1
|
Ibarra-Castaneda N, Lopez-Virgen V, Moy-Lopez N, Gonzalez-Perez O. Permanent tactile sensory loss reduces neuronal activity in the amygdala and ventral hippocampus and alters anxiety-like behaviors. Behav Brain Res 2025; 482:115456. [PMID: 39880100 DOI: 10.1016/j.bbr.2025.115456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/11/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Tactile information from the whiskers (vibrissae) travels through the somatosensory cortex to the entorhinal cortex and the hippocampus, influencing development and psychological well-being. The lack of whiskers affects cognitive functions, spatial memory, neuronal firing, spatial mapping, and neurogenesis in the dorsal hippocampus. Recent studies underline the importance of tactile experiences in emotional health, noting that while tactile stimuli modulate the dorsal hippocampus, the effects of tactile deprivation on anxiety-like behaviors and neural activity in regions like the ventral hippocampus and amygdala are less understood. This study aims to investigate the impact of permanent tactile deprivation on modifying anxiety-like behaviors and c-Fos expression (a marker of neuronal activity) in the dorsolateral and central nucleus of the amygdala and the ventral hippocampus, two regions involved in emotional memory and anxiety. We sectioned the infraorbital nerve, responsible for transmitting whisker information, in CD1 mice to examine how tactile deprivation modifies the behavioral activity in the Elevated Plus Maze and Open-Field Test. Our data revealed a reduction in anxiety-related behaviors post-deprivation, which was linked to a significant decrease in c-Fos expression in the barrel cortex, as well as ventral hippocampus (CA1, dentate gyrus) and dorsolateral, central nucleus of the amygdala, suggesting impaired processing in emotional-regulator brain regions. In conclusion, tactile inputs reduce neuronal activity regulators in brain regions related to emotional regulation, which may trigger possible failures in risk perception or self-protective behaviors associated with the lack of appropriate anxiety responses.
Collapse
Affiliation(s)
- Nereida Ibarra-Castaneda
- Laboratorio de Neurociencias, Facultad de Psicología, Universidad de Colima, Colima 28040, Mexico; Medical Sciences Ph.D. Program. Facultad de Medicina, Universidad de Colima, Colima 28040, Mexico
| | - Veronica Lopez-Virgen
- Laboratorio de Neurociencias, Facultad de Psicología, Universidad de Colima, Colima 28040, Mexico
| | - Norma Moy-Lopez
- Laboratorio de Neurociencias, Facultad de Psicología, Universidad de Colima, Colima 28040, Mexico
| | - Oscar Gonzalez-Perez
- Laboratorio de Neurociencias, Facultad de Psicología, Universidad de Colima, Colima 28040, Mexico.
| |
Collapse
|
2
|
Fischer M, Kukley M. Hidden in the white matter: Current views on interstitial white matter neurons. Neuroscientist 2024:10738584241282969. [PMID: 39365761 DOI: 10.1177/10738584241282969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
The mammalian brain comprises two structurally and functionally distinct compartments: the gray matter (GM) and the white matter (WM). In humans, the WM constitutes approximately half of the brain volume, yet it remains significantly less investigated than the GM. The major cellular elements of the WM are neuronal axons and glial cells. However, the WM also contains cell bodies of the interstitial neurons, estimated to number 10 to 28 million in the adult bat brain, 67 million in Lar gibbon brain, and 450 to 670 million in the adult human brain, representing as much as 1.3%, 2.25%, and 3.5% of all neurons in the cerebral cortex, respectively. Many studies investigated the interstitial WM neurons (IWMNs) using immunohistochemistry, and some information is available regarding their electrophysiological properties. However, the functional role of IWMNs in physiologic and pathologic conditions largely remains unknown. This review aims to provide a concise update regarding the distribution and properties of interstitial WM neurons, highlight possible functions of these cells as debated in the literature, and speculate about other possible functions of the IWMNs and their interactions with glial cells. We hope that our review will inspire new research on IWMNs, which represent an intriguing cell population in the brain.
Collapse
Affiliation(s)
- Maximilian Fischer
- Institut de Neurociències and Departamento Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Maria Kukley
- Achucarro Basque Centre for Neuroscience, Leioa, Spain
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
3
|
Muller M, Pennartz CMA, Bosman CA, Olcese U. A novel task to investigate vibrotactile detection in mice. PLoS One 2023; 18:e0284735. [PMID: 37079581 PMCID: PMC10118142 DOI: 10.1371/journal.pone.0284735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/06/2023] [Indexed: 04/21/2023] Open
Abstract
Throughout the last decades, understanding the neural mechanisms of sensory processing has been a key objective for neuroscientists. Many studies focused on uncovering the microcircuit-level architecture of somatosensation using the rodent whisker system as a model. Although these studies have significantly advanced our understanding of tactile processing, the question remains to what extent the whisker system can provide results translatable to the human somatosensory system. To address this, we developed a restrained vibrotactile detection task involving the limb system in mice. A vibrotactile stimulus was delivered to the hindlimb of head-fixed mice, who were trained to perform a Go/No-go detection task. Mice were able to learn this task with satisfactory performance and with reasonably short training times. In addition, the task we developed is versatile, as it can be combined with diverse neuroscience methods. Thus, this study introduces a novel task to study the neuron-level mechanisms of tactile processing in a system other than the more commonly studied whisker system.
Collapse
Affiliation(s)
- Mariel Muller
- Cognitive and Systems Neuroscience Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, The Netherlands
| | - Cyriel M. A. Pennartz
- Cognitive and Systems Neuroscience Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, The Netherlands
| | - Conrado A. Bosman
- Cognitive and Systems Neuroscience Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, The Netherlands
| | - Umberto Olcese
- Cognitive and Systems Neuroscience Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Brain and Cognition, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Lee J, Urban-Ciecko J, Park E, Zhu M, Myal SE, Margolis DJ, Barth AL. FosGFP expression does not capture a sensory learning-related engram in superficial layers of mouse barrel cortex. Proc Natl Acad Sci U S A 2021; 118:e2112212118. [PMID: 34930843 PMCID: PMC8719899 DOI: 10.1073/pnas.2112212118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2021] [Indexed: 11/18/2022] Open
Abstract
Immediate-early gene (IEG) expression has been used to identify small neural ensembles linked to a particular experience, based on the principle that a selective subset of activated neurons will encode specific memories or behavioral responses. The majority of these studies have focused on "engrams" in higher-order brain areas where more abstract or convergent sensory information is represented, such as the hippocampus, prefrontal cortex, or amygdala. In primary sensory cortex, IEG expression can label neurons that are responsive to specific sensory stimuli, but experience-dependent shaping of neural ensembles marked by IEG expression has not been demonstrated. Here, we use a fosGFP transgenic mouse to longitudinally monitor in vivo expression of the activity-dependent gene c-fos in superficial layers (L2/3) of primary somatosensory cortex (S1) during a whisker-dependent learning task. We find that sensory association training does not detectably alter fosGFP expression in L2/3 neurons. Although training broadly enhances thalamocortical synaptic strength in pyramidal neurons, we find that synapses onto fosGFP+ neurons are not selectively increased by training; rather, synaptic strengthening is concentrated in fosGFP- neurons. Taken together, these data indicate that expression of the IEG reporter fosGFP does not facilitate identification of a learning-specific engram in L2/3 in barrel cortex during whisker-dependent sensory association learning.
Collapse
Affiliation(s)
- Jiseok Lee
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Joanna Urban-Ciecko
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Eunsol Park
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Mo Zhu
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Stephanie E Myal
- University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - David J Margolis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213;
| |
Collapse
|
5
|
Syngap1 regulates experience-dependent cortical ensemble plasticity by promoting in vivo excitatory synapse strengthening. Proc Natl Acad Sci U S A 2021; 118:2100579118. [PMID: 34404727 DOI: 10.1073/pnas.2100579118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
A significant proportion of autism risk genes regulate synapse function, including plasticity, which is believed to contribute to behavioral abnormalities. However, it remains unclear how impaired synapse plasticity contributes to network-level processes linked to adaptive behaviors, such as experience-dependent ensemble plasticity. We found that Syngap1, a major autism risk gene, promoted measures of experience-dependent excitatory synapse strengthening in the mouse cortex, including spike-timing-dependent glutamatergic synaptic potentiation and presynaptic bouton formation. Synaptic depression and bouton elimination were normal in Syngap1 mice. Within cortical networks, Syngap1 promoted experience-dependent increases in somatic neural activity in weakly active neurons. In contrast, plastic changes to highly active neurons from the same ensemble that paradoxically weaken with experience were unaffected. Thus, experience-dependent excitatory synapse strengthening mediated by Syngap1 shapes neuron-specific plasticity within cortical ensembles. We propose that other genes regulate neuron-specific weakening within ensembles, and together, these processes function to redistribute activity within cortical networks during experience.
Collapse
|
6
|
Zeiger WA, Marosi M, Saggi S, Noble N, Samad I, Portera-Cailliau C. Barrel cortex plasticity after photothrombotic stroke involves potentiating responses of pre-existing circuits but not functional remapping to new circuits. Nat Commun 2021; 12:3972. [PMID: 34172735 PMCID: PMC8233353 DOI: 10.1038/s41467-021-24211-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 06/01/2021] [Indexed: 01/14/2023] Open
Abstract
Recovery after stroke is thought to be mediated by adaptive circuit plasticity, whereby surviving neurons assume the roles of those that died. However, definitive longitudinal evidence of neurons changing their response selectivity after stroke is lacking. We sought to directly test whether such functional “remapping” occurs within mouse primary somatosensory cortex after a stroke that destroys the C1 barrel. Using in vivo calcium imaging to longitudinally record sensory-evoked activity under light anesthesia, we did not find any increase in the number of C1 whisker-responsive neurons in the adjacent, spared D3 barrel after stroke. To promote plasticity after stroke, we also plucked all whiskers except C1 (forced use therapy). This led to an increase in the reliability of sensory-evoked responses in C1 whisker-responsive neurons but did not increase the number of C1 whisker-responsive neurons in spared surround barrels over baseline levels. Our results argue against remapping of functionality after barrel cortex stroke, but support a circuit-based mechanism for how rehabilitation may improve recovery. Definitive evidence for functional remapping after stroke remains lacking. Here, the authors performed in vivo intrinsic signal imaging and two-photon calcium imaging of sensory-evoked responses before and after photothrombotic stroke and found no evidence of remapping of lost functionalities to new circuits in peri-infarct cortex.
Collapse
Affiliation(s)
- William A Zeiger
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Máté Marosi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Satvir Saggi
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Natalie Noble
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Isa Samad
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Carlos Portera-Cailliau
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. .,Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Campelo T, Augusto E, Chenouard N, de Miranda A, Kouskoff V, Camus C, Choquet D, Gambino F. AMPAR-Dependent Synaptic Plasticity Initiates Cortical Remapping and Adaptive Behaviors during Sensory Experience. Cell Rep 2020; 32:108097. [PMID: 32877679 PMCID: PMC7487777 DOI: 10.1016/j.celrep.2020.108097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/30/2020] [Accepted: 08/11/2020] [Indexed: 11/28/2022] Open
Abstract
Cortical plasticity improves behaviors and helps recover lost functions after injury. However, the underlying synaptic mechanisms remain unclear. In mice, we show that trimming all but one whisker enhances sensory responses from the spared whisker in the barrel cortex and occludes whisker-mediated synaptic potentiation (w-Pot) in vivo. In addition, whisker-dependent behaviors that are initially impaired by single-whisker experience (SWE) rapidly recover when associated cortical regions remap. Cross-linking the surface GluA2 subunit of AMPA receptors (AMPARs) suppresses the expression of w-Pot, presumably by blocking AMPAR surface diffusion, in mice with all whiskers intact, indicating that synaptic potentiation in vivo requires AMPAR trafficking. We use this approach to demonstrate that w-Pot is required for SWE-mediated strengthening of synaptic inputs and initiates the recovery of previously learned skills during the early phases of SWE. Taken together, our data reveal that w-Pot mediates cortical remapping and behavioral improvement upon partial sensory deafferentation.
Collapse
Affiliation(s)
- Tiago Campelo
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Elisabete Augusto
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Nicolas Chenouard
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Aron de Miranda
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Vladimir Kouskoff
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Come Camus
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4, 33000 Bordeaux, France.
| | - Frédéric Gambino
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France.
| |
Collapse
|
8
|
Lee CR, Najafizadeh L, Margolis DJ. Investigating learning-related neural circuitry with chronic in vivo optical imaging. Brain Struct Funct 2020; 225:467-480. [PMID: 32006147 DOI: 10.1007/s00429-019-02001-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/05/2019] [Indexed: 10/25/2022]
Abstract
Fundamental aspects of brain function, including development, plasticity, learning, and memory, can take place over time scales of days to years. Chronic in vivo imaging of neural activity with cellular resolution is a powerful method for tracking the long-term activity of neural circuits. We review recent advances in our understanding of neural circuit function from diverse brain regions that have been enabled by chronic in vivo cellular imaging. Insight into the neural basis of learning and decision-making, in particular, benefit from the ability to acquire longitudinal data from genetically identified neuronal populations, deep brain areas, and subcellular structures. We propose that combining chronic imaging with further experimental and computational innovations will advance our understanding of the neural circuit mechanisms of brain function.
Collapse
Affiliation(s)
- Christian R Lee
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Laleh Najafizadeh
- Department of Electrical and Computer Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - David J Margolis
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
9
|
Sheng N, Zheng H, Li M, Li M, Wang Z, Peng Y, Yu H, Zhang J. 4,5 caffeoylquinic acid and scutellarin, identified by integrated metabolomics and proteomics approach as the active ingredients of Dengzhan Shengmai, act against chronic cerebral hypoperfusion by regulating glutamatergic and GABAergic synapses. Pharmacol Res 2020; 152:104636. [PMID: 31926275 DOI: 10.1016/j.phrs.2020.104636] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 12/06/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
Abstract
Dengzhan Shengmai (DZSM) is a proprietary Chinese medicine for remarkable curative effect as a treatment of cerebrovascular diseases, such as chronic cerebral hypoperfusion (CCH) and dementia based on evidence-based medicine, which have been widely used in the recovery period of ischemic cerebrovascular diseases. The purpose of this study was to investigate the active substances and mechanism of DZSM against CCH. Integrative metabolomic and proteomic studies were performed to investigate the neuroprotective effect of DZSM based on CCH model rats. The exposed components of DZSM in target brain tissue were analysed by a high-sensitivity HPLC-MS/MS method, and the exposed components were tested on a glutamate-induced neuronal excitatory damage cell model for the verification of active ingredients and mechanism of DZSM. Upon proteomic and metabolomic analysis, we observed a significant response in DZSM therapy from the interconnected neurotransmitter transport pathways including glutamatergic and GABAergic synapses. Additionally, DZSM had a significant regulatory effect on glutamate and GABA-related proteins including vGluT1 and vIAAT, suggested that DZSM could be involved in the vesicle transport of excitatory and inhibitory neurotransmitters in the pre-synaptic membrane. DZSM could also regulated the metabolism of arachidonic acid (AA), phospholipids, lysophospholipids and the expression of phospholipase A2 in post-synaptic membrane. The results of glutamate-induced neuronal excitatory injury cell model experiment for verification of active ingredients and mechanism of DZSM showed that there are five active ingredients, and among them, 4,5 caffeoylquinic acid (4,5-CQA) and scutellarin (SG) could simultaneously affect the GABAergic and glutamatergic synaptic metabolism as well as the related receptors, the NR2b subunit of NMDA and the α1 subunit of GABAA. The active ingredients of DZSM could regulate the over-expression of the NMDA receptor, enhance the expression of the GABAA receptor, resist glutamate-induced neuronal excitatory damage, and finally maintain the balance of excitatory and inhibitory synaptic metabolism dominated by glutamate and GABA. Furtherly, we compared the efficacy of DZSM, 4,5-CQA, SG and the synergistic effect of 4,5-CQA and SG, and the results showed that all the groups significantly improved cell viability compared with the model group (p < 0.001). The western blot results showed that DZSM, 4,5-CQA, SG and 4,5-CQA/SG co-administration groups could significantly regulate the expression of receptors (GABAA α1 and NR2b subunit of NMDA) and synaptic-related proteins, such as Sv2a, Syp, Slc17a7, bin1 and Prkca, respectively. These results proved DZSM and its active ingredients (4,5-CQA and SG) had the effect of regulating glutamatergic and GABAergic synapses. Finally, membrane potential FLIPR assay of 4,5-CQA and SG was used for GABRA1 activity test, and it was found that the two compounds could increase GABA-induced activation of GABRA1 receptor (GABA 10 μM) in a dose-dependent manner with EC50 value of 48.74 μM and 29.77 μM, respectively. Manual patch clamp method was used to record NMDA NR1/NR2B subtype currents, and scutellarin could cause around 10 % blockade at 10 μM (p<0.05 compared with the control group). These studies provided definitive clues of the mechanism for the neuroprotective effect of DZSM for CCH treatment and the active compounds regulating glutamatergic and GABAergic synapses. Additionally, 4,5-CQA and SG might be potential drugs for the treatment of neurodegenerative disease related to CCH.
Collapse
Affiliation(s)
- Ning Sheng
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Hao Zheng
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Min Li
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Menglin Li
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Zhe Wang
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Ying Peng
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Haibo Yu
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| | - Jinlan Zhang
- Institute: State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China.
| |
Collapse
|
10
|
Kloosterboer E, Funke K. Repetitive transcranial magnetic stimulation recovers cortical map plasticity induced by sensory deprivation due to deafferentiation. J Physiol 2019; 597:4025-4051. [PMID: 31145483 PMCID: PMC6852264 DOI: 10.1113/jp277507] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Partial sensory deprivation (deafferentation) by removing whiskers from the rat snout resulted in a reduced responsiveness of related cortical representations. Repetitive transcranial magnetic stimulation (three blocks of intermittent theta-burst) applied for 5 days in combination with sensory exploration restored the normal responsiveness level of the deafferented barrel cortex. However, intracortical inhibition (lateral and recurrent) appeared to be reduced after repetitive transcranial magnetic stimulation, probably as the cause of improved responsiveness. Repetitive transcranial magnetic stimulation also reduced the asymmetry of the lateral spread of sensory activity. ABSTRACT Repetitive transcranial magnetic stimulation (rTMS) modulates human cortical excitability. It has the potential to support recovery to normal cortical function when the excitation-inhibition balance is altered (e.g. after a stroke or loss of sensory input). We tested cortical map plasticity on the basis of sensory responses (local field potentials, LFPs) and expression of neuronal activity marker proteins within the barrel cortex of rats receiving either active or sham rTMS after selective unilateral deafferentation by whiskers plucking. Rats received daily rTMS [intermittent theta-burst (iTBS), active or sham] for 5 days before exploring an enriched environment. Our previous studies indicated a disinhibitory effect of iTBS on cortical activity. Therefore, we also expected disinhibitory effects if deafferentation causes depression of sensory responses. Deafferentation resulted in an acute general reduction of sensory responsiveness and enhanced expression of inhibitory activity markers (GAD67, parvalbumin) in the deafferented hemisphere. Active but not sham-iTBS-rTMS normalized these measures. The stronger caudal-to-frontal horizontal spread of activity across barrels was reduced after deafferentation but not restored after active iTBS, despite generally increased responses. Fitting the LFP data with a computational model of different strengths and types of excitatory and inhibitory connections further revealed an iTBS-induced reduction of lateral and recurrent inhibition as the most probable scenario. Whether the disinhibitory effect of iTBS for the restoration of normal cortical function in the acute phase of depression after deafferentiation is also beneficial in humans remains to be demonstrated. As recently discussed, disinhibition appears to be required to open a window for neuronal plasticity.
Collapse
Affiliation(s)
- Ellen Kloosterboer
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Klaus Funke
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
11
|
Furlanis E, Scheiffele P. Regulation of Neuronal Differentiation, Function, and Plasticity by Alternative Splicing. Annu Rev Cell Dev Biol 2018; 34:451-469. [PMID: 30028642 DOI: 10.1146/annurev-cellbio-100617-062826] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Posttranscriptional mechanisms provide powerful means to expand the coding power of genomes. In nervous systems, alternative splicing has emerged as a fundamental mechanism not only for the diversification of protein isoforms but also for the spatiotemporal control of transcripts. Thus, alternative splicing programs play instructive roles in the development of neuronal cell type-specific properties, neuronal growth, self-recognition, synapse specification, and neuronal network function. Here we discuss the most recent genome-wide efforts on mapping RNA codes and RNA-binding proteins for neuronal alternative splicing regulation. We illustrate how alternative splicing shapes key steps of neuronal development, neuronal maturation, and synaptic properties. Finally, we highlight efforts to dissect the spatiotemporal dynamics of alternative splicing and their potential contribution to neuronal plasticity and the mature nervous system.
Collapse
|
12
|
Audette NJ, Urban-Ciecko J, Matsushita M, Barth AL. POm Thalamocortical Input Drives Layer-Specific Microcircuits in Somatosensory Cortex. Cereb Cortex 2018; 28:1312-1328. [PMID: 28334225 PMCID: PMC6093433 DOI: 10.1093/cercor/bhx044] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/03/2017] [Indexed: 12/11/2022] Open
Abstract
Higher-order thalamic nuclei, such as the posterior medial nucleus (POm) in the somatosensory system or the pulvinar in the visual system, densely innervate the cortex and can influence perception and plasticity. To systematically evaluate how higher-order thalamic nuclei can drive cortical circuits, we investigated cell-type selective responses to POm stimulation in mouse primary somatosensory (barrel) cortex, using genetically targeted whole-cell recordings in acute brain slices. We find that ChR2-evoked thalamic input selectively targets specific cell types in the neocortex, revealing layer-specific modules for the summation and processing of POm input. Evoked activity in pyramidal neurons from deep layers is fast and synchronized by rapid feedforward inhibition from GABAergic parvalbumin-expressing neurons, and activity in superficial layers is weaker and prolonged, facilitated by slow inhibition from GABAergic neurons expressing the 5HT3a receptor. Somatostatin-expressing GABAergic neurons do not receive direct input in either layer and their spontaneous activity is suppressed during POm stimulation. This novel pattern of weak, delayed, thalamus-evoked inhibition in layer 2 suggests a longer integration window for incoming sensory information and may facilitate stimulus detection and plasticity in superficial pyramidal neurons.
Collapse
Affiliation(s)
- Nicholas J Audette
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, USA
| | - Joanna Urban-Ciecko
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, USA
| | - Megumi Matsushita
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, USA
| | - Alison L Barth
- Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Abstract
Most behaviors in mammals are directly or indirectly guided by prior experience and therefore depend on the ability of our brains to form memories. The ability to form an association between an initially possibly neutral sensory stimulus and its behavioral relevance is essential for our ability to navigate in a changing environment. The formation of a memory is a complex process involving many areas of the brain. In this chapter we review classic and recent work that has shed light on the specific contribution of sensory cortical areas to the formation of associative memories. We discuss synaptic and circuit mechanisms that mediate plastic adaptations of functional properties in individual neurons as well as larger neuronal populations forming topographically organized representations. Furthermore, we describe commonly used behavioral paradigms that are used to study the mechanisms of memory formation. We focus on the auditory modality that is receiving increasing attention for the study of associative memory in rodent model systems. We argue that sensory cortical areas may play an important role for the memory-dependent categorical recognition of previously encountered sensory stimuli.
Collapse
Affiliation(s)
- Dominik Aschauer
- Institute of Physiology, Focus Program Translational Neurosciences (FTN), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Simon Rumpel
- Institute of Physiology, Focus Program Translational Neurosciences (FTN), University Medical Center, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
14
|
Abstract
Somatosensory areas containing topographic maps of the body surface are a major feature of parietal cortex. In primates, parietal cortex contains four somatosensory areas, each with its own map, with the primary cutaneous map in area 3b. Rodents have at least three parietal somatosensory areas. Maps are not isomorphic to the body surface, but magnify behaviorally important skin regions, which include the hands and face in primates, and the whiskers in rodents. Within each map, intracortical circuits process tactile information, mediate spatial integration, and support active sensation. Maps may also contain fine-scale representations of touch submodalities, or direction of tactile motion. Functional representations are more overlapping than suggested by textbook depictions of map topography. The whisker map in rodent somatosensory cortex is a canonic system for studying cortical microcircuits, sensory coding, and map plasticity. Somatosensory maps are plastic throughout life in response to altered use or injury. This chapter reviews basic principles and recent findings in primate, human, and rodent somatosensory maps.
Collapse
Affiliation(s)
- Samuel Harding-Forrester
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States
| | - Daniel E Feldman
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, CA, United States.
| |
Collapse
|
15
|
Barlow SM, Maron JL, Alterovitz G, Song D, Wilson BJ, Jegatheesan P, Govindaswami B, Lee J, Rosner AO. Somatosensory Modulation of Salivary Gene Expression and Oral Feeding in Preterm Infants: Randomized Controlled Trial. JMIR Res Protoc 2017; 6:e113. [PMID: 28615158 PMCID: PMC5489710 DOI: 10.2196/resprot.7712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Despite numerous medical advances in the care of at-risk preterm neonates, oral feeding still represents one of the first and most advanced neurological challenges facing this delicate population. Objective, quantitative, and noninvasive assessment tools, as well as neurotherapeutic strategies, are greatly needed in order to improve feeding and developmental outcomes. Pulsed pneumatic orocutaneous stimulation has been shown to improve nonnutritive sucking (NNS) skills in preterm infants who exhibit delayed or disordered nipple feeding behaviors. Separately, the study of the salivary transcriptome in neonates has helped identify biomarkers directly linked to successful neonatal oral feeding behavior. The combination of noninvasive treatment strategies and transcriptomic analysis represents an integrative approach to oral feeding in which rapid technological advances and personalized transcriptomics can safely and noninvasively be brought to the bedside to inform medical care decisions and improve care and outcomes. OBJECTIVE The study aimed to conduct a multicenter randomized control trial (RCT) to combine molecular and behavioral methods in an experimental conceptualization approach to map the effects of PULSED somatosensory stimulation on salivary gene expression in the context of the acquisition of oral feeding habits in high-risk human neonates. The aims of this study represent the first attempt to combine noninvasive treatment strategies and transcriptomic assessments of high-risk extremely preterm infants (EPI) to (1) improve oral feeding behavior and skills, (2) further our understanding of the gene ontology of biologically diverse pathways related to oral feeding, (3) use gene expression data to personalize neonatal care and individualize treatment strategies and timing interventions, and (4) improve long-term developmental outcomes. METHODS A total of 180 extremely preterm infants from three neonatal intensive care units (NICUs) will be randomized to receive either PULSED or SHAM (non-pulsing) orocutaneous intervention simultaneous with tube feedings 3 times per day for 4 weeks, beginning at 30 weeks postconceptional age. Infants will also be assessed 3 times per week for NNS performance, and multiple saliva samples will be obtained each week for transcriptomic analysis, until infants have achieved full oral feeding status. At 18 months corrected age (CA), infants will undergo neurodevelopmental follow-up testing, the results of which will be correlated with feeding outcomes in the neo-and post-natal period and with gene expression data and intervention status. RESULTS The ongoing National Institutes of Health funded randomized controlled trial R01HD086088 is actively recruiting participants. The expected completion date of the study is 2021. CONCLUSIONS Differential salivary gene expression profiles in response to orosensory entrainment intervention are expected to lead to the development of individualized interventions for the diagnosis and management of oral feeding in preterm infants. TRIAL REGISTRATION ClinicalTrials.gov NCT02696343; https://clinicaltrials.gov/ct2/show/NCT02696343 (Archived by WebCite at http://www.webcitation.org/6r5NbJ9Ym).
Collapse
Affiliation(s)
- Steven Michael Barlow
- Center for Brain, Biology, and Behavior, Department of Special Education and Communication Disorders, Biological Systems Engineering, University of Nebraska, Lincoln, NE, United States
| | - Jill Lamanna Maron
- Tufts Medical Center, Division of Neonatology, Department of Pediatrics, Boston, MA, United States
| | - Gil Alterovitz
- Center for Biomedical Informatics, Harvard Medical School, Boston, MA, United States
| | - Dongli Song
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, United States
| | - Bernard Joseph Wilson
- CHI Health St. Elizabeth, Division of Neonatal-Perinatal Medicine, Lincoln, NE, United States
| | - Priya Jegatheesan
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, United States
| | - Balaji Govindaswami
- Division of Neonatology, Department of Pediatrics, Santa Clara Valley Medical Center, San Jose, CA, United States
| | - Jaehoon Lee
- IMMAP, Department of Educational Psychology and Leadership, Texas Tech University, Lubbock, TX, United States
| | - Austin Oder Rosner
- Tufts Medical Center, Division of Neonatology, Department of Pediatrics, Boston, MA, United States
| |
Collapse
|
16
|
Mohammadi E, Shamsizadeh A, Salari E, Fatemi I, Allahtavakoli M, Roohbakhsh A. Effect of TPMPA (GABACreceptor antagonist) on neuronal response properties in rat barrel cortex. Somatosens Mot Res 2017; 34:108-115. [DOI: 10.1080/08990220.2017.1317240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Elham Mohammadi
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Shamsizadeh
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Salari
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Iman Fatemi
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology–Pharmacology Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Ali Roohbakhsh
- Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
17
|
Liguz-Lecznar M, Urban-Ciecko J, Kossut M. Somatostatin and Somatostatin-Containing Neurons in Shaping Neuronal Activity and Plasticity. Front Neural Circuits 2016; 10:48. [PMID: 27445703 PMCID: PMC4927943 DOI: 10.3389/fncir.2016.00048] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/20/2016] [Indexed: 01/27/2023] Open
Abstract
Since its discovery over four decades ago, somatostatin (SOM) receives growing scientific and clinical interest. Being localized in the nervous system in a subset of interneurons somatostatin acts as a neurotransmitter or neuromodulator and its role in the fine-tuning of neuronal activity and involvement in synaptic plasticity and memory formation are widely recognized in the recent literature. Combining transgenic animals with electrophysiological, anatomical and molecular methods allowed to characterize several subpopulations of somatostatin-containing interneurons possessing specific anatomical and physiological features engaged in controlling the output of cortical excitatory neurons. Special characteristic and connectivity of somatostatin-containing neurons set them up as significant players in shaping activity and plasticity of the nervous system. However, somatostatin is not just a marker of particular interneuronal subpopulation. Somatostatin itself acts pre- and postsynaptically, modulating excitability and neuronal responses. In the present review, we combine the knowledge regarding somatostatin and somatostatin-containing interneurons, trying to incorporate it into the current view concerning the role of the somatostatinergic system in cortical plasticity.
Collapse
Affiliation(s)
- Monika Liguz-Lecznar
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology Warsaw, Poland
| | - Joanna Urban-Ciecko
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental BiologyWarsaw, Poland; Department of Biological Sciences and Center for the Neural Basis of Cognition, Carnegie Mellon UniversityPittsburgh, PA, USA
| | - Malgorzata Kossut
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental BiologyWarsaw, Poland; Department of Psychology, University of Social Sciences and Humanities (SWPS)Warsaw, Poland
| |
Collapse
|
18
|
Potter LE, Paylor JW, Suh JS, Tenorio G, Caliaperumal J, Colbourne F, Baker G, Winship I, Kerr BJ. Altered excitatory-inhibitory balance within somatosensory cortex is associated with enhanced plasticity and pain sensitivity in a mouse model of multiple sclerosis. J Neuroinflammation 2016; 13:142. [PMID: 27282914 PMCID: PMC4901403 DOI: 10.1186/s12974-016-0609-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/01/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic neuropathic pain is a common symptom of multiple sclerosis (MS). MOG35-55-induced experimental autoimmune encephalomyelitis (EAE) has been used as an animal model to investigate the mechanisms of pain in MS. Previous studies have implicated sensitization of spinal nociceptive networks in the pathogenesis of pain in EAE. However, the involvement of supraspinal sites of nociceptive integration, such as the primary somatosensory cortex (S1), has not been defined. We therefore examined functional, structural, and immunological alterations in S1 during the early stages of EAE, when pain behaviors first appear. We also assessed the effects of the antidepressant phenelzine (PLZ) on S1 alterations and nociceptive (mechanical) sensitivity in early EAE. PLZ has been shown to restore central nervous system (CNS) tissue concentrations of GABA and the monoamines (5-HT, NA) in EAE. We hypothesized that PLZ treatment would also normalize nociceptive sensitivity in EAE by restoring the balance of excitation and inhibition (E-I) in the CNS. METHODS We used in vivo flavoprotein autofluorescence imaging (FAI) to assess neural ensemble responses in S1 to vibrotactile stimulation of the limbs in early EAE. We also used immunohistochemistry (IHC), and Golgi-Cox staining, to examine synaptic changes and neuroinflammation in S1. Mechanical sensitivity was assessed at the clinical onset of EAE with Von Frey hairs. RESULTS Mice with early EAE exhibited significantly intensified and expanded FAI responses in S1 compared to controls. IHC revealed increased vesicular glutamate transporter (VGLUT1) expression and disrupted parvalbumin+ (PV+) interneuron connectivity in S1 of EAE mice. Furthermore, peri-neuronal nets (PNNs) were significantly reduced in S1. Morphological analysis of excitatory neurons in S1 revealed increased dendritic spine densities. Iba-1+ cortical microglia were significantly elevated early in the disease. Chronic PLZ treatment was found to normalize mechanical thresholds in EAE. PLZ also normalized S1 FAI responses, neuronal morphologies, and cortical microglia numbers and attenuated VGLUT1 reactivity-but did not significantly attenuate the loss of PNNs. CONCLUSIONS These findings implicate a pro-excitatory shift in the E-I balance of the somatosensory CNS, arising early in the pathogenesis EAE and leading to large-scale functional and structural plasticity in S1. They also suggest a novel antinociceptive effect of PLZ treatment.
Collapse
Affiliation(s)
- Liam E Potter
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Department of Anesthesiology and Pain Medicine, University of Alberta, Clinical Sciences Building, 8-120, Edmonton, AB, T6G 2G3, Canada
| | - John W Paylor
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Department of Psychiatry (NRU), University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - Jee Su Suh
- Department of Anesthesiology and Pain Medicine, University of Alberta, Clinical Sciences Building, 8-120, Edmonton, AB, T6G 2G3, Canada
| | - Gustavo Tenorio
- Department of Anesthesiology and Pain Medicine, University of Alberta, Clinical Sciences Building, 8-120, Edmonton, AB, T6G 2G3, Canada
| | - Jayalakshmi Caliaperumal
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Department of Psychology, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Fred Colbourne
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Department of Psychology, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Glen Baker
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Department of Psychiatry (NRU), University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - Ian Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada.,Department of Psychiatry (NRU), University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, T6G 2E1, Canada. .,Department of Pharmacology, University of Alberta, Edmonton, AB, T6E 2H7, Canada. .,Department of Anesthesiology and Pain Medicine, University of Alberta, Clinical Sciences Building, 8-120, Edmonton, AB, T6G 2G3, Canada.
| |
Collapse
|
19
|
Keating P, Rosenior-Patten O, Dahmen JC, Bell O, King AJ. Behavioral training promotes multiple adaptive processes following acute hearing loss. eLife 2016; 5:e12264. [PMID: 27008181 PMCID: PMC4841776 DOI: 10.7554/elife.12264] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 03/23/2016] [Indexed: 11/13/2022] Open
Abstract
The brain possesses a remarkable capacity to compensate for changes in inputs resulting from a range of sensory impairments. Developmental studies of sound localization have shown that adaptation to asymmetric hearing loss can be achieved either by reinterpreting altered spatial cues or by relying more on those cues that remain intact. Adaptation to monaural deprivation in adulthood is also possible, but appears to lack such flexibility. Here we show, however, that appropriate behavioral training enables monaurally-deprived adult humans to exploit both of these adaptive processes. Moreover, cortical recordings in ferrets reared with asymmetric hearing loss suggest that these forms of plasticity have distinct neural substrates. An ability to adapt to asymmetric hearing loss using multiple adaptive processes is therefore shared by different species and may persist throughout the lifespan. This highlights the fundamental flexibility of neural systems, and may also point toward novel therapeutic strategies for treating sensory disorders. DOI:http://dx.doi.org/10.7554/eLife.12264.001 The brain normally compares the timing and intensity of the sounds that reach each ear to work out a sound’s origin. Hearing loss in one ear disrupts these between-ear comparisons, which causes listeners to make errors in this process. With time, however, the brain adapts to this hearing loss and once again learns to localize sounds accurately. Previous research has shown that young ferrets can adapt to hearing loss in one ear in two distinct ways. The ferrets either learn to remap the altered between-ear comparisons, caused by losing hearing in one ear, onto their new locations. Alternatively, the ferrets learn to locate sounds using only their good ear. Each strategy is suited to localizing different types of sound, but it was not known how this adaptive flexibility unfolds over time, whether it persists throughout the lifespan, or whether it is shared by other species. Now, Keating et al. show that, with some coaching, adult humans also adapt to temporary loss of hearing in one ear using the same two strategies. In the experiments, adult humans were trained to localize different kinds of sounds while wearing an earplug in one ear. These sounds were presented from 12 loudspeakers arranged in a horizontal circle around the person being tested. The experiments showed that short periods of behavioral training enable adult humans to adapt to a hearing loss in one ear and recover their ability to localize sounds. Just like the ferrets, adult humans learned to correctly associate altered between-ear comparisons with their new locations and to rely more on the cues from the unplugged ear to locate sound. Which of these adaptive strategies the participants used depended on the frequencies present in the sounds. The cells in the ear and brain that detect and make sense of sound typically respond best to a limited range of frequencies, and so this suggests that each strategy relies on a distinct set of cells. Keating et al. confirmed in ferrets that different brain cells are indeed used to bring about adaptation to hearing loss in one ear using each strategy. These insights may aid the development of new therapies to treat hearing loss. DOI:http://dx.doi.org/10.7554/eLife.12264.002
Collapse
Affiliation(s)
- Peter Keating
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Onayomi Rosenior-Patten
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Johannes C Dahmen
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Olivia Bell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Andrew J King
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Emergence of functional subnetworks in layer 2/3 cortex induced by sequential spikes in vivo. Proc Natl Acad Sci U S A 2016; 113:E1372-81. [PMID: 26903616 DOI: 10.1073/pnas.1513410113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
During cortical circuit development in the mammalian brain, groups of excitatory neurons that receive similar sensory information form microcircuits. However, cellular mechanisms underlying cortical microcircuit development remain poorly understood. Here we implemented combined two-photon imaging and photolysis in vivo to monitor and manipulate neuronal activities to study the processes underlying activity-dependent circuit changes. We found that repeated triggering of spike trains in a randomly chosen group of layer 2/3 pyramidal neurons in the somatosensory cortex triggered long-term plasticity of circuits (LTPc), resulting in the increased probability that the selected neurons would fire when action potentials of individual neurons in the group were evoked. Significant firing pattern changes were observed more frequently in the selected group of neurons than in neighboring control neurons, and the induction was dependent on the time interval between spikes, N-methyl-D-aspartate (NMDA) receptor activation, and Calcium/calmodulin-dependent protein kinase II (CaMKII) activation. In addition, LTPc was associated with an increase of activity from a portion of neighboring neurons with different probabilities. Thus, our results demonstrate that the formation of functional microcircuits requires broad network changes and that its directionality is nonrandom, which may be a general feature of cortical circuit assembly in the mammalian cortex.
Collapse
|
21
|
Abstract
Synaptic neurotransmission is modified at cortical connections throughout life. Varying the amplitude of the postsynaptic response is one mechanism that generates flexible signaling in neural circuits. The timing of the synaptic response may also play a role. Here, we investigated whether weakening and loss of an entire connection between excitatory cortical neurons was foreshadowed in the timing of the postsynaptic response. We made electrophysiological recordings in rat primary somatosensory cortex that was undergoing experience-dependent loss of complete local excitatory connections. The synaptic latency of pyramid-pyramid connections, which typically comprise multiple synapses, was longer and more variable. Connection strength and latency were not correlated. Instead, prolonged latency was more closely related to progression of connection loss. The action potential waveform and axonal conduction velocity were unaffected, suggesting that the altered timing of neurotransmission was attributable to a synaptic mechanism. Modeling studies indicated that increasing the latency and jitter at a subset of synapses reduced the number of action potentials fired by a postsynaptic neuron. We propose that prolonged synaptic latency and diminished temporal precision of neurotransmission are hallmarks of impending loss of a cortical connection.
Collapse
|
22
|
Weiler NC, Collman F, Vogelstein JT, Burns R, Smith SJ. Synaptic molecular imaging in spared and deprived columns of mouse barrel cortex with array tomography. Sci Data 2014; 1:140046. [PMID: 25977797 PMCID: PMC4411012 DOI: 10.1038/sdata.2014.46] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 10/21/2014] [Indexed: 01/26/2023] Open
Abstract
A major question in neuroscience is how diverse subsets of synaptic connections in neural circuits are affected by experience dependent plasticity to form the basis for behavioral learning and memory. Differences in protein expression patterns at individual synapses could constitute a key to understanding both synaptic diversity and the effects of plasticity at different synapse populations. Our approach to this question leverages the immunohistochemical multiplexing capability of array tomography (ATomo) and the columnar organization of mouse barrel cortex to create a dataset comprising high resolution volumetric images of spared and deprived cortical whisker barrels stained for over a dozen synaptic molecules each. These dataset has been made available through the Open Connectome Project for interactive online viewing, and may also be downloaded for offline analysis using web, Matlab, and other interfaces.
Collapse
Affiliation(s)
- Nicholas C Weiler
- Graduate Program in Neurosciences, Stanford University School of Medicine, Stanford, California 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Forrest Collman
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Joshua T Vogelstein
- Department of Statistical Science, Duke University, Durham, North Carolina 27708, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Randal Burns
- Department of Computer Science, The Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Stephen J Smith
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California 94305, USA
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| |
Collapse
|