulation of the early innate control of Listeria monocytogenes infection in vivo. We find that CALHM6 is upregulated in macrophages by pathogen-derived signals and that it relocates from the intracellular compartment to the macrophage-NK cell synapse, facilitating ATP release and controlling the kinetics of NK cell activation. Anti-inflammatory cytokines terminate CALHM6 expression. CALHM6 forms an ion channel when expressed in the plasma membrane of Xenopus oocytes, where channel opening is controlled by a conserved acidic residue, E119. In mammalian cells, CALHM6 is localised to intracellular compartments. Our results contribute to the understanding of neurotransmitter-like signal exchange between immune cells that fine-tunes the timing of innate immune responses.
Collapse
Affiliation(s)
- Sara Danielli
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Zhongming Ma
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eirini Pantazi
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Amrendra Kumar
- Department of Veterans AffairsTennessee Valley Healthcare SystemNashvilleTNUSA
- Department of Pathology, Microbiology, & ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Benjamin Demarco
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Fabian A Fischer
- The Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Usha Paudel
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Jillian Weissenrieder
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Robert J Lee
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Department of Otorhinolaryngology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Sebastian Joyce
- Department of Veterans AffairsTennessee Valley Healthcare SystemNashvilleTNUSA
- Department of Pathology, Microbiology, & ImmunologyVanderbilt University Medical CenterNashvilleTNUSA
| | - J Kevin Foskett
- Department of Physiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Department of Cell and Developmental Biology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | | |
Collapse
|
6
|
Rozhkova IN, Okotrub SV, Brusentsev EY, Uldanova KE, Chuyko EА, Naprimerov VA, Lipina TV, Amstislavskaya TG, Amstislavsky SY. Alterations in the social-conditioned place preference and density of dopaminergic neurons in the ventral tegmental area in Clsnt2-KO mice. Vavilovskii Zhurnal Genet Selektsii 2023; 27:177-184. [PMID: 37063509 PMCID: PMC10090113 DOI: 10.18699/vjgb-23-14] [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: 06/11/2022] [Revised: 10/21/2022] [Accepted: 12/12/2022] [Indexed: 04/18/2023] Open
Abstract
The incidence of autistic spectrum disorders (ASD) constantly increases in the world. Studying the mechanisms underlying ASD as well as searching for new therapeutic targets are crucial tasks. Many researchers agree that autism is a neurodevelopmental disorder. Clstn2-KO mouse strain with a knockout of calsyntenin 2 gene (Clstn2) is model for investigating ASD. This study aims to evaluate the social-conditioned place preference as well as density of dopaminergic (DA) neurons in the ventral tegmental area (VTA), which belongs to the brain reward system, in the males of the Clstn2-KO strain using wild type C57BL/6J males as controls. Social-conditioned place preference test evaluates a reward-dependent component of social behavior. The results of this test revealed differences between the Clstn2-KO and the control males, as the former did not value socializing with the familiar partner, spending equal time in the isolation- and socializing-associated compartments. The Clstn2-KO group entered both compartments more frequently, but spent less time in the socializing-associated compartment compared to the controls. By contrast, the control males of the C57BL/6J strain spent more time in socializing-associated compartment and less time in the compartment that was associated with loneness. At the same time, an increased number of DA and possibly GABA neurons labeled with antibodies against the type 2 dopamine receptor as well as against tyrosine hydroxylase were detected in the VTA of the Clstn2-KO mice. Thus, a change in social-conditioned place preference in Clstn2-KO mice as well as a higher number of neurons expressing type 2 dopamine receptors and tyrosine hydroxylase in the VTA, the key structure of the mesolimbic dopaminergic pathway, were observed.
Collapse
Affiliation(s)
- I N Rozhkova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S V Okotrub
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E Yu Brusentsev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - K E Uldanova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E А Chuyko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - V A Naprimerov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia Novosibirsk State Agricultural University, Novosibirsk, Russia
| | | | - T G Amstislavskaya
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia
| | - S Ya Amstislavsky
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
7
|
Cognitive impairments correlate with increased central nervous system immune activation after allogeneic haematopoietic stem cell transplantation. Leukemia 2023; 37:888-900. [PMID: 36792657 PMCID: PMC10079537 DOI: 10.1038/s41375-023-01840-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Murine studies indicate that, after allogeneic haematopoietic stem cell transplantation (aHSCT), donor-derived macrophages replace damaged microglia and alloreactive T-cells invade the central nervous system (CNS). The clinical relevance of this is unknown. We assessed CNS immune surveillance and metabolic activity involved in neuronal survival, in relation to fatigue and cognitive dysfunction in 25 long-term survivors after aHSCT. Patients with cognitive dysfunction exhibited increased proportions of activated T-cells and CD16 + NK-cells in the cerebrospinal fluid (CSF). Immune cell activation was paralleled with reduced levels of anti-inflammatory factors involved in T-cell suppression (transforming growth factor-β, programmed death ligand-1), NK-cell regulation (poliovirus receptor, nectin-2), and macrophage and microglia activation (CD200, chemokine [C-X3-C motif] ligand-1). Additionally, the CSF mRNA expression pattern was associated with neuroinflammation and oxidative stress. Furthermore, proteomic, and transcriptomic studies demonstrated decreased levels of neuroprotective factors, and an upregulation of apoptosis pathway genes. The kynurenine pathway of tryptophan metabolism was activated in the CNS of all aHSCT patients, resulting in accumulation of neurotoxic and pro-inflammatory metabolites. Cognitive decline and fatigue are overlooked but frequent complications of aHSCT. This study links post-transplant CNS inflammation and neurotoxicity to our previously reported hypoactivation in the prefrontal cortex during cognitive testing, suggesting novel treatment targets.
Collapse
|
8
|
Rozhkova IN, Okotrub SV, Brusentsev EY, Uldanova EE, Chuyko EА, Lipina TV, Amstislavskaya TG, Amstislavsky SY. Neuronal density in the brain cortex and hippocampus in Clsnt2-KO mouse strain modeling autistic spectrum disorder. Vavilovskii Zhurnal Genet Selektsii 2022; 26:365-370. [PMID: 35975241 PMCID: PMC9333157 DOI: 10.18699/vjgb-22-44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/11/2022] [Accepted: 03/01/2022] [Indexed: 11/23/2022] Open
Abstract
Autistic spectrum disorders (ASD) represent conditions starting in childhood, which are characterized by diff iculties with social interaction and communication, as well as non-typical and stereotyping models of behavior. The mechanisms and the origin of these disorders are not yet understood and thus far there is a lack of prophylactic measures for these disorders. The current study aims to estimate neuronal density in the prefrontal cortex and four hippocampal subf ields, i. e. СA1, СA2, СA3, and DG in Clstn2-KO mice as a genetic model of ASD. In addition, the level of
neurogenesis was measured in the DG area of the hippocampus. This mouse strain was obtained by a knockout of the
calsinthenin-2 gene (Clsnt2) in C57BL/6J mice; the latter (wild type) was used as controls. To estimate neuronal density,
serial sections were prepared on a cryotome for the above-mentioned brain structures with the subsequent immunohistochemical
labeling and confocal microscopy; the neuronal marker (anti-NeuN) was used as the primary antibody.
In addition, neurogenesis was estimated in the DG region of the hippocampus; for this purpose, a primary antibody
against doublecortin (anti-DCX) was used. In all cases Goat anti-rabbit IgG was used as the secondary antibody. The
density of neurons in the CA1 region of the hippocampus was lower in Clstn2-KO mice of both sexes as compared with
controls. Moreover, in males of both strains, neuronal density in this region was lower as compared to females. Besides,
the differences between males and females were revealed in two other hippocampal regions. In the CA2 region, a lower
density of neurons was observed in males of both strains, and in the CA3 region, a lower density of neurons was also
observed in males as compared to females but only in C57BL/6J mice. No difference between the studied groups was
revealed in neurogenesis, nor was it in neuronal density in the prefrontal cortex or DG hippocampal region. Our new
f indings indicate that calsyntenin-2 regulates neuronal hippocampal density in subf ield-specif ic manner, suggesting
that the CA1 neuronal subpopulation may represent a cellular target for early-life preventive therapy of ASD.
Collapse
Affiliation(s)
- I. N. Rozhkova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences
| | - S. V. Okotrub
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences; Novosibirsk State University
| | - E. Yu. Brusentsev
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences
| | - E. E. Uldanova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences
| | - E. А. Chuyko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences; Novosibirsk State University
| | | | | | - S. Ya. Amstislavsky
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences
| |
Collapse
|
9
|
Wu J, Zhao Z, Shi Y, He M. Cortical VIP + Interneurons in the Upper and Deeper Layers Are Transcriptionally Distinct. J Mol Neurosci 2022; 72:1779-1795. [PMID: 35708842 DOI: 10.1007/s12031-022-02040-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/07/2022] [Indexed: 12/19/2022]
Abstract
Different interneuron classes have distinct laminar distribution patterns which contribute to the layer-specific organization of cortical microcircuits. However, laminar differences within the same interneuron classes are not well recognized. Despite systematic efforts towards neuron cell-type taxonomy in the neocortex by single-cell transcriptomics, less attention has been driven towards laminar differences in interneurons compared to projection neurons. VIP+ interneurons are the major interneuron class that mostly populate superficial layers and mediate disinhibition. A few reports noted the morphological and electrophysiological differences between VIP+ interneurons residing in layers I-III (upper layer) and layers IV-VI (deeper layer), but little is known about their molecular differences. Here, we delineated the laminar difference in their transcriptome employing single-cell RNA sequencing (scRNAseq) data from public databases. Analysis of 1175 high-quality VIP+ interneurons in the primary visual cortex (VISp) showed that the upper layer and deeper layer VIP+ interneurons are transcriptionally distinct distinguished by genes implicated in synapse organization and regulation of membrane potential. Similar differences are also observed in the anterior lateral motor cortex (ALM) and primary motor cortex (MOp). Cross-comparing between the top 10 differentially expressed genes (DEGs) with Allen Mouse Brain in situ hybridization database, we identified Tac2 and CxCl14 as potential marker genes of upper layer VIP+ interneurons across most cortical regions. Importantly, such expression patterns are conserved in the human brain. Together, we revealed significant laminar differences in transcriptomic profiles within VIP+ interneurons, which provided new insight into their molecular heterogeneity that may contribute to their functional diversity.
Collapse
Affiliation(s)
- Jinyun Wu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhirong Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yun Shi
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Miao He
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurobiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Liu Z, Jiang M, Liakath-Ali K, Sclip A, Ko J, Zhang RS, Südhof TC. Deletion of Calsyntenin-3, an atypical cadherin, suppresses inhibitory synapses but increases excitatory parallel-fiber synapses in cerebellum. eLife 2022; 11:e70664. [PMID: 35420982 PMCID: PMC9064300 DOI: 10.7554/elife.70664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 04/14/2022] [Indexed: 01/11/2023] Open
Abstract
Cadherins contribute to the organization of nearly all tissues, but the functions of several evolutionarily conserved cadherins, including those of calsyntenins, remain enigmatic. Puzzlingly, two distinct, non-overlapping functions for calsyntenins were proposed: As postsynaptic neurexin ligands in synapse formation, or as presynaptic kinesin adaptors in vesicular transport. Here, we show that, surprisingly, acute CRISPR-mediated deletion of calsyntenin-3 in mouse cerebellum in vivo causes a large decrease in inhibitory synapse, but a robust increase in excitatory parallel-fiber synapses in Purkinje cells. As a result, inhibitory synaptic transmission was suppressed, whereas parallel-fiber synaptic transmission was enhanced in Purkinje cells by the calsyntenin-3 deletion. No changes in the dendritic architecture of Purkinje cells or in climbing-fiber synapses were detected. Sparse selective deletion of calsyntenin-3 only in Purkinje cells recapitulated the synaptic phenotype, indicating that calsyntenin-3 acts by a cell-autonomous postsynaptic mechanism in cerebellum. Thus, by inhibiting formation of excitatory parallel-fiber synapses and promoting formation of inhibitory synapses in the same neuron, calsyntenin-3 functions as a postsynaptic adhesion molecule that regulates the excitatory/inhibitory balance in Purkinje cells.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Man Jiang
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Kif Liakath-Ali
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Alessandra Sclip
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and TechnologyDaeguRepublic of Korea
| | - Roger Shen Zhang
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford UniversityStanfordUnited States
- Howard Hughes Medical Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
11
|
Mori K, Koebis M, Nakao K, Kobayashi S, Kiyama Y, Watanabe M, Manabe T, Iino Y, Aiba A. Loss of calsyntenin paralogs disrupts interneuron stability and mouse behavior. Mol Brain 2022; 15:23. [PMID: 35279170 PMCID: PMC8917637 DOI: 10.1186/s13041-022-00909-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/02/2022] [Indexed: 11/26/2022] Open
Abstract
Calsyntenins (CLSTNs) are important synaptic molecules whose molecular functions are not fully understood. Although mutations in calsyntenin (CLSTN) genes have been associated with psychiatric disorders in humans, their function is still unclear. One of the reasons why the function of CLSTNs in the nervous system has not been clarified is the functional redundancy among the three paralogs. Therefore, to investigate the functions of mammalian CLSTNs, we generated triple knockout (TKO) mice lacking all CLSTN paralogs and examined their behavior. The mutant mice tended to freeze in novel environments and exhibited hypersensitivity to stress. Consistent with this, glucose levels under stress were significantly higher in the mutant mice than in the wild-type controls. In particular, phenotypes such as decreased motivation, which had not been reported in single Clstn KO mice, were newly discovered. The TKO mice generated in this study represent an important mouse model for clarifying the function of CLSTN in the future.
Collapse
|
12
|
Caporale N, Leemans M, Birgersson L, Germain PL, Cheroni C, Borbély G, Engdahl E, Lindh C, Bressan RB, Cavallo F, Chorev NE, D'Agostino GA, Pollard SM, Rigoli MT, Tenderini E, Tobon AL, Trattaro S, Troglio F, Zanella M, Bergman Å, Damdimopoulou P, Jönsson M, Kiess W, Kitraki E, Kiviranta H, Nånberg E, Öberg M, Rantakokko P, Rudén C, Söder O, Bornehag CG, Demeneix B, Fini JB, Gennings C, Rüegg J, Sturve J, Testa G. From cohorts to molecules: Adverse impacts of endocrine disrupting mixtures. Science 2022; 375:eabe8244. [PMID: 35175820 DOI: 10.1126/science.abe8244] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Convergent evidence associates exposure to endocrine disrupting chemicals (EDCs) with major human diseases, even at regulation-compliant concentrations. This might be because humans are exposed to EDC mixtures, whereas chemical regulation is based on a risk assessment of individual compounds. Here, we developed a mixture-centered risk assessment strategy that integrates epidemiological and experimental evidence. We identified that exposure to an EDC mixture in early pregnancy is associated with language delay in offspring. At human-relevant concentrations, this mixture disrupted hormone-regulated and disease-relevant regulatory networks in human brain organoids and in the model organisms Xenopus leavis and Danio rerio, as well as behavioral responses. Reinterrogating epidemiological data, we found that up to 54% of the children had prenatal exposures above experimentally derived levels of concern, reaching, for the upper decile compared with the lowest decile of exposure, a 3.3 times higher risk of language delay.
Collapse
Affiliation(s)
- Nicolò Caporale
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.,Human Technopole, V.le Rita Levi-Montalcini, 1, 20157 Milan, Italy
| | - Michelle Leemans
- UMR 7221, Phyma, CNRS-Muséum National d'Histoire Naturelle, Sorbonne Université, 75005 Paris, France
| | - Lina Birgersson
- Department of Biological and Environmental Sciences, University of Gothenburg, 41463 Gothenburg, Sweden
| | - Pierre-Luc Germain
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Cristina Cheroni
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.,Human Technopole, V.le Rita Levi-Montalcini, 1, 20157 Milan, Italy
| | - Gábor Borbély
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden
| | - Elin Engdahl
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Christian Lindh
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, SE-221 85 Lund, Sweden
| | - Raul Bardini Bressan
- Medical Research Council Centre for Regenerative Medicine and Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh, UK
| | - Francesca Cavallo
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Nadav Even Chorev
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Giuseppe Alessandro D'Agostino
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Steven M Pollard
- Medical Research Council Centre for Regenerative Medicine and Edinburgh Cancer Research UK Centre, University of Edinburgh, Edinburgh, UK
| | - Marco Tullio Rigoli
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Erika Tenderini
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Alejandro Lopez Tobon
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Sebastiano Trattaro
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy
| | - Flavia Troglio
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Matteo Zanella
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Åke Bergman
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Department of Environmental Science, Stockholm University, SE-10691 Stockholm, Sweden.,School of Science and Technology, Örebro University, SE-70182 Örebro, Sweden
| | - Pauliina Damdimopoulou
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Maria Jönsson
- Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Wieland Kiess
- Hospital for Children and Adolescents, Department of Women and Child Health, University Hospital, University of Leipzig, 04103 Leipzig, Germany
| | - Efthymia Kitraki
- Lab of Basic Sciences, Faculty of Dentistry, National and Kapodistrian University of Athens, 152 72 Athens, Greece
| | - Hannu Kiviranta
- Department of Health Security, Finnish Institute for Health and Welfare (THL), Kuopio 70210, Finland
| | - Eewa Nånberg
- School of Health Sciences, Örebro University, SE-70182 Örebro, Sweden
| | - Mattias Öberg
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Institute of Environmental Medicine, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Panu Rantakokko
- Department of Health Security, Finnish Institute for Health and Welfare (THL), Kuopio 70210, Finland
| | - Christina Rudén
- Department of Environmental Science, Stockholm University, SE-10691 Stockholm, Sweden
| | - Olle Söder
- Department of Women's and Children's Health, Pediatric Endocrinology Division, Karolinska Institutet and University Hospital, SE-17176 Stockholm, Sweden
| | - Carl-Gustaf Bornehag
- Faculty of Health, Science and Technology, Department of Health Sciences, Karlstad University, SE- 651 88 Karlstad, Sweden.,Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Barbara Demeneix
- UMR 7221, Phyma, CNRS-Muséum National d'Histoire Naturelle, Sorbonne Université, 75005 Paris, France
| | - Jean-Baptiste Fini
- UMR 7221, Phyma, CNRS-Muséum National d'Histoire Naturelle, Sorbonne Université, 75005 Paris, France
| | - Chris Gennings
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joëlle Rüegg
- Swedish Toxicology Sciences Research Center (SWETOX), Södertälje, Sweden.,Department of Organismal Biology, Environmental Toxicology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Joachim Sturve
- Department of Biological and Environmental Sciences, University of Gothenburg, 41463 Gothenburg, Sweden
| | - Giuseppe Testa
- High Definition Disease Modelling Lab, Stem Cell and Organoid Epigenetics, IEO, European Institute of Oncology, IRCCS, 20141 Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, 20122 Milan, Italy.,Human Technopole, V.le Rita Levi-Montalcini, 1, 20157 Milan, Italy
| |
Collapse
|
13
|
Prokopenko D, Morgan SL, Mullin K, Hofmann O, Chapman B, Kirchner R, Alzheimer's Disease Neuroimaging Initiative (ADNI), Amberkar S, Wohlers I, Lange C, Hide W, Bertram L, Tanzi RE. Whole-genome sequencing reveals new Alzheimer's disease-associated rare variants in loci related to synaptic function and neuronal development. Alzheimers Dement 2021; 17:1509-1527. [PMID: 33797837 PMCID: PMC8519060 DOI: 10.1002/alz.12319] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Genome-wide association studies have led to numerous genetic loci associated with Alzheimer's disease (AD). Whole-genome sequencing (WGS) now permits genome-wide analyses to identify rare variants contributing to AD risk. METHODS We performed single-variant and spatial clustering-based testing on rare variants (minor allele frequency [MAF] ≤1%) in a family-based WGS-based association study of 2247 subjects from 605 multiplex AD families, followed by replication in 1669 unrelated individuals. RESULTS We identified 13 new AD candidate loci that yielded consistent rare-variant signals in discovery and replication cohorts (4 from single-variant, 9 from spatial-clustering), implicating these genes: FNBP1L, SEL1L, LINC00298, PRKCH, C15ORF41, C2CD3, KIF2A, APC, LHX9, NALCN, CTNNA2, SYTL3, and CLSTN2. DISCUSSION Downstream analyses of these novel loci highlight synaptic function, in contrast to common AD-associated variants, which implicate innate immunity and amyloid processing. These loci have not been associated previously with AD, emphasizing the ability of WGS to identify AD-associated rare variants, particularly outside of the exome.
Collapse
Affiliation(s)
- Dmitry Prokopenko
- Genetics and Aging Research Unit and The Henry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Sarah L. Morgan
- Department of NeuroscienceSheffield Institute for Translational NeurosciencesUniversity of SheffieldSheffieldUK
- Department of PathologyBeth Israel Deaconess Medical Center330 Brookline AvenueBostonMassachusettsUSA
| | - Kristina Mullin
- Genetics and Aging Research Unit and The Henry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Oliver Hofmann
- Department of Clinical PathologyUniversity of MelbourneMelbourneVICAustralia
| | - Brad Chapman
- Bioinformatics Core, Harvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Rory Kirchner
- Bioinformatics Core, Harvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | | | - Sandeep Amberkar
- Department of NeuroscienceSheffield Institute for Translational NeurosciencesUniversity of SheffieldSheffieldUK
| | - Inken Wohlers
- Lübeck Interdisciplinary Platform for Genome AnalyticsInstitutes of Neurogenetics and CardiogeneticsUniversity of LübeckLübeckGermany
| | - Christoph Lange
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Winston Hide
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeuroscienceSheffield Institute for Translational NeurosciencesUniversity of SheffieldSheffieldUK
- Department of PathologyBeth Israel Deaconess Medical Center330 Brookline AvenueBostonMassachusettsUSA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome AnalyticsInstitutes of Neurogenetics and CardiogeneticsUniversity of LübeckLübeckGermany
- Department of PsychologyUniversity of OsloOsloNorway
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit and The Henry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
14
|
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been shown to modulate neuronal signaling in homeostasis and diseases. In homeostasis, IL-1 regulates sleep and memory formation, whereas in diseases, IL-1 impairs memory and alters affect. Interestingly, IL-1 can cause long-lasting changes in behavior, suggesting IL-1 can alter neuroplasticity. The neuroplastic effects of IL-1 are mediated via its cognate receptor, Interleukin-1 Type 1 Receptor (IL-1R1), and are dependent on the distribution and cell type(s) of IL-1R1 expression. Recent reports found that IL-1R1 expression is restricted to discrete subpopulations of neurons, astrocytes, and endothelial cells and suggest IL-1 can influence neural circuits directly through neuronal IL-1R1 or indirectly via non-neuronal IL-1R1. In this review, we analyzed multiple mechanisms by which IL-1/IL-1R1 signaling might impact neuroplasticity based upon the most up-to-date literature and provided potential explanations to clarify discrepant and confusing findings reported in the past.
Collapse
Affiliation(s)
- Daniel P. Nemeth
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
15
|
Klenova AV, Volodin IA, Volodina EV, Ranneva SV, Amstislavskaya TG, Lipina TV. Vocal and physical phenotypes of calsyntenin2 knockout mouse pups model early-life symptoms of the autism spectrum disorder. Behav Brain Res 2021; 412:113430. [PMID: 34182007 DOI: 10.1016/j.bbr.2021.113430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022]
Abstract
This study discovered a novel acoustic phenotype in Calsyntenin2 deficient knockout (Clstn2-KO) pups in the neurodevelopment period of 5-9 postnatal days (PND 5-9). The narrowband ultrasonic calls (nUSVs) were less complex (mostly one-note, shorter in duration and higher in peak frequency) in Clsnt2-KO than in wild-type (WT) C57BL/6 J pups. The wideband ultrasonic calls (wUSVs) were produced substantially more often by Clstn2-KO than WT pups. The clicks were longer in duration and higher in peak frequency and power quartiles in Clstn2-KO pups. The elevated discomfort due to additional two-minute maternal separation coupled with experimenter's touch, resulted in significantly higher call rates of both nUSVs and clicks in pups of both genotypes and sexes compared to the previous two-minute maternal separation, whereas the call rate of wUSVs was not affected. In Clstn2-KO pups, the prevalence of emission of wUSVs retained at both sex and both degrees of discomfort, thus providing a reliable quantitative acoustic indicator for this genetic line. Besides the acoustic differences, we also detected the increased head-to-body ratio in Clstn2-KO pups. Altogether, this study demonstrated that lack of such synaptic adhesion protein as calsyntenin2 affects neurodevelopment of vocalization in a mouse as a model organism.
Collapse
Affiliation(s)
- Anna V Klenova
- Department of Vertebrate Zoology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
| | - Ilya A Volodin
- Department of Vertebrate Zoology, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia; Department of Behaviour and Behavioural Ecology of Mammals, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, 119071, Russia.
| | - Elena V Volodina
- Department of Behaviour and Behavioural Ecology of Mammals, A.N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, 119071, Russia.
| | - Svetlana V Ranneva
- Institute of Cytology and Genetics, Department of Genetics, Novosibirsk, Russia.
| | - Tamara G Amstislavskaya
- Federal State Budgetary Scientific Institution «Scientific Research Institute of Neurosciences and Medicine» (SRINM), Novosibirsk, 630117, Russia.
| | - Tatiana V Lipina
- Dementia Research Institute at University College London, London, WC1N 3BG, UK.
| |
Collapse
|
16
|
Li N, Cao S, Yu Z, Qiao M, Cheng Y, Shen Y, Song L, Huang X, Yang G, Zhao Y. Perinatal Lead Exposure Alters Calsyntenin-2 and Calsyntenin-3 Expression in the Hippocampus and Causes Learning Deficits in Mice Post-weaning. Biol Trace Elem Res 2021; 199:1414-1424. [PMID: 32557100 DOI: 10.1007/s12011-020-02241-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 06/08/2020] [Indexed: 12/29/2022]
Abstract
Calsyntenin-2 (Clstn2) and calsyntenin-3 (Clstn3) are the members of the cadherin superfamily and function to regulate the postsynaptic activity. Both proteins are known to play an important role in memory and learning. This study was designed to test the hypothesis that exposure of mothers to Pb in drinking water may alter the expression of Clstn2 and Clstn3 in offspring, which contributes to the Pb-induced learning deficiency. Pregnant mice were exposed to Pb in drinking water as Pb acetate from gestation to weaning. At the postnatal day 21, the learning and memory ability of pups was tested by Morris water maze, and the blood and brain tissues from pups were collected for metal and protein analyses. Data showed that perinatal Pb exposure resulted in a dose-dependent increase of Pb concentrations in blood (6-20-fold), hippocampus (2-7-fold), and cerebral cortex (2-8-fold) in offspring, as compared to controls (p < 0.05).The ability of learning and memory was decreased in lead exposure group, as compared to controls (p < 0.05). Both immunofluorescence and Western blot analyses revealed a striking difference in the expression of Clstn2 vs. Clstn3 following perinatal Pb exposure. In pregnant mice exposed to 0.1%, 0.2%, and 0.5% Pb, the expression of Clstn2 in offspring showed a Pb dose-related decrease by 39.2%, 76.5%, and 96.1% in hippocampus and by12.5%, 59.4%, and 78.1% in cerebral cortex, respectively (p < 0.05). In contrast, Clstn3 expression in these offspring brain regions was significantly increased (p < 0.05), after perinatal Pb exposure. The nature of Pb differential effect on Clstn2 and Clstn3 remains unknown. These observations suggest that Clstn2 and Clstn3 may have different roles in synaptic development and differentiation. Pb-induced learning defects may partly relate to the altered expression of calsyntenin proteins.
Collapse
Affiliation(s)
- Ning Li
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China.
| | - Shuai Cao
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| | - Zengli Yu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Mingwu Qiao
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| | - Yongxia Cheng
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| | - Yue Shen
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| | - Lianjun Song
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| | - Xianqing Huang
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| | - Guojun Yang
- Department of Preventive Medicine, Henan Medical College, Henan, 451191, China
| | - Yali Zhao
- College of Food Science and Technology, Henan Agriculture University, AgricultureRoad 63, Zhengzhou, 450002, Henan, China
| |
Collapse
|
17
|
Glutamatergic Dysfunction and Synaptic Ultrastructural Alterations in Schizophrenia and Autism Spectrum Disorder: Evidence from Human and Rodent Studies. Int J Mol Sci 2020; 22:ijms22010059. [PMID: 33374598 PMCID: PMC7793137 DOI: 10.3390/ijms22010059] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The correlation between dysfunction in the glutamatergic system and neuropsychiatric disorders, including schizophrenia and autism spectrum disorder, is undisputed. Both disorders are associated with molecular and ultrastructural alterations that affect synaptic plasticity and thus the molecular and physiological basis of learning and memory. Altered synaptic plasticity, accompanied by changes in protein synthesis and trafficking of postsynaptic proteins, as well as structural modifications of excitatory synapses, are critically involved in the postnatal development of the mammalian nervous system. In this review, we summarize glutamatergic alterations and ultrastructural changes in synapses in schizophrenia and autism spectrum disorder of genetic or drug-related origin, and briefly comment on the possible reversibility of these neuropsychiatric disorders in the light of findings in regular synaptic physiology.
Collapse
|