1
|
Xavier AM, Lin Q, Kang CJ, Cheadle L. A single-cell transcriptomic atlas of sensory-dependent gene expression in developing mouse visual cortex. Development 2025; 152:dev204244. [PMID: 40018816 DOI: 10.1242/dev.204244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Sensory experience drives the maturation of neural circuits during postnatal brain development through molecular mechanisms that remain to be fully elucidated. One likely mechanism involves the sensory-dependent expression of genes that encode direct mediators of circuit remodeling within developing cells. To identify potential drivers of sensory-dependent synaptic development, we generated a single-nucleus RNA sequencing dataset describing the transcriptional responses of cells in the mouse visual cortex to sensory deprivation or to stimulation during a developmental window when visual input is necessary for circuit refinement. We sequenced 118,529 nuclei across 16 neuronal and non-neuronal cell types isolated from control, sensory deprived and sensory stimulated mice, identifying 1268 sensory-induced genes within the developing brain. While experience elicited transcriptomic changes in all cell types, excitatory neurons in layer 2/3 exhibited the most robust changes, and the sensory-induced genes in these cells are poised to strengthen synapse-to-nucleus crosstalk and to promote cell type-specific axon guidance pathways. Altogether, we expect this dataset to significantly broaden our understanding of the molecular mechanisms through which sensory experience shapes neural circuit wiring in the developing brain.
Collapse
Affiliation(s)
- Andre M Xavier
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Qianyu Lin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Chris J Kang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724, USA
| |
Collapse
|
2
|
Zhou C, Gao X, Tan L. Shaping sight: Novel thalamic plasticity channels dLGN feature preference during visual critical period. Neuron 2025; 113:1294-1296. [PMID: 40339563 DOI: 10.1016/j.neuron.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 05/10/2025]
Abstract
Sonoda et al.1 showed that dLGN neurons exhibit long-lasting shifts of tuning preference toward selective features experienced during the classical critical period. They demonstrated that this plasticity results from feedforward-input refinement, revealing a different form of experience-dependent plasticity compared to V1.
Collapse
Affiliation(s)
- Chuying Zhou
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiang Gao
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liming Tan
- Shenzhen Key Laboratory of Neuropsychiatric Modulation, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, the Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
3
|
Pumo G, Rijli FM. Chromatin regulation of neuronal activity-dependent gene programs in circuit formation and plasticity. Curr Opin Neurobiol 2025; 92:103024. [PMID: 40262385 DOI: 10.1016/j.conb.2025.103024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025]
Abstract
Neuronal activity-dependent transcription is crucial for the development and plasticity of neuronal circuits. At the chromatin level, the induction of neuronal activity-regulated genes is orchestrated through various mechanisms, including the deposition of histone modifications at regulatory elements, the binding of transcriptional activators and repressors, chromatin remodeling, and the control of 3D genome architecture. Here, we review our current understanding of how chromatin mechanisms regulate temporally distinct transcriptional waves following neuronal stimulation and allow neurons to mount cell type-specific and stimulus-specific transcriptional responses. We also highlight a specific epigenetic mechanism in developing neurons that maintains immediate early genes (IEGs) in an inactive though poised state, while simultaneously preparing them for rapid activation in response to sensory stimulation. We discuss how chromatin regulation mechanisms play a crucial role in controlling activity-regulated gene expression, enabling the implementation of precise gene expression programs during different stages of neural circuit development and plasticity.
Collapse
Affiliation(s)
- Gabriele Pumo
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, 4058 Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical Research, Fabrikstrasse 24, 4058 Basel, Switzerland; University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Wang P, Hu J, Chen C, Jiang Z, Zhang Y, Lin K, Liao L, Wang X. The immune regulatory mechanism of ketamine-induced psychiatric disorders: A new perspective on drug-induced psychiatric symptoms. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111194. [PMID: 39542202 DOI: 10.1016/j.pnpbp.2024.111194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024]
Abstract
Ketamine, a psychoactive substance strictly regulated by international drug conventions, is classified as a "new type drug" due to its excitatory, hallucinogenic, or inhibitory effects. The etiology of ketamine-induced psychiatric symptoms is multifaceted, with the immune regulatory mechanism being the most prominent among several explanatory theories. In recent years, the interaction between the immune system and nervous system have garnered significant attention in neuropsychiatric disorder research. Notably, the infiltration of peripheral lymphocytes into the central nervous system has emerged as an early hallmark of certain neuropsychiatric disorders. However, a notable gap exists in the current literature, regarding the immune regulatory mechanisms, specifically the peripheral immune alterations, associated with ketamine-induced psychiatric symptoms. To address this void, this article endeavors to provide a comprehensive overview of the pathophysiological processes implicated in psychiatric disorders or symptoms, encompassing those elicited by ketamine. This analysis delves into aspects such as nerve damage, alterations within the central immune system, and the regulation of the peripheral immune system. By emphasizing the intricate crosstalk between the peripheral immune system and the central nervous system, this study sheds light on their collaborative role in the onset and progression of psychiatric diseases or symptoms. This insight offers fresh perspectives on the underlying mechanisms, diagnosis and therapeutic strategies for mental disorders stemming from drug abuse.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Junmei Hu
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Congliang Chen
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Zihan Jiang
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Yu Zhang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Kexin Lin
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Linchuan Liao
- Department of Forensic Toxicological Analysis, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
5
|
Nagappan-Chettiar S, Burbridge TJ, Umemori H. Activity-Dependent Synapse Refinement: From Mechanisms to Molecules. Neuroscientist 2024; 30:673-689. [PMID: 37140155 PMCID: PMC11584027 DOI: 10.1177/10738584231170167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
The refinement of immature neuronal networks into efficient mature ones is critical to nervous system development and function. This process of synapse refinement is driven by the neuronal activity-dependent competition of converging synaptic inputs, resulting in the elimination of weak inputs and the stabilization of strong ones. Neuronal activity, whether in the form of spontaneous activity or experience-evoked activity, is known to drive synapse refinement in numerous brain regions. More recent studies are now revealing the manner and mechanisms by which neuronal activity is detected and converted into molecular signals that appropriately regulate the elimination of weaker synapses and stabilization of stronger ones. Here, we highlight how spontaneous activity and evoked activity instruct neuronal activity-dependent competition during synapse refinement. We then focus on how neuronal activity is transformed into the molecular cues that determine and execute synapse refinement. A comprehensive understanding of the mechanisms underlying synapse refinement can lead to novel therapeutic strategies in neuropsychiatric diseases characterized by aberrant synaptic function.
Collapse
Affiliation(s)
- Sivapratha Nagappan-Chettiar
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J. Burbridge
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
7
|
de Deus JL, Faborode OS, Nandi S. Synaptic Pruning by Microglia: Lessons from Genetic Studies in Mice. Dev Neurosci 2024:1-21. [PMID: 39265565 DOI: 10.1159/000541379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Neural circuits are subjected to refinement throughout life. The dynamic addition and elimination (pruning) of synapses are necessary for maturation of neural circuits and synaptic plasticity. Due to their phagocytic nature, microglia have been considered as the primary mediators of synaptic pruning. Synaptic pruning can strengthen an active synapse by removing excess weaker synapses during development. Inappropriate synaptic pruning can often influence a disease outcome or an injury response. SUMMARY This review offers a focused discussion on microglial roles in synaptic pruning, based on the evidence gathered from genetic manipulations in mice. Genetically labeled microglia and synapses often allow assessment of their interactions in real time. Further manipulations involving synaptically localized molecules, neuronally or glial-derived diffusible factors, and their respective cognate receptors in microglia provide critical evidence in support of a direct role of microglia in synaptic pruning. KEY MESSAGE We discuss microglial contact-dependent "eat-me," "don't-eat-me," and "find-me" signals, as well as recently identified noncontact pruning, under the contexts of neural circuit, brain region, developmental window, and an injury or a disease state.
Collapse
Affiliation(s)
- Junia Lara de Deus
- Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| | | | - Sayan Nandi
- Department of Anatomy, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
8
|
Stebbins K, Somaiya RD, Sabbagh U, Khaksar P, Liang Y, Su J, Fox MA. Retinal Input Is Required for the Maintenance of Neuronal Laminae in the Ventrolateral Geniculate Nucleus. eNeuro 2024; 11:ENEURO.0022-24.2024. [PMID: 39160068 PMCID: PMC11373735 DOI: 10.1523/eneuro.0022-24.2024] [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: 01/17/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
Retinal ganglion cell (RGC) axons provide direct input into several brain regions, including the dorsal lateral geniculate nucleus (dLGN), which is important for image-forming vision, and the ventrolateral geniculate nucleus (vLGN), which is associated with nonimage-forming vision. Through both activity- and morphogen-dependent mechanisms, retinal inputs play important roles in the development of dLGN, including the refinement of retinal projections, morphological development of thalamocortical relay cells (TRCs), timing of corticogeniculate innervation, and recruitment and distribution of inhibitory interneurons. In contrast, little is known about the role of retinal inputs in the development of vLGN. Grossly, vLGN is divided into two domains, the retinorecipient external vLGN (vLGNe) and nonretinorecipient internal vLGN (vLGNi). Studies previously found that vLGNe consists of transcriptionally distinct GABAergic subtypes distributed into at least four adjacent laminae. At present, it remains unclear whether retinal inputs influence the development of these cell-type-specific neuronal laminae in vLGNe. Here, we elucidated the developmental timeline for these laminae in the mouse vLGNe, and results indicate that these laminae are specified at or before birth. We observed that mutant mice without retinal inputs have a normal laminar distribution of GABAergic cells at birth; however, after the first week of postnatal development, these mutants exhibited a dramatic disruption in the laminar organization of inhibitory neurons and clear boundaries between vLGNe and vLGNi. Overall, our results show that while the formation of cell-type-specific layers in mouse vLGNe does not depend on RGC inputs, retinal signals are critical for their maintenance.
Collapse
Affiliation(s)
- Katelyn Stebbins
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia 24061
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia 24016
| | - Rachana Deven Somaiya
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia 24061
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
| | - Ubadah Sabbagh
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, Virginia 24061
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02138
| | - Parsa Khaksar
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia 24016
| | - Yanping Liang
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
| | - Jianmin Su
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia 24061
| | - Michael A Fox
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia 24016
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, California 94720
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, Virginia 24061
- Department of Biology, College of Natural Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
9
|
Akinlaja YO, Nishiyama A. Glial modulation of synapse development and plasticity: oligodendrocyte precursor cells as a new player in the synaptic quintet. Front Cell Dev Biol 2024; 12:1418100. [PMID: 39258226 PMCID: PMC11385347 DOI: 10.3389/fcell.2024.1418100] [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: 04/16/2024] [Accepted: 08/09/2024] [Indexed: 09/12/2024] Open
Abstract
Synaptic communication is an important process in the central nervous system that allows for the rapid and spatially specified transfer of signals. Neurons receive various synaptic inputs and generate action potentials required for information transfer, and these inputs can be excitatory or inhibitory, which collectively determines the output. Non-neuronal cells (glial cells) have been identified as crucial participants in influencing neuronal activity and synaptic transmission, with astrocytes forming tripartite synapses and microglia pruning synapses. While it has been known that oligodendrocyte precursor cells (OPCs) receive neuronal inputs, whether they also influence neuronal activity and synaptic transmission has remained unknown for two decades. Recent findings indicate that OPCs, too, modulate neuronal synapses. In this review, we discuss the roles of different glial cell types at synapses, including the recently discovered involvement of OPCs in synaptic transmission and synapse refinement, and discuss overlapping roles played by multiple glial cell types.
Collapse
Affiliation(s)
- Yetunde O Akinlaja
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Institute of Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
- Institute of Systems Genomics, University of Connecticut, Storrs, CT, United States
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, United States
- Institute of Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, United States
- Institute of Systems Genomics, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
10
|
Joy MT, Carmichael ST. Activity-dependent transcriptional programs in memory regulate motor recovery after stroke. Commun Biol 2024; 7:1048. [PMID: 39183218 PMCID: PMC11345429 DOI: 10.1038/s42003-024-06723-3] [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: 09/15/2023] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
Stroke causes death of brain tissue leading to long-term deficits. Behavioral evidence from neurorehabilitative therapies suggest learning-induced neuroplasticity can lead to beneficial outcomes. However, molecular and cellular mechanisms that link learning and stroke recovery are unknown. We show that in a mouse model of stroke, which exhibits enhanced recovery of function due to genetic perturbations of learning and memory genes, animals display activity-dependent transcriptional programs that are normally active during formation or storage of new memories. The expression of neuronal activity-dependent genes are predictive of recovery and occupy a molecular latent space unique to motor recovery. With motor recovery, networks of activity-dependent genes are co-expressed with their transcription factor targets forming gene regulatory networks that support activity-dependent transcription, that are normally diminished after stroke. Neuronal activity-dependent changes at the circuit level are influenced by interactions with microglia. At the molecular level, we show that enrichment of activity-dependent programs in neurons lead to transcriptional changes in microglia where they differentially interact to support intercellular signaling pathways for axon guidance, growth and synaptogenesis. Together, these studies identify activity-dependent transcriptional programs as a fundamental mechanism for neural repair post-stroke.
Collapse
Affiliation(s)
- Mary T Joy
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
11
|
Wiseman S. In conversation with Lucas Cheadle. Nat Neurosci 2024; 27:1428-1431. [PMID: 39107592 DOI: 10.1038/s41593-024-01719-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
|
12
|
Xavier AM, Lin Q, Kang CJ, Cheadle L. A single-cell transcriptomic atlas of sensory-dependent gene expression in developing mouse visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600673. [PMID: 38979325 PMCID: PMC11230371 DOI: 10.1101/2024.06.25.600673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Sensory experience drives the refinement and maturation of neural circuits during postnatal brain development through molecular mechanisms that remain to be fully elucidated. One likely mechanism involves the sensory-dependent expression of genes that encode direct mediators of circuit remodeling within developing cells. However, while studies in adult systems have begun to uncover crucial roles for sensory-induced genes in modifying circuit connectivity, the gene programs induced by brain cells in response to sensory experience during development remain to be fully characterized. Here, we present a single-nucleus RNA-sequencing dataset describing the transcriptional responses of cells in mouse visual cortex to sensory deprivation or sensory stimulation during a developmental window when visual input is necessary for circuit refinement. We sequenced 118,529 individual nuclei across sixteen neuronal and non-neuronal cortical cell types isolated from control, sensory deprived, and sensory stimulated mice, identifying 1,268 unique sensory-induced genes within the developing brain. To demonstrate the utility of this resource, we compared the architecture and ontology of sensory-induced gene programs between cell types, annotated transcriptional induction and repression events based upon RNA velocity, and discovered Neurexin and Neuregulin signaling networks that underlie cell-cell interactions via CellChat . We find that excitatory neurons, especially layer 2/3 pyramidal neurons, are highly sensitive to sensory stimulation, and that the sensory-induced genes in these cells are poised to strengthen synapse-to-nucleus crosstalk by heightening protein serine/threonine kinase activity. Altogether, we expect this dataset to significantly broaden our understanding of the molecular mechanisms through which sensory experience shapes neural circuit wiring in the developing brain.
Collapse
|
13
|
Li C, Jiang M, Fang Z, Chen Z, Li L, Liu Z, Wang J, Yin X, Wang J, Wu M. Current evidence of synaptic dysfunction after stroke: Cellular and molecular mechanisms. CNS Neurosci Ther 2024; 30:e14744. [PMID: 38727249 PMCID: PMC11084978 DOI: 10.1111/cns.14744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Stroke is an acute cerebrovascular disease in which brain tissue is damaged due to sudden obstruction of blood flow to the brain or the rupture of blood vessels in the brain, which can prompt ischemic or hemorrhagic stroke. After stroke onset, ischemia, hypoxia, infiltration of blood components into the brain parenchyma, and lysed cell fragments, among other factors, invariably increase blood-brain barrier (BBB) permeability, the inflammatory response, and brain edema. These changes lead to neuronal cell death and synaptic dysfunction, the latter of which poses a significant challenge to stroke treatment. RESULTS Synaptic dysfunction occurs in various ways after stroke and includes the following: damage to neuronal structures, accumulation of pathologic proteins in the cell body, decreased fluidity and release of synaptic vesicles, disruption of mitochondrial transport in synapses, activation of synaptic phagocytosis by microglia/macrophages and astrocytes, and a reduction in synapse formation. CONCLUSIONS This review summarizes the cellular and molecular mechanisms related to synapses and the protective effects of drugs or compounds and rehabilitation therapy on synapses in stroke according to recent research. Such an exploration will help to elucidate the relationship between stroke and synaptic damage and provide new insights into protecting synapses and restoring neurologic function.
Collapse
Affiliation(s)
- Chuan Li
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Min Jiang
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| | - Zhi‐Ting Fang
- Department of Pathophysiology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Zhiying Chen
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Li Li
- Department of Intensive Care UnitThe Affiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Ziying Liu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Xiaoping Yin
- Department of NeurologyAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Moxin Wu
- Department of Medical LaboratoryAffiliated Hospital of Jiujiang UniversityJiujiangJiangxiChina
- Jiujiang Clinical Precision Medicine Research CenterJiujiangJiangxiChina
| |
Collapse
|
14
|
Burbridge TJ, Ratliff JM, Dwivedi D, Vrudhula U, Alvarado-Huerta F, Sjulson L, Ibrahim LA, Cheadle L, Fishell G, Batista-Brito R. Disruption of Cholinergic Retinal Waves Alters Visual Cortex Development and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588143. [PMID: 38644996 PMCID: PMC11030223 DOI: 10.1101/2024.04.05.588143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Retinal waves represent an early form of patterned spontaneous neural activity in the visual system. These waves originate in the retina before eye-opening and propagate throughout the visual system, influencing the assembly and maturation of subcortical visual brain regions. However, because it is technically challenging to ablate retina-derived cortical waves without inducing compensatory activity, the role these waves play in the development of the visual cortex remains unclear. To address this question, we used targeted conditional genetics to disrupt cholinergic retinal waves and their propagation to select regions of primary visual cortex, which largely prevented compensatory patterned activity. We find that loss of cholinergic retinal waves without compensation impaired the molecular and synaptic maturation of excitatory neurons located in the input layers of visual cortex, as well as layer 1 interneurons. These perinatal molecular and synaptic deficits also relate to functional changes observed at later ages. We find that the loss of perinatal cholinergic retinal waves causes abnormal visual cortex retinotopy, mirroring changes in the retinotopic organization of gene expression, and additionally impairs the processing of visual information. We further show that retinal waves are necessary for higher order processing of sensory information by impacting the state-dependent activity of layer 1 interneurons, a neuronal type that shapes neocortical state-modulation, as well as for state-dependent gain modulation of visual responses of excitatory neurons. Together, these results demonstrate that a brief targeted perinatal disruption of patterned spontaneous activity alters early cortical gene expression as well as synaptic and physiological development, and compromises both fundamental and, notably, higher-order functions of visual cortex after eye-opening.
Collapse
Affiliation(s)
- Timothy J Burbridge
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115
| | - Jacob M Ratliff
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Deepanjali Dwivedi
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115
| | - Uma Vrudhula
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | | | - Lucas Sjulson
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Psychiatry and Behavioral Sciences, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Leena Ali Ibrahim
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955–6900, KSA
| | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724
| | - Gordon Fishell
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115
| | - Renata Batista-Brito
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Psychiatry and Behavioral Sciences, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Genetics, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| |
Collapse
|
15
|
Ferro A, Arshad A, Boyd L, Stanley T, Berisha A, Vrudhula U, Gomez AM, Borniger JC, Cheadle L. The cytokine receptor Fn14 is a molecular brake on neuronal activity that mediates circadian function in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587786. [PMID: 38617238 PMCID: PMC11014623 DOI: 10.1101/2024.04.02.587786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
To survive, organisms must adapt to a staggering diversity of environmental signals, ranging from sensory information to pathogenic infection, across the lifespan. At the same time, organisms intrinsically generate biological oscillations, such as circadian rhythms, without input from the environment. While the nervous system is well-suited to integrate extrinsic and intrinsic cues, how the brain balances these influences to shape biological function system-wide is not well understood at the molecular level. Here, we demonstrate that the cytokine receptor Fn14, previously identified as a mediator of sensory experience-dependent synaptic refinement during brain development, regulates neuronal activity and function in adult mice in a time-of-day-dependent manner. We show that a subset of excitatory pyramidal (PYR) neurons in the CA1 subregion of the hippocampus increase Fn14 expression when neuronal activity is heightened. Once expressed, Fn14 constrains the activity of these same PYR neurons, suggesting that Fn14 operates as a molecular brake on neuronal activity. Strikingly, differences in PYR neuron activity between mice lacking or expressing Fn14 were most robust at daily transitions between light and dark, and genetic ablation of Fn14 caused aberrations in circadian rhythms, sleep-wake states, and sensory-cued and spatial memory. At the cellular level, microglia contacted fewer, but larger, excitatory synapses in CA1 in the absence of Fn14, suggesting that these brain-resident immune cells may dampen neuronal activity by modifying synaptic inputs onto PYR neurons. Finally, mice lacking Fn14 exhibited heightened susceptibility to chemically induced seizures, implicating Fn14 in disorders characterized by hyperexcitation, such as epilepsy. Altogether, these findings reveal that cytokine receptors that mediates inflammation in the periphery, such as Fn14, can also play major roles in healthy neurological function in the adult brain downstream of both extrinsic and intrinsic cues.
Collapse
Affiliation(s)
- Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Anosha Arshad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
- Department of Neurobiology and Behavior, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA
| | - Leah Boyd
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Tess Stanley
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Adrian Berisha
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Uma Vrudhula
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | - Adrian M. Gomez
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
| | | | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11740, USA
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11740, USA
| |
Collapse
|
16
|
Abstract
It has been more than a century since Pío del Río-Hortega first characterized microglia in histological stains of brain tissue. Since then, significant advances have been made in understanding the role of these resident central nervous system (CNS) macrophages. In particular, it is now known that microglia can sense neural activity and modulate neuronal circuits accordingly. We review the mechanisms by which microglia detect changes in neural activity to then modulate synapse numbers in the developing and mature CNS. This includes responses to both spontaneous and experience-driven neural activity. We further discuss activity-dependent mechanisms by which microglia regulate synaptic function and neural circuit excitability. Together, our discussion provides a comprehensive review of the activity-dependent functions of microglia within neural circuits in the healthy CNS, and highlights exciting new open questions related to understanding more fully microglia as key components and regulators of neural circuits.
Collapse
Affiliation(s)
- Violeta Durán Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
17
|
Okruszko MA, Szabłowski M, Zarzecki M, Michnowska-Kobylińska M, Lisowski Ł, Łapińska M, Stachurska Z, Szpakowicz A, Kamiński KA, Konopińska J. Inflammation and Neurodegeneration in Glaucoma: Isolated Eye Disease or a Part of a Systemic Disorder? - Serum Proteomic Analysis. J Inflamm Res 2024; 17:1021-1037. [PMID: 38370463 PMCID: PMC10874189 DOI: 10.2147/jir.s434989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
Introduction Glaucoma is the most common optic neuropathy and the leading cause of irreversible blindness worldwide, which affects 3.54% of the population aged 40-80 years. Despite numerous published studies, some aspects of glaucoma pathogenesis, serum biomarkers, and their potential link with other diseases remain unclear. Recent articles have proposed that autoimmune, oxidative stress and inflammation may be involved in the pathogenesis of glaucoma. Methods We investigated the serum expression of 92 inflammatory and neurotrophic factors in glaucoma patients. The study group consisted of 26 glaucoma patients and 192 healthy subjects based on digital fundography. Results Patients with glaucoma had significantly lower serum expression of IL-2Rβ, TWEAK, CX3CL1, CD6, CD5, LAP TGF-beta1, LIF-R, TRAIL, NT-3, and CCL23 and significantly higher expression of IL-22Rα1. Conclusion Our results indicate that patients with glaucoma tend to have lower levels of neuroprotective proteins and higher levels of neuroinflammatory proteins, similar to those observed in psychiatric, neurodegenerative and autoimmune diseases, indicating a potential link between these conditions and glaucoma pathogenesis.
Collapse
Affiliation(s)
| | - Maciej Szabłowski
- Department of Ophthalmology, Medical University of Bialystok, Białystok, 15-089, Poland
| | - Mateusz Zarzecki
- Department of Ophthalmology, Medical University of Bialystok, Białystok, 15-089, Poland
| | | | - Łukasz Lisowski
- Department of Ophthalmology, Medical University of Bialystok, Białystok, 15-089, Poland
| | - Magda Łapińska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Zofia Stachurska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Anna Szpakowicz
- Department of Cardiology, Medical University of Bialystok, Białystok, Poland
| | - Karol Adam Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, Białystok, Poland
| | - Joanna Konopińska
- Department of Ophthalmology, Medical University of Bialystok, Białystok, 15-089, Poland
| |
Collapse
|
18
|
Stebbins K, Somaiya RD, Sabbagh U, Liang Y, Su J, Fox MA. Retinal input is required for the maintenance of neuronal laminae in the ventral lateral geniculate nucleus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575402. [PMID: 38293194 PMCID: PMC10827117 DOI: 10.1101/2024.01.12.575402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Retinal ganglion cell (RGC) axons provide direct input into several nuclei of the mouse visual thalamus, including the dorsal lateral geniculate nucleus (dLGN), which is important for classical image-forming vision, and the ventral lateral geniculate nucleus (vLGN), which is associated with non-image-forming vision. Through both activity- and morphogen-dependent mechanisms, retinal inputs play important roles in the development of dLGN, including the refinement of retinal projections, morphological development of thalamocortical relay cells (TRCs), the timing of corticogeniculate innervation, and the recruitment of inhibitory interneurons from progenitor zones. In contrast, little is known about the role of retinal inputs in the development of vLGN. Grossly, vLGN is divided into two domains, the retinorecipient external vLGN (vLGNe) and the non-retinorecipient internal vLGN (vLGNi). We previously found that vLGNe consists of transcriptionally distinct GABAergic subtypes that are distributed into at least four adjacent laminae. At present, it remains unclear whether retinal inputs influence the development of these cell-specific neuronal laminae in vLGNe. Here, we elucidated the developmental timeline for the formation and maintenance of these laminae in the mouse vLGNe and results indicate that these laminae are specified at or before birth, well before eye-opening and the emergence of experience-dependent visual activity. We observed that mutant mice without retinal inputs have a normal laminar distribution of GABAergic cells at birth; however, after the first week of postnatal development, these mutants exhibited a dramatic disruption in the laminar organization of inhibitory neurons and clear boundaries between vLGNe and vLGNi. Overall, our results show that while the formation of cell type-specific layers in vLGNe does not depend on RGC inputs, retinal signals are critical for their maintenance.
Collapse
|
19
|
Tzeng CP, Whitwam T, Boxer LD, Li E, Silberfeld A, Trowbridge S, Mei K, Lin C, Shamah R, Griffith EC, Renthal W, Chen C, Greenberg ME. Activity-induced MeCP2 phosphorylation regulates retinogeniculate synapse refinement. Proc Natl Acad Sci U S A 2023; 120:e2310344120. [PMID: 37871205 PMCID: PMC10623012 DOI: 10.1073/pnas.2310344120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/25/2023] [Indexed: 10/25/2023] Open
Abstract
Mutations in MECP2 give rise to Rett syndrome (RTT), an X-linked neurodevelopmental disorder that results in broad cognitive impairments in females. While the exact etiology of RTT symptoms remains unknown, one possible explanation for its clinical presentation is that loss of MECP2 causes miswiring of neural circuits due to defects in the brain's capacity to respond to changes in neuronal activity and sensory experience. Here, we show that MeCP2 is phosphorylated at four residues in the mouse brain (S86, S274, T308, and S421) in response to neuronal activity, and we generate a quadruple knock-in (QKI) mouse line in which all four activity-dependent sites are mutated to alanines to prevent phosphorylation. QKI mice do not display overt RTT phenotypes or detectable gene expression changes in two brain regions. However, electrophysiological recordings from the retinogeniculate synapse of QKI mice reveal that while synapse elimination is initially normal at P14, it is significantly compromised at P20. Notably, this phenotype is distinct from the synapse refinement defect previously reported for Mecp2 null mice, where synapses initially refine but then regress after the third postnatal week. We thus propose a model in which activity-induced phosphorylation of MeCP2 is critical for the proper timing of retinogeniculate synapse maturation specifically during the early postnatal period.
Collapse
Affiliation(s)
| | - Tess Whitwam
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
- Program in Neuroscience, Harvard Medical School, Boston, MA02115
| | - Lisa D. Boxer
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Emmy Li
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | | | - Sara Trowbridge
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Kevin Mei
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Cindy Lin
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Rebecca Shamah
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Eric C. Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - William Renthal
- Department of Neurobiology, Harvard Medical School, Boston, MA02115
| | - Chinfei Chen
- Department of Neurology, F.M. Kirby Neurobiology Center, Children’s Hospital, Boston, MA02115
| | | |
Collapse
|
20
|
Fang LP, Bai X. Oligodendrocyte precursor cells: the multitaskers in the brain. Pflugers Arch 2023; 475:1035-1044. [PMID: 37401986 PMCID: PMC10409806 DOI: 10.1007/s00424-023-02837-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023]
Abstract
In the central nervous system, oligodendrocyte precursor cells (OPCs) are recognized as the progenitors responsible for the generation of oligodendrocytes, which play a critical role in myelination. Extensive research has shed light on the mechanisms underlying OPC proliferation and differentiation into mature myelin-forming oligodendrocytes. However, recent advances in the field have revealed that OPCs have multiple functions beyond their role as progenitors, exerting control over neural circuits and brain function through distinct pathways. This review aims to provide a comprehensive understanding of OPCs by first introducing their well-established features. Subsequently, we delve into the emerging roles of OPCs in modulating brain function in both healthy and diseased states. Unraveling the cellular and molecular mechanisms by which OPCs influence brain function holds great promise for identifying novel therapeutic targets for central nervous system diseases.
Collapse
Affiliation(s)
- Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| |
Collapse
|
21
|
Tzeng CP, Whitwam T, Boxer LD, Li E, Silberfeld A, Trowbridge S, Mei K, Lin C, Shamah R, Griffith EC, Renthal W, Chen C, Greenberg ME. Activity-Induced MeCP2 Phosphorylation Regulates Retinogeniculate Synapse Refinement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547549. [PMID: 37461668 PMCID: PMC10349931 DOI: 10.1101/2023.07.03.547549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Mutations in MECP2 give rise to Rett syndrome (RTT), an X-linked neurodevelopmental disorder that results in broad cognitive impairments in females. While the exact etiology of RTT symptoms remains unknown, one possible explanation for its clinical presentation is that loss of MeCP2 causes miswiring of neural circuits due to defects in the brain's capacity to respond to changes in neuronal activity and sensory experience. Here we show that MeCP2 is phosphorylated at four residues in the brain (S86, S274, T308, and S421) in response to neuronal activity, and we generate a quadruple knock-in (QKI) mouse line in which all four activity-dependent sites are mutated to alanines to prevent phosphorylation. QKI mice do not display overt RTT phenotypes or detectable gene expression changes in two brain regions. However, electrophysiological recordings from the retinogeniculate synapse of QKI mice reveal that while synapse elimination is initially normal at P14, it is significantly compromised at P20. Notably, this phenotype is distinct from that previously reported for Mecp2 null mice, where synapses initially refine but then regress after the third postnatal week. We thus propose a model in which activity-induced phosphorylation of MeCP2 is critical for the proper timing of retinogeniculate synapse maturation specifically during the early postnatal period. SIGNIFICANCE STATEMENT Rett syndrome (RTT) is an X-linked neurodevelopmental disorder that predominantly affects girls. RTT is caused by loss of function mutations in a single gene MeCP2. Girls with RTT develop normally during their first year of life, but then experience neurological abnormalities including breathing and movement difficulties, loss of speech, and seizures. This study investigates the function of the MeCP2 protein in the brain, and how MeCP2 activity is modulated by sensory experience in early life. Evidence is presented that sensory experience affects MeCP2 function, and that this is required for synaptic pruning in the brain. These findings provide insight into MeCP2 function, and clues as to what goes awry in the brain when the function of MeCP2 is disrupted.
Collapse
|
22
|
Whitelaw BS, Stoessel MB, Majewska AK. Movers and shakers: Microglial dynamics and modulation of neural networks. Glia 2023; 71:1575-1591. [PMID: 36533844 PMCID: PMC10729610 DOI: 10.1002/glia.24323] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Microglia are multifaceted cells that act as immune sentinels, with important roles in pathological events, but also as integral contributors to the normal development and function of neural circuits. In the last decade, our understanding of the contributions these cells make to synaptic health and dysfunction has expanded at a dizzying pace. Here we review the known mechanisms that govern the dynamics of microglia allowing these motile cells to interact with synapses, and recruit microglia to specific sites on neurons. We then review the molecular signals that may underlie the function of microglia in synaptic remodeling. The emerging picture from the literature suggests that microglia are highly sensitive cells, reacting to neuronal signals with dynamic and specific actions tuned to the need of specific synapses and networks.
Collapse
Affiliation(s)
- Brendan Steven Whitelaw
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Mark Blohm Stoessel
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Ania Katarzyna Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| |
Collapse
|
23
|
Zipp F, Bittner S, Schafer DP. Cytokines as emerging regulators of central nervous system synapses. Immunity 2023; 56:914-925. [PMID: 37163992 PMCID: PMC10233069 DOI: 10.1016/j.immuni.2023.04.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 05/12/2023]
Abstract
Cytokines are key messengers by which immune cells communicate, and they drive many physiological processes, including immune and inflammatory responses. Early discoveries demonstrated that cytokines, such as the interleukin family members and TNF-α, regulate synaptic scaling and plasticity. Still, we continue to learn more about how these traditional immune system cytokines affect neuronal structure and function. Different cytokines shape synaptic function on multiple levels ranging from fine-tuning neurotransmission, to regulating synapse number, to impacting global neuronal networks and complex behavior. These recent findings have cultivated an exciting and growing field centered on the importance of immune system cytokines for regulating synapse and neural network structure and function. Here, we highlight the latest findings related to cytokines in the central nervous system and their regulation of synapse structure and function. Moreover, we explore how these mechanisms are becoming increasingly important to consider in diseases-especially those with a large neuroinflammatory component.
Collapse
Affiliation(s)
- Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
24
|
Nagappan-Chettiar S, Yasuda M, Johnson-Venkatesh EM, Umemori H. The molecular signals that regulate activity-dependent synapse refinement in the brain. Curr Opin Neurobiol 2023; 79:102692. [PMID: 36805716 PMCID: PMC10023433 DOI: 10.1016/j.conb.2023.102692] [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: 09/07/2022] [Revised: 12/11/2022] [Accepted: 01/10/2023] [Indexed: 02/19/2023]
Abstract
The formation of appropriate synaptic connections is critical for the proper functioning of the brain. Early in development, neurons form a surplus of immature synapses. To establish efficient, functional neural networks, neurons selectively stabilize active synapses and eliminate less active ones. This process is known as activity-dependent synapse refinement. Defects in this process have been implicated in neuropsychiatric disorders such as schizophrenia and autism. Here we review the manner and mechanisms by which synapse elimination is regulated through activity-dependent competition. We propose a theoretical framework for the molecular mechanisms of synapse refinement, in which three types of signals regulate the refinement. We then describe the identity of these signals and discuss how multiple molecular signals interact to achieve appropriate synapse refinement in the brain.
Collapse
Affiliation(s)
- Sivapratha Nagappan-Chettiar
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. https://twitter.com/sivapratha
| | - Masahiro Yasuda
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Erin M Johnson-Venkatesh
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Whitney IE, Butrus S, Dyer MA, Rieke F, Sanes JR, Shekhar K. Vision-Dependent and -Independent Molecular Maturation of Mouse Retinal Ganglion Cells. Neuroscience 2023; 508:153-173. [PMID: 35870562 PMCID: PMC10809145 DOI: 10.1016/j.neuroscience.2022.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/20/2022] [Accepted: 07/13/2022] [Indexed: 01/17/2023]
Abstract
The development and connectivity of retinal ganglion cells (RGCs), the retina's sole output neurons, are patterned by activity-independent transcriptional programs and activity-dependent remodeling. To inventory the molecular correlates of these influences, we applied high-throughput single-cell RNA sequencing (scRNA-seq) to mouse RGCs at six embryonic and postnatal ages. We identified temporally regulated modules of genes that correlate with, and likely regulate, multiple phases of RGC development, ranging from differentiation and axon guidance to synaptic recognition and refinement. Some of these genes are expressed broadly while others, including key transcription factors and recognition molecules, are selectively expressed by one or a few of the 45 transcriptomically distinct types defined previously in adult mice. Next, we used these results as a foundation to analyze the transcriptomes of RGCs in mice lacking visual experience due to dark rearing from birth or to mutations that ablate either bipolar or photoreceptor cells. 98.5% of visually deprived (VD) RGCs could be unequivocally assigned to a single RGC type based on their transcriptional profiles, demonstrating that visual activity is dispensable for acquisition and maintenance of RGC type identity. However, visual deprivation significantly reduced the transcriptomic distinctions among RGC types, implying that activity is required for complete RGC maturation or maintenance. Consistent with this notion, transcriptomic alternations in VD RGCs significantly overlapped with gene modules found in developing RGCs. Our results provide a resource for mechanistic analyses of RGC differentiation and maturation, and for investigating the role of activity in these processes.
Collapse
Affiliation(s)
- Irene E Whitney
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Salwan Butrus
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Karthik Shekhar
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Helen Wills Neuroscience Institute, California Institute for Quantitative Biosciences, QB3, Center for Computational Biology, University of California, Berkeley, CA 94720, USA; Biological Systems Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
26
|
Huang Z. A Function of Amyloid-β in Mediating Activity-Dependent Axon/Synapse Competition May Unify Its Roles in Brain Physiology and Pathology. J Alzheimers Dis 2023; 92:29-57. [PMID: 36710681 PMCID: PMC10023438 DOI: 10.3233/jad-221042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Amyloid-β protein precursor (AβPP) gives rise to amyloid-β (Aβ), a peptide at the center of Alzheimer's disease (AD). AβPP, however, is also an ancient molecule dating back in evolution to some of the earliest forms of metazoans. This suggests a possible ancestral function that may have been obscured by those that evolve later. Based on literature from the functions of Aβ/AβPP in nervous system development, plasticity, and disease, to those of anti-microbial peptides (AMPs) in bacterial competition as well as mechanisms of cell competition uncovered first by Drosophila genetics, I propose that Aβ/AβPP may be part of an ancient mechanism employed in cell competition, which is subsequently co-opted during evolution for the regulation of activity-dependent neural circuit development and plasticity. This hypothesis is supported by foremost the high similarities of Aβ to AMPs, both of which possess unique, opposite (i.e., trophic versus toxic) activities as monomers and oligomers. A large body of data further suggests that the different Aβ oligomeric isoforms may serve as the protective and punishment signals long predicted to mediate activity-dependent axonal/synaptic competition in the developing nervous system and that the imbalance in their opposite regulation of innate immune and glial cells in the brain may ultimately underpin AD pathogenesis. This hypothesis can not only explain the diverse roles observed of Aβ and AβPP family molecules, but also provide a conceptual framework that can unify current hypotheses on AD. Furthermore, it may explain major clinical observations not accounted for and identify approaches for overcoming shortfalls in AD animal modeling.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
27
|
Clarkson BDS, Grund E, David K, Johnson RK, Howe CL. ISGylation is induced in neurons by demyelination driving ISG15-dependent microglial activation. J Neuroinflammation 2022; 19:258. [PMID: 36261842 PMCID: PMC9583544 DOI: 10.1186/s12974-022-02618-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/07/2022] [Indexed: 11/22/2022] Open
Abstract
The causes of grey matter pathology and diffuse neuron injury in MS remain incompletely understood. Axonal stress signals arising from white matter lesions has been suggested to play a role in initiating this diffuse grey matter pathology. Therefore, to identify the most upstream transcriptional responses in neurons arising from demyelinated axons, we analyzed the transcriptome of actively translating neuronal transcripts in mouse models of demyelinating disease. Among the most upregulated genes, we identified transcripts associated with the ISGylation pathway. ISGylation refers to the covalent attachment of the ubiquitin-like molecule interferon stimulated gene (ISG) 15 to lysine residues on substrates targeted by E1 ISG15-activating enzyme, E2 ISG15-conjugating enzymes and E3 ISG15-protein ligases. We further confirmed that ISG15 expression is increased in MS cortical and deep gray matter. Upon investigating the functional impact of neuronal ISG15 upregulation, we noted that ISG15 expression was associated changes in neuronal extracellular vesicle protein and miRNA cargo. Specifically, extracellular vesicle-associated miRNAs were skewed toward increased frequency of proinflammatory and neurotoxic miRNAs and decreased frequency of anti-inflammatory and neuroprotective miRNAs. Furthermore, we found that ISG15 directly activated microglia in a CD11b-dependent manner and that microglial activation was potentiated by treatment with EVs from neurons expressing ISG15. Further study of the role of ISG15 and ISGylation in neurons in MS and neurodegenerative diseases is warranted.
Collapse
Affiliation(s)
- Benjamin D. S. Clarkson
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XDepartment of Laboratory Medicine and Pathology, Mayo Clinic, Guggenheim 1521C, 200 First Street SW, Rochester, MN 55905 USA
| | - Ethan Grund
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XMayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and Mayo Clinic Medical Scientist Training Program, MN 55905 Rochester, USA
| | - Kenneth David
- grid.418935.20000 0004 0436 053XConcordia College, Moorhead, MN USA
| | - Renee K. Johnson
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA
| | - Charles L. Howe
- grid.66875.3a0000 0004 0459 167XDepartment of Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XDivision of Experimental Neurology, Mayo Clinic, Rochester, MN 55905 USA ,grid.66875.3a0000 0004 0459 167XCenter for Multiple Sclerosis and Autoimmune Neurology, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
28
|
Auguste YSS, Ferro A, Kahng JA, Xavier AM, Dixon JR, Vrudhula U, Nichitiu AS, Rosado D, Wee TL, Pedmale UV, Cheadle L. Oligodendrocyte precursor cells engulf synapses during circuit remodeling in mice. Nat Neurosci 2022; 25:1273-1278. [PMID: 36171430 PMCID: PMC9534756 DOI: 10.1038/s41593-022-01170-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/18/2022] [Indexed: 01/13/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes throughout life, but the functions of OPCs are not limited to oligodendrogenesis. Here we show that OPCs contribute to thalamocortical presynapse elimination in the developing and adult mouse visual cortex. OPC-mediated synapse engulfment increases in response to sensory experience during neural circuit refinement. Our data suggest that OPCs may regulate synaptic connectivity in the brain independently of oligodendrogenesis.
Collapse
Affiliation(s)
| | - Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jessica A Kahng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Andre M Xavier
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Uma Vrudhula
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Daniele Rosado
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Tse-Luen Wee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
29
|
Abstract
TWEAK (tumor necrosis factor-like weak inducer of apoptosis) is a member of the TNF superfamily that controls a multitude of cellular events including proliferation, migration, differentiation, apoptosis, angiogenesis, and inflammation. TWEAK control of these events is via an expanding list of intracellular signalling pathways which include NF-κB, ERK/MAPK, Notch, EGFR and AP-1. Two receptors have been identified for TWEAK - Fn14, which targets the membrane bound form of TWEAK, and CD163, which scavenges the soluble form of TWEAK. TWEAK appears to elicit specific events based on the receptor to which it binds, tissue type in which it is expressed, specific extrinsic conditions, and the presence of other cytokines. TWEAK signalling is protective in healthy tissues, but in chronic inflammatory states become detrimental to the tissue. Consistent data show a role for the TWEAK/FN14/CD163 axis in metabolic disease, chronic autoimmune diseases, and acute ischaemic stroke. Low circulating concentrations of soluble TWEAK are predictive of poor cardiovascular outcomes in those with and without diabetes. This review details the current understanding of the TWEAK/Fn14/CD163 axis as one of the chief regulators of immune signalling and its cell-specific role in metabolic disease development and progression.
Collapse
Affiliation(s)
- Wiktoria Ratajczak
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, Altnagelvin Hospital Campus, C-TRIC Building Glenshane Road, Derry/Londonderry, Northern Ireland, UK
| | - Sarah D Atkinson
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, Altnagelvin Hospital Campus, C-TRIC Building Glenshane Road, Derry/Londonderry, Northern Ireland, UK
| | - Catriona Kelly
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, Altnagelvin Hospital Campus, C-TRIC Building Glenshane Road, Derry/Londonderry, Northern Ireland, UK.
| |
Collapse
|
30
|
Pumo GM, Kitazawa T, Rijli FM. Epigenetic and Transcriptional Regulation of Spontaneous and Sensory Activity Dependent Programs During Neuronal Circuit Development. Front Neural Circuits 2022; 16:911023. [PMID: 35664458 PMCID: PMC9158562 DOI: 10.3389/fncir.2022.911023] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Spontaneous activity generated before the onset of sensory transduction has a key role in wiring developing sensory circuits. From axonal targeting, to synapse formation and elimination, to the balanced integration of neurons into developing circuits, this type of activity is implicated in a variety of cellular processes. However, little is known about its molecular mechanisms of action, especially at the level of genome regulation. Conversely, sensory experience-dependent activity implements well-characterized transcriptional and epigenetic chromatin programs that underlie heterogeneous but specific genomic responses that shape both postnatal circuit development and neuroplasticity in the adult. In this review, we focus on our knowledge of the developmental processes regulated by spontaneous activity and the underlying transcriptional mechanisms. We also review novel findings on how chromatin regulates the specificity and developmental induction of the experience-dependent program, and speculate their relevance for our understanding of how spontaneous activity may act at the genomic level to instruct circuit assembly and prepare developing neurons for sensory-dependent connectivity refinement and processing.
Collapse
Affiliation(s)
- Gabriele M. Pumo
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department Biozentrum, University of Basel, Basel, Switzerland
| | - Taro Kitazawa
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Filippo M. Rijli
- Laboratory of Neurodevelopmental Epigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Department Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Midorikawa M. Pathway-specific maturation of presynaptic functions of the somatosensory thalamus. Neurosci Res 2022; 181:1-8. [DOI: 10.1016/j.neures.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023]
|
32
|
Duménieu M, Marquèze-Pouey B, Russier M, Debanne D. Mechanisms of Plasticity in Subcortical Visual Areas. Cells 2021; 10:3162. [PMID: 34831385 PMCID: PMC8621502 DOI: 10.3390/cells10113162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/10/2023] Open
Abstract
Visual plasticity is classically considered to occur essentially in the primary and secondary cortical areas. Subcortical visual areas such as the dorsal lateral geniculate nucleus (dLGN) or the superior colliculus (SC) have long been held as basic structures responsible for a stable and defined function. In this model, the dLGN was considered as a relay of visual information travelling from the retina to cortical areas and the SC as a sensory integrator orienting body movements towards visual targets. However, recent findings suggest that both dLGN and SC neurons express functional plasticity, adding unexplored layers of complexity to their previously attributed functions. The existence of neuronal plasticity at the level of visual subcortical areas redefines our approach of the visual system. The aim of this paper is therefore to review the cellular and molecular mechanisms for activity-dependent plasticity of both synaptic transmission and cellular properties in subcortical visual areas.
Collapse
Affiliation(s)
| | | | | | - Dominique Debanne
- INSERM, Aix-Marseille Université, UNIS, 13015 Marseille, France; (M.D.); (B.M.-P.); (M.R.)
| |
Collapse
|
33
|
Li T, Yu D, Oak HC, Zhu B, Wang L, Jiang X, Molday RS, Kriegstein A, Piao X. Phospholipid-flippase chaperone CDC50A is required for synapse maintenance by regulating phosphatidylserine exposure. EMBO J 2021; 40:e107915. [PMID: 34585770 PMCID: PMC8561630 DOI: 10.15252/embj.2021107915] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
Synaptic refinement is a critical physiological process that removes excess synapses to establish and maintain functional neuronal circuits. Recent studies have shown that focal exposure of phosphatidylserine (PS) on synapses acts as an "eat me" signal to mediate synaptic pruning. However, the molecular mechanism underlying PS externalization at synapses remains elusive. Here, we find that murine CDC50A, a chaperone of phospholipid flippases, localizes to synapses, and that its expression depends on neuronal activity. Cdc50a knockdown leads to phosphatidylserine exposure at synapses and subsequent erroneous synapse removal by microglia partly via the GPR56 pathway. Taken together, our data support that CDC50A safeguards synapse maintenance by regulating focal phosphatidylserine exposure at synapses.
Collapse
Affiliation(s)
- Tao Li
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Diankun Yu
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Hayeon C Oak
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Beika Zhu
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Li Wang
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Xueqiao Jiang
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| | - Robert S Molday
- Department of Biochemistry and Molecular BiologyUniversity of British ColumbiaVancouverBCCanada
| | - Arnold Kriegstein
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Department of NeurologyUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Xianhua Piao
- Weill Institute for NeuroscienceUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Newborn Brain Research InstituteUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell ResearchUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
- Division of NeonatologyDepartment of PediatricsUniversity of California, San Francisco (UCSF)San FranciscoCAUSA
| |
Collapse
|
34
|
Bakken TE, van Velthoven CTJ, Menon V, Hodge RD, Yao Z, Nguyen TN, Graybuck LT, Horwitz GD, Bertagnolli D, Goldy J, Yanny AM, Garren E, Parry S, Casper T, Shehata SI, Barkan ER, Szafer A, Levi BP, Dee N, Smith KA, Sunkin SM, Bernard A, Phillips J, Hawrylycz MJ, Koch C, Murphy GJ, Lein E, Zeng H, Tasic B. Single-cell and single-nucleus RNA-seq uncovers shared and distinct axes of variation in dorsal LGN neurons in mice, non-human primates, and humans. eLife 2021; 10:e64875. [PMID: 34473054 PMCID: PMC8412930 DOI: 10.7554/elife.64875] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/18/2021] [Indexed: 12/11/2022] Open
Abstract
Abundant evidence supports the presence of at least three distinct types of thalamocortical (TC) neurons in the primate dorsal lateral geniculate nucleus (dLGN) of the thalamus, the brain region that conveys visual information from the retina to the primary visual cortex (V1). Different types of TC neurons in mice, humans, and macaques have distinct morphologies, distinct connectivity patterns, and convey different aspects of visual information to the cortex. To investigate the molecular underpinnings of these cell types, and how these relate to differences in dLGN between human, macaque, and mice, we profiled gene expression in single nuclei and cells using RNA-sequencing. These efforts identified four distinct types of TC neurons in the primate dLGN: magnocellular (M) neurons, parvocellular (P) neurons, and two types of koniocellular (K) neurons. Despite extensively documented morphological and physiological differences between M and P neurons, we identified few genes with significant differential expression between transcriptomic cell types corresponding to these two neuronal populations. Likewise, the dominant feature of TC neurons of the adult mouse dLGN is high transcriptomic similarity, with an axis of heterogeneity that aligns with core vs. shell portions of mouse dLGN. Together, these data show that transcriptomic differences between principal cell types in the mature mammalian dLGN are subtle relative to the observed differences in morphology and cortical projection targets. Finally, alignment of transcriptome profiles across species highlights expanded diversity of GABAergic neurons in primate versus mouse dLGN and homologous types of TC neurons in primates that are distinct from TC neurons in mouse.
Collapse
Affiliation(s)
| | | | - Vilas Menon
- Allen Institute for Brain ScienceSeattleUnited States
- Department of Neurology, Columbia University Medical CenterNew YorkUnited States
| | | | - Zizhen Yao
- Allen Institute for Brain ScienceSeattleUnited States
| | | | | | - Gregory D Horwitz
- Washington National Primate Research Center and Department of Physiology and Biophysics, University of WashingtonSeattleUnited States
| | | | - Jeff Goldy
- Allen Institute for Brain ScienceSeattleUnited States
| | | | - Emma Garren
- Allen Institute for Brain ScienceSeattleUnited States
| | - Sheana Parry
- Allen Institute for Brain ScienceSeattleUnited States
| | - Tamara Casper
- Allen Institute for Brain ScienceSeattleUnited States
| | | | | | - Aaron Szafer
- Allen Institute for Brain ScienceSeattleUnited States
| | - Boaz P Levi
- Allen Institute for Brain ScienceSeattleUnited States
| | - Nick Dee
- Allen Institute for Brain ScienceSeattleUnited States
| | | | | | - Amy Bernard
- Allen Institute for Brain ScienceSeattleUnited States
| | - John Phillips
- Allen Institute for Brain ScienceSeattleUnited States
| | | | - Christof Koch
- Allen Institute for Brain ScienceSeattleUnited States
| | - Gabe J Murphy
- Allen Institute for Brain ScienceSeattleUnited States
| | - Ed Lein
- Allen Institute for Brain ScienceSeattleUnited States
| | - Hongkui Zeng
- Allen Institute for Brain ScienceSeattleUnited States
| | | |
Collapse
|
35
|
Dziabis JE, Bilbo SD. Microglia and Sensitive Periods in Brain Development. Curr Top Behav Neurosci 2021; 53:55-78. [PMID: 34463934 DOI: 10.1007/7854_2021_242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
From embryonic neuronal migration to adolescent circuit refinement, the immune system plays an essential role throughout central nervous system (CNS) development. Immune signaling molecules serve as a common language between the immune system and CNS, allowing them to work together to modulate brain function both in health and disease. As the resident CNS macrophage, microglia comprise the majority of immune cells in the brain. Much like their peripheral counterparts, microglia survey their environment for pathology, clean up debris, and propagate inflammatory responses when necessary. Beyond this, recent studies have highlighted that microglia perform a number of complex tasks during neural development, from directing neuronal and axonal positioning to pruning synapses, receptors, and even whole cells. In this chapter, we discuss this literature within the framework that immune activation during discrete windows of neural development can profoundly impact brain function long-term, and thus the risk of neurodevelopmental and neuropsychiatric disorders. In this chapter, we review three sensitive developmental periods - embryonic wiring, early postnatal synaptic pruning, and adolescent circuit refinement - in order to highlight the diversity of functions that microglia perform in building a brain. In reviewing this literature, it becomes obvious that timing matters, perhaps more so than the nature of the immune activation itself; largely conserved patterns of microglial response to diverse insults result in different functional impacts depending on the stage of brain maturation at the time of the challenge.
Collapse
Affiliation(s)
- Julia E Dziabis
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA.,Department of Neurobiology, Duke University, Durham, NC, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA. .,Department of Neurobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
36
|
Ferro A, Auguste YSS, Cheadle L. Microglia, Cytokines, and Neural Activity: Unexpected Interactions in Brain Development and Function. Front Immunol 2021; 12:703527. [PMID: 34276699 PMCID: PMC8281303 DOI: 10.3389/fimmu.2021.703527] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 12/01/2022] Open
Abstract
Intercellular signaling molecules such as cytokines and their receptors enable immune cells to communicate with one another and their surrounding microenvironments. Emerging evidence suggests that the same signaling pathways that regulate inflammatory responses to injury and disease outside of the brain also play powerful roles in brain development, plasticity, and function. These observations raise the question of how the same signaling molecules can play such distinct roles in peripheral tissues compared to the central nervous system, a system previously thought to be largely protected from inflammatory signaling. Here, we review evidence that the specialized roles of immune signaling molecules such as cytokines in the brain are to a large extent shaped by neural activity, a key feature of the brain that reflects active communication between neurons at synapses. We discuss the known mechanisms through which microglia, the resident immune cells of the brain, respond to increases and decreases in activity by engaging classical inflammatory signaling cascades to assemble, remodel, and eliminate synapses across the lifespan. We integrate evidence from (1) in vivo imaging studies of microglia-neuron interactions, (2) developmental studies across multiple neural circuits, and (3) molecular studies of activity-dependent gene expression in microglia and neurons to highlight the specific roles of activity in defining immune pathway function in the brain. Given that the repurposing of signaling pathways across different tissues may be an important evolutionary strategy to overcome the limited size of the genome, understanding how cytokine function is established and maintained in the brain could lead to key insights into neurological health and disease.
Collapse
Affiliation(s)
| | | | - Lucas Cheadle
- Neuroscience Department, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| |
Collapse
|
37
|
An activity-dependent determinant of synapse elimination in the mammalian brain. Neuron 2021; 109:1333-1349.e6. [PMID: 33770504 DOI: 10.1016/j.neuron.2021.03.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/26/2021] [Accepted: 03/04/2021] [Indexed: 01/06/2023]
Abstract
To establish functional neural circuits in the brain, synaptic connections are refined by neural activity during development, where active connections are maintained and inactive ones are eliminated. However, the molecular signals that regulate synapse refinement remain to be elucidated. When we inactivate a subset of neurons in the mouse cingulate cortex, their callosal connections are eliminated through activity-dependent competition. Using this system, we identify JAK2 tyrosine kinase as a key regulator of inactive synapse elimination. We show that JAK2 is necessary and sufficient for elimination of inactive connections; JAK2 is activated at inactive synapses in response to signals from other active synapses; STAT1, a substrate of JAK2, mediates inactive synapse elimination; JAK2 signaling is critical for physiological refinement of synapses during normal development; and JAK2 regulates synapse refinement in multiple brain regions. We propose that JAK2 is an activity-dependent switch that serves as a determinant of inactive synapse elimination.
Collapse
|
38
|
Nagy D, Ennis KA, Wei R, Su SC, Hinckley CA, Gu RF, Gao B, Massol RH, Ehrenfels C, Jandreski L, Thomas AM, Nelson A, Gyoneva S, Hajós M, Burkly LC. Developmental synaptic regulator, TWEAK/Fn14 signaling, is a determinant of synaptic function in models of stroke and neurodegeneration. Proc Natl Acad Sci U S A 2021; 118:e2001679118. [PMID: 33526652 PMCID: PMC8017933 DOI: 10.1073/pnas.2001679118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Identifying molecular mediators of neural circuit development and/or function that contribute to circuit dysfunction when aberrantly reengaged in neurological disorders is of high importance. The role of the TWEAK/Fn14 pathway, which was recently reported to be a microglial/neuronal axis mediating synaptic refinement in experience-dependent visual development, has not been explored in synaptic function within the mature central nervous system. By combining electrophysiological and phosphoproteomic approaches, we show that TWEAK acutely dampens basal synaptic transmission and plasticity through neuronal Fn14 and impacts the phosphorylation state of pre- and postsynaptic proteins in adult mouse hippocampal slices. Importantly, this is relevant in two models featuring synaptic deficits. Blocking TWEAK/Fn14 signaling augments synaptic function in hippocampal slices from amyloid-beta-overexpressing mice. After stroke, genetic or pharmacological inhibition of TWEAK/Fn14 signaling augments basal synaptic transmission and normalizes plasticity. Our data support a glial/neuronal axis that critically modifies synaptic physiology and pathophysiology in different contexts in the mature brain and may be a therapeutic target for improving neurophysiological outcomes.
Collapse
Affiliation(s)
- Dávid Nagy
- Clinical Sciences, Biogen, Cambridge, MA 02142
- Biogen Postdoctoral Scientist Program, Cellular Physiology, Biogen, Cambridge, MA 02142
| | - Katelin A Ennis
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Ru Wei
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Susan C Su
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | | | - Rong-Fang Gu
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Benbo Gao
- Chemical Biology and Proteomics, Biogen, Cambridge, MA 02142
| | - Ramiro H Massol
- Translational Cellular Sciences, Biogen, Cambridge, MA 02142
| | - Chris Ehrenfels
- Translational Cellular Sciences, Biogen, Cambridge, MA 02142
| | | | - Ankur M Thomas
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Ashley Nelson
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Stefka Gyoneva
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142
| | - Mihály Hajós
- Clinical Sciences, Biogen, Cambridge, MA 02142
- Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520
| | - Linda C Burkly
- Genetic and Neurodevelopmental Disease Research, Biogen, Cambridge, MA 02142;
| |
Collapse
|
39
|
Keck T. Microglia Tweak Retinogeniculate Pathways during Visual Circuit Refinement. Neuron 2020; 108:397-399. [PMID: 33181071 DOI: 10.1016/j.neuron.2020.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cheadle et al. reveal that microglia expressing TWEAK facilitate synapse elimination through a novel, non-phagocytic mechanism in the retinogeniculate pathway during visual circuit development. This mechanism is experience-dependent and occurs through the local binding of TWEAK to postsynaptic Fn14.
Collapse
Affiliation(s)
- Tara Keck
- Department of Neuroscience, Physiology and Pharmacology, University College London, 21 University St., London WC1E 6DE, UK.
| |
Collapse
|
40
|
Liang L, Chen C. Organization, Function, and Development of the Mouse Retinogeniculate Synapse. Annu Rev Vis Sci 2020; 6:261-285. [DOI: 10.1146/annurev-vision-121219-081753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Visual information is encoded in distinct retinal ganglion cell (RGC) types in the eye tuned to specific features of the visual space. These streams of information project to the visual thalamus, the first station of the image-forming pathway. In the mouse, this connection between RGCs and thalamocortical neurons, the retinogeniculate synapse, has become a powerful experimental model for understanding how circuits in the thalamus are constructed to process these incoming lines of information. Using modern molecular and genetic tools, recent studies have suggested a more complex circuit organization than was previously understood. In this review, we summarize the current understanding of the structural and functional organization of the retinogeniculate synapse in the mouse. We discuss a framework by which a seemingly complex circuit can effectively integrate and parse information to downstream stations of the visual pathway. Finally, we review how activity and visual experience can sculpt this exquisite connectivity.
Collapse
Affiliation(s)
- Liang Liang
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neuroscience, Yale University, New Haven, Connecticut 06520, USA
| | - Chinfei Chen
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| |
Collapse
|
41
|
Cheadle L, Rivera SA, Phelps JS, Ennis KA, Stevens B, Burkly LC, Lee WCA, Greenberg ME. Sensory Experience Engages Microglia to Shape Neural Connectivity through a Non-Phagocytic Mechanism. Neuron 2020; 108:451-468.e9. [PMID: 32931754 DOI: 10.1016/j.neuron.2020.08.002] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 05/10/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
Sensory experience remodels neural circuits in the early postnatal brain through mechanisms that remain to be elucidated. Applying a new method of ultrastructural analysis to the retinogeniculate circuit, we find that visual experience alters the number and structure of synapses between the retina and the thalamus. These changes require vision-dependent transcription of the receptor Fn14 in thalamic relay neurons and the induction of its ligand TWEAK in microglia. Fn14 functions to increase the number of bulbous spine-associated synapses at retinogeniculate connections, likely contributing to the strengthening of the circuit that occurs in response to visual experience. However, at retinogeniculate connections near TWEAK-expressing microglia, TWEAK signals via Fn14 to restrict the number of bulbous spines on relay neurons, leading to the elimination of a subset of connections. Thus, TWEAK and Fn14 represent an intercellular signaling axis through which microglia shape retinogeniculate connectivity in response to sensory experience.
Collapse
Affiliation(s)
- Lucas Cheadle
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Samuel A Rivera
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Jasper S Phelps
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Katelin A Ennis
- Research and Early Development, Biogen, 115 Broadway, Cambridge, MA 04142, USA
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Linda C Burkly
- Research and Early Development, Biogen, 115 Broadway, Cambridge, MA 04142, USA
| | - Wei-Chung Allen Lee
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Michael E Greenberg
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
42
|
Abstract
Binocular vision depends on retinal ganglion cell (RGC) axon projection either to the same side or to the opposite side of the brain. In this article, we review the molecular mechanisms for decussation of RGC axons, with a focus on axon guidance signaling at the optic chiasm and ipsi- and contralateral axon organization in the optic tract prior to and during targeting. The spatial and temporal features of RGC neurogenesis that give rise to ipsilateral and contralateral identity are described. The albino visual system is highlighted as an apt comparative model for understanding RGC decussation, as albinos have a reduced ipsilateral projection and altered RGC neurogenesis associated with perturbed melanogenesis in the retinal pigment epithelium. Understanding the steps for RGC specification into ipsi- and contralateral subtypes will facilitate differentiation of stem cells into RGCs with proper navigational abilities for effective axon regeneration and correct targeting of higher-order visual centers.
Collapse
Affiliation(s)
- Carol Mason
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10027, USA; .,Department of Neuroscience, Columbia University, New York, NY 10027, USA.,Department of Ophthalmology, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA;
| | - Nefeli Slavi
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA;
| |
Collapse
|
43
|
Huh CYL, Abdelaal K, Salinas KJ, Gu D, Zeitoun J, Figueroa Velez DX, Peach JP, Fowlkes CC, Gandhi SP. Long-term Monocular Deprivation during Juvenile Critical Period Disrupts Binocular Integration in Mouse Visual Thalamus. J Neurosci 2020; 40:585-604. [PMID: 31767678 PMCID: PMC6961993 DOI: 10.1523/jneurosci.1626-19.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/06/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023] Open
Abstract
Study of the neural deficits caused by mismatched binocular vision in early childhood has predominantly focused on circuits in the primary visual cortex (V1). Recent evidence has revealed that neurons in mouse dorsolateral geniculate nucleus (dLGN) can undergo rapid ocular dominance plasticity following monocular deprivation (MD). It remains unclear, however, whether the long-lasting deficits attributed to MD during the critical period originate in the thalamus. Using in vivo two-photon Ca2+ imaging of dLGN afferents in superficial layers of V1 in female and male mice, we demonstrate that 14 d MD during the critical period leads to a chronic loss of binocular dLGN inputs while sparing response strength and spatial acuity. Importantly, MD leads to profoundly mismatched visual tuning properties in remaining binocular dLGN afferents. Furthermore, MD impairs binocular modulation, reducing facilitation of responses of both binocular and monocular dLGN inputs during binocular viewing. As predicted by our findings in thalamic inputs, Ca2+ imaging from V1 neurons revealed spared spatial acuity but impaired binocularity in L4 neurons. V1 L2/3 neurons in contrast displayed deficits in both binocularity and spatial acuity. Our data demonstrate that critical-period MD produces long-lasting disruptions in binocular integration beginning in early binocular circuits in dLGN, whereas spatial acuity deficits first arise from circuits further downstream in V1. Our findings indicate that the development of normal binocular vision and spatial acuity depend upon experience-dependent refinement of distinct stages in the mammalian visual system.SIGNIFICANCE STATEMENT Abnormal binocular vision and reduced acuity are hallmarks of amblyopia, a disorder that affects 2%-5% of the population. It is widely thought that the neural deficits underlying amblyopia begin in the circuits of primary visual cortex. Using in vivo two-photon calcium imaging of thalamocortical axons in mice, we show that depriving one eye of input during a critical period in development chronically impairs binocular integration in thalamic inputs to primary visual cortex. In contrast, visual acuity is spared in thalamic inputs. These findings shed new light on the role for developmental mechanisms in the thalamus in establishing binocular vision and may have critical implications for amblyopia.
Collapse
Affiliation(s)
| | | | | | - Diyue Gu
- Donald Bren School of Information & Computer Sciences
| | | | | | - John P Peach
- Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218
| | | | - Sunil P Gandhi
- Department of Neurobiology and Behavior,
- Center for Neurobiology of Learning and Memory, University of California, Irvine, California 92697, and
| |
Collapse
|
44
|
The metabotropic glutamate receptor subtype 1 regulates development and maintenance of lemniscal synaptic connectivity in the somatosensory thalamus. PLoS One 2019; 14:e0226820. [PMID: 31881077 PMCID: PMC6934304 DOI: 10.1371/journal.pone.0226820] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/05/2019] [Indexed: 11/19/2022] Open
Abstract
The metabotropic glutamate receptor subtype 1 (mGluR1) is a major subtype of group I mGluRs, which contributes to the development and plasticity of synapses in the brain. In the sensory thalamus, the thalamocortical neuron receives sensory afferents and massive feedback input from corticothalamic (CT) fibers. Notably, mGluR1 is more concentrated in CT synapses in the sensory thalamus. In the visual thalamus, mGluR1 maintains mature afferent synaptic connectivity. However, it is unknown whether mGluR1 contributes to strengthening of immature synapses or weakening of excess synapses during development and whether mGluR1 at CT synapses heterosynaptically regulates the development or refinement of afferent synapses. Here we investigated the effects of knocking out the gene encoding mGluR1 or pharmacologically blocking cortical activity on the development and maintenance of lemniscal synapses, i.e., the somatosensory afferent synapses, in the ventral posteromedial somatosensory thalamus. mGluR1-knockout (KO) mice exhibited delayed developmental strengthening as well as incomplete elimination and remodeling after maturation of lemniscal synapses. Similar to the phenotypes exhibited by mGluR1-KO mice, pharmacological blockade of somatosensory cortical activity from P12 or P21 for 1 week in wild-type mice perturbed elimination or maintenance of lemniscal synapses, respectively. The same manipulation in mGluR1-KO mice failed to induce additional abnormalities in lemniscal synaptic connectivity. These results suggest that activation of mGluR1, driven by CT input, regulates multiple stages of the development of lemniscal synapses, including strengthening, refinement, and maintenance in the somatosensory thalamus.
Collapse
|
45
|
Bennett FC, Molofsky AV. The immune system and psychiatric disease: a basic science perspective. Clin Exp Immunol 2019; 197:294-307. [PMID: 31125426 PMCID: PMC6693968 DOI: 10.1111/cei.13334] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 12/13/2022] Open
Abstract
Mental illness exerts a major burden on human health, yet evidence-based treatments are rudimentary due to a limited understanding of the underlying pathologies. Clinical studies point to roles for the immune system in psychiatric diseases, while basic science has revealed that the brain has an active and multi-cellular resident immune system that interacts with peripheral immunity and impacts behavior. In this perspective, we highlight evidence of immune involvement in human psychiatric disease and review data from animal models that link immune signaling to neuronal function and behavior. We propose a conceptual framework for linking advances in basic neuroimmunology to their potential relevance for psychiatric diseases, based on the subtypes of immune responses defined in peripheral tissues. Our goal is to identify novel areas of focus for future basic and translational studies that may reveal the potential of the immune system for diagnosing and treating mental illnesses.
Collapse
Affiliation(s)
- F. C. Bennett
- Department of Psychiatry, Perelman School of MedicineUniversity of Pennsylvania, The Children's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - A. V. Molofsky
- Department of Psychiatry and Weill Institute for NeurosciencesUniversity of CaliforniaSan FranciscoSan FranciscoCAUSA
| |
Collapse
|
46
|
Yap EL, Greenberg ME. Activity-Regulated Transcription: Bridging the Gap between Neural Activity and Behavior. Neuron 2019; 100:330-348. [PMID: 30359600 DOI: 10.1016/j.neuron.2018.10.013] [Citation(s) in RCA: 400] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 10/02/2018] [Accepted: 10/05/2018] [Indexed: 12/21/2022]
Abstract
Gene transcription is the process by which the genetic codes of organisms are read and interpreted as a set of instructions for cells to divide, differentiate, migrate, and mature. As cells function in their respective niches, transcription further allows mature cells to interact dynamically with their external environment while reliably retaining fundamental information about past experiences. In this Review, we provide an overview of the field of activity-dependent transcription in the vertebrate brain and highlight contemporary work that ranges from studies of activity-dependent chromatin modifications to plasticity mechanisms underlying adaptive behaviors. We identify key gaps in knowledge and propose integrated approaches toward a deeper understanding of how activity-dependent transcription promotes the refinement and plasticity of neural circuits for cognitive function.
Collapse
Affiliation(s)
- Ee-Lynn Yap
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Program in Neuroscience, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael E Greenberg
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
47
|
Abstract
Functional neural circuits of mature animals are shaped during postnatal development by eliminating early-formed redundant synapses and strengthening of necessary connections. In the nervous system of newborn animals, redundant synapses are only transient features of the circuit. During subsequent postnatal development, some synapses are strengthened whereas other redundant connections are weakened and eventually eliminated. In this review, we introduce recent studies on the mechanisms of developmental remodeling of climbing fiber-to-Purkinje cell synapses in the cerebellum and synapses from the retina to neurons in the dorsal lateral geniculate nucleus of the visual thalamus (retinogeniculate synapses). These are the two representative models of developmental synapse remodeling in the brain and they share basic principles, including dependency on neural activity. However, recent studies have disclosed that, in several respects, the two models use different molecules and strategies to establish mature synaptic connectivity. We describe similarities and differences between the two models and discuss remaining issues to be tackled in the future in order to understand the general schemes of developmental synapse remodeling.
Collapse
Affiliation(s)
- Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
| | - Takaki Watanabe
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
48
|
Developmental Remodeling of Thalamic Interneurons Requires Retinal Signaling. J Neurosci 2019; 39:3856-3866. [PMID: 30842249 DOI: 10.1523/jneurosci.2224-18.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/04/2019] [Accepted: 02/26/2019] [Indexed: 02/06/2023] Open
Abstract
The dorsal lateral geniculate nucleus (dLGN) of the mouse is a model system to study the development of thalamic circuitry. Most studies focus on relay neurons of dLGN, yet little is known about the development of the other principal cell type, intrinsic interneurons. Here we examined whether the structure and function of interneurons relies on retinal signaling. We took a loss-of-function approach and crossed GAD67-GFP mice, which express GFP in dLGN interneurons, with math5 nulls (math5-/-), mutants that lack retinal ganglion cells and retinofugal projections. In vitro recordings and 3-D reconstructions of biocytin-filled interneurons at different postnatal ages showed their development is a multistaged process involving migration, arbor remodeling, and synapse formation. Arbor remodeling begins during the second postnatal week, after migration to and dispersion within dLGN is complete. This phase includes a period of exuberant branching where arbors grow in number, complexity, and field size. Such growth is followed by branch pruning and stabilization, as interneurons adopt a bipolar architecture. The absence of retinal signaling disrupts this process. The math5-/- interneurons fail to branch and prune, and instead maintain a simple, sparse architecture. To test how such defects influence connectivity with dLGN relay neurons, we used DHPG [(RS)-3,5-dihydroxyphenylglycine], the mGluR1,5 agonist that targets F2 terminals. This led to substantial increases in IPSC activity among WT relay neurons but had little impact in math5-/- mice. Together, these data suggest that retinal signaling is needed to support the arbor elaboration and synaptic connectivity of dLGN interneurons.SIGNIFICANCE STATEMENT Presently, our understanding about the development of the dorsal lateral geniculate nucleus is limited to circuits involving excitatory thalamocortical relay neurons. Here we show that the other principal cell type, intrinsic interneurons, has a multistaged developmental plan that relies on retinal innervation. These findings indicate that signaling from the periphery guides the maturation of interneurons and the establishment of inhibitory thalamic circuits.
Collapse
|