1
|
Agner SC, Brier LM, Hill JD, Liu EY, Bice A, Rahn RM, Chen S, Culver JP, Klein RS. Zika virus encephalitis causes transient reduction of functional cortical connectivity. NEUROPHOTONICS 2025; 12:S14603. [PMID: 39610883 PMCID: PMC11603678 DOI: 10.1117/1.nph.12.s1.s14603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024]
Abstract
Significance Determining the long-term cognitive impact of infections is clinically challenging. Using functional cortical connectivity, we demonstrate that interhemispheric cortical connectivity is decreased in individuals with acute Zika virus (ZIKV) encephalitis. This correlates with decreased presynaptic terminals in the somatosensory cortex. During recovery from ZIKV infection, presynaptic terminals recover, which is associated with recovered interhemispheric connectivity. This supports the contribution of synapses in the cortex to functional networks in the brain, which can be detected by widefield optical imaging. Although myeloid cell and astrocyte numbers are still increased during recovery, RNA transcription of multiple proinflammatory cytokines that increase during acute infection decreases to levels comparable to mock-infected mice during recovery. These findings also suggest that the immune response and cytokine-mediated neuroinflammation play significant roles in the integrity of brain networks during and after viral encephalitis. Aim We hypothesized that widefield optical imaging would allow us to assess functional cortical network disruption by ZIKV, including hippocampal-cortical networks. Approach We use widefield optical imaging to measure cortical functional connectivity (FC) in mice during acute infection with, and recovery from, intracranial infection with a mouse-adapted strain of ZIKV. Results Acute ZIKV infection leads to high levels of myeloid cell activation, with loss of neurons and presynaptic termini in the cerebral cortex and associated loss of FC primarily within the somatosensory cortex. During recovery, neuron numbers, synapses, and FC recover to levels near those of healthy mice. However, hippocampal injury and impaired spatial cognition persist. The magnitude of activated myeloid cells during acute infection predicted both recovery of synapses and the degree of FC recovery after recovery from ZIKV infection. Conclusions These findings suggest that a robust inflammatory response may contribute to the health of functional brain networks after recovery from infection.
Collapse
Affiliation(s)
- Shannon C. Agner
- Washington University School of Medicine, Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, Missouri, United States
- Washington University School of Medicine, Department of Neurology, St. Louis, Missouri, United States
| | - Lindsey M. Brier
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri, United States
| | - Jeremy D. Hill
- Washington University School of Medicine, Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, Missouri, United States
- Washington University School of Medicine, Department of Medicine, St. Louis, Missouri, United States
| | - Ethan Y. Liu
- Washington University School of Medicine, Department of Neurology, St. Louis, Missouri, United States
| | - Annie Bice
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri, United States
| | - Rachel M. Rahn
- Washington University School of Medicine, St. Louis, Missouri, United States
| | - Shengxuan Chen
- Washington University School of Medicine, St. Louis, Missouri, United States
| | - Joseph P. Culver
- Washington University School of Medicine, Department of Radiology, St. Louis, Missouri, United States
- Washington University School of Medicine, St. Louis, Missouri, United States
- Washington University School of Medicine, Departments of Physics, Biomedical Engineering, and Electrical and Systems Engineering, St. Louis, Missouri, United States
| | - Robyn S. Klein
- Washington University School of Medicine, Center for Neuroimmunology and Neuroinfectious Diseases, St. Louis, Missouri, United States
- Western University, Departments of Medicine, Microbiology & Immunology, Western Institute of Neuroscience, London, Ontario, Canada
| |
Collapse
|
2
|
Gerasimov E, Pchitskaya E, Vlasova O, Bezprozvanny I. Dynamic changes in the hippocampal neuronal circuits activity following acute stress revealed by miniature fluorescence microscopy imaging. Mol Brain 2024; 17:92. [PMID: 39695833 DOI: 10.1186/s13041-024-01168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Coordinated activity of neuronal ensembles is a basis for information processing in the brain. Recent development of miniscope imaging technology enabled recordings of neuronal circuits activity in vivo in freely behaving animals. Acute stress is believed to affect various hippocampal functions, especially memory. In the current study, we utilized miniscope imaging to investigate the hippocampal neuronal circuits properties in a mouse as function of time and immediately in response to an acute stress, induced by passive restraint, 3 h and 10 days after. Comprehensive quantitative analysis of network activity changes at the neuronal ensembles level revealed highly stable neuronal activity parameters, which exhibited a rapid and robust shift in response to acute stress stimulation. This shift was accompanied by the restructuring of the pairwise-correlated neuronal pairs. Remarkably, we discovered that ensembles activity characteristics returned to the initial state following recovery period, demonstrating hippocampal homeostatic stability at the neuronal circuits level. Obtained results provide an evidence about hippocampal neuronal ensembles activity in response to acute stress over time.
Collapse
Affiliation(s)
- Evgenii Gerasimov
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia.
| | - Ekaterina Pchitskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia
| | - Olga Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia
| | - Ilya Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya St. 29, 195220, St. Petersburg, Russia.
| |
Collapse
|
3
|
Schlett K, Oueslati Morales CO, Bencsik N, Hausser A. Getting smart - Deciphering the neuronal functions of protein kinase D. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119812. [PMID: 39147241 DOI: 10.1016/j.bbamcr.2024.119812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
Protein kinase D (PKD) is a family of serine/threonine kinases that play important roles in various signalling pathways in cells, including neuronal cells. In the nervous system, PKD has been shown to be involved in learning and memory formation by regulating neurotransmitter release, neurite outgrowth and dendrite development, synapse formation and synaptic plasticity. In addition, PKD has been implicated in pain perception or neuroprotection during oxidative stress. Dysregulation of PKD expression and activity has been linked to several neurological disorders, including autism and epilepsy. In this review, we summarize the current knowledge on the function of the PKD family members in neuronal cells, including the spatial regulation of their downstream signalling pathways. We will further discuss the potential role of PKD in the pathogenesis of neurological disorders.
Collapse
Affiliation(s)
- Katalin Schlett
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Carlos O Oueslati Morales
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Norbert Bencsik
- Neuronal Cell Biology Group, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Angelika Hausser
- Membrane Trafficking and Signalling Group, Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
4
|
Jürgensen AM, Sakagiannis P, Schleyer M, Gerber B, Nawrot MP. Prediction error drives associative learning and conditioned behavior in a spiking model of Drosophila larva. iScience 2024; 27:108640. [PMID: 38292165 PMCID: PMC10824792 DOI: 10.1016/j.isci.2023.108640] [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: 03/01/2023] [Revised: 11/10/2023] [Accepted: 12/01/2023] [Indexed: 02/01/2024] Open
Abstract
Predicting reinforcement from sensory cues is beneficial for goal-directed behavior. In insect brains, underlying associations between cues and reinforcement, encoded by dopaminergic neurons, are formed in the mushroom body. We propose a spiking model of the Drosophila larva mushroom body. It includes a feedback motif conveying learned reinforcement expectation to dopaminergic neurons, which can compute prediction error as the difference between expected and present reinforcement. We demonstrate that this can serve as a driving force in learning. When combined with synaptic homeostasis, our model accounts for theoretically derived features of acquisition and loss of associations that depend on the intensity of the reinforcement and its temporal proximity to the cue. From modeling olfactory learning over the time course of behavioral experiments and simulating the locomotion of individual larvae toward or away from odor sources in a virtual environment, we conclude that learning driven by prediction errors can explain larval behavior.
Collapse
Affiliation(s)
- Anna-Maria Jürgensen
- Computational Systems Neuroscience, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| | - Panagiotis Sakagiannis
- Computational Systems Neuroscience, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| | - Michael Schleyer
- Leibniz Institute for Neurobiology (LIN), Department of Genetics, 39118 Magdeburg, Germany
- Institute for the Advancement of Higher Education, Faculty of Science, Hokkaido University, Sapporo 060-08080, Japan
| | - Bertram Gerber
- Leibniz Institute for Neurobiology (LIN), Department of Genetics, 39118 Magdeburg, Germany
- Institute for Biology, Otto-von-Guericke University, 39120 Magdeburg, Germany
- Center for Brain and Behavioral Sciences (CBBS), Otto-von-Guericke University, 39118 Magdeburg, Germany
| | - Martin Paul Nawrot
- Computational Systems Neuroscience, Institute of Zoology, University of Cologne, 50674 Cologne, Germany
| |
Collapse
|
5
|
Agner SC, Brier LM, Hill J, Liu E, Bice A, Rahn RM, Culver JP, Klein RS. Myeloid cell activation during Zika virus encephalitis predicts recovery of functional cortical connectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.06.547991. [PMID: 37461558 PMCID: PMC10350085 DOI: 10.1101/2023.07.06.547991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Neurologic complications of Zika virus (ZIKV) infection across the lifespan have been described during outbreaks in Southeast Asia, South America, and Central America since 2016. In the adult CNS ZIKV tropism for neurons is tightly linked to its effects, with neuronal loss within the hippocampus during acute infection and protracted synapse loss during recovery, which is associated with cognitive deficits. The effects of ZIKV on cortical networks have not been evaluated. Although animal behavior assays have been used previously to model cognitive impairment, in vivo brain imaging can provide orthogonal information regarding the health of brain networks in real time, providing a tool to translate findings in animal models to humans. In this study, we use widefield optical imaging to measure cortical functional connectivity (FC) in mice during acute infection with, and recovery from, intracranial infection with a mouse-adapted strain of ZIKV. Acute ZIKV infection leads to high levels of myeloid cell activation, with loss of neurons and presynaptic termini in the cerebral cortex and associated loss of FC primarily within the somatosensory cortex. During recovery, neuron numbers, synapses and FC recover to levels near those of healthy mice. However, hippocampal injury and impaired spatial cognition persist. The magnitude of activated myeloid cells during acute infection predicted both recovery of synapses and the degree of FC recovery after recovery from ZIKV infection. These findings suggest that a robust inflammatory response may contribute to the health of functional brain networks after recovery from infection.
Collapse
Affiliation(s)
- Shannon C. Agner
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lindsey M. Brier
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeremy Hill
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ethan Liu
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Annie Bice
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel M. Rahn
- Departments of Physics, Biomedical Engineering, and Electrical and Systems Engineering, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph P. Culver
- Departments of Physics, Biomedical Engineering, and Electrical and Systems Engineering, Washington University School of Medicine, St. Louis, MO, USA
| | - Robyn S. Klein
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Medicine, Pathology & Immunology, and Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Charbonneau JA, Bennett JL, Chau K, Bliss-Moreau E. Reorganization in the macaque interoceptive-allostatic network following anterior cingulate cortex damage. Cereb Cortex 2023; 33:4334-4349. [PMID: 36066407 PMCID: PMC10110454 DOI: 10.1093/cercor/bhac346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/14/2022] Open
Abstract
Accumulating evidence indicates that the adult brain is capable of significant structural change following damage-a capacity once thought to be largely limited to developing brains. To date, most existing research on adult plasticity has focused on how exteroceptive sensorimotor networks compensate for damage to preserve function. Interoceptive networks-those that represent and process sensory information about the body's internal state-are now recognized to be critical for a wide range of physiological and psychological functions from basic energy regulation to maintaining a sense of self, but the extent to which these networks remain plastic in adulthood has not been established. In this report, we used detailed histological analyses to pinpoint precise changes to gray matter volume in the interoceptive-allostatic network in adult rhesus monkeys (Macaca mulatta) who received neurotoxic lesions of the anterior cingulate cortex (ACC) and neurologically intact control monkeys. Relative to controls, monkeys with ACC lesions had significant and selective unilateral expansion of the ventral anterior insula and significant relative bilateral expansion of the lateral nucleus of the amygdala. This work demonstrates the capacity for neuroplasticity in the interoceptive-allostatic network which, given that changes included expansion rather than atrophy, is likely to represent an adaptive response following damage.
Collapse
Affiliation(s)
- Joey A Charbonneau
- Neuroscience Graduate Program, University of California Davis, 1544 Newton Court, Davis, CA 95618, United States
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Jeffrey L Bennett
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, 2230 Stockton Blvd, Sacramento, CA 95817, United States
- The MIND Institute, University of California Davis, 2825 50th Street, Sacramento, CA 95817, United States
| | - Kevin Chau
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
| | - Eliza Bliss-Moreau
- California National Primate Research Center, University of California Davis, One Shields Avenue, Davis, CA 95616, United States
- Department of Psychology, University of California Davis, 135 Young Hall One Shields Avenue, Davis, CA 95616, United States
| |
Collapse
|
7
|
Schwarz K, Schmitz F. Synapse Dysfunctions in Multiple Sclerosis. Int J Mol Sci 2023; 24:ijms24021639. [PMID: 36675155 PMCID: PMC9862173 DOI: 10.3390/ijms24021639] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory disease of the central nervous system (CNS) affecting nearly three million humans worldwide. In MS, cells of an auto-reactive immune system invade the brain and cause neuroinflammation. Neuroinflammation triggers a complex, multi-faceted harmful process not only in the white matter but also in the grey matter of the brain. In the grey matter, neuroinflammation causes synapse dysfunctions. Synapse dysfunctions in MS occur early and independent from white matter demyelination and are likely correlates of cognitive and mental symptoms in MS. Disturbed synapse/glia interactions and elevated neuroinflammatory signals play a central role. Glutamatergic excitotoxic synapse damage emerges as a major mechanism. We review synapse/glia communication under normal conditions and summarize how this communication becomes malfunctional during neuroinflammation in MS. We discuss mechanisms of how disturbed glia/synapse communication can lead to synapse dysfunctions, signaling dysbalance, and neurodegeneration in MS.
Collapse
|
8
|
Kanishka, Jha SK. Compensatory cognition in neurological diseases and aging: A review of animal and human studies. AGING BRAIN 2023; 3:100061. [PMID: 36911258 PMCID: PMC9997140 DOI: 10.1016/j.nbas.2022.100061] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/06/2022] [Accepted: 12/12/2022] [Indexed: 12/27/2022] Open
Abstract
Specialized individual circuits in the brain are recruited for specific functions. Interestingly, multiple neural circuitries continuously compete with each other to acquire the specialized function. However, the dominant among them compete and become the central neural network for that particular function. For example, the hippocampal principal neural circuitries are the dominant networks among many which are involved in learning processes. But, in the event of damage to the principal circuitry, many times, less dominant networks compensate for the primary network. This review highlights the psychopathologies of functional loss and the aspects of functional recuperation in the absence of the hippocampus.
Collapse
Affiliation(s)
- Kanishka
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Sushil K Jha
- School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
9
|
Cordella F, Ferrucci L, D’Antoni C, Ghirga S, Brighi C, Soloperto A, Gigante Y, Ragozzino D, Bezzi P, Di Angelantonio S. Human iPSC-Derived Cortical Neurons Display Homeostatic Plasticity. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111884. [PMID: 36431019 PMCID: PMC9696876 DOI: 10.3390/life12111884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Maintaining the excitability of neurons and circuits is fundamental for healthy brain functions. The global compensatory increase in excitatory synaptic strength, in response to decreased activity, is one of the main homeostatic mechanisms responsible for such regulation. This type of plasticity has been extensively characterized in rodents in vivo and in vitro, but few data exist on human neurons maturation. We have generated an in vitro cortical model system, based on differentiated human-induced pluripotent stem cells, chronically treated with tetrodotoxin, to investigate homeostatic plasticity at different developmental stages. Our findings highlight the presence of homeostatic plasticity in human cortical networks and show that the changes in synaptic strength are due to both pre- and post-synaptic mechanisms. Pre-synaptic plasticity involves the potentiation of neurotransmitter release machinery, associated to an increase in synaptic vesicle proteins expression. At the post-synaptic level, we report an increase in the expression of post-synaptic density proteins, involved in glutamatergic receptor anchoring. These results extend our understanding of neuronal homeostasis and reveal the developmental regulation of its expression in human cortical networks. Since induced pluripotent stem cell-derived neurons can be obtained from patients with neurodevelopmental and neurodegenerative diseases, our platform offers a versatile model for assessing human neural plasticity under physiological and pathological conditions.
Collapse
Affiliation(s)
- Federica Cordella
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Laura Ferrucci
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara D’Antoni
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Silvia Ghirga
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Carlo Brighi
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- CrestOptics S.p.A., Via di Torre Rossa 66, 00165 Rome, Italy
| | - Alessandro Soloperto
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Ylenia Gigante
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- D-Tails s.r.l., Via di Torre Rossa 66, 00165 Rome, Italy
| | - Davide Ragozzino
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy
| | - Paola Bezzi
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, 1015 Lausanne, Switzerland
- Correspondence: or (P.B.); or (S.D.A.)
| | - Silvia Di Angelantonio
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Center for Life Nano- & Neuro-Science of Istituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
- D-Tails s.r.l., Via di Torre Rossa 66, 00165 Rome, Italy
- Correspondence: or (P.B.); or (S.D.A.)
| |
Collapse
|
10
|
Chan ES, Ge Y, So YW, Bai YF, Liu L, Wang YT. Allosteric potentiation of GABAA receptor single-channel conductance by netrin-1 during neuronal-excitation-induced inhibitory synaptic homeostasis. Cell Rep 2022; 41:111584. [DOI: 10.1016/j.celrep.2022.111584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/13/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
|
11
|
Fitzpatrick MJ, Kerschensteiner D. Homeostatic plasticity in the retina. Prog Retin Eye Res 2022; 94:101131. [PMID: 36244950 DOI: 10.1016/j.preteyeres.2022.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 02/07/2023]
Abstract
Vision begins in the retina, whose intricate neural circuits extract salient features of the environment from the light entering our eyes. Neurodegenerative diseases of the retina (e.g., inherited retinal degenerations, age-related macular degeneration, and glaucoma) impair vision and cause blindness in a growing number of people worldwide. Increasing evidence indicates that homeostatic plasticity (i.e., the drive of a neural system to stabilize its function) can, in principle, preserve retinal function in the face of major perturbations, including neurodegeneration. Here, we review the circumstances and events that trigger homeostatic plasticity in the retina during development, sensory experience, and disease. We discuss the diverse mechanisms that cooperate to compensate and the set points and outcomes that homeostatic retinal plasticity stabilizes. Finally, we summarize the opportunities and challenges for unlocking the therapeutic potential of homeostatic plasticity. Homeostatic plasticity is fundamental to understanding retinal development and function and could be an important tool in the fight to preserve and restore vision.
Collapse
|
12
|
Wong NF, Xu-Friedman MA. Induction of Activity-Dependent Plasticity at Auditory Nerve Synapses. J Neurosci 2022; 42:6211-6220. [PMID: 35790402 PMCID: PMC9374128 DOI: 10.1523/jneurosci.0666-22.2022] [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: 04/05/2022] [Revised: 05/26/2022] [Accepted: 06/25/2022] [Indexed: 11/21/2022] Open
Abstract
Exposure to nontraumatic noise in vivo drives long-lasting changes in auditory nerve synapses, which may influence hearing, but the induction mechanisms are not known. We mimicked activity in acute slices of the cochlear nucleus from mice of both sexes by treating them with high potassium, after which voltage-clamp recordings from bushy cells indicated that auditory nerve synapses had reduced EPSC amplitude, quantal size, and vesicle release probability (P r). The effects of high potassium were prevented by blockers of nitric oxide (NO) synthase and protein kinase A. Treatment with the NO donor, PAPA-NONOate, also decreased P r, suggesting NO plays a central role in inducing synaptic changes. To identify the source of NO, we activated auditory nerve fibers specifically using optogenetics. Strobing for 2 h led to decreased EPSC amplitude and P r, which was prevented by antagonists against ionotropic glutamate receptors and NO synthase. This suggests that the activation of AMPA and NMDA receptors in postsynaptic targets of auditory nerve fibers drives release of NO, which acts retrogradely to cause long-term changes in synaptic function in auditory nerve synapses. This may provide insight into preventing or treating disorders caused by noise exposure.SIGNIFICANCE STATEMENT Auditory nerve fibers undergo long-lasting changes in synaptic properties in response to noise exposure in vivo, which may contribute to changes in hearing. Here, we investigated the cellular mechanisms underlying induction of synaptic changes using high potassium and optogenetic stimulation in vitro and identified important signaling pathways using pharmacology. Our results suggest that auditory nerve activity drives postsynaptic depolarization through AMPA and NMDA receptors, leading to the release of nitric oxide, which acts retrogradely to regulate presynaptic neurotransmitter release. These experiments revealed that auditory nerve synapses are unexpectedly sensitive to activity and can show dramatic, long-lasting changes in a few hours that could affect hearing.
Collapse
Affiliation(s)
- Nicole F Wong
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| | - Matthew A Xu-Friedman
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York 14260
| |
Collapse
|
13
|
Dubes S, Soula A, Benquet S, Tessier B, Poujol C, Favereaux A, Thoumine O, Letellier M. miR
‐124‐dependent tagging of synapses by synaptopodin enables input‐specific homeostatic plasticity. EMBO J 2022; 41:e109012. [PMID: 35875872 PMCID: PMC9574720 DOI: 10.15252/embj.2021109012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 06/11/2022] [Accepted: 06/27/2022] [Indexed: 12/26/2022] Open
Abstract
Homeostatic synaptic plasticity is a process by which neurons adjust their synaptic strength to compensate for perturbations in neuronal activity. Whether the highly diverse synapses on a neuron respond uniformly to the same perturbation remains unclear. Moreover, the molecular determinants that underlie synapse‐specific homeostatic synaptic plasticity are unknown. Here, we report a synaptic tagging mechanism in which the ability of individual synapses to increase their strength in response to activity deprivation depends on the local expression of the spine‐apparatus protein synaptopodin under the regulation of miR‐124. Using genetic manipulations to alter synaptopodin expression or regulation by miR‐124, we show that synaptopodin behaves as a “postsynaptic tag” whose translation is derepressed in a subpopulation of synapses and allows for nonuniform homeostatic strengthening and synaptic AMPA receptor stabilization. By genetically silencing individual connections in pairs of neurons, we demonstrate that this process operates in an input‐specific manner. Overall, our study shifts the current view that homeostatic synaptic plasticity affects all synapses uniformly to a more complex paradigm where the ability of individual synapses to undergo homeostatic changes depends on their own functional and biochemical state.
Collapse
Affiliation(s)
- Sandra Dubes
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Anaïs Soula
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Sébastien Benquet
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Béatrice Tessier
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Christel Poujol
- University of Bordeaux CNRS INSERM Bordeaux Imaging Center BIC UMS 3420, US 4 Bordeaux France
| | - Alexandre Favereaux
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Olivier Thoumine
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| | - Mathieu Letellier
- University of Bordeaux CNRS Interdisciplinary Institute for Neuroscience UMR 5297 Bordeaux France
| |
Collapse
|
14
|
Long J, Lu F, Yang S, Zhang Q, Chen X, Pang Y, Wang M, He B, Liu H, Duan X, Chen H, Ye S, Chen H. Different functional connectivity optimal frequency in autism compared with healthy controls and the relationship with social communication deficits: Evidence from gene expression and behavior symptom analyses. Hum Brain Mapp 2022; 44:258-268. [PMID: 35822559 PMCID: PMC9783427 DOI: 10.1002/hbm.26011] [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: 02/24/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 02/05/2023] Open
Abstract
Studies have reported that different brain regions/connections possess distinct frequency properties, which are related to brain function. Previous studies have proposed altered brain activity frequency and frequency-specific functional connectivity (FC) patterns in autism spectrum disorder (ASD), implying the varied dominant frequency of FC in ASD. However, the difference of the dominant frequency of FC between ASD and healthy controls (HCs) remains unclear. In the present study, the dominant frequency of FC was measured by FC optimal frequency, which was defined as the intermediate of the frequency bin at which the FC strength could reach the maximum. A multivariate pattern analysis was conducted to determine whether the FC optimal frequency in ASD differs from that in HCs. Partial least squares regression (PLSR) and enrichment analyses were conducted to determine the relationship between the FC optimal frequency difference of ASD/HCs and cortical gene expression. PLSR analyses were also performed to explore the relationship between FC optimal frequency and the clinical symptoms of ASD. Results showed a significant difference of FC optimal frequency between ASD and HCs. Some genes whose cortical expression patterns are related to the FC optimal frequency difference of ASD/HCs were enriched for social communication problems. Meanwhile, the FC optimal frequency in ASD was significantly related to social communication symptoms. These results may help us understand the neuro-mechanism of the social communication deficits in ASD.
Collapse
Affiliation(s)
- Jinjin Long
- School of MedicineGuizhou UniversityGuiyangChina,Guiyang Hospital of StomatologyGuiyangChina
| | - Fengmei Lu
- Key laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology and Center for Information in BioMedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | | | | | - Xue Chen
- School of MedicineGuizhou UniversityGuiyangChina
| | - Yajing Pang
- School of Electrical EngineeringZhengzhou UniversityZhengzhouChina
| | - Min Wang
- Bioinformatics and BioMedical Bigdata Mining Laboratory, School of Big HealthGuizhou Medical UniversityChina
| | - Bifang He
- School of MedicineGuizhou UniversityGuiyangChina
| | - Heng Liu
- Department of Radiology, Affiliated Hospital of Zunyi Medical UniversityMedical Imaging Center of Guizhou ProvinceZunyiChina
| | - Xujun Duan
- Key laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology and Center for Information in BioMedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Huafu Chen
- Key laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology and Center for Information in BioMedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Shaobing Ye
- The People's Hospital of Kaizhou DistrictChongqingChina
| | - Heng Chen
- School of MedicineGuizhou UniversityGuiyangChina,Key laboratory for NeuroInformation of Ministry of Education, School of Life Science and Technology and Center for Information in BioMedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
15
|
Constraints on Persistent Activity in a Biologically Detailed Network Model of the Prefrontal Cortex with Heterogeneities. Prog Neurobiol 2022; 215:102287. [DOI: 10.1016/j.pneurobio.2022.102287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 02/25/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022]
|
16
|
Imaging the effect of ketamine on synaptic density (SV2A) in the living brain. Mol Psychiatry 2022; 27:2273-2281. [PMID: 35165397 PMCID: PMC9133063 DOI: 10.1038/s41380-022-01465-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 12/28/2022]
Abstract
The discovery of ketamine as a rapid and robust antidepressant marks the beginning of a new era in the treatment of psychiatric disorders. Ketamine is thought to produce rapid and sustained antidepressant effects through restoration of lost synaptic connections. We investigated this hypothesis in humans for the first time using positron emission tomography (PET) and [11C]UCB-J-a radioligand that binds to the synaptic vesicle protein 2A (SV2A) and provides an index of axon terminal density. Overall, we did not find evidence of a measurable effect on SV2A density 24 h after a single administration of ketamine in non-human primates, healthy controls (HCs), or individuals with major depressive disorder (MDD) and/or posttraumatic stress disorder (PTSD), despite a robust reduction in symptoms. A post-hoc, exploratory analysis suggests that patients with lower SV2A density at baseline may exhibit increased SV2A density 24 h after ketamine. This increase in SV2A was associated with a reduction in depression severity, as well as an increase in dissociative symptoms. These initial findings suggest that a restoration of synaptic connections in patients with lower SV2A at baseline may underlie ketamine's therapeutic effects, however, this needs replication in a larger sample. Further work is needed to build on these initial findings and further establish the nuanced pre- and post-synaptic mechanisms underpinning ketamine's therapeutic effects.
Collapse
|
17
|
Shavit-Stein E, Berkowitz S, Gofrit SG, Altman K, Weinberg N, Maggio N. Neurocoagulation from a Mechanistic Point of View in the Central Nervous System. Semin Thromb Hemost 2022; 48:277-287. [PMID: 35052009 DOI: 10.1055/s-0041-1741569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coagulation mechanisms are critical for maintaining homeostasis in the central nervous system (CNS). Thrombin, an important player of the coagulation cascade, activates protease activator receptors (PARs), members of the G-protein coupled receptor family. PAR1 is located on neurons and glia. Following thrombin activation, PAR1 signals through the extracellular signal-regulated kinase pathway, causing alterations in neuronal glutamate release and astrocytic morphological changes. Similarly, the anticoagulation factor activated protein C (aPC) can cleave PAR1, following interaction with the endothelial protein C receptor. Both thrombin and aPC are expressed on endothelial cells and pericytes in the blood-brain barrier (BBB). Thrombin-induced PAR1 activation increases cytosolic Ca2+ concentration in brain vessels, resulting in nitric oxide release and increasing F-actin stress fibers, damaging BBB integrity. aPC also induces PAR1 activation and preserves BBB vascular integrity via coupling to sphingosine 1 phosphate receptors. Thrombin-induced PAR1 overactivation and BBB disruption are evident in CNS pathologies. During epileptic seizures, BBB disruption promotes thrombin penetration. Thrombin induces PAR1 activation and potentiates N-methyl-D-aspartate receptors, inducing glutamate-mediated hyperexcitability. Specific PAR1 inhibition decreases status epilepticus severity in vivo. In stroke, the elevation of brain thrombin levels further compromises BBB integrity, with direct parenchymal damage, while systemic factor Xa inhibition improves neurological outcomes. In multiple sclerosis (MS), brain thrombin inhibitory capacity correlates with clinical presentation. Both thrombin inhibition by hirudin and the use of recombinant aPC improve disease severity in an MS animal model. This review presents the mechanisms underlying the effects of coagulation on the physiology and pathophysiology of the CNS.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Keren Altman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nitai Weinberg
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
18
|
Barnes SJ, Keller GB, Keck T. Homeostatic regulation through strengthening of neuronal network-correlated synaptic inputs. eLife 2022; 11:81958. [PMID: 36515269 PMCID: PMC9803349 DOI: 10.7554/elife.81958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Homeostatic regulation is essential for stable neuronal function. Several synaptic mechanisms of homeostatic plasticity have been described, but the functional properties of synapses involved in homeostasis are unknown. We used longitudinal two-photon functional imaging of dendritic spine calcium signals in visual and retrosplenial cortices of awake adult mice to quantify the sensory deprivation-induced changes in the responses of functionally identified spines. We found that spines whose activity selectively correlated with intrinsic network activity underwent tumor necrosis factor alpha (TNF-α)-dependent homeostatic increases in their response amplitudes, but spines identified as responsive to sensory stimulation did not. We observed an increase in the global sensory-evoked responses following sensory deprivation, despite the fact that the identified sensory inputs did not strengthen. Instead, global sensory-evoked responses correlated with the strength of network-correlated inputs. Our results suggest that homeostatic regulation of global responses is mediated through changes to intrinsic network-correlated inputs rather than changes to identified sensory inputs thought to drive sensory processing.
Collapse
Affiliation(s)
- Samuel J Barnes
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, Hammersmith Hospital CampusLondonUnited Kingdom,UK Dementia Research Institute at Imperial CollegeLondonUnited Kingdom
| | - Georg B Keller
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Tara Keck
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| |
Collapse
|
19
|
Park E, Lau AG, Arendt KL, Chen L. FMRP Interacts with RARα in Synaptic Retinoic Acid Signaling and Homeostatic Synaptic Plasticity. Int J Mol Sci 2021; 22:ijms22126579. [PMID: 34205274 PMCID: PMC8235556 DOI: 10.3390/ijms22126579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
The fragile X syndrome (FXS) is an X-chromosome-linked neurodevelopmental disorder with severe intellectual disability caused by inactivation of the fragile X mental retardation 1 (FMR1) gene and subsequent loss of the fragile X mental retardation protein (FMRP). Among the various types of abnormal synaptic function and synaptic plasticity phenotypes reported in FXS animal models, defective synaptic retinoic acid (RA) signaling and subsequent defective homeostatic plasticity have emerged as a major synaptic dysfunction. However, the mechanism underlying the defective synaptic RA signaling in the absence of FMRP is unknown. Here, we show that RARα, the RA receptor critically involved in synaptic RA signaling, directly interacts with FMRP. This interaction is enhanced in the presence of RA. Blocking the interaction between FMRP and RARα with a small peptide corresponding to the critical binding site in RARα abolishes RA-induced increases in excitatory synaptic transmission, recapitulating the phenotype seen in the Fmr1 knockout mouse. Taken together, these data suggest that not only are functional FMRP and RARα necessary for RA-dependent homeostatic synaptic plasticity, but that the interaction between these two proteins is essential for proper transcription-independent RA signaling. Our results may provide further mechanistic understanding into FXS synaptic pathophysiology.
Collapse
|
20
|
The BAD-BAX-Caspase-3 Cascade Modulates Synaptic Vesicle Pools via Autophagy. J Neurosci 2021; 41:1174-1190. [PMID: 33303681 DOI: 10.1523/jneurosci.0969-20.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022] Open
Abstract
The BAD-BAX-caspase-3 cascade is a canonical apoptosis pathway. Macroautophagy ("autophagy" hereinafter) is a process by which organelles and aggregated proteins are delivered to lysosomes for degradation. Here, we report a new function of the BAD-BAX-caspase-3 cascade and autophagy in the control of synaptic vesicle pools. We found that, in hippocampal neurons of male mice, the BAD-BAX-caspase-3 pathway regulates autophagy, which in turn limits the size of synaptic vesicle pools and influences the kinetics of activity-induced depletion and recovery of synaptic vesicle pools. Moreover, the caspase-autophagy pathway is engaged by fear conditioning to facilitate associative fear learning and memory. This work identifies a new mechanism for controlling synaptic vesicle pools, and a novel, nonapoptotic, presynaptic function of the BAD-BAX-caspase-3 cascade.SIGNIFICANCE STATEMENT Despite the importance of synaptic vesicles for neurons, little is known about how the size of synaptic vesicle pools is maintained under basal conditions and regulated by neural activity. This study identifies a new mechanism for the control of synaptic vesicle pools, and a new, nonapoptotic function of the BAD-BAX-caspase-3 pathway in presynaptic terminals. Additionally, it indicates that autophagy is not only a homeostatic mechanism to maintain the integrity of cells and tissues, but also a process engaged by neural activity to regulate synaptic vesicle pools for optimal synaptic responses, learning, and memory.
Collapse
|
21
|
Optogenetic Imaging of Protein Activity Using Two-Photon Fluorescence Lifetime Imaging Microscopy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1293:295-308. [PMID: 33398821 DOI: 10.1007/978-981-15-8763-4_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Spatiotemporal dynamics of cellular proteins, including protein-protein interactions and conformational changes, is essential for understanding cellular functions such as synaptic plasticity, cell motility, and cell division. One of the best ways to understand the mechanisms of signal transduction is to visualize protein activity with high spatiotemporal resolution in living cells within tissues. Optogenetic probes such as fluorescent proteins, in combination with Förster Resonance Energy Transfer (FRET) techniques, enable the measurement of protein-protein interactions and conformational changes in response to signaling events in living cells. Of the various FRET detection systems, two-photon fluorescence lifetime imaging microscopy (2pFLIM) is one of the methods best suited to monitoring FRET in subcellular compartments of living cells located deep within tissues, such as brain slices. This review will introduce the principle of 2pFLIM-FRET and the use of chromoproteins for imaging intracellular protein activities and protein-protein interactions. Also, we will discuss two examples of 2pFLIM-FRET application: imaging actin polymerization in synapses of hippocampal neurons in brain sections and detecting small GTPase Cdc42 activity in astrocytes.
Collapse
|
22
|
Muret D, Makin TR. The homeostatic homunculus: rethinking deprivation-triggered reorganisation. Curr Opin Neurobiol 2020; 67:115-122. [PMID: 33248404 DOI: 10.1016/j.conb.2020.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/16/2022]
Abstract
While amputation was considered a prominent model for cortical reorganisation, recent evidence highlights persistent representation of the missing hand. We offer a new perspective on the literature of amputation-triggered sensorimotor plasticity, by emphasising the need for homeostasis and emerging evidence of latent activity distributed across the homunculus. We argue that deprivation uncovers pre-existing latent activity, which can manifest as remapping, but that since this activity was already there, remapping could in some instances correspond to functional stability of the system rather than reorganisation. Adaptive behaviour and Hebbian-like plasticity may also play crucial roles in maintaining the functional organisation of the homunculus when deprivation occurs in adulthood or in early development. Collectively, we suggest that the brain's need for stability may underlie several key phenotypes for brain remapping, previously interpreted as consequential to reorganisation. Nevertheless, reorganisation may still be possible, especially when cortical changes contribute to the stability of the system.
Collapse
Affiliation(s)
- Dollyane Muret
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom
| | - Tamar R Makin
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom.
| |
Collapse
|
23
|
Khlghatyan J, Evstratova A, Bozoyan L, Chamberland S, Chatterjee D, Marakhovskaia A, Soares Silva T, Toth K, Mongrain V, Beaulieu J. Fxr1 regulates sleep and synaptic homeostasis. EMBO J 2020; 39:e103864. [PMID: 32893934 PMCID: PMC7604579 DOI: 10.15252/embj.2019103864] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
The fragile X autosomal homolog 1 (Fxr1) is regulated by lithium and has been GWAS-associated with schizophrenia and insomnia. Homeostatic regulation of synaptic strength is essential for the maintenance of brain functions and involves both cell-autonomous and system-level processes such as sleep. We examined the contribution of Fxr1 to cell-autonomous homeostatic synaptic scaling and neuronal responses to sleep loss, using a combination of gene overexpression and Crispr/Cas9-mediated somatic knockouts to modulate gene expression. Our findings indicate that Fxr1 is downregulated during both scaling and sleep deprivation via a glycogen synthase kinase 3 beta (GSK3β)-dependent mechanism. In both conditions, downregulation of Fxr1 is essential for the homeostatic modulation of surface AMPA receptors and synaptic strength. Preventing the downregulation of Fxr1 during sleep deprivation results in altered EEG signatures. Furthermore, sequencing of neuronal translatomes revealed the contribution of Fxr1 to changes induced by sleep deprivation. These findings uncover a role of Fxr1 as a shared signaling hub between cell-autonomous homeostatic plasticity and system-level responses to sleep loss, with potential implications for neuropsychiatric illnesses and treatments.
Collapse
Affiliation(s)
- Jivan Khlghatyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
Department of NeuroscienceNovartis Institutes for Biomedical ResearchCambridgeMAUSA
| | - Alesya Evstratova
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Lusine Bozoyan
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Simon Chamberland
- Department of Psychiatry and NeuroscienceFaculty of MedicineUniversité LavalQuébec‐CityQCCanada
- Present address:
NYU Neuroscience InstituteLangone Medical CenterNew York UniversityNew YorkNYUSA
| | | | | | - Tiago Soares Silva
- Department of Pharmacology & ToxicologyUniversity of TorontoTorontoONCanada
| | - Katalin Toth
- Department of Cellular and Molecular MedicineFaculty of MedicineUniversity of OttawaOttawaONCanada
| | - Valerie Mongrain
- Department of NeuroscienceUniversité de Montréal and Center for Advanced Research in Sleep MedicineHôpital du Sacré‐Coeur de Montréal (CIUSSS‐NIM)MontrealQCCanada
| | | |
Collapse
|
24
|
Activity-Dependent Global Downscaling of Evoked Neurotransmitter Release across Glutamatergic Inputs in Drosophila. J Neurosci 2020; 40:8025-8041. [PMID: 32928887 DOI: 10.1523/jneurosci.0349-20.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022] Open
Abstract
Within mammalian brain circuits, activity-dependent synaptic adaptations, such as synaptic scaling, stabilize neuronal activity in the face of perturbations. Stability afforded through synaptic scaling involves uniform scaling of quantal amplitudes across all synaptic inputs formed on neurons, as well as on the postsynaptic side. It remains unclear whether activity-dependent uniform scaling also operates within peripheral circuits. We tested for such scaling in a Drosophila larval neuromuscular circuit, where the muscle receives synaptic inputs from different motoneurons. We used motoneuron-specific genetic manipulations to increase the activity of only one motoneuron and recordings of postsynaptic currents from inputs formed by the different motoneurons. We discovered an adaptation which caused uniform downscaling of evoked neurotransmitter release across all inputs through decreases in release probabilities. This "presynaptic downscaling" maintained the relative differences in neurotransmitter release across all inputs around a homeostatic set point, caused a compensatory decrease in synaptic drive to the muscle affording robust and stable muscle activity, and was induced within hours. Presynaptic downscaling was associated with an activity-dependent increase in Drosophila vesicular glutamate transporter expression. Activity-dependent uniform scaling can therefore manifest also on the presynaptic side to produce robust and stable circuit outputs. Within brain circuits, uniform downscaling on the postsynaptic side is implicated in sleep- and memory-related processes. Our results suggest that evaluation of such processes might be broadened to include uniform downscaling on the presynaptic side.SIGNIFICANCE STATEMENT To date, compensatory adaptations which stabilise target cell activity through activity-dependent global scaling have been observed only within central circuits, and on the postsynaptic side. Considering that maintenance of stable activity is imperative for the robust function of the nervous system as a whole, we tested whether activity-dependent global scaling could also manifest within peripheral circuits. We uncovered a compensatory adaptation which causes global scaling within a peripheral circuit and on the presynaptic side through uniform downscaling of evoked neurotransmitter release. Unlike in central circuits, uniform scaling maintains functionality over a wide, rather than a narrow, operational range, affording robust and stable activity. Activity-dependent global scaling therefore operates on both the presynaptic and postsynaptic sides to maintain target cell activity.
Collapse
|
25
|
Camporesi E, Nilsson J, Brinkmalm A, Becker B, Ashton NJ, Blennow K, Zetterberg H. Fluid Biomarkers for Synaptic Dysfunction and Loss. Biomark Insights 2020; 15:1177271920950319. [PMID: 32913390 PMCID: PMC7444114 DOI: 10.1177/1177271920950319] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Synapses are the site for brain communication where information is transmitted between neurons and stored for memory formation. Synaptic degeneration is a global and early pathogenic event in neurodegenerative disorders with reduced levels of pre- and postsynaptic proteins being recognized as a core feature of Alzheimer's disease (AD) pathophysiology. Together with AD, other neurodegenerative and neurodevelopmental disorders show altered synaptic homeostasis as an important pathogenic event, and due to that, they are commonly referred to as synaptopathies. The exact mechanisms of synapse dysfunction in the different diseases are not well understood and their study would help understanding the pathogenic role of synaptic degeneration, as well as differences and commonalities among them and highlight candidate synaptic biomarkers for specific disorders. The assessment of synaptic proteins in cerebrospinal fluid (CSF), which can reflect synaptic dysfunction in patients with cognitive disorders, is a keen area of interest. Substantial research efforts are now directed toward the investigation of CSF synaptic pathology to improve the diagnosis of neurodegenerative disorders at an early stage as well as to monitor clinical progression. In this review, we will first summarize the pathological events that lead to synapse loss and then discuss the available data on established (eg, neurogranin, SNAP-25, synaptotagmin-1, GAP-43, and α-syn) and emerging (eg, synaptic vesicle glycoprotein 2A and neuronal pentraxins) CSF biomarkers for synapse dysfunction, while highlighting possible utilities, disease specificity, and technical challenges for their detection.
Collapse
Affiliation(s)
- Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Nilsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ann Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Bruno Becker
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- King’s College London, Institute of Psychiatry, Psychology & Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
- Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
| |
Collapse
|
26
|
Bachmann C, Tetzlaff T, Duarte R, Morrison A. Firing rate homeostasis counteracts changes in stability of recurrent neural networks caused by synapse loss in Alzheimer's disease. PLoS Comput Biol 2020; 16:e1007790. [PMID: 32841234 PMCID: PMC7505475 DOI: 10.1371/journal.pcbi.1007790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/21/2020] [Accepted: 03/17/2020] [Indexed: 11/19/2022] Open
Abstract
The impairment of cognitive function in Alzheimer's disease is clearly correlated to synapse loss. However, the mechanisms underlying this correlation are only poorly understood. Here, we investigate how the loss of excitatory synapses in sparsely connected random networks of spiking excitatory and inhibitory neurons alters their dynamical characteristics. Beyond the effects on the activity statistics, we find that the loss of excitatory synapses on excitatory neurons reduces the network's sensitivity to small perturbations. This decrease in sensitivity can be considered as an indication of a reduction of computational capacity. A full recovery of the network's dynamical characteristics and sensitivity can be achieved by firing rate homeostasis, here implemented by an up-scaling of the remaining excitatory-excitatory synapses. Mean-field analysis reveals that the stability of the linearised network dynamics is, in good approximation, uniquely determined by the firing rate, and thereby explains why firing rate homeostasis preserves not only the firing rate but also the network's sensitivity to small perturbations.
Collapse
Affiliation(s)
- Claudia Bachmann
- Institute of Neuroscience and Medicine (INM-6) and Institute for Advanced Simulation (IAS-6) and JARA BRAIN Institute I, Jülich Research Centre, Jülich, Germany
| | - Tom Tetzlaff
- Institute of Neuroscience and Medicine (INM-6) and Institute for Advanced Simulation (IAS-6) and JARA BRAIN Institute I, Jülich Research Centre, Jülich, Germany
| | - Renato Duarte
- Institute of Neuroscience and Medicine (INM-6) and Institute for Advanced Simulation (IAS-6) and JARA BRAIN Institute I, Jülich Research Centre, Jülich, Germany
| | - Abigail Morrison
- Institute of Neuroscience and Medicine (INM-6) and Institute for Advanced Simulation (IAS-6) and JARA BRAIN Institute I, Jülich Research Centre, Jülich, Germany
- Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
27
|
Verhage M, Sørensen JB. SNAREopathies: Diversity in Mechanisms and Symptoms. Neuron 2020; 107:22-37. [PMID: 32559416 DOI: 10.1016/j.neuron.2020.05.036] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/29/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Neuronal SNAREs and their key regulators together drive synaptic vesicle exocytosis and synaptic transmission as a single integrated membrane fusion machine. Human pathogenic mutations have now been reported for all eight core components, but patients are diagnosed with very different neurodevelopmental syndromes. We propose to unify these syndromes, based on etiology and mechanism, as "SNAREopathies." Here, we review the strikingly diverse clinical phenomenology and disease severity and the also remarkably diverse genetic mechanisms. We argue that disease severity generally scales with functional redundancy and, conversely, that the large effect of mutations in some SNARE genes is the price paid for extensive integration and exceptional specialization. Finally, we discuss how subtle differences in components being rate limiting in different types of neurons helps to explain the main symptoms.
Collapse
Affiliation(s)
- Matthijs Verhage
- Department of Functional Genomics, Vrije Universiteit (VU) Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands; Department of Clinical Genetics, UMC Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands.
| | - Jakob B Sørensen
- Department of Neuroscience, University of Copenhagen, 2200 Copenhagen N, Denmark.
| |
Collapse
|
28
|
Differential Scaling of Synaptic Molecules within Functional Zones of an Excitatory Synapse during Homeostatic Plasticity. eNeuro 2020; 7:ENEURO.0407-19.2020. [PMID: 32184300 PMCID: PMC7189482 DOI: 10.1523/eneuro.0407-19.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 11/29/2022] Open
Abstract
Homeostatic scaling is a form of synaptic plasticity where individual synapses scale their strengths to compensate for global suppression or elevation of neuronal activity. This process can be studied by measuring miniature EPSP (mEPSP) amplitudes and frequencies following the regulation of activity in neuronal cultures. Here, we demonstrate a quantitative approach to characterize multiplicative synaptic scaling using immunolabelling of hippocampal neuronal cultures treated with tetrodotoxin (TTX) or bicuculline to extract scaling factors for various synaptic proteins. This approach allowed us to directly examine the scaling of presynaptic and postsynaptic scaffolding molecules along with neurotransmitter receptors in primary cultures from mouse and rat hippocampal neurons. We show robust multiplicative scaling of synaptic scaffolding molecules namely, Shank2, PSD95, Bassoon, and AMPA receptor subunits and quantify their scaling factors. We use super-resolution microscopy to calculate scaling factors of surface expressed GluA2 within functional zones of the synapse and show that there is differential and correlated scaling of GluA2 levels within the spine, the postsynaptic density (PSD), and the perisynaptic regions. Our method opens a novel paradigm to quantify relative molecular changes of synaptic proteins within distinct subsynaptic compartments from a large number of synapses in response to alteration of neuronal activity, providing anatomic insights into the intricacies of variability in strength of individual synapses.
Collapse
|
29
|
Dörrbaum AR, Alvarez-Castelao B, Nassim-Assir B, Langer JD, Schuman EM. Proteome dynamics during homeostatic scaling in cultured neurons. eLife 2020; 9:e52939. [PMID: 32238265 PMCID: PMC7117909 DOI: 10.7554/elife.52939] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/22/2020] [Indexed: 12/11/2022] Open
Abstract
Protein turnover, the net result of protein synthesis and degradation, enables cells to remodel their proteomes in response to internal and external cues. Previously, we analyzed protein turnover rates in cultured brain cells under basal neuronal activity and found that protein turnover is influenced by subcellular localization, protein function, complex association, cell type of origin, and by the cellular environment (Dörrbaum et al., 2018). Here, we advanced our experimental approach to quantify changes in protein synthesis and degradation, as well as the resulting changes in protein turnover or abundance in rat primary hippocampal cultures during homeostatic scaling. Our data demonstrate that a large fraction of the neuronal proteome shows changes in protein synthesis and/or degradation during homeostatic up- and down-scaling. More than half of the quantified synaptic proteins were regulated, including pre- as well as postsynaptic proteins with diverse molecular functions.
Collapse
Affiliation(s)
- Aline Ricarda Dörrbaum
- Max Planck Institute for Brain Research, Frankfurt, Germany
- Goethe University Frankfurt, Faculty of Biological Sciences, Frankfurt, Germany
| | | | | | - Julian D Langer
- Max Planck Institute for Brain Research, Frankfurt, Germany
- Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt, Germany
| |
Collapse
|
30
|
Dubes S, Favereaux A, Thoumine O, Letellier M. miRNA-Dependent Control of Homeostatic Plasticity in Neurons. Front Cell Neurosci 2019; 13:536. [PMID: 31866828 PMCID: PMC6906196 DOI: 10.3389/fncel.2019.00536] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/19/2019] [Indexed: 11/13/2022] Open
Abstract
Homeostatic plasticity is a form of plasticity in which neurons compensate for changes in neuronal activity through the control of key physiological parameters such as the number and the strength of their synaptic inputs and intrinsic excitability. Recent studies revealed that miRNAs, which are small non-coding RNAs repressing mRNA translation, participate in this process by controlling the translation of multiple effectors such as glutamate transporters, receptors, signaling molecules and voltage-gated ion channels. In this review, we present and discuss the role of miRNAs in both cell-wide and compartmentalized forms of homeostatic plasticity as well as their implication in pathological processes associated with homeostatic failure.
Collapse
Affiliation(s)
- Sandra Dubes
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Alexandre Favereaux
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Olivier Thoumine
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - Mathieu Letellier
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| |
Collapse
|
31
|
Palaniyappan L. Inefficient neural system stabilization: a theory of spontaneous resolutions and recurrent relapses in psychosis. J Psychiatry Neurosci 2019; 44:367-383. [PMID: 31245961 PMCID: PMC6821513 DOI: 10.1503/jpn.180038] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 02/07/2019] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
A striking feature of psychosis is its heterogeneity. Presentations of psychosis vary from transient symptoms with no functional consequence in the general population to a tenacious illness at the other extreme, with a wide range of variable trajectories in between. Even among patients with schizophrenia, who are diagnosed on the basis of persistent deterioration, marked variation is seen in response to treatment, frequency of relapses and degree of eventual recovery. Existing theoretical accounts of psychosis focus almost exclusively on how symptoms are initially formed, with much less emphasis on explaining their variable course. In this review, I present an account that links several existing notions of the biology of psychosis with the variant clinical trajectories. My aim is to incorporate perspectives of systems neuroscience in a staging framework to explain the individual variations in illness course that follow the onset of psychosis.
Collapse
Affiliation(s)
- Lena Palaniyappan
- From the Department of Psychiatry and Robarts Research Institute, University of Western Ontario and Lawson Health Research Institute, London, Ont., Canada
| |
Collapse
|
32
|
Emerging Roles of Synapse Organizers in the Regulation of Critical Periods. Neural Plast 2019; 2019:1538137. [PMID: 31565044 PMCID: PMC6745111 DOI: 10.1155/2019/1538137] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/09/2019] [Accepted: 07/25/2019] [Indexed: 01/10/2023] Open
Abstract
Experience remodels cortical connectivity during developmental windows called critical periods. Experience-dependent regulation of synaptic strength during these periods establishes circuit functions that are stabilized as critical period plasticity wanes. These processes have been extensively studied in the developing visual cortex, where critical period opening and closure are orchestrated by the assembly, maturation, and strengthening of distinct synapse types. The synaptic specificity of these processes points towards the involvement of distinct molecular pathways. Attractive candidates are pre- and postsynaptic transmembrane proteins that form adhesive complexes across the synaptic cleft. These synapse-organizing proteins control synapse development and maintenance and modulate structural and functional properties of synapses. Recent evidence suggests that they have pivotal roles in the onset and closure of the critical period for vision. In this review, we describe roles of synapse-organizing adhesion molecules in the regulation of visual critical period plasticity and we discuss the potential they offer to restore circuit functions in amblyopia and other neurodevelopmental disorders.
Collapse
|
33
|
Wang G, Zhong J, Guttieres D, Man HY. Non-scaling regulation of AMPA receptors in homeostatic synaptic plasticity. Neuropharmacology 2019; 158:107700. [PMID: 31283924 DOI: 10.1016/j.neuropharm.2019.107700] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/10/2019] [Accepted: 07/04/2019] [Indexed: 11/28/2022]
Abstract
Homeostatic synaptic plasticity (HSP) as an activity-dependent negative feedback regulation of synaptic strength plays important roles in the maintenance of neuronal and neural circuitry stability. A primary cellular substrate for HSP expression is alterations in synaptic accumulation of glutamatergic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR). It is widely believed that during HSP, AMPAR accumulation changes with the same proportion at each synapse of a neuron, a process known as synaptic scaling. However, direct evidence on AMPAR synaptic scaling remains largely lacking. Here we report a direct examination of inactivity-induced homeostatic scaling of AMPAR at individual synapse by live-imaging. Surprisingly, instead of uniform up-scaling, a scattered pattern of changes in synaptic AMPAR was observed in cultured rat hippocampal neurons. While the majority of synapses showed up-regulation after activity inhibition, a reduction of AMPAR could be detected in certain synapses. More importantly, among the up-regulated synapses, a wide range of AMPAR changes was observed in synapses of the same neuron. We also found that synapses with higher levels of pre-existing AMPAR tend to be up-regulated by lesser extents, whereas the locations of synapses relative to the soma seem not affecting AMPAR scaling strengths. In addition, we observed strong competition between neighboring synapses during HSP. These results reveal that synaptic AMPAR may not be scaled during HSP, suggesting novel molecular mechanisms for information processing and storage at synapses.
Collapse
Affiliation(s)
- Guan Wang
- Department of Biology, Boston University, Boston, MA, USA; School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jia Zhong
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA; Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
34
|
Letellier M, Levet F, Thoumine O, Goda Y. Differential role of pre- and postsynaptic neurons in the activity-dependent control of synaptic strengths across dendrites. PLoS Biol 2019; 17:e2006223. [PMID: 31166943 PMCID: PMC6576792 DOI: 10.1371/journal.pbio.2006223] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/17/2019] [Accepted: 05/17/2019] [Indexed: 01/07/2023] Open
Abstract
Neurons receive a large number of active synaptic inputs from their many presynaptic partners across their dendritic tree. However, little is known about how the strengths of individual synapses are controlled in balance with other synapses to effectively encode information while maintaining network homeostasis. This is in part due to the difficulty in assessing the activity of individual synapses with identified afferent and efferent connections for a synapse population in the brain. Here, to gain insights into the basic cellular rules that drive the activity-dependent spatial distribution of pre- and postsynaptic strengths across incoming axons and dendrites, we combine patch-clamp recordings with live-cell imaging of hippocampal pyramidal neurons in dissociated cultures and organotypic slices. Under basal conditions, both pre- and postsynaptic strengths cluster on single dendritic branches according to the identity of the presynaptic neurons, thus highlighting the ability of single dendritic branches to exhibit input specificity. Stimulating a single presynaptic neuron induces input-specific and dendritic branchwise spatial clustering of presynaptic strengths, which accompanies a widespread multiplicative scaling of postsynaptic strengths in dissociated cultures and heterosynaptic plasticity at distant synapses in organotypic slices. Our study provides evidence for a potential homeostatic mechanism by which the rapid changes in global or distant postsynaptic strengths compensate for input-specific presynaptic plasticity.
Collapse
Affiliation(s)
- Mathieu Letellier
- RIKEN Brain Science Institute, Wako, Saitama, Japan
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, France
- * E-mail: (ML); (YG)
| | - Florian Levet
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, France
- Bordeaux Imaging Center, University of Bordeaux, Bordeaux, France
- Bordeaux Imaging Center, CNRS UMS 3420, Bordeaux, France
- Bordeaux Imaging Center, INSERM US04, Bordeaux, France
| | - Olivier Thoumine
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, Centre National de la Recherche Scientifique (CNRS) UMR 5297, Bordeaux, France
| | - Yukiko Goda
- RIKEN Center for Brain Science, Wako, Saitama, Japan
- * E-mail: (ML); (YG)
| |
Collapse
|
35
|
Sheppard O, Coleman MP, Durrant CS. Lipopolysaccharide-induced neuroinflammation induces presynaptic disruption through a direct action on brain tissue involving microglia-derived interleukin 1 beta. J Neuroinflammation 2019; 16:106. [PMID: 31103036 PMCID: PMC6525970 DOI: 10.1186/s12974-019-1490-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/29/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Systemic inflammation has been linked to synapse loss and cognitive decline in human patients and animal models. A role for microglial release of pro-inflammatory cytokines has been proposed based on in vivo and primary culture studies. However, mechanisms are hard to study in vivo as specific microglial ablation is challenging and the extracellular fluid cannot be sampled without invasive methods. Primary cultures have different limitations as the intricate multicellular architecture in the brain is not fully reproduced. It is essential to confirm proposed brain-specific mechanisms of inflammatory synapse loss directly in brain tissue. Organotypic hippocampal slice cultures (OHSCs) retain much of the in vivo neuronal architecture, synaptic connections and diversity of cell types whilst providing convenient access to manipulate and sample the culture medium and observe cellular reactions. METHODS OHSCs were generated from P6-P9 C57BL/6 mice. Inflammation was induced via addition of lipopolysaccharide (LPS), and cultures were analysed for changes in synaptic proteins, gene expression and protein secretion. Microglia were selectively depleted using clodronate, and the effect of IL1β was assessed using a specific neutralising monoclonal antibody. RESULTS LPS treatment induced loss of the presynaptic protein synaptophysin without altering PSD95 or Aβ protein levels. Depletion of microglia prior to LPS application prevented the loss of synaptophysin, whilst microglia depletion after the inflammatory insult was partially effective, although less so than pre-emptive treatment, indicating a time-critical window in which microglia can induce synaptic damage. IL1β protein and mRNA were increased after LPS addition, with these effects also prevented by microglia depletion. Direct application of IL1β to OHSCs resulted in synaptophysin loss whilst pre-treatment with IL1β neutralising antibody prior to LPS addition prevented a significant loss of synaptophysin but may also impact basal synaptic levels. CONCLUSIONS The loss of synaptophysin in this system confirms LPS can act directly within brain tissue to disrupt synapses, and we show that microglia are the relevant cellular target when all major CNS cell types are present. By overcoming limitations of primary culture and in vivo work, our study strengthens the evidence for a key role of microglia-derived IL1β in synaptic dysfunction after inflammatory insult.
Collapse
Affiliation(s)
- Olivia Sheppard
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.,Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Claire S Durrant
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D Adrian Building, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK. .,Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, CB22 3AT, UK.
| |
Collapse
|
36
|
Synaptic retinoic acid receptor signaling mediates mTOR-dependent metaplasticity that controls hippocampal learning. Proc Natl Acad Sci U S A 2019; 116:7113-7122. [PMID: 30782829 DOI: 10.1073/pnas.1820690116] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Homeostatic synaptic plasticity is a stabilizing mechanism engaged by neural circuits in response to prolonged perturbation of network activity. The non-Hebbian nature of homeostatic synaptic plasticity is thought to contribute to network stability by preventing "runaway" Hebbian plasticity at individual synapses. However, whether blocking homeostatic synaptic plasticity indeed induces runaway Hebbian plasticity in an intact neural circuit has not been explored. Furthermore, how compromised homeostatic synaptic plasticity impacts animal learning remains unclear. Here, we show in mice that the experience of an enriched environment (EE) engaged homeostatic synaptic plasticity in hippocampal circuits, thereby reducing excitatory synaptic transmission. This process required RARα, a nuclear retinoic acid receptor that doubles as a cytoplasmic retinoic acid-induced postsynaptic regulator of protein synthesis. Blocking RARα-dependent homeostatic synaptic plasticity during an EE experience by ablating RARα signaling induced runaway Hebbian plasticity, as evidenced by greatly enhanced long-term potentiation (LTP). As a consequence, RARα deletion in hippocampal circuits during an EE experience resulted in enhanced spatial learning but suppressed learning flexibility. In the absence of RARα, moreover, EE experience superactivated mammalian target of rapamycin (mTOR) signaling, causing a shift in protein translation that enhanced the expression levels of AMPA-type glutamate receptors. Treatment of mice with the mTOR inhibitor rapamycin during an EE experience not only restored normal AMPA-receptor expression levels but also reversed the increases in runaway Hebbian plasticity and learning after hippocampal RARα deletion. Thus, our findings reveal an RARα- and mTOR-dependent mechanism by which homeostatic plasticity controls Hebbian plasticity and learning.
Collapse
|
37
|
Susi G, Antón Toro L, Canuet L, López ME, Maestú F, Mirasso CR, Pereda E. A Neuro-Inspired System for Online Learning and Recognition of Parallel Spike Trains, Based on Spike Latency, and Heterosynaptic STDP. Front Neurosci 2018; 12:780. [PMID: 30429767 PMCID: PMC6220070 DOI: 10.3389/fnins.2018.00780] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/09/2018] [Indexed: 11/17/2022] Open
Abstract
Humans perform remarkably well in many cognitive tasks including pattern recognition. However, the neuronal mechanisms underlying this process are not well understood. Nevertheless, artificial neural networks, inspired in brain circuits, have been designed and used to tackle spatio-temporal pattern recognition tasks. In this paper we present a multi-neuronal spike pattern detection structure able to autonomously implement online learning and recognition of parallel spike sequences (i.e., sequences of pulses belonging to different neurons/neural ensembles). The operating principle of this structure is based on two spiking/synaptic neurocomputational characteristics: spike latency, which enables neurons to fire spikes with a certain delay and heterosynaptic plasticity, which allows the own regulation of synaptic weights. From the perspective of the information representation, the structure allows mapping a spatio-temporal stimulus into a multi-dimensional, temporal, feature space. In this space, the parameter coordinate and the time at which a neuron fires represent one specific feature. In this sense, each feature can be considered to span a single temporal axis. We applied our proposed scheme to experimental data obtained from a motor-inhibitory cognitive task. The results show that out method exhibits similar performance compared with other classification methods, indicating the effectiveness of our approach. In addition, its simplicity and low computational cost suggest a large scale implementation for real time recognition applications in several areas, such as brain computer interface, personal biometrics authentication, or early detection of diseases.
Collapse
Affiliation(s)
- Gianluca Susi
- UCM-UPM Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Dipartimento di Ingegneria Civile e Ingegneria Informatica, Università di Roma 'Tor Vergata', Rome, Italy
| | - Luis Antón Toro
- UCM-UPM Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Departamento de Psicología Experimental, Procesos Cognitivos y Logopedia, Facultad de Psicología, Universidad Complutense de Madrid, Madrid, Spain
| | - Leonides Canuet
- UCM-UPM Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Departamento de Psicología Clinica, Psicobiología y Metodología, Universidad de La Laguna, La Laguna, Spain
| | - Maria Eugenia López
- UCM-UPM Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Departamento de Psicología Experimental, Procesos Cognitivos y Logopedia, Facultad de Psicología, Universidad Complutense de Madrid, Madrid, Spain
| | - Fernando Maestú
- UCM-UPM Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Departamento de Psicología Experimental, Procesos Cognitivos y Logopedia, Facultad de Psicología, Universidad Complutense de Madrid, Madrid, Spain
| | - Claudio R Mirasso
- Instituto de Fisica Interdisciplinar y Sistemas Complejos, CSIC-UIB, Campus Universitat de les Illes Balears, Palma de Mallorca, Spain
| | - Ernesto Pereda
- UCM-UPM Laboratory of Cognitive and Computational Neuroscience, Center for Biomedical Technology, Technical University of Madrid, Madrid, Spain.,Departamento de Ingeniería Industrial, Escuela Superior de Ingeniería y Tecnología & IUNE, Universidad de La Laguna, La Laguna, Spain
| |
Collapse
|
38
|
Removal of area CA3 from hippocampal slices induces postsynaptic plasticity at Schaffer collateral synapses that normalizes CA1 pyramidal cell discharge. Neurosci Lett 2018; 678:55-61. [PMID: 29738844 DOI: 10.1016/j.neulet.2018.05.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/24/2018] [Accepted: 05/04/2018] [Indexed: 11/23/2022]
Abstract
Neural networks that undergo acute insults display remarkable reorganization. This injury related plasticity is thought to permit recovery of function in the face of damage that cannot be reversed. Previously, an increase in the transmission strength at Schaffer collateral to CA1 pyramidal cell synapses was observed after long-term activity reduction in organotypic hippocampal slices. Here we report that, following acute preparation of adult rat hippocampal slices and surgical removal of area CA3, input to area CA1 was reduced and Schaffer collateral synapses underwent functional strengthening. This increase in synaptic strength was limited to Schaffer collateral inputs (no alteration to temporoammonic synapses) and acted to normalize postsynaptic discharge, supporting a homeostatic or compensatory response. Short-term plasticity was not altered, but an increase in immunohistochemical labeling of GluA1 subunits was observed in the stratum radiatum (but not stratum moleculare), suggesting increased numbers of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors and a postsynaptic locus of expression. Combined, these data support the idea that, in response to the reduction in presynaptic activity caused by removal of area CA3, Schaffer collateral synapses undergo a relatively rapid increase in functional efficacy likely supported by insertion of more AMPARs, which maintains postsynaptic excitability in CA1 pyramidal neurons. This novel fast compensatory plasticity exhibits properties that would allow it to maintain optimal network activity levels in the hippocampus, a brain structure lauded for its ongoing experience-dependent malleability.
Collapse
|
39
|
Microscopic Study of Nervous System Plasticity: Interactions of Sympathetic Nerves with Neurons of Intraocular Hippocampal Transplants. Bull Exp Biol Med 2018; 164:680-684. [DOI: 10.1007/s10517-018-4058-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Indexed: 10/17/2022]
|
40
|
Homeostatic Plasticity in the Hippocampus Facilitates Memory Extinction. Cell Rep 2018; 22:1451-1461. [DOI: 10.1016/j.celrep.2018.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 11/25/2017] [Accepted: 01/09/2018] [Indexed: 11/19/2022] Open
|
41
|
Yee AX, Hsu YT, Chen L. A metaplasticity view of the interaction between homeostatic and Hebbian plasticity. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0155. [PMID: 28093549 DOI: 10.1098/rstb.2016.0155] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2016] [Indexed: 01/25/2023] Open
Abstract
Hebbian and homeostatic plasticity are two major forms of plasticity in the nervous system: Hebbian plasticity provides a synaptic basis for associative learning, whereas homeostatic plasticity serves to stabilize network activity. While achieving seemingly very different goals, these two types of plasticity interact functionally through overlapping elements in their respective mechanisms. Here, we review studies conducted in the mammalian central nervous system, summarize known circuit and molecular mechanisms of homeostatic plasticity, and compare these mechanisms with those that mediate Hebbian plasticity. We end with a discussion of 'local' homeostatic plasticity and the potential role of local homeostatic plasticity as a form of metaplasticity that modulates a neuron's future capacity for Hebbian plasticity.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
Affiliation(s)
- Ada X Yee
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Yu-Tien Hsu
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Lu Chen
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| |
Collapse
|
42
|
Yiannakas A, Rosenblum K. The Insula and Taste Learning. Front Mol Neurosci 2017; 10:335. [PMID: 29163022 PMCID: PMC5676397 DOI: 10.3389/fnmol.2017.00335] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/03/2017] [Indexed: 12/29/2022] Open
Abstract
The sense of taste is a key component of the sensory machinery, enabling the evaluation of both the safety as well as forming associations regarding the nutritional value of ingestible substances. Indicative of the salience of the modality, taste conditioning can be achieved in rodents upon a single pairing of a tastant with a chemical stimulus inducing malaise. This robust associative learning paradigm has been heavily linked with activity within the insular cortex (IC), among other regions, such as the amygdala and medial prefrontal cortex. A number of studies have demonstrated taste memory formation to be dependent on protein synthesis at the IC and to correlate with the induction of signaling cascades involved in synaptic plasticity. Taste learning has been shown to require the differential involvement of dopaminergic GABAergic, glutamatergic, muscarinic neurotransmission across an extended taste learning circuit. The subsequent activation of downstream protein kinases (ERK, CaMKII), transcription factors (CREB, Elk-1) and immediate early genes (c-fos, Arc), has been implicated in the regulation of the different phases of taste learning. This review discusses the relevant neurotransmission, molecular signaling pathways and genetic markers involved in novel and aversive taste learning, with a particular focus on the IC. Imaging and other studies in humans have implicated the IC in the pathophysiology of a number of cognitive disorders. We conclude that the IC participates in circuit-wide computations that modulate the interception and encoding of sensory information, as well as the formation of subjective internal representations that control the expression of motivated behaviors.
Collapse
Affiliation(s)
- Adonis Yiannakas
- Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
| | - Kobi Rosenblum
- Sagol Department of Neuroscience, University of Haifa, Haifa, Israel
- Center for Gene Manipulation in the Brain, University of Haifa, Haifa, Israel
| |
Collapse
|
43
|
Malviya M, Barman S, Golombeck KS, Planagumà J, Mannara F, Strutz-Seebohm N, Wrzos C, Demir F, Baksmeier C, Steckel J, Falk KK, Gross CC, Kovac S, Bönte K, Johnen A, Wandinger KP, Martín-García E, Becker AJ, Elger CE, Klöcker N, Wiendl H, Meuth SG, Hartung HP, Seebohm G, Leypoldt F, Maldonado R, Stadelmann C, Dalmau J, Melzer N, Goebels N. NMDAR encephalitis: passive transfer from man to mouse by a recombinant antibody. Ann Clin Transl Neurol 2017; 4:768-783. [PMID: 29159189 PMCID: PMC5682115 DOI: 10.1002/acn3.444] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/11/2022] Open
Abstract
Objective Autoimmune encephalitis is most frequently associated with anti‐NMDAR autoantibodies. Their pathogenic relevance has been suggested by passive transfer of patients' cerebrospinal fluid (CSF) in mice in vivo. We aimed to analyze the intrathecal plasma cell repertoire, identify autoantibody‐producing clones, and characterize their antibody signatures in recombinant form. Methods Patients with recent onset typical anti‐NMDAR encephalitis were subjected to flow cytometry analysis of the peripheral and intrathecal immune response before, during, and after immunotherapy. Recombinant human monoclonal antibodies (rhuMab) were cloned and expressed from matching immunoglobulin heavy‐ (IgH) and light‐chain (IgL) amplicons of clonally expanded intrathecal plasma cells (cePc) and tested for their pathogenic relevance. Results Intrathecal accumulation of B and plasma cells corresponded to the clinical course. The presence of cePc with hypermutated antigen receptors indicated an antigen‐driven intrathecal immune response. Consistently, a single recombinant human GluN1‐specific monoclonal antibody, rebuilt from intrathecal cePc, was sufficient to reproduce NMDAR epitope specificity in vitro. After intraventricular infusion in mice, it accumulated in the hippocampus, decreased synaptic NMDAR density, and caused severe reversible memory impairment, a key pathogenic feature of the human disease, in vivo. Interpretation A CNS‐specific humoral immune response is present in anti‐NMDAR encephalitis specifically targeting the GluN1 subunit of the NMDAR. Using reverse genetics, we recovered the typical intrathecal antibody signature in recombinant form, and proved its pathogenic relevance by passive transfer of disease symptoms from man to mouse, providing the critical link between intrathecal immune response and the pathogenesis of anti‐NMDAR encephalitis as a humorally mediated autoimmune disease.
Collapse
Affiliation(s)
- Manish Malviya
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany.,Present address: Centre Physiopathologie de Toulouse-Purpan Université Toulouse III Toulouse France
| | - Sumanta Barman
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany
| | | | - Jesús Planagumà
- Institut d'Investigació Biomèdica August Pi i Sunyer University of Barcelona Barcelona Spain
| | - Francesco Mannara
- Institut d'Investigació Biomèdica August Pi i Sunyer University of Barcelona Barcelona Spain.,Laboratori de Neurofarmacologia Universitat Pompeu Fabra Facultat de Ciències de la Salut i de la Vida Barcelona Spain
| | | | - Claudia Wrzos
- Institute of Neuropathology University of Göttingen Göttingen Germany
| | - Fatih Demir
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany.,Present address: Forschungszentrum Jülich Jülich Germany
| | - Christine Baksmeier
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany
| | - Julia Steckel
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany
| | - Kim Kristin Falk
- Institute of Clinical Chemistry and Department of Neurology University Hospital of Schleswig-Holstein Lübeck/Kiel Schleswig-Holstein Germany
| | | | - Stjepana Kovac
- Department of Neurology University of Münster Münster Germany
| | - Kathrin Bönte
- Department of Neurology University of Münster Münster Germany
| | - Andreas Johnen
- Department of Neurology University of Münster Münster Germany
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry and Department of Neurology University Hospital of Schleswig-Holstein Lübeck/Kiel Schleswig-Holstein Germany
| | - Elena Martín-García
- Laboratori de Neurofarmacologia Universitat Pompeu Fabra Facultat de Ciències de la Salut i de la Vida Barcelona Spain
| | - Albert J Becker
- Department of Neuropathology University of Bonn Bonn Germany
| | | | - Nikolaj Klöcker
- Institute of Neurophysiology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany
| | - Heinz Wiendl
- Department of Neurology University of Münster Münster Germany
| | - Sven G Meuth
- Department of Neurology University of Münster Münster Germany
| | - Hans-Peter Hartung
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH) University of Münster Münster Germany
| | - Frank Leypoldt
- Institute of Clinical Chemistry and Department of Neurology University Hospital of Schleswig-Holstein Lübeck/Kiel Schleswig-Holstein Germany
| | - Rafael Maldonado
- Laboratori de Neurofarmacologia Universitat Pompeu Fabra Facultat de Ciències de la Salut i de la Vida Barcelona Spain
| | | | - Josep Dalmau
- Institut d'Investigació Biomèdica August Pi i Sunyer University of Barcelona Barcelona Spain.,Catalan Institution for Research and Advanced Studies Barcelona Spain.,Department of Neurology University of Pennsylvania Philadelphia Pennsylvania
| | - Nico Melzer
- Department of Neurology University of Münster Münster Germany
| | - Norbert Goebels
- Department of Neurology Medical Faculty Heinrich Heine University Düsseldorf Düsseldorf Germany
| |
Collapse
|
44
|
Chang CW, Wilkerson JR, Hale CF, Gibson JR, Huber KM. Distinct stages of synapse elimination are induced by burst firing of CA1 neurons and differentially require MEF2A/D. eLife 2017; 6:26278. [PMID: 28901289 PMCID: PMC5608508 DOI: 10.7554/elife.26278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 09/05/2017] [Indexed: 12/26/2022] Open
Abstract
Experience and activity refine cortical circuits through synapse elimination, but little is known about the activity patterns and downstream molecular mechanisms that mediate this process. We used optogenetics to drive individual mouse CA1 hippocampal neurons to fire in theta frequency bursts to understand how cell autonomous, postsynaptic activity leads to synapse elimination. Brief (1 hr) periods of postsynaptic bursting selectively depressed AMPA receptor (R) synaptic transmission, or silenced excitatory synapses, whereas more prolonged (24 hr) firing depressed both AMPAR and NMDAR EPSCs and eliminated spines, indicative of a synapse elimination. Both synapse silencing and elimination required de novo transcription, but only silencing required the activity-dependent transcription factors MEF2A/D. Burst firing induced MEF2A/D-dependent induction of the target gene Arc which contributed to synapse silencing and elimination. This work reveals new and distinct forms of activity and transcription-dependent synapse depression and suggests that these processes can occur independently.
Collapse
Affiliation(s)
- Chia-Wei Chang
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
| | - Julia R Wilkerson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
| | - Carly F Hale
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jay R Gibson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kimberly M Huber
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
45
|
Hoxha E, Lippiello P, Scelfo B, Tempia F, Ghirardi M, Miniaci MC. Maturation, Refinement, and Serotonergic Modulation of Cerebellar Cortical Circuits in Normal Development and in Murine Models of Autism. Neural Plast 2017; 2017:6595740. [PMID: 28894610 PMCID: PMC5574313 DOI: 10.1155/2017/6595740] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/06/2017] [Accepted: 07/25/2017] [Indexed: 12/13/2022] Open
Abstract
The formation of the complex cerebellar cortical circuits follows different phases, with initial synaptogenesis and subsequent processes of refinement guided by a variety of mechanisms. The regularity of the cellular and synaptic organization of the cerebellar cortex allowed detailed studies of the structural plasticity mechanisms underlying the formation of new synapses and retraction of redundant ones. For the attainment of the monoinnervation of the Purkinje cell by a single climbing fiber, several signals are involved, including electrical activity, contact signals, homosynaptic and heterosynaptic interaction, calcium transients, postsynaptic receptors, and transduction pathways. An important role in this developmental program is played by serotonergic projections that, acting on temporally and spatially regulated postsynaptic receptors, induce and modulate the phases of synaptic formation and maturation. In the adult cerebellar cortex, many developmental mechanisms persist but play different roles, such as supporting synaptic plasticity during learning and formation of cerebellar memory traces. A dysfunction at any stage of this process can lead to disorders of cerebellar origin, which include autism spectrum disorders but are not limited to motor deficits. Recent evidence in animal models links impairment of Purkinje cell function with autism-like symptoms including sociability deficits, stereotyped movements, and interspecific communication by vocalization.
Collapse
Affiliation(s)
- Eriola Hoxha
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Torino, Italy
- Department of Neuroscience, University of Torino, Torino, Italy
| | | | - Bibiana Scelfo
- Department of Neuroscience, University of Torino, Torino, Italy
| | - Filippo Tempia
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Torino, Italy
- Department of Neuroscience, University of Torino, Torino, Italy
- National Institute of Neuroscience (INN), Torino, Italy
| | | | | |
Collapse
|
46
|
Werner CT, Murray CH, Reimers JM, Chauhan NM, Woo KKY, Molla HM, Loweth JA, Wolf ME. Trafficking of calcium-permeable and calcium-impermeable AMPA receptors in nucleus accumbens medium spiny neurons co-cultured with prefrontal cortex neurons. Neuropharmacology 2016; 116:224-232. [PMID: 27993521 DOI: 10.1016/j.neuropharm.2016.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/09/2016] [Accepted: 12/15/2016] [Indexed: 11/27/2022]
Abstract
AMPA receptor (AMPAR) transmission onto medium spiny neurons (MSNs) of the adult rat nucleus accumbens (NAc) is normally dominated by GluA2-containing, Ca2+-impermeable AMPAR (CI-AMPARs). However, GluA2-lacking, Ca2+-permeable AMPA receptors (CP-AMPARs) accumulate after prolonged withdrawal from extended-access cocaine self-administration and thereafter their activation is required for the intensified (incubated) cue-induced cocaine craving that characterizes prolonged withdrawal from such regimens. These findings suggest the existence of mechanisms in NAc MSNs that differentially regulate CI-AMPARs and CP-AMPARs. Here, we compared trafficking of GluA1A2 CI-AMPARs and homomeric GluA1 CP-AMPARs using immunocytochemical assays in cultured NAc MSNs plated with prefrontal cortical neurons to restore excitatory inputs. We began by evaluating constitutive internalization of surface receptors and found that this occurs more rapidly for CP-AMPARs. Next, we studied receptor insertion into the membrane; combined with past results, the present findings suggest that activation of protein kinase A accelerates insertion of both CP-AMPARs and CI-AMPARs. We also studied constitutive cycling (net loss of receptors from the membrane under conditions where internalization and recycling are both occurring). Interestingly, although CP-AMPARs exhibit faster constitutive internalization, they cycle at similar rates as CI-AMPARs, suggesting faster reinsertion of CP-AMPARs. In studies of synaptic scaling, long-term (24 h) activity blockade increased surface expression and cycling rates of CI-AMPARs but not CP-AMPARs, whereas long-term increases in activity produced more pronounced scaling down of CI-AMPARs than CP-AMPARs but did not alter receptor cycling. These findings can be used to evaluate and generate hypotheses regarding AMPAR plasticity in the rat NAc following cocaine exposure.
Collapse
Affiliation(s)
- Craig T Werner
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Conor H Murray
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Jeremy M Reimers
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Niravkumar M Chauhan
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Kenneth K Y Woo
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Hanna M Molla
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Jessica A Loweth
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Marina E Wolf
- Department of Neuroscience, The Chicago Medical School at Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| |
Collapse
|
47
|
Regional Regulation of Purkinje Cell Dendritic Spines by Integrins and Eph/Ephrins. PLoS One 2016; 11:e0158558. [PMID: 27518800 PMCID: PMC4982633 DOI: 10.1371/journal.pone.0158558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/19/2016] [Indexed: 02/02/2023] Open
Abstract
Climbing fibres and parallel fibres compete for dendritic space on Purkinje cells in the cerebellum. Normally, climbing fibres populate the proximal dendrites, where they suppress the multiple small spines typical of parallel fibres, leading to their replacement by the few large spines that contact climbing fibres. Previous work has shown that ephrins acting via EphA4 are a signal for this change in spine type and density. We have used an in vitro culture model in which to investigate the ephrin effect on Purkinje cell dendritic spines and the role of integrins in these changes. We found that integrins α3, α5 and β4 are present in many of the dendritic spines of cultured Purkinje cells. pFAK, the main downstream signalling molecule from integrins, has a similar distribution, although the intenstity of pFAK staining and the percentage of pFAK+ spines was consistently higher in the proximal dendrites. Activating integrins with Mg2+ led to an increase in the intensity of pFAK staining and an increase in the proportion of pFAK+ spines in both the proximal and distal dendrites, but no change in spine length, density or morphology. Blocking integrin binding with an RGD-containing peptide led to a reduction in spine length, with more stubby spines on both proximal and distal dendrites. Treatment of the cultures with ephrinA3-Fc chimera suppressed dendritic spines specifically on the proximal dendrites and there was also a decrease of pFAK in spines on this domain. This effect was blocked by simultaneous activation of integrins with Mn2+. We conclude that Eph/ephrin signaling regulates proximal dendritic spines in Purkinje cells by inactivating integrin downstream signalling.
Collapse
|
48
|
Kozol RA, Abrams AJ, James DM, Buglo E, Yan Q, Dallman JE. Function Over Form: Modeling Groups of Inherited Neurological Conditions in Zebrafish. Front Mol Neurosci 2016; 9:55. [PMID: 27458342 PMCID: PMC4935692 DOI: 10.3389/fnmol.2016.00055] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/23/2016] [Indexed: 12/11/2022] Open
Abstract
Zebrafish are a unique cell to behavior model for studying the basic biology of human inherited neurological conditions. Conserved vertebrate genetics and optical transparency provide in vivo access to the developing nervous system as well as high-throughput approaches for drug screens. Here we review zebrafish modeling for two broad groups of inherited conditions that each share genetic and molecular pathways and overlap phenotypically: neurodevelopmental disorders such as Autism Spectrum Disorders (ASD), Intellectual Disability (ID) and Schizophrenia (SCZ), and neurodegenerative diseases, such as Cerebellar Ataxia (CATX), Hereditary Spastic Paraplegia (HSP) and Charcot-Marie Tooth Disease (CMT). We also conduct a small meta-analysis of zebrafish orthologs of high confidence neurodevelopmental disorder and neurodegenerative disease genes by looking at duplication rates and relative protein sizes. In the past zebrafish genetic models of these neurodevelopmental disorders and neurodegenerative diseases have provided insight into cellular, circuit and behavioral level mechanisms contributing to these conditions. Moving forward, advances in genetic manipulation, live imaging of neuronal activity and automated high-throughput molecular screening promise to help delineate the mechanistic relationships between different types of neurological conditions and accelerate discovery of therapeutic strategies.
Collapse
Affiliation(s)
- Robert A. Kozol
- Department of Biology, University of MiamiCoral Gables, FL, USA
| | - Alexander J. Abrams
- Department of Human Genetics, John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation, University of MiamiMiami, FL, USA
| | - David M. James
- Department of Biology, University of MiamiCoral Gables, FL, USA
| | - Elena Buglo
- Department of Human Genetics, John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation, University of MiamiMiami, FL, USA
| | - Qing Yan
- Department of Biology, University of MiamiCoral Gables, FL, USA
| | | |
Collapse
|
49
|
Karabanov A, Ziemann U, Hamada M, George MS, Quartarone A, Classen J, Massimini M, Rothwell J, Siebner HR. Consensus Paper: Probing Homeostatic Plasticity of Human Cortex With Non-invasive Transcranial Brain Stimulation. Brain Stimul 2016; 8:993-1006. [PMID: 26598772 DOI: 10.1016/j.brs.2015.06.017] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Homeostatic plasticity is thought to stabilize neural activity around a set point within a physiologically reasonable dynamic range. Over the last ten years, a wide range of non-invasive transcranial brain stimulation (NTBS) techniques have been used to probe homeostatic control of cortical plasticity in the intact human brain. Here, we review different NTBS approaches to study homeostatic plasticity on a systems level and relate the findings to both, physiological evidence from in vitro studies and to a theoretical framework of homeostatic function. We highlight differences between homeostatic and other non-homeostatic forms of plasticity and we examine the contribution of sleep in restoring synaptic homeostasis. Finally, we discuss the growing number of studies showing that abnormal homeostatic plasticity may be associated to a range of neuropsychiatric diseases.
Collapse
|
50
|
Astrocytes regulate heterogeneity of presynaptic strengths in hippocampal networks. Proc Natl Acad Sci U S A 2016; 113:E2685-94. [PMID: 27118849 DOI: 10.1073/pnas.1523717113] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Dendrites are neuronal structures specialized for receiving and processing information through their many synaptic inputs. How input strengths are modified across dendrites in ways that are crucial for synaptic integration and plasticity remains unclear. We examined in single hippocampal neurons the mechanism of heterosynaptic interactions and the heterogeneity of synaptic strengths of pyramidal cell inputs. Heterosynaptic presynaptic plasticity that counterbalances input strengths requires N-methyl-d-aspartate receptors (NMDARs) and astrocytes. Importantly, this mechanism is shared with the mechanism for maintaining highly heterogeneous basal presynaptic strengths, which requires astrocyte Ca(2+) signaling involving NMDAR activation, astrocyte membrane depolarization, and L-type Ca(2+) channels. Intracellular infusion of NMDARs or Ca(2+)-channel blockers into astrocytes, conditionally ablating the GluN1 NMDAR subunit, or optogenetically hyperpolarizing astrocytes with archaerhodopsin promotes homogenization of convergent presynaptic inputs. Our findings support the presence of an astrocyte-dependent cellular mechanism that enhances the heterogeneity of presynaptic strengths of convergent connections, which may help boost the computational power of dendrites.
Collapse
|