1
|
Martinez-Navarrete G, Castaño-Castaño S, Morales-Navas M, Nieto-Escámez F, Sánchez-Santed F, Fernandez E. Impact of transcranial Direct Current Stimulation on stereoscopic vision and retinal structure in adult amblyopic rodents. Eye Brain 2024; 16:75-88. [PMID: 39498234 PMCID: PMC11533879 DOI: 10.2147/eb.s474573] [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: 04/19/2024] [Accepted: 09/23/2024] [Indexed: 11/07/2024] Open
Abstract
Purpose The impact of visual deprivation on retinal structure is widely debated. Experimental models, like monocular deprivation through lid suture, provide insights into the consequences of lacking visual experience during development. This deprivation delays primary visual cortex (CV1) maturation due to improper neural connection consolidation, which remains plastic beyond the critical period. However, few studies have used Optical Coherence Tomography (OCT) to investigate structural alterations in the retina of animal models following monocular deprivation. Instead, some studies have focused on the ganglion cell layer using post-mortem histological techniques in amblyopia models induced by monocular deprivation. Methods In this study, we used Cliff test to assess stereoscopic vision and spectral domain optical coherence tomography (SD-OCT) to evaluate retinal changes in an in vivo model of visual deprivation treated with Transcranial Direct Current Stimulation (tDCS). Results The depth perception test initially revealed differences between individuals with amblyopia and the control group. However, after 8 tDCS sessions, amblyopic subjects matched the control group's performance, which remained stable Additionally, significant changes were observed in retinal structures post-tDCS treatment. Specifically, the thickness of the Nerve Fiber Layer + Ganglion Cell Layer + Inner Plexiform Layer (NFL+GCL+IPL) increased significantly in amblyopic eyes (p<0.001). Moreover, significant retinal thickening, including the Nerve Fiber Layer + Ganglion Cell Layer + Inner Plexiform Layer (NFL+GCL+IPL) and the entire retina, was observed post-tDCS treatment (p<0.05), highlighting the critical role of tDCS in ameliorating amblyopia. Additionally, treated animals exhibited reduced thickness in the Inner Nuclear Layer (INL) and Outer Nuclear Layer (ONL). Conclusion tDCS treatment effectively restores amblyopic individuals' stereoscopic vision, aligning their performance with controls, while impacting retinal structure, highlighting its potential in ameliorating amblyopia's visual deficits.
Collapse
Affiliation(s)
- Gema Martinez-Navarrete
- Neuroprosthesis and Neuroengineering Research Group, Institute of Bioengineering (IB), University Miguel Hernández (UMH), Elche, 03020, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - Sergio Castaño-Castaño
- Department Psychology, University of Oviedo, Plaza Feijoo S/N, Oviedo, 33003, Spain
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain
- Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Miguel Morales-Navas
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
| | - Francisco Nieto-Escámez
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
- Neuropsychological Evaluation and Rehabilitation Center (CERNEP), Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
| | - Fernando Sánchez-Santed
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
| | - Eduardo Fernandez
- Neuroprosthesis and Neuroengineering Research Group, Institute of Bioengineering (IB), University Miguel Hernández (UMH), Elche, 03020, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
2
|
Xin W, Kaneko M, Roth RH, Zhang A, Nocera S, Ding JB, Stryker MP, Chan JR. Oligodendrocytes and myelin limit neuronal plasticity in visual cortex. Nature 2024; 633:856-863. [PMID: 39169185 PMCID: PMC11424474 DOI: 10.1038/s41586-024-07853-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 07/19/2024] [Indexed: 08/23/2024]
Abstract
Developmental myelination is a protracted process in the mammalian brain1. One theory for why oligodendrocytes mature so slowly posits that myelination may stabilize neuronal circuits and temper neuronal plasticity as animals age2-4. We tested this theory in the visual cortex, which has a well-defined critical period for experience-dependent neuronal plasticity5. During adolescence, visual experience modulated the rate of oligodendrocyte maturation in visual cortex. To determine whether oligodendrocyte maturation in turn regulates neuronal plasticity, we genetically blocked oligodendrocyte differentiation and myelination in adolescent mice. In adult mice lacking adolescent oligodendrogenesis, a brief period of monocular deprivation led to a significant decrease in visual cortex responses to the deprived eye, reminiscent of the plasticity normally restricted to adolescence. This enhanced functional plasticity was accompanied by a greater turnover of dendritic spines and coordinated reductions in spine size following deprivation. Furthermore, inhibitory synaptic transmission, which gates experience-dependent plasticity at the circuit level, was diminished in the absence of adolescent oligodendrogenesis. These results establish a critical role for oligodendrocytes in shaping the maturation and stabilization of cortical circuits and support the concept of developmental myelination acting as a functional brake on neuronal plasticity.
Collapse
Affiliation(s)
- Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| | - Megumi Kaneko
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Richard H Roth
- Departments of Neurosurgery and Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Albert Zhang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Sonia Nocera
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jun B Ding
- Departments of Neurosurgery and Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael P Stryker
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jonah R Chan
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Takahata T. Development of ocular dominance columns across rodents and other species: revisiting the concept of critical period plasticity. Front Neural Circuits 2024; 18:1402700. [PMID: 39036421 PMCID: PMC11258045 DOI: 10.3389/fncir.2024.1402700] [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: 03/18/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
The existence of cortical columns, regarded as computational units underlying both lower and higher-order information processing, has long been associated with highly evolved brains, and previous studies suggested their absence in rodents. However, recent discoveries have unveiled the presence of ocular dominance columns (ODCs) in the primary visual cortex (V1) of Long-Evans rats. These domains exhibit continuity from layer 2 through layer 6, confirming their identity as genuine ODCs. Notably, ODCs are also observed in Brown Norway rats, a strain closely related to wild rats, suggesting the physiological relevance of ODCs in natural survival contexts, although they are lacking in albino rats. This discovery has enabled researchers to explore the development and plasticity of cortical columns using a multidisciplinary approach, leveraging studies involving hundreds of individuals-an endeavor challenging in carnivore and primate species. Notably, developmental trajectories differ depending on the aspect under examination: while the distribution of geniculo-cortical afferent terminals indicates matured ODCs even before eye-opening, consistent with prevailing theories in carnivore/primate studies, examination of cortical neuron spiking activities reveals immature ODCs until postnatal day 35, suggesting delayed maturation of functional synapses which is dependent on visual experience. This developmental gap might be recognized as 'critical period' for ocular dominance plasticity in previous studies. In this article, I summarize cross-species differences in ODCs and geniculo-cortical network, followed by a discussion on the development, plasticity, and evolutionary significance of rat ODCs. I discuss classical and recent studies on critical period plasticity in the venue where critical period plasticity might be a component of experience-dependent development. Consequently, this series of studies prompts a paradigm shift in our understanding of species conservation of cortical columns and the nature of plasticity during the classical critical period.
Collapse
|
4
|
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
|
5
|
Le L, Miyanishi K, Tanaka J, Majewska AK. Microglial Regulation of Sleep and Wakefulness. ADVANCES IN NEUROBIOLOGY 2024; 37:243-260. [PMID: 39207696 DOI: 10.1007/978-3-031-55529-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sleep serves a multitude of roles in brain maturation and function. Although the neural networks involved in sleep regulation have been extensively characterized, it is increasingly recognized that neurons are not the sole conductor orchestrating the rhythmic cycle of sleep and wakefulness. In the central nervous system, microglia have emerged as an important player in sleep regulation. Within the last two decades, microglia have gained substantial attention for carrying out numerous nonimmune tasks that are crucial for brain development and function by co-opting similar mechanisms used in their conventional immune functions. Here, we highlight the importance of microglia in sleep regulation with recent findings reporting an arrhythmic sleep/wake cycle in the absence of microglia. Although the underlying mechanisms for such regulation are still being uncovered, it is likely that microglial contributions to the rhythmic control of the sleep/wake cycle come from their influence on synaptic strength and neuronal activity. We review the current literature to provide speculative signaling pathways and suggest key questions for future research. Advancing our knowledge of the mechanistic contribution of microglia to sleep regulation will not only further our insight into this critical biological process but also be instrumental in providing novel therapeutic strategies for sleep disorders.
Collapse
Affiliation(s)
- Linh Le
- Department of Neuroscience, Del Monte Institute of Neuroscience, Center for Visual Science, Neuroscience Graduate Program, University of Rochester, Rochester, NY, USA
| | - Kazuya Miyanishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan.
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute of Neuroscience, Center for Visual Science, Neuroscience Graduate Program, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
6
|
Rudolph S, Badura A, Lutzu S, Pathak SS, Thieme A, Verpeut JL, Wagner MJ, Yang YM, Fioravante D. Cognitive-Affective Functions of the Cerebellum. J Neurosci 2023; 43:7554-7564. [PMID: 37940582 PMCID: PMC10634583 DOI: 10.1523/jneurosci.1451-23.2023] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 11/10/2023] Open
Abstract
The cerebellum, traditionally associated with motor coordination and balance, also plays a crucial role in various aspects of higher-order function and dysfunction. Emerging research has shed light on the cerebellum's broader contributions to cognitive, emotional, and reward processes. The cerebellum's influence on autonomic function further highlights its significance in regulating motivational and emotional states. Perturbations in cerebellar development and function have been implicated in various neurodevelopmental disorders, including autism spectrum disorder and attention deficit hyperactivity disorder. An increasing appreciation for neuropsychiatric symptoms that arise from cerebellar dysfunction underscores the importance of elucidating the circuit mechanisms that underlie complex interactions between the cerebellum and other brain regions for a comprehensive understanding of complex behavior. By briefly discussing new advances in mapping cerebellar function in affective, cognitive, autonomic, and social processing and reviewing the role of the cerebellum in neuropathology beyond the motor domain, this Mini-Symposium review aims to provide a broad perspective of cerebellar intersections with the limbic brain in health and disease.
Collapse
Affiliation(s)
- Stephanie Rudolph
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Aleksandra Badura
- Department of Neuroscience, Erasmus MC Rotterdam, Rotterdam, 3015 GD, The Netherlands
| | - Stefano Lutzu
- Department of Neuroscience, Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York 10461
| | - Salil Saurav Pathak
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
| | - Andreas Thieme
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, D-45147, Germany
| | - Jessica L Verpeut
- Department of Psychology, Arizona State University, Tempe, Arizona 85287
| | - Mark J Wagner
- National Institute of Neurological Disorders & Stroke, National Institutes of Health, Bethesda, Maryland 20814
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, Minnesota 55812
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Diasynou Fioravante
- Center for Neuroscience, University of California-Davis, Davis, California 95618
- Department of Neurobiology, Physiology and Behavior, University of California-Davis, Davis, California 95618
| |
Collapse
|
7
|
Xin W, Kaneko M, Roth RH, Zhang A, Nocera S, Ding JB, Stryker MP, Chan JR. Adolescent oligodendrogenesis and myelination restrict experience-dependent neuronal plasticity in adult visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.29.560231. [PMID: 37808666 PMCID: PMC10557765 DOI: 10.1101/2023.09.29.560231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
BACKGROUND Developmental myelination is a protracted process in the mammalian brain. One theory for why oligodendrocytes mature so slowly posits that myelination may stabilize neuronal circuits and temper neuronal plasticity as animals age. We tested this hypothesis in the visual cortex, which has a well-defined critical period for experience-dependent neuronal plasticity. OBJECTIVES/METHODS To prevent myelin progression, we conditionally deleted Myrf, a transcription factor necessary for oligodendrocyte maturation, from oligodendrocyte precursor cells (Myrf cKO) in adolescent mice. To induce experience-dependent plasticity, adult control and Myrf cKO mice were monocularly deprived by eyelid suture. Functional and structural neuronal plasticity in the visual cortex were assessed in vivo by intrinsic signal optical imaging and longitudinal two photon imaging of dendritic spines, respectively. RESULTS During adolescence, visual experience modulated the rate of oligodendrocyte maturation in visual cortex. Myrf deletion from oligodendrocyte precursors during adolescence led to inhibition of oligodendrocyte maturation and myelination that persisted into adulthood. Following monocular deprivation, visual cortex activity in response to visual stimulation of the deprived eye remained stable in adult control mice, as expected for post-critical period animals. By contrast, visual cortex responses to the deprived eye decreased significantly following monocular deprivation in adult Myrf cKO mice, reminiscent of the plasticity observed in adolescent mice. Furthermore, visual cortex neurons in adult Myrf cKO mice had fewer dendritic spines and a higher level of spine turnover. Finally, monocular deprivation induced spatially coordinated spine size decreases in adult Myrf cKO, but not control, mice. CONCLUSIONS These results demonstrate a critical role for oligodendrocytes in shaping the maturation and stabilization of cortical circuits and support the concept of myelin acting as a brake on neuronal plasticity during development.
Collapse
Affiliation(s)
- Wendy Xin
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| | - Megumi Kaneko
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco
| | - Richard H Roth
- Departments of Neurosurgery and Neurology and Neurological Science, Stanford University
| | - Albert Zhang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| | - Sonia Nocera
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| | - Jun B Ding
- Departments of Neurosurgery and Neurology and Neurological Science, Stanford University
| | - Michael P Stryker
- Department of Physiology, Kavli Institute for Fundamental Neuroscience and Weill Institute for Neurosciences, University of California San Francisco
| | - Jonah R Chan
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco
| |
Collapse
|
8
|
Wang M, Yu X. Experience-dependent structural plasticity of pyramidal neurons in the developing sensory cortices. Curr Opin Neurobiol 2023; 81:102724. [PMID: 37068383 DOI: 10.1016/j.conb.2023.102724] [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: 08/25/2022] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 04/19/2023]
Abstract
Sensory experience regulates the structural and functional wiring of neuronal circuits, during development and throughout adulthood. Here, we review current knowledge of how experience affects structural plasticity of pyramidal neurons in the sensory cortices. We discuss the pros and cons of existing labeling approaches, as well as what structural parameters are most plastic. We further discuss how recent advances in sparse labeling of specific neuronal subtypes, as well as development of techniques that allow fast, high resolution imaging in large fields, would enable future studies to address currently unanswered questions in the field of structural plasticity.
Collapse
Affiliation(s)
- Miao Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and PKU-IDG/McGovern Institute, Peking University, Beijing 100871, China.
| | - Xiang Yu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and PKU-IDG/McGovern Institute, Peking University, Beijing 100871, China; Autism Research Center of Peking University Health Science Center, Beijing 100191, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
9
|
Wang G, Tu Y, Hou P, Li P, Liu L. Regulatory role of the p38 MAPK/ATF2 signaling pathway in visual function and visual cortical plasticity in mice with monocular deprivation. Neurosci Lett 2023:137353. [PMID: 37393009 DOI: 10.1016/j.neulet.2023.137353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/24/2023] [Accepted: 06/15/2023] [Indexed: 07/03/2023]
Abstract
BACKGROUND This study aimed to examine the role of the p38 mitogen-activated protein kinase (MAPK)/ activating transcription factor 2 (ATF2) signaling in visual function impairment and visual cortical plasticity in mice with monocular deprivation (MD). METHODS Visual behavioral tests, including visual water task, visual cliff test, and flash visual evoked potential, were performed on each group. We studied the density of dendritic spines and the synaptic ultrastructure by Golgi staining and transmission electron microscope. We performed Western blot and immunohistochemistry and detected the expression of ATF2, PSD-95, p38 MAPK, and phosphor-p38 MAPK in the left visual cortex. RESULTS In the MD + SB group, the visual acuity in deprived eyes substantially improved, the impairment of visual depth perception was alleviated, and the P wave amplitude and C/I ratio increased. The density of dendritic spines and the numerical density of synapses increased significantly, the width of the synaptic cleft decreased significantly, and the length of the active synaptic zone and the thickness of post-synaptic density (PSD) increased substantially. The protein expression of phosphor-p38 MAPK decreased, whereas that of PSD-95 and ATF2 increased significantly. CONCLUSIONS Inhibiting the phosphorylation of p38 MAPK and negative feedback upregulated ATF2 expression, alleviated damage to visual function, and protected against synaptic plasticity in mice with MD.
Collapse
Affiliation(s)
- Guiqu Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, 37 Guoxue Xiang, Chengdu, 610041, China; Department of Ophthalmology, the Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, 646000, China
| | - Yanqiong Tu
- Department of Ophthalmology, West China Hospital, Sichuan University, 37 Guoxue Xiang, Chengdu, 610041, China
| | - Peixian Hou
- Department of Ophthalmology, West China Hospital, Sichuan University, 37 Guoxue Xiang, Chengdu, 610041, China
| | - Pinxiong Li
- Department of Radiology, the Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou 646000, China
| | - Longqian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, 37 Guoxue Xiang, Chengdu, 610041, China; Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Woo AM, Sontheimer H. Interactions between astrocytes and extracellular matrix structures contribute to neuroinflammation-associated epilepsy pathology. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1198021. [PMID: 39086689 PMCID: PMC11285605 DOI: 10.3389/fmmed.2023.1198021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/31/2023] [Indexed: 08/02/2024]
Abstract
Often considered the "housekeeping" cells of the brain, astrocytes have of late been rising to the forefront of neurodegenerative disorder research. Identified as crucial components of a healthy brain, it is undeniable that when astrocytes are dysfunctional, the entire brain is thrown into disarray. We offer epilepsy as a well-studied neurological disorder in which there is clear evidence of astrocyte contribution to diseases as evidenced across several different disease models, including mouse models of hippocampal sclerosis, trauma associated epilepsy, glioma-associated epilepsy, and beta-1 integrin knockout astrogliosis. In this review we suggest that astrocyte-driven neuroinflammation, which plays a large role in the pathology of epilepsy, is at least partially modulated by interactions with perineuronal nets (PNNs), highly structured formations of the extracellular matrix (ECM). These matrix structures affect synaptic placement, but also intrinsic neuronal properties such as membrane capacitance, as well as ion buffering in their immediate milieu all of which alters neuronal excitability. We propose that the interactions between PNNs and astrocytes contribute to the disease progression of epilepsy vis a vis neuroinflammation. Further investigation and alteration of these interactions to reduce the resultant neuroinflammation may serve as a potential therapeutic target that provides an alternative to the standard anti-seizure medications from which patients are so frequently unable to benefit.
Collapse
Affiliation(s)
- AnnaLin M. Woo
- Neuroscience Graduate Program, Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| | - Harald Sontheimer
- Neuroscience Department, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
11
|
Ashtari M, Cook P, Lipin M, Yu Y, Ying GS, Maguire A, Bennett J, Gee J, Zhang H. Dynamic structural remodeling of the human visual system prompted by bilateral retinal gene therapy. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 4:100089. [PMID: 37397812 PMCID: PMC10313860 DOI: 10.1016/j.crneur.2023.100089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/03/2023] [Accepted: 05/01/2023] [Indexed: 07/04/2023] Open
Abstract
The impact of changes in visual input on neuronal circuitry is complex and much of our knowledge on human brain plasticity of the visual systems comes from animal studies. Reinstating vision in a group of patients with low vision through retinal gene therapy creates a unique opportunity to dynamically study the underlying process responsible for brain plasticity. Historically, increases in the axonal myelination of the visual pathway has been the biomarker for brain plasticity. Here, we demonstrate that to reach the long-term effects of myelination increase, the human brain may undergo demyelination as part of a plasticity process. The maximum change in dendritic arborization of the primary visual cortex and the neurite density along the geniculostriate tracks occurred at three months (3MO) post intervention, in line with timing for the peak changes in postnatal synaptogenesis within the visual cortex reported in animal studies. The maximum change at 3MO for both the gray and white matter significantly correlated with patients' clinical responses to light stimulations called full field sensitivity threshold (FST). Our results shed a new light on the underlying process of brain plasticity by challenging the concept of increase myelination being the hallmark of brain plasticity and instead reinforcing the idea of signal speed optimization as a dynamic process for brain plasticity.
Collapse
Affiliation(s)
- Manzar Ashtari
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania, Philadelphia, PA, 19104, United States
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Philip Cook
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Mikhail Lipin
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Yinxi Yu
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Gui-Shuang Ying
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Albert Maguire
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania, Philadelphia, PA, 19104, United States
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Jean Bennett
- Center for Advanced Retinal and Ocular Therapeutics (CAROT), University of Pennsylvania, Philadelphia, PA, 19104, United States
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - James Gee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, United States
| | - Hui Zhang
- Centre for Medical Image Computing, University College London, London, United Kingdom
| |
Collapse
|
12
|
Henton A, Zhao Y, Tzounopoulos T. A Role for KCNQ Channels on Cell Type-Specific Plasticity in Mouse Auditory Cortex after Peripheral Damage. J Neurosci 2023; 43:2277-2290. [PMID: 36813573 PMCID: PMC10072297 DOI: 10.1523/jneurosci.1070-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Damage to sensory organs triggers compensatory plasticity mechanisms in sensory cortices. These plasticity mechanisms result in restored cortical responses, despite reduced peripheral input, and contribute to the remarkable recovery of perceptual detection thresholds to sensory stimuli. Overall, peripheral damage is associated with a reduction of cortical GABAergic inhibition; however, less is known about changes in intrinsic properties and the underlying biophysical mechanisms. To study these mechanisms, we used a model of noise-induced peripheral damage in male and female mice. We uncovered a rapid, cell type-specific reduction in the intrinsic excitability of parvalbumin-expressing neurons (PVs) in layer (L) 2/3 of auditory cortex. No changes in the intrinsic excitability of either L2/3 somatostatin-expressing or L2/3 principal neurons (PNs) were observed. The decrease in L2/3 PV excitability was observed 1, but not 7, d after noise exposure, and was evidenced by a hyperpolarization of the resting membrane potential, depolarization of the action potential threshold, and reduction in firing frequency in response to depolarizing current. To uncover the underlying biophysical mechanisms, we recorded potassium currents. We found an increase in KCNQ potassium channel activity in L2/3 PVs of auditory cortex 1 d after noise exposure, associated with a hyperpolarizing shift in the minimal voltage activation of KCNQ channels. This increase contributes to the decreased intrinsic excitability of PVs. Our results highlight cell-type- and channel-specific mechanisms of plasticity after noise-induced hearing loss and will aid in understanding the pathologic processes involved in hearing loss and hearing loss-related disorders, such as tinnitus and hyperacusis.SIGNIFICANCE STATEMENT Noise-induced damage to the peripheral auditory system triggers central plasticity that compensates for the reduced peripheral input. The mechanisms of this plasticity are not fully understood. In the auditory cortex, this plasticity likely contributes to the recovery of sound-evoked responses and perceptual hearing thresholds. Importantly, other functional aspects of hearing do not recover, and peripheral damage may also lead to maladaptive plasticity-related disorders, such as tinnitus and hyperacusis. Here, after noise-induced peripheral damage, we highlight a rapid, transient, and cell type-specific reduction in the excitability of layer 2/3 parvalbumin-expressing neurons, which is due, at least in part, to increased KCNQ potassium channel activity. These studies may highlight novel strategies for enhancing perceptual recovery after hearing loss and mitigating hyperacusis and tinnitus.
Collapse
Affiliation(s)
- Amanda Henton
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Center for Neuroscience, University of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Yanjun Zhao
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
13
|
Rasia-Filho AA, Calcagnotto ME, von Bohlen Und Halbach O. Glial Cell Modulation of Dendritic Spine Structure and Synaptic Function. ADVANCES IN NEUROBIOLOGY 2023; 34:255-310. [PMID: 37962798 DOI: 10.1007/978-3-031-36159-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Glia comprise a heterogeneous group of cells involved in the structure and function of the central and peripheral nervous system. Glial cells are found from invertebrates to humans with morphological specializations related to the neural circuits in which they are embedded. Glial cells modulate neuronal functions, brain wiring and myelination, and information processing. For example, astrocytes send processes to the synaptic cleft, actively participate in the metabolism of neurotransmitters, and release gliotransmitters, whose multiple effects depend on the targeting cells. Human astrocytes are larger and more complex than their mice and rats counterparts. Astrocytes and microglia participate in the development and plasticity of neural circuits by modulating dendritic spines. Spines enhance neuronal connectivity, integrate most postsynaptic excitatory potentials, and balance the strength of each input. Not all central synapses are engulfed by astrocytic processes. When that relationship occurs, a different pattern for thin and large spines reflects an activity-dependent remodeling of motile astrocytic processes around presynaptic and postsynaptic elements. Microglia are equally relevant for synaptic processing, and both glial cells modulate the switch of neuroendocrine secretion and behavioral display needed for reproduction. In this chapter, we provide an overview of the structure, function, and plasticity of glial cells and relate them to synaptic maturation and modulation, also involving neurotrophic factors. Together, neurons and glia coordinate synaptic transmission in both normal and abnormal conditions. Neglected over decades, this exciting research field can unravel the complexity of species-specific neural cytoarchitecture as well as the dynamic region-specific functional interactions between diverse neurons and glial subtypes.
Collapse
Affiliation(s)
- Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
14
|
Lépine M, Douceau S, Devienne G, Prunotto P, Lenoir S, Regnauld C, Pouettre E, Piquet J, Lebouvier L, Hommet Y, Maubert E, Agin V, Lambolez B, Cauli B, Ali C, Vivien D. Parvalbumin interneuron-derived tissue-type plasminogen activator shapes perineuronal net structure. BMC Biol 2022; 20:218. [PMID: 36199089 PMCID: PMC9535866 DOI: 10.1186/s12915-022-01419-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 09/27/2022] [Indexed: 11/18/2022] Open
Abstract
Background Perineuronal nets (PNNs) are specialized extracellular matrix structures mainly found around fast-spiking parvalbumin (FS-PV) interneurons. In the adult, their degradation alters FS-PV-driven functions, such as brain plasticity and memory, and altered PNN structures have been found in neurodevelopmental and central nervous system disorders such as Alzheimer’s disease, leading to interest in identifying targets able to modify or participate in PNN metabolism. The serine protease tissue-type plasminogen activator (tPA) plays multifaceted roles in brain pathophysiology. However, its cellular expression profile in the brain remains unclear and a possible role in matrix plasticity through PNN remodeling has never been investigated. Result By combining a GFP reporter approach, immunohistology, electrophysiology, and single-cell RT-PCR, we discovered that cortical FS-PV interneurons are a source of tPA in vivo. We found that mice specifically lacking tPA in FS-PV interneurons display denser PNNs in the somatosensory cortex, suggesting a role for tPA from FS-PV interneurons in PNN remodeling. In vitro analyses in primary cultures of mouse interneurons also showed that tPA converts plasminogen into active plasmin, which in turn, directly degrades aggrecan, a major structural chondroitin sulfate proteoglycan (CSPG) in PNNs. Conclusions We demonstrate that tPA released from FS-PV interneurons in the central nervous system reduces PNN density through CSPG degradation. The discovery of this tPA-dependent PNN remodeling opens interesting insights into the control of brain plasticity. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01419-8.
Collapse
Affiliation(s)
- Matthieu Lépine
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Sara Douceau
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Gabrielle Devienne
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Paul Prunotto
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Sophie Lenoir
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Caroline Regnauld
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Elsa Pouettre
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Juliette Piquet
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Laurent Lebouvier
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Yannick Hommet
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Eric Maubert
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Véronique Agin
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France
| | - Bertrand Lambolez
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Bruno Cauli
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS), Sorbonne Université UM119, CNRS UMR8246, INSERM U1130, 75005, Paris, France
| | - Carine Ali
- Normandie Univ, UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Institut Blood and Brain @ Caen Normandie, Cyceron, Bd Becquerel, BP 5229-14074, 14000, Caen, France.
| | - Denis Vivien
- Department of clinical research, CHU de Caen Normandie, Caen, France
| |
Collapse
|
15
|
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: 66] [Impact Index Per Article: 33.0] [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
|
16
|
Rahn RM, Brier LM, Bice AR, Reisman MD, Dougherty JD, Culver JP. Functional Connectivity of the Developing Mouse Cortex. Cereb Cortex 2022; 32:1755-1768. [PMID: 34498678 PMCID: PMC9016285 DOI: 10.1093/cercor/bhab312] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/14/2022] Open
Abstract
Cross-sectional studies have established a variety of structural, synaptic, and cell physiological changes corresponding to critical periods in cortical development. However, the emergence of functional connectivity (FC) in development has not been fully characterized, and hemodynamic-based measures are vulnerable to any neurovascular coupling changes occurring in parallel. We therefore used optical fluorescence imaging to trace longitudinal calcium FC in the awake, resting-state mouse cortex at 5 developmental timepoints beginning at postnatal day 15 (P15) and ending in early adulthood at P60. Calcium FC displayed coherent functional maps as early as P15, and FC significantly varied in connections between many regions across development, with the developmental trajectory's shape specific to the functional region. Evaluating 325 seed-seed connections, we found that there was a significant increase in FC between P15 and P22 over the majority of the cortex as well as bilateral connectivity and node degree differences in frontal, motor, and retrosplenial cortices after P22. A rebalancing of inter- and intrahemispheric FC and local-distal FC dominance was also observed during development. This longitudinal developmental calcium FC study therefore provides a resource dataset to the field and identifies periods of dynamic change which cross-sectional studies may target for examination of disease states.
Collapse
Affiliation(s)
- Rachel M Rahn
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lindsey M Brier
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Annie R Bice
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew D Reisman
- Department of Physics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph P Culver
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Physics, Washington University in St. Louis, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
17
|
Steinecke A, Bolton MM, Taniguchi H. Neuromodulatory control of inhibitory network arborization in the developing postnatal neocortex. SCIENCE ADVANCES 2022; 8:eabe7192. [PMID: 35263136 PMCID: PMC8906727 DOI: 10.1126/sciadv.abe7192] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
Interregional neuronal communication is pivotal to instructing and adjusting cortical circuit assembly. Subcortical neuromodulatory systems project long-range axons to the cortex and affect cortical processing. However, their roles and signaling mechanisms in cortical wiring remain poorly understood. Here, we explored whether and how the cholinergic system regulates inhibitory axonal ramification of neocortical chandelier cells (ChCs), which control spike generation by innervating axon initial segments of pyramidal neurons. We found that acetylcholine (ACh) signaling through nicotinic ACh receptors (nAChRs) and downstream T-type voltage-dependent calcium (Ca2+) channels cell-autonomously controls axonal arborization in developing ChCs through regulating filopodia initiation. This signaling axis shapes the basal Ca2+ level range in varicosities where filopodia originate. Furthermore, the normal development of ChC axonal arbors requires proper levels of activity in subcortical cholinergic neurons. Thus, the cholinergic system regulates inhibitory network arborization in the developing neocortex and may tune cortical circuit properties depending on early-life experiences.
Collapse
Affiliation(s)
- André Steinecke
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - McLean M. Bolton
- Disorders of Neural Circuit Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Hiroki Taniguchi
- Development and Function of Inhibitory Neural Circuits, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| |
Collapse
|
18
|
Chronic Monocular Deprivation Reveals MMP9-Dependent and -Independent Aspects of Murine Visual System Plasticity. Int J Mol Sci 2022; 23:ijms23052438. [PMID: 35269580 PMCID: PMC8909986 DOI: 10.3390/ijms23052438] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
The deletion of matrix metalloproteinase MMP9 is combined here with chronic monocular deprivation (cMD) to identify the contributions of this proteinase to plasticity in the visual system. Calcium imaging of supragranular neurons of the binocular region of primary visual cortex (V1b) of wild-type mice revealed that cMD initiated at eye opening significantly decreased the strength of deprived-eye visual responses to all stimulus contrasts and spatial frequencies. cMD did not change the selectivity of V1b neurons for the spatial frequency, but orientation selectivity was higher in low spatial frequency-tuned neurons, and orientation and direction selectivity were lower in high spatial frequency-tuned neurons. Constitutive deletion of MMP9 did not impact the stimulus selectivity of V1b neurons, including ocular preference and tuning for spatial frequency, orientation, and direction. However, MMP9-/- mice were completely insensitive to plasticity engaged by cMD, such that the strength of the visual responses evoked by deprived-eye stimulation was maintained across all stimulus contrasts, orientations, directions, and spatial frequencies. Other forms of experience-dependent plasticity, including stimulus selective response potentiation, were normal in MMP9-/- mice. Thus, MMP9 activity is dispensable for many forms of activity-dependent plasticity in the mouse visual system, but is obligatory for the plasticity engaged by cMD.
Collapse
|
19
|
Lupori L, Cornuti S, Mazziotti R, Borghi E, Ottaviano E, Cas MD, Sagona G, Pizzorusso T, Tognini P. The gut microbiota of environmentally enriched mice regulates visual cortical plasticity. Cell Rep 2022; 38:110212. [PMID: 35021093 DOI: 10.1016/j.celrep.2021.110212] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Exposing animals to an enriched environment (EE) has dramatic effects on brain structure, function, and plasticity. The poorly known "EE-derived signals'' mediating the EE effects are thought to be generated within the central nervous system. Here, we shift the focus to the body periphery, revealing that gut microbiota signals are crucial for EE-driven plasticity. Developmental analysis reveals striking differences in intestinal bacteria composition between EE and standard rearing (ST) mice, as well as enhanced levels of short-chain fatty acids (SCFA) in EE mice. Depleting the microbiota of EE mice with antibiotics strongly decreases SCFA and prevents activation of adult ocular dominance plasticity, spine dynamics, and microglia rearrangement. SCFA treatment in ST mice mimics EE induction of ocular dominance plasticity and microglial remodeling. Remarkably, transferring the microbiota of EE mice to ST recipients activates adult ocular dominance plasticity. Thus, experience-dependent changes in gut microbiota regulate brain plasticity.
Collapse
Affiliation(s)
| | - Sara Cornuti
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Raffaele Mazziotti
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
| | | | - Michele Dei Cas
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
| | - Giulia Sagona
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Tommaso Pizzorusso
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA University of Florence, 50100 Florence, Italy; Institute of Neuroscience, National Research Council, 56124 Pisa, Italy
| | - Paola Tognini
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
20
|
Chen CC, Brumberg JC. Sensory Experience as a Regulator of Structural Plasticity in the Developing Whisker-to-Barrel System. Front Cell Neurosci 2022; 15:770453. [PMID: 35002626 PMCID: PMC8739903 DOI: 10.3389/fncel.2021.770453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
Cellular structures provide the physical foundation for the functionality of the nervous system, and their developmental trajectory can be influenced by the characteristics of the external environment that an organism interacts with. Historical and recent works have determined that sensory experiences, particularly during developmental critical periods, are crucial for information processing in the brain, which in turn profoundly influence neuronal and non-neuronal cortical structures that subsequently impact the animals' behavioral and cognitive outputs. In this review, we focus on how altering sensory experience influences normal/healthy development of the central nervous system, particularly focusing on the cerebral cortex using the rodent whisker-to-barrel system as an illustrative model. A better understanding of structural plasticity, encompassing multiple aspects such as neuronal, glial, and extra-cellular domains, provides a more integrative view allowing for a deeper appreciation of how all aspects of the brain work together as a whole.
Collapse
Affiliation(s)
- Chia-Chien Chen
- Department of Psychology, Queens College City University of New York, Flushing, NY, United States.,Department of Neuroscience, Duke Kunshan University, Suzhou, China
| | - Joshua C Brumberg
- Department of Psychology, Queens College City University of New York, Flushing, NY, United States.,The Biology (Neuroscience) and Psychology (Behavioral and Cognitive Neuroscience) PhD Programs, The Graduate Center, The City University of New York, New York, NY, United States
| |
Collapse
|
21
|
Faust TE, Gunner G, Schafer DP. Mechanisms governing activity-dependent synaptic pruning in the developing mammalian CNS. Nat Rev Neurosci 2021; 22:657-673. [PMID: 34545240 PMCID: PMC8541743 DOI: 10.1038/s41583-021-00507-y] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2021] [Indexed: 02/08/2023]
Abstract
Almost 60 years have passed since the initial discovery by Hubel and Wiesel that changes in neuronal activity can elicit developmental rewiring of the central nervous system (CNS). Over this period, we have gained a more comprehensive picture of how both spontaneous neural activity and sensory experience-induced changes in neuronal activity guide CNS circuit development. Here we review activity-dependent synaptic pruning in the mammalian CNS, which we define as the removal of a subset of synapses, while others are maintained, in response to changes in neural activity in the developing nervous system. We discuss the mounting evidence that immune and cell-death molecules are important mechanistic links by which changes in neural activity guide the pruning of specific synapses, emphasizing the role of glial cells in this process. Finally, we discuss how these developmental pruning programmes may go awry in neurodevelopmental disorders of the human CNS, focusing on autism spectrum disorder and schizophrenia. Together, our aim is to give an overview of how the field of activity-dependent pruning research has evolved, led to exciting new questions and guided the identification of new, therapeutically relevant mechanisms that result in aberrant circuit development in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Georgia Gunner
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
22
|
Osborne BF, Beamish SB, Schwarz JM. The effects of early-life immune activation on microglia-mediated neuronal remodeling and the associated ontogeny of hippocampal-dependent learning in juvenile rats. Brain Behav Immun 2021; 96:239-255. [PMID: 34126173 PMCID: PMC8319153 DOI: 10.1016/j.bbi.2021.06.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/11/2021] [Accepted: 06/07/2021] [Indexed: 10/21/2022] Open
Abstract
Many neurodevelopmental disorders and associated learning deficits have been linked to early-life immune activation or ongoing immune dysregulation (Laskaris et al., 2016; O'Connor et al., 2014; Frick et al., 2013). Neuroscientists have begun to understand how the maturation of neural circuits allows for the emergence of cognitive and learning behaviors; yet we know very little about how these developing neural circuits are perturbed by certain events, including risk-factors such as early-life immune activation and immune dysregulation. To answer these questions, we examined the impact of early-life immune activation on the emergence of hippocampal-dependent learning in juvenile male and female rats using a well-characterized hippocampal-dependent learning task and we investigated the corresponding, dynamic multicellular interactions in the hippocampus that may contribute to these learning deficits. We found that even low levels of immune activation can result in hippocampal-depedent learning deficits days later, but only when this activation occurs during a sensitive period of development. The initial immune response and associated cytokine production in the hippocampus resolved within 24 h, several days prior to the observed learning deficit, but notably the initial immune response was followed by altered microglial-neuronal communication and synapse remodeling that changed the structure of hippocampal neurons during this period of juvenile brain development. We conclude that immune activation or dysregulation during a sensitive period of hippocampal development can precipitate the emergence of learning deficits via a multi-cellular process that may be initiated by, but not the direct result of the initial cytokine response. SIGNIFICANCE STATEMENT: Many neurodevelopmental disorders have been linked to early-life immune activation or immune dysregulation; however, very little is known about how dynamic changes in neuroimmune cells mediate the transition from normal brain function to the early stages of cognitive disorders, or how changes in immune signaling are subsequently integrated into developing neuronal networks. The current experiments examined the consequences of immune activation on the cellular and molecular changes that accompany the emergence of learning deficits during a sensitive period of hippocampal development. These findings have the potential to significantly advance our understanding of how early-life immune activation or dysregulation can result in the emergence of cognitive and learning deficits that are the largest source of years lived with disability in humans.
Collapse
Affiliation(s)
- Brittany F. Osborne
- University of Delaware, Department of Psychological & Brain Sciences, 108 Wolf Hall, Newark, DE, 19716, USA
| | - Sarah B. Beamish
- University of Delaware, Department of Psychological & Brain Sciences, 108 Wolf Hall, Newark, DE, 19716, USA
| | - Jaclyn M. Schwarz
- University of Delaware, Department of Psychological & Brain Sciences, 108 Wolf Hall, Newark, DE, 19716, USA
| |
Collapse
|
23
|
Kirchner JH, Gjorgjieva J. Emergence of local and global synaptic organization on cortical dendrites. Nat Commun 2021; 12:4005. [PMID: 34183661 PMCID: PMC8239006 DOI: 10.1038/s41467-021-23557-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023] Open
Abstract
Synaptic inputs on cortical dendrites are organized with remarkable subcellular precision at the micron level. This organization emerges during early postnatal development through patterned spontaneous activity and manifests both locally where nearby synapses are significantly correlated, and globally with distance to the soma. We propose a biophysically motivated synaptic plasticity model to dissect the mechanistic origins of this organization during development and elucidate synaptic clustering of different stimulus features in the adult. Our model captures local clustering of orientation in ferret and receptive field overlap in mouse visual cortex based on the receptive field diameter and the cortical magnification of visual space. Including action potential back-propagation explains branch clustering heterogeneity in the ferret and produces a global retinotopy gradient from soma to dendrite in the mouse. Therefore, by combining activity-dependent synaptic competition and species-specific receptive fields, our framework explains different aspects of synaptic organization regarding stimulus features and spatial scales.
Collapse
Affiliation(s)
- Jan H. Kirchner
- grid.419505.c0000 0004 0491 3878Computation in Neural Circuits Group, Max Planck Institute for Brain Research, Frankfurt, Germany ,grid.6936.a0000000123222966School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Julijana Gjorgjieva
- grid.419505.c0000 0004 0491 3878Computation in Neural Circuits Group, Max Planck Institute for Brain Research, Frankfurt, Germany ,grid.6936.a0000000123222966School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
24
|
Bragg-Gonzalo L, De León Reyes NS, Nieto M. Genetic and activity dependent-mechanisms wiring the cortex: Two sides of the same coin. Semin Cell Dev Biol 2021; 118:24-34. [PMID: 34030948 DOI: 10.1016/j.semcdb.2021.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 01/17/2023]
Abstract
The cerebral cortex is responsible for the higher-order functions of the brain such as planning, cognition, or social behaviour. It provides us with the capacity to interact with and transform our world. The substrates of cortical functions are complex neural circuits that arise during development from the dynamic remodelling and progressive specialization of immature undefined networks. Here, we review the genetic and activity-dependent mechanisms of cortical wiring focussing on the importance of their interaction. Cortical circuits emerge from an initial set of neuronal types that engage in sequential forms of embryonic and postnatal activity. Such activities further complement the cells' genetic programs, increasing neuronal diversity and modifying the electrical properties while promoting selective connectivity. After a temporal window of enhanced plasticity, the main features of mature circuits are established. Failures in these processes can lead to neurodevelopmental disorders whose treatment remains elusive. However, a deeper dissection of cortical wiring will pave the way for innovative therapies.
Collapse
Affiliation(s)
- L Bragg-Gonzalo
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain
| | - N S De León Reyes
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain; Instituto de Neurociencias de Alicante, CSIC-UMH, 03550 San Juan de Alicante, Spain
| | - M Nieto
- Department of Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, (CNB-CSIC) Campus de Cantoblanco, Darwin 3, 28049 Madrid, Spain.
| |
Collapse
|
25
|
Implications of Extended Inhibitory Neuron Development. Int J Mol Sci 2021; 22:ijms22105113. [PMID: 34066025 PMCID: PMC8150951 DOI: 10.3390/ijms22105113] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/23/2022] Open
Abstract
A prolonged developmental timeline for GABA (γ-aminobutyric acid)-expressing inhibitory neurons (GABAergic interneurons) is an amplified trait in larger, gyrencephalic animals. In several species, the generation, migration, and maturation of interneurons take place over several months, in some cases persisting after birth. The late integration of GABAergic interneurons occurs in a region-specific pattern, especially during the early postnatal period. These changes can contribute to the formation of functional connectivity and plasticity, especially in the cortical regions responsible for higher cognitive tasks. In this review, we discuss GABAergic interneuron development in the late gestational and postnatal forebrain. We propose the protracted development of interneurons at each stage (neurogenesis, neuronal migration, and network integration), as a mechanism for increased complexity and cognitive flexibility in larger, gyrencephalic brains. This developmental feature of interneurons also provides an avenue for environmental influences to shape neural circuit formation.
Collapse
|
26
|
Yusifov R, Tippmann A, Staiger JF, Schlüter OM, Löwel S. Spine dynamics of PSD-95-deficient neurons in the visual cortex link silent synapses to structural cortical plasticity. Proc Natl Acad Sci U S A 2021; 118:e2022701118. [PMID: 33649238 PMCID: PMC7958355 DOI: 10.1073/pnas.2022701118] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Critical periods (CPs) are time windows of heightened brain plasticity during which experience refines synaptic connections to achieve mature functionality. At glutamatergic synapses on dendritic spines of principal cortical neurons, the maturation is largely governed by postsynaptic density protein-95 (PSD-95)-dependent synaptic incorporation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors into nascent AMPA-receptor silent synapses. Consequently, in mouse primary visual cortex (V1), impaired silent synapse maturation in PSD-95-deficient neurons prevents the closure of the CP for juvenile ocular dominance plasticity (jODP). A structural hallmark of jODP is increased spine elimination, induced by brief monocular deprivation (MD). However, it is unknown whether impaired silent synapse maturation facilitates spine elimination and also preserves juvenile structural plasticity. Using two-photon microscopy, we assessed spine dynamics in apical dendrites of layer 2/3 pyramidal neurons (PNs) in binocular V1 during ODP in awake adult mice. Under basal conditions, spine formation and elimination ratios were similar between PSD-95 knockout (KO) and wild-type (WT) mice. However, a brief MD affected spine dynamics only in KO mice, where MD doubled spine elimination, primarily affecting newly formed spines, and caused a net reduction in spine density similar to what has been observed during jODP in WT mice. A similar increase in spine elimination after MD occurred if PSD-95 was knocked down in single PNs of layer 2/3. Thus, structural plasticity is dictated cell autonomously by PSD-95 in vivo in awake mice. Loss of PSD-95 preserves hallmark features of spine dynamics in jODP into adulthood, revealing a functional link of PSD-95 for experience-dependent synapse maturation and stabilization during CPs.
Collapse
Affiliation(s)
- Rashad Yusifov
- Department of Systems Neuroscience, Johann Friedrich Blumenbach Institut für Zoologie und Anthropologie, Universität Göttingen, D-37075 Göttingen, Germany
- Collaborative Research Center 889, Universität Göttingen, D-37075 Göttingen, Germany
- Campus Institute for Dynamics of Biological Networks, Universität Göttingen, D-37075 Göttingen, Germany
| | - Anja Tippmann
- Department of Systems Neuroscience, Johann Friedrich Blumenbach Institut für Zoologie und Anthropologie, Universität Göttingen, D-37075 Göttingen, Germany
- Campus Institute for Dynamics of Biological Networks, Universität Göttingen, D-37075 Göttingen, Germany
| | - Jochen F Staiger
- Collaborative Research Center 889, Universität Göttingen, D-37075 Göttingen, Germany
- Institute for Neuroanatomy, University Medical Center, Universität Göttingen, D-37075 Göttingen, Germany
| | - Oliver M Schlüter
- Collaborative Research Center 889, Universität Göttingen, D-37075 Göttingen, Germany
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Universität Göttingen, D-37075 Göttingen, Germany
| | - Siegrid Löwel
- Department of Systems Neuroscience, Johann Friedrich Blumenbach Institut für Zoologie und Anthropologie, Universität Göttingen, D-37075 Göttingen, Germany;
- Collaborative Research Center 889, Universität Göttingen, D-37075 Göttingen, Germany
- Campus Institute for Dynamics of Biological Networks, Universität Göttingen, D-37075 Göttingen, Germany
| |
Collapse
|
27
|
Gan X, Chopp M, Xin H, Wang F, Golembieski W, Lu M, He L, Liu Z. Targeted tPA overexpression in denervated spinal motor neurons promotes stroke recovery in mice. J Cereb Blood Flow Metab 2021; 41:92-104. [PMID: 31987011 PMCID: PMC7747163 DOI: 10.1177/0271678x20901686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Our previous studies demonstrated that axonal remodeling of the corticospinal tract (CST) contributes to neurological recovery after stroke in rodents. The present study employed a novel non-invasive peripheral approach, to over-express tPA in denervated spinal motor neurons via recombinant adeno-associated virus (AAV) intramuscular injection in transgenic mice subjected to permanent middle cerebral artery occlusion (MCAo), in which the CST axons are specifically and completely labeled with yellow fluorescent protein (YFP). One day after surgery, mice were randomly selected to receive saline, AAV5-RFP, or tPA (1 × 1010 viral particles) injected into the stroke-impaired forelimb muscles (n = 10/group). Functional deficits and recovery were monitored with foot-fault and single pellet reaching tests. At day 28 after MCAo, mice received intramuscular injection of PRV-614-mRFP (1.52 × 107 pfu) as above, and were euthanized four days later. Compared with saline or AAV-RFP-treated mice, AAV-tPA significantly enhanced behavioral recovery (p < 0.01, both tests), as well as increased CST axonal density in the denervated gray matter of the cervical cord (p < 0.001), and RFP-positive pyramidal neurons in both ipsilesional and contralesional cortices (p < 0.001). Behavioral outcomes were significantly correlated to neural remodeling (p < 0.05). Our results provide a fundamental basis for the development of therapeutic approaches aimed at promoting corticospinal innervation for stroke treatment.
Collapse
Affiliation(s)
- Xinling Gan
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China.,Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.,Department of Physics, Oakland University, Rochester, MI, USA
| | - Hongqi Xin
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Fengjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | - Mei Lu
- Biostatistics and Research Epidemiology, Henry Ford Health System, Detroit, MI, USA
| | - Li He
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
28
|
Wada Y, Maekawa M, Ohnishi T, Balan S, Matsuoka S, Iwamoto K, Iwayama Y, Ohba H, Watanabe A, Hisano Y, Nozaki Y, Toyota T, Shimogori T, Itokawa M, Kobayashi T, Yoshikawa T. Peroxisome proliferator-activated receptor α as a novel therapeutic target for schizophrenia. EBioMedicine 2020; 62:103130. [PMID: 33279456 PMCID: PMC7728824 DOI: 10.1016/j.ebiom.2020.103130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The pathophysiology of schizophrenia, a major psychiatric disorder, remains elusive. In this study, the role of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor (RXR) families, belonging to the ligand-activated nuclear receptor superfamily, in schizophrenia, was analyzed. METHODS The PPAR/RXR family genes were screened by exploiting molecular inversion probe (MIP)-based targeted next-generation sequencing (NGS) using the samples of 1,200 Japanese patients with schizophrenia. The results were compared with the whole-genome sequencing databases of the Japanese cohort (ToMMo) and the gnomAD. To reveal the relationship between PPAR/RXR dysfunction and schizophrenia, Ppara KO mice and fenofibrate (a clinically used PPARα agonist)-administered mice were assessed by performing behavioral, histological, and RNA-seq analyses. FINDINGS Our findings indicate that c.209-2delA, His117Gln, Arg141Cys, and Arg226Trp of the PPARA gene are risk variants for schizophrenia. The c.209-2delA variant generated a premature termination codon. The three missense variants significantly decreased the activity of PPARα as a transcription factor in vitro. The Ppara KO mice exhibited schizophrenia-relevant phenotypes, including behavioral deficits and impaired synaptogenesis in the cerebral cortex. Oral administration of fenofibrate alleviated spine pathology induced by phencyclidine, an N-methyl-d-aspartate (NMDA) receptor antagonist. Furthermore, pre-treatment with fenofibrate suppressed the sensitivity of mice to another NMDA receptor antagonist, MK-801. RNA-seq analysis revealed that PPARα regulates the expression of synaptogenesis signaling pathway-related genes. INTERPRETATION The findings of this study indicate that the mechanisms underlying schizophrenia pathogenesis involve PPARα-regulated transcriptional machinery and modulation of synapse physiology. Hence, PPARα can serve as a novel therapeutic target for schizophrenia.
Collapse
Affiliation(s)
- Yuina Wada
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan.
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | | | - Kazuya Iwamoto
- Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Hisako Ohba
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Akiko Watanabe
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Yasuko Hisano
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Yayoi Nozaki
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan
| | - Tomomi Shimogori
- Laboratory for Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Masanari Itokawa
- Center for Medical Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Tetsuyuki Kobayashi
- Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan; Department of Biological Science, Graduate School of Humanities and Science, Ochanomizu University, Tokyo 112-8610, Japan.
| |
Collapse
|
29
|
Kasamatsu T, Imamura K. Ocular dominance plasticity: Molecular mechanisms revisited. J Comp Neurol 2020; 528:3039-3074. [PMID: 32737874 DOI: 10.1002/cne.25001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/14/2022]
Abstract
Ocular dominance plasticity (ODP) is a type of cortical plasticity operating in visual cortex of mammals that are endowed with binocular vision based on the competition-driven disparity. Earlier, a molecular mechanism was proposed that catecholamines play an important role in the maintenance of ODP in kittens. Having survived the initial test, the hypothesis was further advanced to identify noradrenaline (NA) as a key factor that regulates ODP in the immature cortex. Later, the ODP-promoting effect of NA is extended to the adult with age-related limitations. Following the enhanced NA availability, the chain events downstream lead to the β-adrenoreceptor-induced cAMP accumulation, which in turn activates the protein kinase A. Eventually, the protein kinase translocates to the cell nucleus to activate cAMP responsive element binding protein (CREB). CREB is a cellular transcription factor that controls the transcription of various genes, underpinning neuronal plasticity and long-term memory. In the advent of molecular genetics in that various types of new tools have become available with relative ease, ODP research has lightly adopted in the rodent model the original concepts and methodologies. Here, after briefly tracing the strategic maturation of our quest, the review moves to the later development of the field, with the emphasis placed around the following issues: (a) Are we testing ODP per se? (b) What does monocular deprivation deprive of the immature cortex? (c) The critical importance of binocular competition, (d) What is the adult plasticity? (e) Excitation-Inhibition balance in local circuits, and (f) Species differences in the animal models.
Collapse
Affiliation(s)
- Takuji Kasamatsu
- Smith-Kettlewell Eye Research Institute, San Francisco, California, USA
| | - Kazuyuki Imamura
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi-shi, Gunma, Japan
| |
Collapse
|
30
|
Nakanishi H, Ni J, Nonaka S, Hayashi Y. Microglial circadian clock regulation of microglial structural complexity, dendritic spine density and inflammatory response. Neurochem Int 2020; 142:104905. [PMID: 33217515 DOI: 10.1016/j.neuint.2020.104905] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
Cortical microglia exhibit a ramified shape during sleep, while they have a hyper-ramified shape during wakefulness, which is characterized by their longer processes with increased branching points. The microglial molecular circadian clock regulates expressions of both cathepsin S (CatS) and P2Y12 receptors in the brain with a peak at zeitgeber time 14 (2 h after beginning of the dark phase). We postulated that these two microglia-specific molecules contribute to diurnal alterations of microglial shapes and neuronal activities in the cerebral cortex. During wakefulness, CatS secreted from cortical microglia may be involved in P2Y12 receptor-dependent process extension. Secreted CatS subsequently degrades the perineuronal nets, initiating the downscaling of both spine density and synaptic strength of cortical neurons toward the beginning of sleep. The downscaling of both spine density and synaptic strength of cortical neurons during sleep could improve signal-to-noise, which would benefit memory consolidation, or allow for new learning to occur during subsequent waking. Furthermore, disruption of CatS induces the sleep disturbance and impaired social interaction in mice. Moreover, the microglial clock system disruption may also play a role in the early pathogenesis of Alzheimer's disease. The reduced expression of BMAL1 in cortical microglia caused by oligomeric amyloid β may induce the increased presence of inflammatory phenotype through a reduction in RORα, which in turn reduced IκBα and enhanced NF-κB activation. These observations suggest that the microglial clock system disruption contribute to pathogeneses of sleep disturbance, impaired social interaction and cognitive impairment. Therefore, the growing understanding of the microglial circadian molecular clock might aid in the development of novel pharmacological interventions against both neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan.
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Saori Nonaka
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
31
|
Baroncelli L, Lunghi C. Neuroplasticity of the visual cortex: in sickness and in health. Exp Neurol 2020; 335:113515. [PMID: 33132181 DOI: 10.1016/j.expneurol.2020.113515] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 01/18/2023]
Abstract
Brain plasticity refers to the ability of synaptic connections to adapt their function and structure in response to experience, including environmental changes, sensory deprivation and injuries. Plasticity is a distinctive, but not exclusive, property of the developing nervous system. This review introduces the concept of neuroplasticity and describes classic paradigms to illustrate cellular and molecular mechanisms underlying synapse modifiability. Then, we summarize a growing number of studies showing that the adult cerebral cortex retains a significant degree of plasticity highlighting how the identification of strategies to enhance the plastic potential of the adult brain could pave the way for the development of novel therapeutic approaches aimed at treating amblyopia and other neurodevelopmental disorders. Finally, we analyze how the visual system adjusts to neurodegenerative conditions leading to blindness and we discuss the crucial role of spared plasticity in the visual system for sight recovery.
Collapse
Affiliation(s)
- Laura Baroncelli
- Institute of Neuroscience, National Research Council (CNR), I-56124 Pisa, Italy; Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, I-56128 Pisa, Italy.
| | - Claudia Lunghi
- Laboratoire des systèmes perceptifs, Département d'études cognitives, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| |
Collapse
|
32
|
Napoli D, Lupori L, Mazziotti R, Sagona G, Bagnoli S, Samad M, Sacramento EK, Kirkpartick J, Putignano E, Chen S, Terzibasi Tozzini E, Tognini P, Baldi P, Kwok JC, Cellerino A, Pizzorusso T. MiR-29 coordinates age-dependent plasticity brakes in the adult visual cortex. EMBO Rep 2020; 21:e50431. [PMID: 33026181 DOI: 10.15252/embr.202050431] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Visual cortical circuits show profound plasticity during early life and are later stabilized by molecular "brakes" limiting excessive rewiring beyond a critical period. The mechanisms coordinating the expression of these factors during the transition from development to adulthood remain unknown. We found that miR-29a expression in the visual cortex dramatically increases with age, but it is not experience-dependent. Precocious high levels of miR-29a blocked ocular dominance plasticity and caused an early appearance of perineuronal nets. Conversely, inhibition of miR-29a in adult mice using LNA antagomirs activated ocular dominance plasticity, reduced perineuronal nets, and restored their juvenile chemical composition. Activated adult plasticity had the typical functional and proteomic signature of critical period plasticity. Transcriptomic and proteomic studies indicated that miR-29a manipulation regulates the expression of plasticity brakes in specific cortical circuits. These data indicate that miR-29a is a regulator of the plasticity brakes promoting age-dependent stabilization of visual cortical connections.
Collapse
Affiliation(s)
- Debora Napoli
- BIO@SNS Lab, Scuola Normale Superiore, Pisa, Italy.,Institute of Neuroscience, National Research Council, Pisa, Italy
| | | | - Raffaele Mazziotti
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA University of Florence, Florence, Italy
| | - Giulia Sagona
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA University of Florence, Florence, Italy.,Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
| | - Sara Bagnoli
- BIO@SNS Lab, Scuola Normale Superiore, Pisa, Italy
| | - Muntaha Samad
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | | | - Joanna Kirkpartick
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Elena Putignano
- Institute of Neuroscience, National Research Council, Pisa, Italy
| | - Siwei Chen
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | | | - Paola Tognini
- BIO@SNS Lab, Scuola Normale Superiore, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Pierre Baldi
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California, Irvine, CA, USA
| | - Jessica Cf Kwok
- School of Biomedical Sciences, University of Leeds, Leeds, UK.,Institute of Experimental Medicine, Czech Academy of Science, Prague, Czech Republic
| | - Alessandro Cellerino
- BIO@SNS Lab, Scuola Normale Superiore, Pisa, Italy.,Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Tommaso Pizzorusso
- BIO@SNS Lab, Scuola Normale Superiore, Pisa, Italy.,Institute of Neuroscience, National Research Council, Pisa, Italy.,Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA University of Florence, Florence, Italy
| |
Collapse
|
33
|
Tanaka S, Miyashita M, Wakabayashi N, O'Hashi K, Tani T, Ribot J. Development and Reorganization of Orientation Representation in the Cat Visual Cortex: Experience-Dependent Synaptic Rewiring in Early Life. Front Neuroinform 2020; 14:41. [PMID: 32973480 PMCID: PMC7468406 DOI: 10.3389/fninf.2020.00041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/28/2020] [Indexed: 11/13/2022] Open
Abstract
To date, numerous mathematical models have been proposed on the basis of some types of Hebbian synaptic learning to account for the activity-dependent development of orientation maps as well as neuronal orientation selectivity. These models successfully reproduced orientation map-like spatial patterns. Nevertheless, we still have questions: (1) How does synaptic rewiring occur in the visual cortex during the formation of orderly orientation maps in early life? (2) How does visual experience contribute to the maturation of orientation selectivity of visual cortical neurons and reorganize orientation maps? (3) How does the sensitive period for orientation plasticity end? In this study, we performed animal experiments and mathematical modeling to understand the mechanisms underlying synaptic rewiring for experience-dependent formation and reorganization of orientation maps. At first, we visualized orientation maps from the intrinsic signal optical imaging in area 17 of kittens reared under single-orientation exposure through cylindrical-lens-fitted goggles. The experiments revealed that the degree of expansion of cortical domains representing the experienced orientation depends on the age at which the single-orientation exposure starts. As a result, we obtained the sensitive period profile for orientation plasticity. Next, we refined our previously proposed mathematical model for the activity-dependent self-organization of thalamo-cortical inputs on the assumption that rewiring is caused by the competitive interactions among transient synaptic contacts on the same dendritic spine. Although various kinds of molecules have been reported to be involved in such interactions, we attempt to build a mathematical model to describe synaptic rewiring focusing on brain-derived neurotrophic factor (BDNF) and its related molecules. Performing computer simulations based on the refined model, we successfully reproduced orientation maps reorganized in kittens reared under single-orientation exposure as well as normal visual experience. We also reproduced the experimentally obtained sensitive period profile for orientation plasticity. The excellent agreement between experimental observations and theoretical reproductions suggests that the BDNF-induced competitive interaction among synaptic contacts from different axons on the same spine is an important factor for the experience-dependent formation and reorganization of orientation selectivity and orientation maps.
Collapse
Affiliation(s)
- Shigeru Tanaka
- Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, Chofu, Japan
| | - Masanobu Miyashita
- Department of Control and Computer Engineering, National Institute of Technology, Numazu College, Numazu, Japan
| | - Nodoka Wakabayashi
- Power Plant Engineering, Engineering & Maintenance Center, All Nippon Airways Co., Ltd., Tokyo, Japan
| | - Kazunori O'Hashi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Toshiki Tani
- Laboratory for Molecular Analysis of Higher Brain Functions, RIKEN Center for Brain Science, Wako, Japan
| | - Jérôme Ribot
- Centre for Interdisciplinary Research in Biology, Collège de France, Paris, France
| |
Collapse
|
34
|
REM sleep promotes experience-dependent dendritic spine elimination in the mouse cortex. Nat Commun 2020; 11:4819. [PMID: 32968048 PMCID: PMC7511313 DOI: 10.1038/s41467-020-18592-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 08/26/2020] [Indexed: 01/10/2023] Open
Abstract
In many parts of the nervous system, experience-dependent refinement of neuronal circuits predominantly involves synapse elimination. The role of sleep in this process remains unknown. We investigated the role of sleep in experience-dependent dendritic spine elimination of layer 5 pyramidal neurons in the visual (V1) and frontal association cortex (FrA) of 1-month-old mice. We found that monocular deprivation (MD) or auditory-cued fear conditioning (FC) caused rapid spine elimination in V1 or FrA, respectively. MD- or FC-induced spine elimination was significantly reduced after total sleep or REM sleep deprivation. Total sleep or REM sleep deprivation also prevented MD- and FC-induced reduction of neuronal activity in response to visual or conditioned auditory stimuli. Furthermore, dendritic calcium spikes increased substantially during REM sleep, and the blockade of these calcium spikes prevented MD- and FC-induced spine elimination. These findings reveal an important role of REM sleep in experience-dependent synapse elimination and neuronal activity reduction. Sleep plays an important role in learning and memory. Here the authors show that experience dependent elimination of spines is attenuated by REM sleep deprivation.
Collapse
|
35
|
Reh RK, Dias BG, Nelson CA, Kaufer D, Werker JF, Kolb B, Levine JD, Hensch TK. Critical period regulation across multiple timescales. Proc Natl Acad Sci U S A 2020; 117:23242-23251. [PMID: 32503914 PMCID: PMC7519216 DOI: 10.1073/pnas.1820836117] [Citation(s) in RCA: 230] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Brain plasticity is dynamically regulated across the life span, peaking during windows of early life. Typically assessed in the physiological range of milliseconds (real time), these trajectories are also influenced on the longer timescales of developmental time (nurture) and evolutionary time (nature), which shape neural architectures that support plasticity. Properly sequenced critical periods of circuit refinement build up complex cognitive functions, such as language, from more primary modalities. Here, we consider recent progress in the biological basis of critical periods as a unifying rubric for understanding plasticity across multiple timescales. Notably, the maturation of parvalbumin-positive (PV) inhibitory neurons is pivotal. These fast-spiking cells generate gamma oscillations associated with critical period plasticity, are sensitive to circadian gene manipulation, emerge at different rates across brain regions, acquire perineuronal nets with age, and may be influenced by epigenetic factors over generations. These features provide further novel insight into the impact of early adversity and neurodevelopmental risk factors for mental disorders.
Collapse
Affiliation(s)
- Rebecca K Reh
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Brian G Dias
- Division of Behavioral Neuroscience and Psychiatric Disorders, Yerkes National Primate Research Center, Atlanta, GA 30322
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30329
| | - Charles A Nelson
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Graduate School of Education, Harvard University, Cambridge, MA 02138
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720
- Department of Integrative Biology, University of California, Berkeley, CA 94720
| | - Janet F Werker
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Bryan Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Joel D Levine
- Department of Biology, University of Toronto at Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Takao K Hensch
- Boston Children's Hospital, Harvard Medical School, Boston, MA 02115;
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, Cambridge, MA 02138
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo 113-0033, Japan
| |
Collapse
|
36
|
Maekawa M, Ohnishi T, Toyoshima M, Shimamoto-Mitsuyama C, Hamazaki K, Balan S, Wada Y, Esaki K, Takagai S, Tsuchiya KJ, Nakamura K, Iwata Y, Nara T, Iwayama Y, Toyota T, Nozaki Y, Ohba H, Watanabe A, Hisano Y, Matsuoka S, Tsujii M, Mori N, Matsuzaki H, Yoshikawa T. A potential role of fatty acid binding protein 4 in the pathophysiology of autism spectrum disorder. Brain Commun 2020; 2:fcaa145. [PMID: 33225276 PMCID: PMC7667725 DOI: 10.1093/braincomms/fcaa145] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by difficulties in social communication and interaction, as well as repetitive and characteristic patterns of behaviour. Although the pathogenesis of autism spectrum disorder is unknown, being overweight or obesity during infancy and low weight at birth are known as risks, suggesting a metabolic aspect. In this study, we investigated adipose tissue development as a pathophysiological factor of autism spectrum disorder by examining the serum levels of adipokines and other metabolic markers in autism spectrum disorder children (n = 123) and typically developing children (n = 92) at 4–12 years of age. Among multiple measures exhibiting age-dependent trajectories, the leptin levels displayed different trajectory patterns between autism spectrum disorder and typically developing children, supporting an adipose tissue-dependent mechanism of autism spectrum disorder. Of particular interest, the levels of fatty acid binding protein 4 (FABP4) were significantly lower in autism spectrum disorder children than in typically developing subjects, at preschool age (4–6 years old: n = 21 for autism spectrum disorder and n = 26 for typically developing). The receiver operating characteristic curve analysis discriminated autism spectrum disorder children from typically developing children with a sensitivity of 94.4% and a specificity of 75.0%. We re-sequenced the exons of the FABP4 gene in a Japanese cohort comprising 659 autism spectrum disorder and 1000 control samples, and identified two rare functional variants in the autism spectrum disorder group. The Trp98Stop, one of the two variants, was transmitted to the proband from his mother with a history of depression. The disruption of the Fabp4 gene in mice evoked autism spectrum disorder-like behavioural phenotypes and increased spine density on apical dendrites of pyramidal neurons, which has been observed in the postmortem brains of autism spectrum disorder subjects. The Fabp4 knockout mice had an altered fatty acid composition in the cortex. Collectively, these results suggest that an ‘adipo-brain axis’ may underlie the pathophysiology of autism spectrum disorder, with FABP4 as a potential molecule for use as a biomarker.
Collapse
Affiliation(s)
- Motoko Maekawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
- Correspondence to: Motoko Maekawa, Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako-city, Saitama 351-0198, Japan. E-mail:
| | - Tetsuo Ohnishi
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Manabu Toyoshima
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | | | - Kei Hamazaki
- Department of Public Health, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Shabeesh Balan
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Yuina Wada
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Kayoko Esaki
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Shu Takagai
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Kenji J Tsuchiya
- Research Center for Child Mental Development, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Kazuhiko Nakamura
- Department of Psychiatry, Hirosaki University School of Medicine, Aomori, Japan
| | - Yasuhide Iwata
- Department of Psychiatry and Neurology, Fukude Nishi Hospital, Shizuoka, Japan
| | - Takahiro Nara
- Department of Rehabilitation, Miyagi Children's Hospital, Miyagi, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Yayoi Nozaki
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Hisako Ohba
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Akiko Watanabe
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Yasuko Hisano
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
| | - Shigeru Matsuoka
- Department of Clinical Pharmacology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masatsugu Tsujii
- School of Contemporary Sociology, Chukyo University, Aichi, Japan
| | - Norio Mori
- Department of Psychiatry and Neurology, Fukude Nishi Hospital, Shizuoka, Japan
| | - Hideo Matsuzaki
- Research Center for Child Mental Development, University of Fukui, Fukui, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Saitama, Japan
- Correspondence may also be addressed to: Takeo Yoshikawa. E-mail:
| |
Collapse
|
37
|
Frantz MG, Crouse EC, Sokhadze G, Ikrar T, Stephany CÉ, Nguyen C, Xu X, McGee AW. Layer 4 Gates Plasticity in Visual Cortex Independent of a Canonical Microcircuit. Curr Biol 2020; 30:2962-2973.e5. [PMID: 32589913 PMCID: PMC7919382 DOI: 10.1016/j.cub.2020.05.067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/23/2020] [Accepted: 05/20/2020] [Indexed: 01/09/2023]
Abstract
Disrupting binocular vision during a developmental critical period can yield enduring changes to ocular dominance (OD) in primary visual cortex (V1). Here we investigated how this experience-dependent plasticity is coordinated within the laminar circuitry of V1 by deleting separately in each cortical layer (L) a gene required to close the critical period, nogo-66 receptor (ngr1). Deleting ngr1 in excitatory neurons in L4, but not in L2/3, L5, or L6, prevented closure of the critical period, and adult mice remained sensitive to brief monocular deprivation. Intracortical disinhibition, but not thalamocortical disinhibition, accompanied this OD plasticity. Both juvenile wild-type mice and adult mice lacking ngr1 in L4 displayed OD plasticity that advanced more rapidly L4 than L2/3 or L5. Interestingly, blocking OD plasticity in L2/3 with the drug AM-251 did not impair OD plasticity in L5. We propose that L4 restricts disinhibition and gates OD plasticity independent of a canonical cortical microcircuit.
Collapse
Affiliation(s)
- Michael G Frantz
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Emily C Crouse
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Guela Sokhadze
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Taruna Ikrar
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Céleste-Élise Stephany
- Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA
| | - Collins Nguyen
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA.
| | - Aaron W McGee
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA; Developmental Neuroscience Program, Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027, USA.
| |
Collapse
|
38
|
Zhan Z, Wu Y, Liu Z, Quan Y, Li D, Huang Y, Yang S, Wu K, Huang L, Yu M. Reduced Dendritic Spines in the Visual Cortex Contralateral to the Optic Nerve Crush Eye in Adult Mice. Invest Ophthalmol Vis Sci 2020; 61:55. [PMID: 32866269 PMCID: PMC7463183 DOI: 10.1167/iovs.61.10.55] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/31/2020] [Indexed: 01/19/2023] Open
Abstract
Purpose To determine alteration of dendritic spines and associated changes in the primary visual cortex (V1 region) related to unilateral optic nerve crush (ONC) in adult mice. Methods Adult unilateral ONC mice were established. Retinal nerve fiber layer (RNFL) thickness was measured by spectral-domain optical coherence tomography. Visual function was estimated by flash visual evoked potentials (FVEPs). Dendritic spines were observed in the V1 region contralateral to the ONC eye by two-photon imaging in vivo. The neurons, reactive astrocytes, oligodendrocytes, and activated microglia were assessed by NeuN, glial fibrillary acidic protein, CNPase, and CD68 in immunohistochemistry, respectively. Tropomyosin receptor kinase B (TrkB) and the markers in TrkB trafficking were estimated using western blotting and co-immunoprecipitation. Transmission electron microscopy and western blotting were used to evaluate autophagy. Results The amplitude and latency of FVEPs were decreased and delayed at 3 days, 1 week, 2 weeks, and 4 weeks after ONC, and RNFL thickness was decreased at 2 and 4 weeks after ONC. Dendritic spines were reduced in the V1 region contralateral to the ONC eye at 2, 3, and 4 weeks after ONC, with an unchanged number of neurons. Reactive astrocyte staining was increased at 2 and 4 weeks after ONC, but oligodendrocyte and activated microglia staining remained unchanged. TrkB was reduced with changes in the major trafficking proteins, and enhanced autophagy was observed in the V1 region contralateral to the ONC eye. Conclusions Dendritic spines were reduced in the V1 region contralateral to the ONC eye in adult mice. Reactive astrocytes and decreased TrkB may be associated with the reduced dendritic spines.
Collapse
Affiliation(s)
- Zongyi Zhan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yali Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zitian Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yadan Quan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Deling Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiru Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shana Yang
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kaili Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lianyan Huang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Minbin Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
Takeuchi C, Ishikawa M, Sawano T, Shin Y, Mizuta N, Hasegawa S, Tanaka R, Tsuboi Y, Nakatani J, Sugiura H, Yamagata K, Tanaka H. Dendritic Spine Density is Increased in Arcadlin-deleted Mouse Hippocampus. Neuroscience 2020; 442:296-310. [DOI: 10.1016/j.neuroscience.2020.06.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/12/2020] [Accepted: 06/26/2020] [Indexed: 11/28/2022]
|
40
|
Laliberté G, Othman R, Vaucher E. Mesoscopic Mapping of Stimulus-Selective Response Plasticity in the Visual Pathways Modulated by the Cholinergic System. Front Neural Circuits 2020; 14:38. [PMID: 32719589 PMCID: PMC7350895 DOI: 10.3389/fncir.2020.00038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/22/2020] [Indexed: 11/13/2022] Open
Abstract
The cholinergic potentiation of visual conditioning enhances visual acuity and discrimination of the trained stimulus. To determine if this also induces long-term plastic changes on cortical maps and connectivity in the visual cortex and higher associative areas, mesoscopic calcium imaging was performed in head-fixed awake GCaMP6s adult mice before and after conditioning. The conditioned stimulus (0.03 cpd, 30°, 100% contrast, 1 Hz-drifting gratings) was presented 10 min daily for a week. Saline or Donepezil (DPZ, 0.3 mg/kg, s.c.), a cholinesterase inhibitor that potentiates cholinergic transmission, were injected prior to each conditioning session and compared to a sham-conditioned group. Cortical maps of resting state and evoked response to the monocular presentation of conditioned or non-conditioned stimulus (30°, 50 and 75% contrast; 90°, 50, 75, and 100% contrast) were established. Amplitude, duration, and latency of the peak response, as well as size of activation were measured in the primary visual cortex (V1), secondary visual areas (AL, A, AM, PM, LM, RL), retrosplenial cortex (RSC), and higher cortical areas. Visual stimulation increased calcium signaling in all primary and secondary visual areas, the RSC, but no other cortices. There were no significant effects of sham-conditioning or conditioning alone, but DPZ treatment during conditioning significantly decreased the integrated neuronal activity of superficial layers evoked by the conditioned stimulus in V1, AL, PM, and LM. The activity of downstream cortical areas was not changed. The size of the activated area was decreased in V1 and PM, and the signal-to-noise ratio was decreased in AL and PM. Interestingly, signal correlation was seen only between V1, the ventral visual pathway, and the RSC, and was decreased by DPZ administration. The resting state activity was slightly correlated and rarely affected by treatments, except between binocular and monocular V1 in both hemispheres. In conclusion, cholinergic potentiation of visual conditioning induced change in visual processing in the superficial cortical layers. This effect might be a key mechanism in the establishment of the fine cortical tuning in response to the conditioned visual stimulus.
Collapse
Affiliation(s)
- Guillaume Laliberté
- Laboratoire de Neurobiologie de la Cognition Visuelle, École d'Optométrie, Université de Montréal, Montréal, QC, Canada
| | - Rahmeh Othman
- Laboratoire de Neurobiologie de la Cognition Visuelle, École d'Optométrie, Université de Montréal, Montréal, QC, Canada.,Départment de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Elvire Vaucher
- Laboratoire de Neurobiologie de la Cognition Visuelle, École d'Optométrie, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
41
|
MEF2C and HDAC5 regulate Egr1 and Arc genes to increase dendritic spine density and complexity in early enriched environment. Neuronal Signal 2020; 4:NS20190147. [PMID: 32714604 PMCID: PMC7378308 DOI: 10.1042/ns20190147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 01/16/2023] Open
Abstract
We investigated the effects of environmental enrichment during critical period of early postnatal life and how it interplays with the epigenome to affect experience-dependent visual cortical plasticity. Mice raised in an EE from birth to during CP have increased spine density and dendritic complexity in the visual cortex. EE upregulates synaptic plasticity genes, Arc and Egr1, and a transcription factor MEF2C. We also observed an increase in MEF2C binding to the promoters of Arc and Egr1. In addition, pups raised in EE show a reduction in HDAC5 and its binding to promoters of Mef2c, Arc and Egr1 genes. With an overexpression of Mef2c, neurite outgrowth increased in complexity. Our results suggest a possible underlying molecular mechanism of EE, acting through MEF2C and HDAC5, which drive Arc and Egr1. This could lead to the observed increased dendritic spine density and complexity induced by early EE.
Collapse
|
42
|
Xu W, Löwel S, Schlüter OM. Silent Synapse-Based Mechanisms of Critical Period Plasticity. Front Cell Neurosci 2020; 14:213. [PMID: 32765222 PMCID: PMC7380267 DOI: 10.3389/fncel.2020.00213] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/17/2020] [Indexed: 01/08/2023] Open
Abstract
Critical periods are postnatal, restricted time windows of heightened plasticity in cortical neural networks, during which experience refines principal neuron wiring configurations. Here, we propose a model with two distinct types of synapses, innate synapses that establish rudimentary networks with innate function, and gestalt synapses that govern the experience-dependent refinement process. Nascent gestalt synapses are constantly formed as AMPA receptor-silent synapses which are the substrates for critical period plasticity. Experience drives the unsilencing and stabilization of gestalt synapses, as well as synapse pruning. This maturation process changes synapse patterning and consequently the functional architecture of cortical excitatory networks. Ocular dominance plasticity (ODP) in the primary visual cortex (V1) is an established experimental model for cortical plasticity. While converging evidence indicates that the start of the critical period for ODP is marked by the maturation of local inhibitory circuits, recent results support our model that critical periods end through the progressive maturation of gestalt synapses. The cooperative yet opposing function of two postsynaptic signaling scaffolds of excitatory synapses, PSD-93 and PSD-95, governs the maturation of gestalt synapses. Without those proteins, networks do not progress far beyond their innate functionality, resulting in rather impaired perception. While cortical networks remain malleable throughout life, the cellular mechanisms and the scope of critical period and adult plasticity differ. Critical period ODP is initiated with the depression of deprived eye responses in V1, whereas adult ODP is characterized by an initial increase in non-deprived eye responses. Our model proposes the gestalt synapse-based mechanism for critical period ODP, and also predicts a different mechanism for adult ODP based on the sparsity of nascent gestalt synapses at that age. Under our model, early life experience shapes the boundaries (the gestalt) for network function, both for its optimal performance as well as for its pathological state. Thus, reintroducing nascent gestalt synapses as plasticity substrates into adults may improve the network gestalt to facilitate functional recovery.
Collapse
Affiliation(s)
- Weifeng Xu
- Department of Neuroscience, Brown University, Providence, RI, United States
- Carney Institute for Brain Science, Brown University, Providence, RI, United States
| | - Siegrid Löwel
- Department of Systems Neuroscience, Johann-Friedrich-Blumenbach Institute for Zoology & Anthropology, University of Göttingen, Göttingen, Germany
- Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Oliver M. Schlüter
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
43
|
Kalish BT, Barkat TR, Diel EE, Zhang EJ, Greenberg ME, Hensch TK. Single-nucleus RNA sequencing of mouse auditory cortex reveals critical period triggers and brakes. Proc Natl Acad Sci U S A 2020; 117:11744-11752. [PMID: 32404418 PMCID: PMC7261058 DOI: 10.1073/pnas.1920433117] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Auditory experience drives neural circuit refinement during windows of heightened brain plasticity, but little is known about the genetic regulation of this developmental process. The primary auditory cortex (A1) of mice exhibits a critical period for thalamocortical connectivity between postnatal days P12 and P15, during which tone exposure alters the tonotopic topography of A1. We hypothesized that a coordinated, multicellular transcriptional program governs this window for patterning of the auditory cortex. To generate a robust multicellular map of gene expression, we performed droplet-based, single-nucleus RNA sequencing (snRNA-seq) of A1 across three developmental time points (P10, P15, and P20) spanning the tonotopic critical period. We also tone-reared mice (7 kHz pips) during the 3-d critical period and collected A1 at P15 and P20. We identified and profiled both neuronal (glutamatergic and GABAergic) and nonneuronal (oligodendrocytes, microglia, astrocytes, and endothelial) cell types. By comparing normal- and tone-reared mice, we found hundreds of genes across cell types showing altered expression as a result of sensory manipulation during the critical period. Functional voltage-sensitive dye imaging confirmed GABA circuit function determines critical period onset, while Nogo receptor signaling is required for its closure. We further uncovered previously unknown effects of developmental tone exposure on trajectories of gene expression in interneurons, as well as candidate genes that might execute tonotopic plasticity. Our single-nucleus transcriptomic resource of developing auditory cortex is thus a powerful discovery platform with which to identify mediators of tonotopic plasticity.
Collapse
Affiliation(s)
- Brian T Kalish
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA 02115
| | - Tania R Barkat
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- Center for Brain Science, Harvard University, Cambridge, MA 02138
| | - Erin E Diel
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- Center for Brain Science, Harvard University, Cambridge, MA 02138
| | | | | | - Takao K Hensch
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138;
- Center for Brain Science, Harvard University, Cambridge, MA 02138
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
- Child Brain Development, Canadian Institute for Advanced Research, Toronto, ON M5G 1M1, Canada
| |
Collapse
|
44
|
Cheyne JE, Montgomery JM. The cellular and molecular basis of in vivo synaptic plasticity in rodents. Am J Physiol Cell Physiol 2020; 318:C1264-C1283. [PMID: 32320288 DOI: 10.1152/ajpcell.00416.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Plasticity within the neuronal networks of the brain underlies the ability to learn and retain new information. The initial discovery of synaptic plasticity occurred by measuring synaptic strength in vivo, applying external stimulation and observing an increase in synaptic strength termed long-term potentiation (LTP). Many of the molecular pathways involved in LTP and other forms of synaptic plasticity were subsequently uncovered in vitro. Over the last few decades, technological advances in recording and imaging in live animals have seen many of these molecular mechanisms confirmed in vivo, including structural changes both pre- and postsynaptically, changes in synaptic strength, and changes in neuronal excitability. A well-studied aspect of neuronal plasticity is the capacity of the brain to adapt to its environment, gained by comparing the brains of deprived and experienced animals in vivo, and in direct response to sensory stimuli. Multiple in vivo studies have also strongly linked plastic changes to memory by interfering with the expression of plasticity and by manipulating memory engrams. Plasticity in vivo also occurs in the absence of any form of external stimulation, i.e., during spontaneous network activity occurring with brain development. However, there is still much to learn about how plasticity is induced during natural learning and how this is altered in neurological disorders.
Collapse
Affiliation(s)
- Juliette E Cheyne
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Department of Physiology and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
45
|
Yu KW, Wang CJ, Wu Y, Wang YY, Wang NH, Kuang SY, Liu G, Xie HY, Jiang CY, Wu JF. An enriched environment increases the expression of fibronectin type III domain-containing protein 5 and brain-derived neurotrophic factor in the cerebral cortex of the ischemic mouse brain. Neural Regen Res 2020; 15:1671-1677. [PMID: 32209771 PMCID: PMC7437579 DOI: 10.4103/1673-5374.276339] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Many studies have shown that fibronectin type III domain-containing protein 5 (FDNC5) and brain-derived neurotrophic factor (BDNF) play vital roles in plasticity after brain injury. An enriched environment refers to an environment that provides animals with multi-sensory stimulation and movement opportunities. An enriched environment has been shown to promote the regeneration of nerve cells, synapses, and blood vessels in the animal brain after cerebral ischemia; however, the exact mechanisms have not been clarified. This study aimed to determine whether an enriched environment could improve neurobehavioral functions after the experimental inducement of cerebral ischemia and whether neurobehavioral outcomes were associated with the expression of FDNC5 and BDNF. This study established ischemic mouse models using permanent middle cerebral artery occlusion (pMCAO) on the left side. On postoperative day 1, the mice were randomly assigned to either enriched environment or standard housing condition groups. Mice in the standard housing condition group were housed and fed under standard conditions. Mice in the enriched environment group were housed in a large cage, containing various toys, and fed with a standard diet. Sham-operated mice received the same procedure, but without artery occlusion, and were housed and fed under standard conditions. On postoperative days 7 and 14, a beam-walking test was used to assess coordination, balance, and spatial learning. On postoperative days 16–20, a Morris water maze test was used to assess spatial learning and memory. On postoperative day 15, the expression levels of FDNC5 and BDNF proteins in the ipsilateral cerebral cortex were analyzed by western blot assay. The results showed that compared with the standard housing condition group, the motor balance and coordination functions (based on beam-walking test scores 7 and 14 days after operation), spatial learning abilities (based on the spatial learning scores from the Morris water maze test 16–19 days after operation), and memory abilities (based on the memory scores of the Morris water maze test 20 days after operation) of the enriched environment group improved significantly. In addition, the expression levels of FDNC5 and BDNF proteins in the ipsilateral cerebral cortex increased in the enriched environment group compared with those in the standard housing condition group. Furthermore, the Pearson correlation coefficient showed that neurobehavioral functions were positively associated with the expression levels of FDNC5 and BDNF (r = 0.587 and r = 0.840, respectively). These findings suggest that an enriched environment upregulates FDNC5 protein expression in the ipsilateral cerebral cortex after cerebral ischemia, which then activates BDNF protein expression, improving neurological function. BDNF protein expression was positively correlated with improved neurological function. The experimental protocols were approved by the Institutional Animal Care and Use Committee of Fudan University, China (approval Nos. 20160858A232, 20160860A234) on February 24, 2016.
Collapse
Affiliation(s)
- Ke-Wei Yu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuan-Jie Wang
- Department of Rehabilitation Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yi Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu-Yang Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Nian-Hong Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shen-Yi Kuang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Hong-Yu Xie
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Cong-Yu Jiang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jun-Fa Wu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Murase S, Winkowski D, Liu J, Kanold PO, Quinlan EM. Homeostatic regulation of perisynaptic matrix metalloproteinase 9 (MMP9) activity in the amblyopic visual cortex. eLife 2019; 8:52503. [PMID: 31868167 PMCID: PMC6961978 DOI: 10.7554/elife.52503] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/19/2019] [Indexed: 01/07/2023] Open
Abstract
Dark exposure (DE) followed by light reintroduction (LRx) reactivates robust synaptic plasticity in adult mouse primary visual cortex (V1), which allows subsequent recovery from amblyopia. Previously we showed that perisynaptic proteolysis by MMP9 mediates the enhancement of plasticity by LRx in binocular adult mice (Murase et al., 2017). However, it was unknown if a visual system compromised by amblyopia could engage this pathway. Here we show that LRx to adult amblyopic mice induces perisynaptic MMP2/9 activity and extracellular matrix (ECM) degradation in deprived and non-deprived V1. Indeed, LRx restricted to the amblyopic eye is sufficient to induce robust MMP2/9 activity at thalamo-cortical synapses and ECM degradation in deprived V1. Two-photon live imaging demonstrates that the history of visual experience regulates MMP2/9 activity in V1, and that DE lowers the threshold for the proteinase activation. The homeostatic reduction of the MMP2/9 activation threshold by DE enables visual input from the amblyopic pathway to trigger robust perisynaptic proteolysis.
Collapse
Affiliation(s)
- Sachiko Murase
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Dan Winkowski
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Ji Liu
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| | - Elizabeth M Quinlan
- Department of Biology, University of Maryland, College Park, United States.,Neuroscience Cognitive Sciences Program, University of Maryland, College Park, United States
| |
Collapse
|
47
|
Patel DC, Tewari BP, Chaunsali L, Sontheimer H. Neuron-glia interactions in the pathophysiology of epilepsy. Nat Rev Neurosci 2019; 20:282-297. [PMID: 30792501 DOI: 10.1038/s41583-019-0126-4] [Citation(s) in RCA: 234] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epilepsy is a neurological disorder afflicting ~65 million people worldwide. It is caused by aberrant synchronized firing of populations of neurons primarily due to imbalance between excitatory and inhibitory neurotransmission. Hence, the historical focus of epilepsy research has been neurocentric. However, the past two decades have enjoyed an explosion of research into the role of glia in supporting and modulating neuronal activity, providing compelling evidence of glial involvement in the pathophysiology of epilepsy. The mechanisms by which glia, particularly astrocytes and microglia, may contribute to epilepsy and consequently could be harnessed therapeutically are discussed in this Review.
Collapse
Affiliation(s)
- Dipan C Patel
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA
| | - Bhanu P Tewari
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA
| | - Lata Chaunsali
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA
| | - Harald Sontheimer
- Fralin Biomedical Research Institute, Glial Biology in Health, Disease, and Cancer Center, Roanoke, VA, USA. .,School of Neuroscience, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
48
|
Ocular Dominance Plasticity in Binocular Primary Visual Cortex Does Not Require C1q. J Neurosci 2019; 40:769-783. [PMID: 31801811 DOI: 10.1523/jneurosci.1011-19.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 11/21/2022] Open
Abstract
C1q, the initiator of the classical complement cascade, mediates synapse elimination in the postnatal mouse dorsolateral geniculate nucleus of the thalamus and sensorimotor cortex. Here, we asked whether C1q plays a role in experience-dependent synaptic refinement in the visual system at later stages of development. The binocular zone of primary visual cortex (V1b) undergoes spine loss and changes in neuronal responsiveness following the closure of one eye during a defined critical period [a process referred to as ocular dominance plasticity (ODP)]. We therefore hypothesized that ODP would be impaired in the absence of C1q, and that V1b development would also be abnormal without C1q-mediated synapse elimination. However, when we examined several features of V1b development in mice lacking C1q, we found that the densities of most spine populations on basal and proximal apical dendrites, as well as firing rates and ocular dominance, were normal. C1q was only transiently required for the development of spines on apical, but not basal, secondary dendrites. Dendritic morphologies were also unaffected. Although we did not observe the previously described spine loss during ODP in either genotype, our results reveal that the animals lacking C1q had normal shifts in neuronal responsiveness following eye closure. Experiments were performed in both male and female mice. These results suggest that the development and plasticity of the mouse V1b is grossly normal in the absence of C1q.SIGNIFICANCE STATEMENT These findings illustrate that the development and experience-dependent plasticity of V1b is mostly normal in the absence of C1q, even though C1q has previously been shown to be required for developmental synapse elimination in the mouse visual thalamus as well as sensorimotor cortex. The V1b phenotypes in mice lacking C1q are more similar to the mild defects previously observed in the hippocampus of these mice, emphasizing that the contribution of C1q to synapse elimination appears to be dependent on context.
Collapse
|
49
|
Xu ZX, Tan JW, Xu H, Hill CJ, Ostrovskaya O, Martemyanov KA, Xu B. Caspase-2 promotes AMPA receptor internalization and cognitive flexibility via mTORC2-AKT-GSK3β signaling. Nat Commun 2019; 10:3622. [PMID: 31399584 PMCID: PMC6689033 DOI: 10.1038/s41467-019-11575-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 07/23/2019] [Indexed: 01/22/2023] Open
Abstract
Caspase-2 is the most evolutionarily conserved member in the caspase family of proteases and is constitutively expressed in most cell types including neurons; however, its physiological function remains largely unknown. Here we report that caspase-2 plays a critical role in synaptic plasticity and cognitive flexibility. We found that caspase-2 deficiency led to deficits in dendritic spine pruning, internalization of AMPA receptors and long-term depression. Our results indicate that caspase-2 degrades Rictor, a key mTOR complex 2 (mTORC2) component, to inhibit Akt activation, which leads to enhancement of the GSK3β activity and thereby long-term depression. Furthermore, we found that mice lacking caspase-2 displayed elevated levels of anxiety, impairment in reversal water maze learning, and little memory loss over time. These results not only uncover a caspase-2-mTORC2-Akt-GSK3β signaling pathway, but also suggest that caspase-2 is important for memory erasing and normal behaviors by regulating synaptic number and transmission.
Collapse
Affiliation(s)
- Zhi-Xiang Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Ji-Wei Tan
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Haifei Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Cassandra J Hill
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Olga Ostrovskaya
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA.
| |
Collapse
|
50
|
Protease-independent action of tissue plasminogen activator in brain plasticity and neurological recovery after ischemic stroke. Proc Natl Acad Sci U S A 2019; 116:9115-9124. [PMID: 30996120 DOI: 10.1073/pnas.1821979116] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that tissue plasminogen activator (tPA), currently the only FDA-approved medication for ischemic stroke, exerts important biological actions on the CNS besides its well-known thrombolytic effect. In this study, we investigated the role of tPA on primary neurons in culture and on brain recovery and plasticity after ischemic stroke in mice. Treatment with recombinant tPA stimulated axonal growth in culture, an effect independent of its protease activity and achieved through epidermal growth factor receptor (EGFR) signaling. After permanent focal cerebral ischemia, tPA knockout mice developed more severe sensorimotor and cognitive deficits and greater axonal and myelin injury than wild-type mice, suggesting that endogenously expressed tPA promotes long-term neurological recovery after stroke. In tPA knockout mice, intranasal administration of recombinant tPA protein 6 hours poststroke and 7 more times at 2 d intervals mitigated white matter injury, improved axonal conduction, and enhanced neurological recovery. Consistent with the proaxonal growth effects observed in vitro, exogenous tPA delivery increased poststroke axonal sprouting of corticobulbar and corticospinal tracts, which might have contributed to restoration of neurological functions. Notably, recombinant mutant tPA-S478A lacking protease activity (but retaining the EGF-like domain) was as effective as wild-type tPA in rescuing neurological functions in tPA knockout stroke mice. These findings demonstrate that tPA improves long-term functional outcomes in a clinically relevant stroke model, likely by promoting brain plasticity through EGFR signaling. Therefore, treatment with the protease-dead recombinant tPA-S478A holds particular promise as a neurorestorative therapy, as the risk for triggering intracranial hemorrhage is eliminated and tPA-S478A can be delivered intranasally hours after stroke.
Collapse
|