1
|
Dong WK. Modulation of multisensory nociceptive neurons in monkey cortical area 7b and behavioral correlates. J Neurophysiol 2024; 132:544-569. [PMID: 38985936 PMCID: PMC11427044 DOI: 10.1152/jn.00377.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/12/2024] Open
Abstract
Wide-range thermoreceptive neurons (WRT-EN) in monkey cortical area 7b that encoded innocuous and nocuous cutaneous thermal and threatening visuosensory stimulation with high fidelity were studied to identify their multisensory integrative response properties. Emphasis was given to characterizing the spatial and temporal effects of threatening visuosensory input on the thermal stimulus-response properties of these multisensory nociceptive neurons. Threatening visuosensory stimulation was most efficacious in modulating thermal evoked responses when presented as a downward ("looming"), spatially congruent, approaching and closely proximal target in relation to the somatosensory receptive field. Both temporal alignment and misalignment of spatially aligned threatening visual and thermal stimulation significantly increased mean discharge frequencies above those evoked by thermal stimulation alone, particularly at near noxious (43°C) and mildly noxious (45°C) temperatures. The enhanced multisensory discharge frequencies were equivalent to the discharge frequency evoked by overtly noxious thermal stimulation alone at 47°C (monkey pain tolerance threshold). A significant increase in behavioral mean escape frequency with shorter escape latency was evoked by multisensory stimulation at near noxious temperature (43°C), which was equivalent to that evoked by noxious stimulation alone (47°C). The remarkable concordance of elevating both neural discharge and escape frequency from a nonnociceptive and prepain level by near noxious thermal stimulation to a nociceptive and pain level by multisensory visual and near noxious thermal stimulation and integration is an elegantly designed defensive neural mechanism that in effect lowers both nociceptive response and pain thresholds to preemptively engage nocifensive behavior and, consequently, avert impending and actual injurious noxious thermal stimulation.NEW & NOTEWORTHY Multisensory nociceptive neurons in cortical area 7b are engaged in integration of threatening visuosensory and a wide range of innocuous and nocuous somatosensory (thermoreceptive) inputs. The enhancement of neuronal activity and escape behavior in monkey by multisensory integration is consistent and supportive of human psychophysical studies. The spatial features of visuosensory stimulation in peripersonal space in relation to somatic stimulation in personal space are critical to multisensory integration, nociception, nocifensive behavior, and pain.
Collapse
Affiliation(s)
- Willie K Dong
- Department of Anesthesiology and Pain Medicine, School of Medicine, University of Washington, Seattle, Washington, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
| |
Collapse
|
2
|
Lu WH, Chang TT, Chang YM, Liu YH, Lin CH, Suen CS, Hwang MJ, Huang YS. CPEB2-activated axonal translation of VGLUT2 mRNA promotes glutamatergic transmission and presynaptic plasticity. J Biomed Sci 2024; 31:69. [PMID: 38992696 PMCID: PMC11241979 DOI: 10.1186/s12929-024-01061-2] [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: 11/13/2023] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity. METHODS Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles. RESULTS Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles. CONCLUSIONS We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.
Collapse
Affiliation(s)
- Wen-Hsin Lu
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Tzu-Tung Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yi-Hsiang Liu
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Chia-Hsuan Lin
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, 11529, Taiwan
| | - Ching-Shu Suen
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Ming-Jing Hwang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan.
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, 11529, Taiwan.
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
3
|
Alkadhi KA. Synaptic Plasticity and Cognitive Ability in Experimental Adult-Onset Hypothyroidism. J Pharmacol Exp Ther 2024; 389:150-162. [PMID: 38508752 DOI: 10.1124/jpet.123.001887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
Adult-onset hypothyroidism impairs normal brain function. Research on animal models of hypothyroidism has revealed critical information on how deficiency of thyroid hormones impacts the electrophysiological and molecular functions of the brain, which leads to the well known cognitive impairment in untreated hypothyroid patients. Currently, such information can only be obtained from experiments on animal models of hypothyroidism. This review summarizes important research findings that pertain to understanding the clinical cognitive consequences of hypothyroidism, which will provide a better guiding path for therapy of hypothyroidism. SIGNIFICANCE STATEMENT: Cognitive impairment occurs during adult-onset hypothyroidism in both humans and animal models. Findings from animal studies validate clinical findings showing impaired long-term potentiation, decreased CaMKII, and increased calcineurin. Such findings can only be gleaned from animal experiments to show how hypothyroidism produces clinical symptoms.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| |
Collapse
|
4
|
Tao-Cheng JH, Moreira SL, Winters CA, Reese TS, Dosemeci A. Modification of the synaptic cleft under excitatory conditions. Front Synaptic Neurosci 2023; 15:1239098. [PMID: 37840571 PMCID: PMC10568020 DOI: 10.3389/fnsyn.2023.1239098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
The synaptic cleft is the extracellular part of the synapse, bridging the pre- and postsynaptic membranes. The geometry and molecular organization of the cleft is gaining increased attention as an important determinant of synaptic efficacy. The present study by electron microscopy focuses on short-term morphological changes at the synaptic cleft under excitatory conditions. Depolarization of cultured hippocampal neurons with high K+ results in an increased frequency of synaptic profiles with clefts widened at the periphery (open clefts), typically exhibiting patches of membranes lined by postsynaptic density, but lacking associated presynaptic membranes (18.0% open clefts in high K+ compared to 1.8% in controls). Similarly, higher frequencies of open clefts were observed in adult brain upon a delay of perfusion fixation to promote excitatory/ischemic conditions. Inhibition of basal activity in cultured neurons through the application of TTX results in the disappearance of open clefts whereas application of NMDA increases their frequency (19.0% in NMDA vs. 5.3% in control and 2.6% in APV). Depletion of extracellular Ca2+ with EGTA also promotes an increase in the frequency of open clefts (16.6% in EGTA vs. 4.0% in controls), comparable to that by depolarization or NMDA, implicating dissociation of Ca2+-dependent trans-synaptic bridges. Dissociation of transsynaptic bridges under excitatory conditions may allow perisynaptic mobile elements, such as AMPA receptors to enter the cleft. In addition, peripheral opening of the cleft would facilitate neurotransmitter clearance and thus may have a homeostatic and/or protective function.
Collapse
Affiliation(s)
- Jung-Hwa Tao-Cheng
- NINDS Electron Microscopy Facility, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Sandra L. Moreira
- NINDS Electron Microscopy Facility, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Christine A. Winters
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Thomas S. Reese
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Ayse Dosemeci
- Laboratory of Neurobiology, National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
5
|
Li L, Wang S, Duan X, Wang Z, Chang KC. Targeted Chemical Processing Initiating Biosome Action-Potential-Matched Artificial Synapses for the Brain-Machine Interface. ACS APPLIED MATERIALS & INTERFACES 2023; 15:40753-40761. [PMID: 37585625 DOI: 10.1021/acsami.3c07684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
A great gap still exists between artificial synapses and their biological counterparts in operation voltage or stimulation duration. Here, an artificial synaptic device based on a thin-film transistor with an operating voltage (-50-50 mV) analogous to biological action potential is developed by targeted chemical processing with the help of supercritical fluids. Chemical molecules [hexamethyldisilazane (HMDS)] are elaborately chosen and brought into the target interface to form charge receptors through supercritical processing. These charge receptors with the ability of capturing electrons mimic neurotransmitter receptors in terms of mechanism and constitute key players accounting for the synaptic behaviors. The relatively lower electrical barrier height contributes to an action-potential-matched operating voltage and considerably low power consumption (∼1 pJ/synaptic event), minimizing the divide with biological synapse for a seamless linkage to the biosystem or brain-machine interface. The stable synaptic behaviors also lead to near-ideal accuracy in pattern recognition. Moreover, this methodology that introduces chemical groups into a target interface can be viewed as a platform technology that could be adapted to other conventional devices with suitable chemical molecules to reach designed synaptic behaviors. This environmentally friendly and low-temperature processing method, which can be performed even after device fabrication, has the potential to play an important role in the future development of bionic devices.
Collapse
Affiliation(s)
- Lei Li
- School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, People's Republic of China
| | - Shidong Wang
- School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, People's Republic of China
| | - Xinqing Duan
- School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, People's Republic of China
| | - Zewen Wang
- School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, People's Republic of China
| | - Kuan-Chang Chang
- School of Electronic and Computer Engineering, Peking University Shenzhen Graduate School, Shenzhen, Guangdong 518055, People's Republic of China
| |
Collapse
|
6
|
Calis D, Hess M, Marchetta P, Singer W, Modro J, Nelissen E, Prickaerts J, Sandner P, Lukowski R, Ruth P, Knipper M, Rüttiger L. Acute deletion of the central MR/GR steroid receptor correlates with changes in LTP, auditory neural gain, and GC-A cGMP signaling. Front Mol Neurosci 2023; 16:1017761. [PMID: 36873102 PMCID: PMC9983609 DOI: 10.3389/fnmol.2023.1017761] [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: 08/12/2022] [Accepted: 01/18/2023] [Indexed: 02/19/2023] Open
Abstract
The complex mechanism by which stress can affect sensory processes such as hearing is still poorly understood. In a previous study, the mineralocorticoid (MR) and/or glucocorticoid receptor (GR) were deleted in frontal brain regions but not cochlear regions using a CaMKIIα-based tamoxifen-inducible Cre ERT2/loxP approach. These mice exhibit either a diminished (MRTMXcKO) or disinhibited (GRTMXcKO) auditory nerve activity. In the present study, we observed that mice differentially were (MRTMXcKO) or were not (GRTMXcKO) able to compensate for altered auditory nerve activity in the central auditory pathway. As previous findings demonstrated a link between central auditory compensation and memory-dependent adaptation processes, we analyzed hippocampal paired-pulse facilitation (PPF) and long-term potentiation (LTP). To determine which molecular mechanisms may impact differences in synaptic plasticity, we analyzed Arc/Arg3.1, known to control AMPA receptor trafficking, as well as regulators of tissue perfusion and energy consumption (NO-GC and GC-A). We observed that the changes in PPF of MRTMXcKOs mirrored the changes in their auditory nerve activity, whereas changes in the LTP of MRTMXcKOs and GRTMXcKOs mirrored instead the changes in their central compensation capacity. Enhanced GR expression levels in MRTMXcKOs suggest that MRs typically suppress GR expression. We observed that hippocampal LTP, GC-A mRNA expression levels, and ABR wave IV/I ratio were all enhanced in animals with elevated GR (MRTMXcKOs) but were all lower or not mobilized in animals with impaired GR expression levels (GRTMXcKOs and MRGRTMXcKOs). This suggests that GC-A may link LTP and auditory neural gain through GR-dependent processes. In addition, enhanced NO-GC expression levels in MR, GR, and MRGRTMXcKOs suggest that both receptors suppress NO-GC; on the other hand, elevated Arc/Arg3.1 levels in MRTMXcKOs and MRGRTMXcKOs but not GRTMXcKOs suggest that MR suppresses Arc/Arg3.1 expression levels. Conclusively, MR through GR inhibition may define the threshold for hemodynamic responses for LTP and auditory neural gain associated with GC-A.
Collapse
Affiliation(s)
- Dila Calis
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Morgan Hess
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Philine Marchetta
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Julian Modro
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Ellis Nelissen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, Netherlands
| | - Peter Sandner
- Bayer Health Care Pharmaceuticals, Global Drug Discovery Pharma Research Centre Wuppertal, Wuppertal, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Institute of Pharmacy, Pharmacology, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Quintanilla J, Jia Y, Lauterborn JC, Pruess BS, Le AA, Cox CD, Gall CM, Lynch G, Gunn BG. Novel types of frequency filtering in the lateral perforant path projections to dentate gyrus. J Physiol 2022; 600:3865-3896. [PMID: 35852108 PMCID: PMC9513824 DOI: 10.1113/jp283012] [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: 06/02/2022] [Accepted: 06/26/2022] [Indexed: 11/08/2022] Open
Abstract
Despite its evident importance to learning theory and models, the manner in which the lateral perforant path (LPP) transforms signals from entorhinal cortex to hippocampus is not well understood. The present studies measured synaptic responses in the dentate gyrus (DG) of adult mouse hippocampal slices during different patterns of LPP stimulation. Theta (5 Hz) stimulation produced a modest within-train facilitation that was markedly enhanced at the level of DG output. Gamma (50 Hz) activation resulted in a singular pattern with initial synaptic facilitation being followed by a progressively greater depression. DG output was absent after only two pulses. Reducing release probability with low extracellular calcium instated frequency facilitation to gamma stimulation while long-term potentiation, which increases release by LPP terminals, enhanced within-train depression. Relatedly, per terminal concentrations of VGLUT2, a vesicular glutamate transporter associated with high release probability, were much greater in the LPP than in CA3-CA1 connections. Attempts to circumvent the potent gamma filter using a series of short (three-pulse) 50 Hz trains spaced by 200 ms were only partially successful: composite responses were substantially reduced after the first burst, an effect opposite to that recorded in field CA1. The interaction between bursts was surprisingly persistent (>1.0 s). Low calcium improved throughput during theta/gamma activation but buffering of postsynaptic calcium did not. In all, presynaptic specializations relating to release probability produce an unusual but potent type of frequency filtering in the LPP. Patterned burst input engages a different type of filter with substrates that are also likely to be located presynaptically. KEY POINTS: The lateral perforant path (LPP)-dentate gyrus (DG) synapse operates as a low-pass filter, where responses to a train of 50 Hz, γ frequency activation are greatly suppressed. Activation with brief bursts of γ frequency information engages a secondary filter that persists for prolonged periods (lasting seconds). Both forms of LPP frequency filtering are influenced by presynaptic, as opposed to postsynaptic, processes; this contrasts with other hippocampal synapses. LPP frequency filtering is modified by the unique presynaptic long-term potentiation at this synapse. Computational simulations indicate that presynaptic factors associated with release probability and vesicle recycling may underlie the potent LPP-DG frequency filtering.
Collapse
Affiliation(s)
- Julian Quintanilla
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Yousheng Jia
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Julie C Lauterborn
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Benedict S Pruess
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Aliza A Le
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Conor D Cox
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| | - Christine M Gall
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
- Departments of Neurobiology & Behavior, University of California, Irvine, CA, USA
| | - Gary Lynch
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
- Departments of Psychiatry & Human Behavior, University of California, Irvine, CA, USA
| | - Benjamin G Gunn
- Departments of Anatomy & Neurobiology, University of California, Irvine, CA, USA
| |
Collapse
|
8
|
Savitska D, Hess M, Calis D, Marchetta P, Harasztosi C, Fink S, Eckert P, Ruth P, Rüttiger L, Knipper M, Singer W. Stress Affects Central Compensation of Neural Responses to Cochlear Synaptopathy in a cGMP-Dependent Way. Front Neurosci 2022; 16:864706. [PMID: 35968392 PMCID: PMC9372611 DOI: 10.3389/fnins.2022.864706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
In light of the increasing evidence supporting a link between hearing loss and dementia, it is critical to gain a better understanding of the nature of this relationship. We have previously observed that following cochlear synaptopathy, the temporal auditory processing (e.g., auditory steady state responses, ASSRs), is sustained when reduced auditory input is centrally compensated. This central compensation process was linked to elevated hippocampal long-term potentiation (LTP). We further observed that, independently of age, central responsiveness to cochlear synaptopathy can differ, resulting in either a low or high capacity to compensate for the reduced auditory input. Lower central compensation resulted in poorer temporal auditory processing, reduced hippocampal LTP, and decreased recruitment of activity-dependent brain-derived neurotrophic factor (BDNF) expression in hippocampal regions (low compensators). Higher central compensation capacity resulted in better temporal auditory processing, higher LTP responses, and increased activity-dependent BDNF expression in hippocampal regions. Here, we aimed to identify modifying factors that are potentially responsible for these different central responses. Strikingly, a poorer central compensation capacity was linked to lower corticosterone levels in comparison to those of high compensators. High compensators responded to repeated placebo injections with elevated blood corticosterone levels, reduced auditory brainstem response (ABR) wave I amplitude, reduced inner hair cell (IHC) ribbon number, diminished temporal processing, reduced LTP responses, and decreased activity-dependent hippocampal BDNF expression. In contrast, the same stress exposure through injection did not elevate blood corticosterone levels in low compensators, nor did it reduce IHC ribbons, ABR wave I amplitude, ASSR, LTP, or BDNF expression as seen in high compensators. Interestingly, in high compensators, the stress-induced responses, such as a decline in ABR wave I amplitude, ASSR, LTP, and BDNF could be restored through the "memory-enhancing" drug phosphodiesterase 9A inhibitor (PDE9i). In contrast, the same treatment did not improve these aspects in low compensators. Thus, central compensation of age-dependent cochlear synaptopathy is a glucocorticoid and cyclic guanosine-monophosphate (cGMP)-dependent neuronal mechanism that fails upon a blunted stress response.
Collapse
Affiliation(s)
- Daria Savitska
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Morgan Hess
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Dila Calis
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Philine Marchetta
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Csaba Harasztosi
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Stefan Fink
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Philipp Eckert
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Centre, Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Conserved patterns of functional organization between cortex and thalamus in mice. Proc Natl Acad Sci U S A 2022; 119:e2201481119. [PMID: 35588455 DOI: 10.1073/pnas.2201481119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
SignificanceNeuroanatomical tracing provides just a partial picture of information flow in the brain, because excitatory synapses are not all equal. Some strongly drive postsynaptic targets to transfer information, whereas others weakly modulate their responsiveness. Here, we show conserved patterns of synaptic function across somatosensory and visual thalamocortical circuits in mice involving higher-order thalamic nuclei. These nuclei serve as hubs in transthalamic or cortico-thalamo-cortical pathways. We report that feedforward transthalamic circuits in the somatosensory and visual systems operate to efficiently transmit information, whereas feedback transthalamic circuits act to modulate their target areas. These patterns may generalize to other brain systems and show how methods of synapse physiology and molecular biology can inform the exploration of brain circuitry and information processing.
Collapse
|
10
|
Romero-Barragán MT, Gruart A, Delgado-García JM. Transsynaptic Long-Term Potentiation in the Hippocampus of Behaving Mice. Front Synaptic Neurosci 2022; 13:811806. [PMID: 35126083 PMCID: PMC8810508 DOI: 10.3389/fnsyn.2021.811806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/31/2021] [Indexed: 11/17/2022] Open
Abstract
Long-term potentiation (LTP) is an experimental procedure that shares certain mechanisms with neuronal learning and memory processes and represents a well-known example of synaptic plasticity. LTP consists of an increase of the synaptic response to a control stimulus following the presentation of a high-frequency stimulation (HFS) train to an afferent pathway. This technique is studied mostly in the hippocampus due to the latter’s high susceptibility and its laminar nature which facilitates the location of defined synapses. Although most preceding studies have been performed in vitro, we have developed an experimental approach to carry out these experiments in alert behaving animals. The main goal of this study was to confirm the existence of synaptic changes in strength in synapses that are post-synaptic to the one presented with the HFS. We recorded field excitatory post-synaptic potentials (fEPSPs) evoked in five hippocampal synapses, from both hemispheres, of adult male mice. HFS was presented to the perforant pathway (PP). We characterized input/output curves, paired-pulse stimulation, and LTP of these synapses. We also performed depth-profile recordings to determine differences in fEPSP latencies. Collected data indicate that the five selected synapses have similar basic electrophysiological properties, a fact that enables an easier comparison of LTP characteristics. Importantly, we observed the presence of significant LTP in the contralateral CA1 (cCA1) area following the control stimulation of non-HFS-activated pathways. These results indicate that LTP appears as a physiological process present in synapses located far away from the HFS-stimulated afferent pathway.
Collapse
|
11
|
Bian H, Goh YY, Liu Y, Ling H, Xie L, Liu X. Stimuli-Responsive Memristive Materials for Artificial Synapses and Neuromorphic Computing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006469. [PMID: 33837601 DOI: 10.1002/adma.202006469] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Neuromorphic computing holds promise for building next-generation intelligent systems in a more energy-efficient way than the conventional von Neumann computing architecture. Memristive hardware, which mimics biological neurons and synapses, offers high-speed operation and low power consumption, enabling energy- and area-efficient, brain-inspired computing. Here, recent advances in memristive materials and strategies that emulate synaptic functions for neuromorphic computing are highlighted. The working principles and characteristics of biological neurons and synapses, which can be mimicked by memristive devices, are presented. Besides device structures and operation with different external stimuli such as electric, magnetic, and optical fields, how memristive materials with a rich variety of underlying physical mechanisms can allow fast, reliable, and low-power neuromorphic applications is also discussed. Finally, device requirements are examined and a perspective on challenges in developing memristive materials for device engineering and computing science is given.
Collapse
Affiliation(s)
- Hongyu Bian
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
| | - Yi Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 119077, Singapore
| | - Yuxia Liu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Center for Functional Materials, National University of Singapore Suzhou Research Institute, Suzhou, 215123, China
| | - Haifeng Ling
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Linghai Xie
- Key Laboratory for Organic Electronics and Information Displays and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore, 117543, Singapore
- Center for Functional Materials, National University of Singapore Suzhou Research Institute, Suzhou, 215123, China
| |
Collapse
|
12
|
Sánchez-Hidalgo AC, Arias-Aragón F, Romero-Barragán MT, Martín-Cuevas C, Delgado-García JM, Martinez-Mir A, Scholl FG. Selective expression of the neurexin substrate for presenilin in the adult forebrain causes deficits in associative memory and presynaptic plasticity. Exp Neurol 2021; 347:113896. [PMID: 34662541 DOI: 10.1016/j.expneurol.2021.113896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/27/2021] [Accepted: 10/10/2021] [Indexed: 01/25/2023]
Abstract
Presenilins (PS) form the active subunit of the gamma-secretase complex, which mediates the proteolytic clearance of a broad variety of type-I plasma membrane proteins. Loss-of-function mutations in PSEN1/2 genes are the leading cause of familial Alzheimer's disease (fAD). However, the PS/gamma-secretase substrates relevant for the neuronal deficits associated with a loss of PS function are not completely known. The members of the neurexin (Nrxn) family of presynaptic plasma membrane proteins are candidates to mediate aspects of the synaptic and memory deficits associated with a loss of PS function. Previous work has shown that fAD-linked PS mutants or inactivation of PS by genetic and pharmacological approaches failed to clear Nrxn C-terminal fragments (NrxnCTF), leading to its abnormal accumulation at presynaptic terminals. Here, we generated transgenic mice that selectively recreate the presynaptic accumulation of NrxnCTF in adult forebrain neurons, leaving unaltered the function of PS/gamma-secretase complex towards other substrates. Behavioral characterization identified selective impairments in NrxnCTF mice, including decreased fear-conditioning memory. Electrophysiological recordings in medial prefrontal cortex-basolateral amygdala (mPFC-BLA) of behaving mice showed normal synaptic transmission and uncovered specific defects in synaptic facilitation. These data functionally link the accumulation of NrxnCTF with defects in associative memory and short-term synaptic plasticity, pointing at impaired clearance of NrxnCTF as a new mediator in AD.
Collapse
Affiliation(s)
- Ana C Sánchez-Hidalgo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain
| | - Francisco Arias-Aragón
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain
| | | | - Celia Martín-Cuevas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain
| | | | - Amalia Martinez-Mir
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain
| | - Francisco G Scholl
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Avda. Manuel Siurot s/n, Sevilla 41013, Spain; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Avda. Sánchez Pizjuán, 4, Sevilla 41009, Spain.
| |
Collapse
|
13
|
Soutar CN, Grenier P, Patel A, Kabitsis PP, Olmstead MC, Bailey CDC, Dringenberg HC. Brain-Generated 17β-Estradiol Modulates Long-Term Synaptic Plasticity in the Primary Auditory Cortex of Adult Male Rats. Cereb Cortex 2021; 32:2140-2155. [PMID: 34628498 DOI: 10.1093/cercor/bhab345] [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: 11/13/2022] Open
Abstract
Neuron-derived 17β-estradiol (E2) alters synaptic transmission and plasticity in brain regions with endocrine and non-endocrine functions. Investigations into a modulatory role of E2 in synaptic activity and plasticity have mainly focused on the rodent hippocampal formation. In songbirds, E2 is synthesized by auditory forebrain neurons and promotes auditory signal processing and memory for salient acoustic stimuli; however, the modulatory effects of E2 on memory-related synaptic plasticity mechanisms have not been directly examined in the auditory forebrain. We investigated the effects of bidirectional E2 manipulations on synaptic transmission and long-term potentiation (LTP) in the rat primary auditory cortex (A1). Immunohistochemistry revealed widespread neuronal expression of the E2 biosynthetic enzyme aromatase in multiple regions of the rat sensory and association neocortex, including A1. In A1, E2 application reduced the threshold for in vivo LTP induction at layer IV synapses, whereas pharmacological suppression of E2 production by aromatase inhibition abolished LTP induction at layer II/III synapses. In acute A1 slices, glutamate and γ-aminobutyric acid (GABA) receptor-mediated currents were sensitive to E2 manipulations in a layer-specific manner. These findings demonstrate that locally synthesized E2 modulates synaptic transmission and plasticity in A1 and suggest potential mechanisms by which E2 contributes to auditory signal processing and memory.
Collapse
Affiliation(s)
- Chloe N Soutar
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Patrick Grenier
- Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Ashutosh Patel
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Pauline P Kabitsis
- Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Mary C Olmstead
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada.,Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Craig D C Bailey
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Hans C Dringenberg
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario K7L 3N6, Canada.,Department of Psychology, Queen's University, Kingston, Ontario K7L 3N6, Canada
| |
Collapse
|
14
|
Sancho L, Contreras M, Allen NJ. Glia as sculptors of synaptic plasticity. Neurosci Res 2021; 167:17-29. [PMID: 33316304 PMCID: PMC8513541 DOI: 10.1016/j.neures.2020.11.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022]
Abstract
Glial cells are non-neuronal cells in the nervous system that are crucial for proper brain development and function. Three major classes of glia in the central nervous system (CNS) include astrocytes, microglia and oligodendrocytes. These cells have dynamic morphological and functional properties and constantly surveil neural activity throughout life, sculpting synaptic plasticity. Astrocytes form part of the tripartite synapse with neurons and perform many homeostatic functions essential to proper synaptic function including clearing neurotransmitter and regulating ion balance; they can modify these properties, in addition to additional mechanisms such as gliotransmitter release, to influence short- and long-term plasticity. Microglia, the resident macrophage of the CNS, monitor synaptic activity and can eliminate synapses by phagocytosis or modify synapses by release of cytokines or neurotrophic factors. Oligodendrocytes regulate speed of action potential conduction and efficiency of information exchange through the formation of myelin, having important consequences for the plasticity of neural circuits. A deeper understanding of how glia modulate synaptic and circuit plasticity will further our understanding of the ongoing changes that take place throughout life in the dynamic environment of the CNS.
Collapse
Affiliation(s)
- Laura Sancho
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Minerva Contreras
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
15
|
Cannady R, Nguyen T, Padula AE, Rinker JA, Lopez MF, Becker HC, Woodward JJ, Mulholland PJ. Interaction of chronic intermittent ethanol and repeated stress on structural and functional plasticity in the mouse medial prefrontal cortex. Neuropharmacology 2021; 182:108396. [PMID: 33181147 PMCID: PMC7942177 DOI: 10.1016/j.neuropharm.2020.108396] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/05/2020] [Accepted: 11/06/2020] [Indexed: 01/27/2023]
Abstract
Stress is a risk factor that plays a considerable role in the development and maintenance of alcohol (ethanol) abuse and relapse. Preclinical studies examining ethanol-stress interactions have demonstrated elevated ethanol drinking, cognitive deficits, and negative affective behaviors in mice. However, the neural adaptations in prefrontal cortical regions that drive these aberrant behaviors produced by ethanol-stress interactions are unknown. In this study, male C57BL/6J mice were exposed to chronic intermittent ethanol (CIE) and repeated forced swim stress (FSS). After two cycles of CIE x FSS, brain slices containing the prelimbic (PrL) and infralimbic (IfL) cortex were prepared for analysis of adaptations in dendritic spines and synaptic plasticity. In the PrL cortex, total spine density was increased in mice exposed to CIE. Immediately following induction of long-term potentiation (LTP), the fEPSP slope was increased in the PrL of CIE x FSS treated mice, indicative of a presynaptic adaptation on post-tetanic potentiation (PTP). In the IfL cortex, CIE exposure regardless of FSS experience resulted in an increase in spine density. FSS alone or when combined with CIE exposure increased PTP following LTP induction. Repeated FSS episodes increased IfL cortical paired-pulse facilitation, a second measure of presynaptic plasticity. In summary, CIE exposure resulted in structural adaptations while repeated stress exposure drove metaplastic changes in presynaptic function, demonstrating distinct morphological and functional changes in PrL and IfL cortical neurons. Thus, the structural and functional adaptations may be one mechanism underlying the development of excessive drinking and cognitive deficits associated with ethanol-stress interactions.
Collapse
Affiliation(s)
- Reginald Cannady
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA; Department of Biology, College of Science and Technology, North Carolina Agricultural & Technical State University, 1601 East Market Street, Barnes Hall 215, Greensboro, NC, 27411, USA
| | - Tiffany Nguyen
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Audrey E Padula
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Jennifer A Rinker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Marcelo F Lopez
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Howard C Becker
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - John J Woodward
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Patrick J Mulholland
- Department of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA.
| |
Collapse
|
16
|
Electrical Properties and Biological Synaptic Simulation of Ag/MXene/SiO2/Pt RRAM Devices. ELECTRONICS 2020. [DOI: 10.3390/electronics9122098] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Utilizing electronic devices to emulate biological synapses for the construction of artificial neural networks has provided a feasible research approach for the future development of artificial intelligence systems. Until now, different kinds of electronic devices have been proposed in the realization of biological synapse functions. However, the device stability and the power consumption are major challenges for future industrialization applications. Herein, an electronic synapse of MXene/SiO2 structure-based resistive random-access memory (RRAM) devices has been designed and fabricated by taking advantage of the desirable properties of SiO2 and 2D MXene material. The proposed RRAM devices, Ag/MXene/SiO2/Pt, exhibit the resistance switching characteristics where both the volatile and nonvolatile behaviors coexist in a single device. These intriguing features of the Ag/MXene/SiO2/Pt devices make them more applicable for emulating biological synaptic plasticity. Additionally, the conductive mechanisms of the Ag/MXene/SiO2/Pt RRAM devices have been discussed on the basis of our experimental results.
Collapse
|
17
|
Choi S, Yang J, Wang G. Emerging Memristive Artificial Synapses and Neurons for Energy-Efficient Neuromorphic Computing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2004659. [PMID: 33006204 DOI: 10.1002/adma.202004659] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/12/2020] [Indexed: 06/11/2023]
Abstract
Memristors have recently attracted significant interest due to their applicability as promising building blocks of neuromorphic computing and electronic systems. The dynamic reconfiguration of memristors, which is based on the history of applied electrical stimuli, can mimic both essential analog synaptic and neuronal functionalities. These can be utilized as the node and terminal devices in an artificial neural network. Consequently, the ability to understand, control, and utilize fundamental switching principles and various types of device architectures of the memristor is necessary for achieving memristor-based neuromorphic hardware systems. Herein, a wide range of memristors and memristive-related devices for artificial synapses and neurons is highlighted. The device structures, switching principles, and the applications of essential synaptic and neuronal functionalities are sequentially presented. Moreover, recent advances in memristive artificial neural networks and their hardware implementations are introduced along with an overview of the various learning algorithms. Finally, the main challenges of the memristive synapses and neurons toward high-performance and energy-efficient neuromorphic computing are briefly discussed. This progress report aims to be an insightful guide for the research on memristors and neuromorphic-based computing.
Collapse
Affiliation(s)
- Sanghyeon Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jehyeon Yang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Gunuk Wang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
18
|
Local Design Principles at Hippocampal Synapses Revealed by an Energy-Information Trade-Off. eNeuro 2020; 7:ENEURO.0521-19.2020. [PMID: 32847867 PMCID: PMC7540928 DOI: 10.1523/eneuro.0521-19.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/01/2022] Open
Abstract
Synapses across different brain regions display distinct structure-function relationships. We investigated the interplay of fundamental design constraints that shape the transmission properties of the excitatory CA3-CA1 pyramidal cell connection, a prototypic synapse for studying the mechanisms of learning in the mammalian hippocampus. This small synapse is characterized by probabilistic release of transmitter, which is markedly facilitated in response to naturally occurring trains of action potentials. Based on a physiologically motivated computational model of the rat CA3 presynaptic terminal, we show how unreliability and short-term dynamics of vesicular release work together to regulate the trade-off of information transfer versus energy use. We propose that individual CA3-CA1 synapses are designed to operate near the maximum possible capacity of information transmission in an efficient manner. Experimental measurements reveal a wide range of vesicular release probabilities at hippocampal synapses, which may be a necessary consequence of long-term plasticity and homeostatic mechanisms that manifest as presynaptic modifications of the release probability. We show that the timescales and magnitude of short-term plasticity (STP) render synaptic information transfer nearly independent of differences in release probability. Thus, individual synapses transmit optimally while maintaining a heterogeneous distribution of presynaptic strengths indicative of synaptically-encoded memory representations. Our results support the view that organizing principles that are evident on higher scales of neural organization percolate down to the design of an individual synapse.
Collapse
|
19
|
Kapur M, Ganguly A, Nagy G, Adamson SI, Chuang JH, Frankel WN, Ackerman SL. Expression of the Neuronal tRNA n-Tr20 Regulates Synaptic Transmission and Seizure Susceptibility. Neuron 2020; 108:193-208.e9. [PMID: 32853550 DOI: 10.1016/j.neuron.2020.07.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/07/2020] [Accepted: 07/19/2020] [Indexed: 12/31/2022]
Abstract
The mammalian genome has hundreds of nuclear-encoded tRNAs, but the contribution of individual tRNA genes to cellular and organismal function remains unknown. Here, we demonstrate that mutations in a neuronally enriched arginine tRNA, n-Tr20, increased seizure threshold and altered synaptic transmission. n-Tr20 expression also modulated seizures caused by an epilepsy-linked mutation in Gabrg2, a gene encoding a GABAA receptor subunit. Loss of n-Tr20 altered translation initiation by activating the integrated stress response and suppressing mTOR signaling, the latter of which may contribute to altered neurotransmission in mutant mice. Deletion of a highly expressed isoleucine tRNA similarly altered these signaling pathways in the brain, suggesting that regulation of translation initiation is a conserved response to tRNA loss. Our data indicate that loss of a single member of a tRNA family results in multiple cellular phenotypes, highlighting the disease-causing potential of tRNA mutations.
Collapse
Affiliation(s)
- Mridu Kapur
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Archan Ganguly
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gabor Nagy
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Scott I Adamson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Wayne N Frankel
- Institute for Genomic Medicine, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Susan L Ackerman
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Section of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
20
|
Downregulation of parvalbumin expression in the prefrontal cortex during adolescence causes enduring prefrontal disinhibition in adulthood. Neuropsychopharmacology 2020; 45:1527-1535. [PMID: 32403119 PMCID: PMC7360578 DOI: 10.1038/s41386-020-0709-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 01/10/2023]
Abstract
The expression of the calcium binding protein parvalbumin (PV) has been observed in several cortical regions during development in a temporal pattern consistent with increased afferent-dependent activity. In the prefrontal cortex (PFC), PV expression appears last and continues to substantially increase throughout adolescence, yet the significance of this increase remains unclear. Because of the expression of PV in fast-spiking GABAergic interneurons, we hypothesized that PV upregulation during adolescence is necessary to sustain the increase in GABAergic activity observed in the PFC during this period. To test this hypothesis, we utilized an RNAi strategy to directly downregulate PV levels in the PFC during adolescence and examined its impact on prefrontal GABAergic function, plasticity, and associated behaviors during adulthood. The data indicate that a mere 25% reduction of adult PV levels in the PFC was sufficient to reduce local GABAergic transmission onto pyramidal neurons, disrupt prefrontal excitatory-inhibitory balance, and alter processing of afferent information from the ventral hippocampus. Accordingly, these animals displayed an impairment in the level of extinction learning of a trace fear conditioning response, a behavioral paradigm that requires intact PFC-ventral hippocampus connectivity. These results indicate the PV upregulation observed in the PFC during adolescence is necessary for refinement of prefrontal GABAergic function, the absence of which results in immature afferent processing and a hypofunctional state. Importantly, these results suggest there is a critical window of plasticity during which PV upregulation supports the acquisition of mature GABAergic phenotype necessary to sustain adult PFC functions.
Collapse
|
21
|
Patzke C, Brockmann MM, Dai J, Gan KJ, Grauel MK, Fenske P, Liu Y, Acuna C, Rosenmund C, Südhof TC. Neuromodulator Signaling Bidirectionally Controls Vesicle Numbers in Human Synapses. Cell 2020; 179:498-513.e22. [PMID: 31585084 DOI: 10.1016/j.cell.2019.09.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/28/2019] [Accepted: 09/06/2019] [Indexed: 10/25/2022]
Abstract
Neuromodulators bind to pre- and postsynaptic G protein-coupled receptors (GPCRs), are able to quickly change intracellular cyclic AMP (cAMP) and Ca2+ levels, and are thought to play important roles in neuropsychiatric and neurodegenerative diseases. Here, we discovered in human neurons an unanticipated presynaptic mechanism that acutely changes synaptic ultrastructure and regulates synaptic communication. Activation of neuromodulator receptors bidirectionally controlled synaptic vesicle numbers within nerve terminals. This control correlated with changes in the levels of cAMP-dependent protein kinase A-mediated phosphorylation of synapsin-1. Using a conditional deletion approach, we reveal that the neuromodulator-induced control of synaptic vesicle numbers was largely dependent on synapsin-1. We propose a mechanism whereby non-phosphorylated synapsin-1 "latches" synaptic vesicles to presynaptic clusters at the active zone. cAMP-dependent phosphorylation of synapsin-1 then removes the vesicles. cAMP-independent dephosphorylation of synapsin-1 in turn recruits vesicles. Synapsin-1 thereby bidirectionally regulates synaptic vesicle numbers and modifies presynaptic neurotransmitter release as an effector of neuromodulator signaling in human neurons.
Collapse
Affiliation(s)
- Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA.
| | - Marisa M Brockmann
- Institut für Neurophysiologie, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Jinye Dai
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Kathlyn J Gan
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - M Katharina Grauel
- Institut für Neurophysiologie, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Pascal Fenske
- Institut für Neurophysiologie, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Yu Liu
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Claudio Acuna
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - Christian Rosenmund
- Institut für Neurophysiologie, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| |
Collapse
|
22
|
Corsetti V, Borreca A, Latina V, Giacovazzo G, Pignataro A, Krashia P, Natale F, Cocco S, Rinaudo M, Malerba F, Florio R, Ciarapica R, Coccurello R, D’Amelio M, Ammassari-Teule M, Grassi C, Calissano P, Amadoro G. Passive immunotherapy for N-truncated tau ameliorates the cognitive deficits in two mouse Alzheimer's disease models. Brain Commun 2020; 2:fcaa039. [PMID: 32954296 PMCID: PMC7425324 DOI: 10.1093/braincomms/fcaa039] [Citation(s) in RCA: 27] [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: 10/24/2019] [Revised: 02/05/2020] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and neuropathological studies have shown that tau pathology better correlates with the severity of dementia than amyloid plaque burden, making tau an attractive target for the cure of Alzheimer's disease. We have explored whether passive immunization with the 12A12 monoclonal antibody (26-36aa of tau protein) could improve the Alzheimer's disease phenotype of two well-established mouse models, Tg2576 and 3xTg mice. 12A12 is a cleavage-specific monoclonal antibody which selectively binds the pathologically relevant neurotoxic NH226-230 fragment (i.e. NH2htau) of tau protein without cross-reacting with its full-length physiological form(s). We found out that intravenous administration of 12A12 monoclonal antibody into symptomatic (6 months old) animals: (i) reaches the hippocampus in its biologically active (antigen-binding competent) form and successfully neutralizes its target; (ii) reduces both pathological tau and amyloid precursor protein/amyloidβ metabolisms involved in early disease-associated synaptic deterioration; (iii) improves episodic-like type of learning/memory skills in hippocampal-based novel object recognition and object place recognition behavioural tasks; (iv) restores the specific up-regulation of the activity-regulated cytoskeleton-associated protein involved in consolidation of experience-dependent synaptic plasticity; (v) relieves the loss of dendritic spine connectivity in pyramidal hippocampal CA1 neurons; (vi) rescues the Alzheimer's disease-related electrophysiological deficits in hippocampal long-term potentiation at the CA3-CA1 synapses; and (vii) mitigates the neuroinflammatory response (reactive gliosis). These findings indicate that the 20-22 kDa NH2-terminal tau fragment is crucial target for Alzheimer's disease therapy and prospect immunotherapy with 12A12 monoclonal antibody as safe (normal tau-preserving), beneficial approach in contrasting the early Amyloidβ-dependent and independent neuropathological and cognitive alterations in affected subjects.
Collapse
Affiliation(s)
| | - Antonella Borreca
- Humanitas University Laboratory of Pharmacology and Brain Pathology, Neuro Center, 20089 Milan, Italy
- Institute of Neuroscience, 20129 Milan, Italy
| | | | | | | | - Paraskevi Krashia
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | - Francesca Natale
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Sara Cocco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Rinaudo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Rita Florio
- European Brain Research Institute (EBRI), 00161 Rome, Italy
| | | | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Institute for Complex Systems (ISC), CNR, 00185 Rome, Italy
| | - Marcello D’Amelio
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | | | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT)–National Research Council (CNR), 00133 Rome, Italy
| |
Collapse
|
23
|
Müller-Thomsen L, Borgmann D, Morcinek K, Schröder S, Dengler B, Moser N, Neumaier F, Schneider T, Schröder H, Huggenberger S. Consequences of hyperphosphorylated tau on the morphology and excitability of hippocampal neurons in aged tau transgenic mice. Neurobiol Aging 2020; 93:109-123. [PMID: 32278495 DOI: 10.1016/j.neurobiolaging.2020.03.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
The intracellular accumulation of hyperphosphorylated tau characterizes many neurodegenerative diseases such as Alzheimer's disease and frontotemporal dementia. A critical role for tau is supported by studies in transgenic mouse models expressing the P301L mutation with accumulation of hyperphosphorylated human tau in hippocampal pyramidal neurons of aged mice. Especially, the somatodendritic mislocalization of hyperphosphorylated tau seems to affect the neuronal network of the hippocampus. To show the consequences of aggregation of hyperphosphorylated tau within hippocampal neurons of aged mice, the CA1 pyramidal cells were analyzed morphologically and electrophysiologically. Here we demonstrate in the P301L pR5 mouse model that hyperphosphorylated tau leads to an increase in stubby spines and filopodia, as well as a decrease in total dendritic length of hippocampal pyramidal neurons due to a decrease in apical dendritic length and nodes. This atrophy is in line with the significant reduction in CA1 long-term potentiation. Furthermore, mutant tau induced a depolarized threshold for action potential initiation and an increased current of inward rectifying potassium channels, which should lead, together with the long-term potentiation decrease, to a decreased excitability of CA1 neurons.
Collapse
Affiliation(s)
| | - Diba Borgmann
- Department II of Anatomy, University of Cologne, Cologne, Germany
| | - Kerstin Morcinek
- Department II of Anatomy, University of Cologne, Cologne, Germany
| | - Sophia Schröder
- Department II of Anatomy, University of Cologne, Cologne, Germany
| | - Brigitte Dengler
- Department II of Anatomy, University of Cologne, Cologne, Germany
| | - Natasha Moser
- Department II of Anatomy, University of Cologne, Cologne, Germany
| | - Felix Neumaier
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Toni Schneider
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | | | - Stefan Huggenberger
- Department II of Anatomy, University of Cologne, Cologne, Germany; Institute of Anatomy and Clinical Morphology, Faculty of Health, Witten/Herdecke University, Witten, Germany
| |
Collapse
|
24
|
Djemil S, Chen X, Zhang Z, Lee J, Rauf M, Pak DTS, Dzakpasu R. Activation of nicotinic acetylcholine receptors induces potentiation and synchronization within in vitro hippocampal networks. J Neurochem 2019; 153:468-484. [PMID: 31821553 DOI: 10.1111/jnc.14938] [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: 09/08/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/08/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) are known to play a role in cognitive functions of the hippocampus, such as memory consolidation. Given that they conduct Ca2+ and are capable of regulating the release of glutamate and γ-aminobutyric acid (GABA) within the hippocampus, thereby shifting the excitatory-inhibitory ratio, we hypothesized that the activation of nAChRs will result in the potentiation of hippocampal networks and alter synchronization. We used nicotine as a tool to investigate the impact of activation of nAChRs on neuronal network dynamics in primary embryonic rat hippocampal cultures prepared from timed-pregnant Sprague-Dawley rats. We perturbed cultured hippocampal networks with increasing concentrations of bath-applied nicotine and performed network extracellular recordings of action potentials using a microelectrode array. We found that nicotine modulated network dynamics in a concentration-dependent manner; it enhanced firing of action potentials as well as facilitated bursting activity. In addition, we used pharmacological agents to determine the contributions of discrete nAChR subtypes to the observed network dynamics. We found that β4-containing nAChRs are necessary for the observed increases in spiking, bursting, and synchrony, while the activation of α7 nAChRs augments nicotine-mediated network potentiation but is not necessary for its manifestation. We also observed that antagonists of N-methyl-D-aspartate receptors (NMDARs) and group I metabotropic glutamate receptors (mGluRs) partially blocked the effects of nicotine. Furthermore, nicotine exposure promoted autophosphorylation of Ca2+ /calmodulin-dependent kinase II (CaMKII) and serine 831 phosphorylation of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit GluA1. These results suggest that nicotinic receptors induce potentiation and synchronization of hippocampal networks and glutamatergic synaptic transmission. Findings from this work highlight the impact of cholinergic signaling in generating network-wide potentiation in the form of enhanced spiking and bursting dynamics that coincide with molecular correlates of memory such as increased phosphorylation of CaMKII and GluA1. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.
Collapse
Affiliation(s)
- Sarra Djemil
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Xin Chen
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Ziyue Zhang
- Department of Physics, Georgetown University, Washington, DC, USA
| | - Jisoo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mikael Rauf
- Department of Human Science, Georgetown University Medical Center, Washington, DC, USA
| | - Daniel T S Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Rhonda Dzakpasu
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA.,Department of Physics, Georgetown University, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
25
|
Tang J, Yuan F, Shen X, Wang Z, Rao M, He Y, Sun Y, Li X, Zhang W, Li Y, Gao B, Qian H, Bi G, Song S, Yang JJ, Wu H. Bridging Biological and Artificial Neural Networks with Emerging Neuromorphic Devices: Fundamentals, Progress, and Challenges. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902761. [PMID: 31550405 DOI: 10.1002/adma.201902761] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/16/2019] [Indexed: 05/08/2023]
Abstract
As the research on artificial intelligence booms, there is broad interest in brain-inspired computing using novel neuromorphic devices. The potential of various emerging materials and devices for neuromorphic computing has attracted extensive research efforts, leading to a large number of publications. Going forward, in order to better emulate the brain's functions, its relevant fundamentals, working mechanisms, and resultant behaviors need to be re-visited, better understood, and connected to electronics. A systematic overview of biological and artificial neural systems is given, along with their related critical mechanisms. Recent progress in neuromorphic devices is reviewed and, more importantly, the existing challenges are highlighted to hopefully shed light on future research directions.
Collapse
Affiliation(s)
- Jianshi Tang
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, 100084, China
| | - Fang Yuan
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
| | - Xinke Shen
- Tsinghua Laboratory of Brain and Intelligence and Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zhongrui Wang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Mingyi Rao
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Yuanyuan He
- Tsinghua Laboratory of Brain and Intelligence and Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yuhao Sun
- Tsinghua Laboratory of Brain and Intelligence and Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Xinyi Li
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, 100084, China
| | - Wenbin Zhang
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
| | - Yijun Li
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
| | - Bin Gao
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, 100084, China
| | - He Qian
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, 100084, China
| | - Guoqiang Bi
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Sen Song
- Tsinghua Laboratory of Brain and Intelligence and Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - J Joshua Yang
- Department of Electrical and Computer Engineering, University of Massachusetts, Amherst, MA, 01003, USA
| | - Huaqiang Wu
- Institute of Microelectronics, Beijing Innovation Center for Future Chips (ICFC), Tsinghua University, Beijing, 100084, China
- Beijing National Research Center for Information Science and Technology (BNRist), Tsinghua University, Beijing, 100084, China
| |
Collapse
|
26
|
Response Adaptation in Barrel Cortical Neurons Facilitates Stimulus Detection during Rhythmic Whisker Stimulation in Anesthetized Mice. eNeuro 2019; 6:eN-NWR-0471-18. [PMID: 30957014 PMCID: PMC6449164 DOI: 10.1523/eneuro.0471-18.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/28/2019] [Accepted: 03/08/2019] [Indexed: 11/21/2022] Open
Abstract
Rodents use rhythmic whisker movements at frequencies between 4 and 12 Hz to sense the environment that will be disturbed when the animal touches an object. The aim of this work is to study the response adaptation to rhythmic whisker stimulation trains at 4 Hz in the barrel cortex and the sensitivity of cortical neurons to changes in the timing of the stimulation pattern. Longitudinal arrays of four iridium oxide electrodes were used to obtain single-unit recordings in supragranular, granular, and infragranular neurons in urethane anesthetized mice. The stimulation protocol consisted in a stimulation train of three air puffs (20 ms duration each) in which the time interval between the first and the third stimuli was fixed (500 ms) and the time interval between the first and the second stimuli changed (regular: 250 ms; “accelerando”: 375 ms; or “decelerando” stimulation train: 125 ms interval). Cortical neurons adapted strongly their response to regular stimulation trains. Response adaptation was reduced when accelerando or decelerando stimulation trains were applied. This facilitation of the shifted stimulus was mediated by activation of NMDA receptors because the effect was blocked by AP5. The facilitation was not observed in thalamic nuclei. Facilitation increased during periods of EEG activation induced by systemic application of IGF-I, probably by activation of NMDA receptors, as well. We suggest that response adaptation is the outcome of an intrinsic cortical information processing aimed at contributing to improve the detection of “unexpected” stimuli that disturbed the rhythmic behavior of exploration.
Collapse
|
27
|
Li D, Jing D, Liu Z, Chen Y, Huang F, Behnisch T. Enhanced Expression of Secreted α-Klotho in the Hippocampus Alters Nesting Behavior and Memory Formation in Mice. Front Cell Neurosci 2019; 13:133. [PMID: 31001090 PMCID: PMC6454015 DOI: 10.3389/fncel.2019.00133] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/18/2019] [Indexed: 12/30/2022] Open
Abstract
The klotho gene family consists of α-, β-, and γ-Klotho, which encode type I single-pass transmembrane proteins with large extracellular domains. α-Klotho exists as a full-length membrane-bound and as a soluble form after cleavage of the extracellular domain. Due to gene splicing, a short extracellular Klotho form can be expressed and secreted. Inactivation of α-Klotho leads to a phenotype that resembles accelerated aging, as the expression level of the α-Klotho protein in the hippocampal formation of mice decreases with age. Here, we show that intrahippocampal viral expression of secreted human α-Klotho alters social behavior and memory formation. Interestingly, overexpression of secreted human α-Klotho in the CA1 changed the nest-building behavior and improved object recognition, object location and passive avoidance memory. Moreover, α-Klotho overexpression increased hippocampal synaptic transmission in response to standardized stimulation strengths, altered paired-pulse facilitation of synaptic transmission, and enhanced activity-dependent synaptic plasticity. These results indicate that memory formation benefits from an augmented level of secreted α-Klotho.
Collapse
Affiliation(s)
- Dongxue Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Dongqing Jing
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ziyang Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Chen
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Fang Huang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Thomas Behnisch
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Active Zone Proteins RIM1αβ Are Required for Normal Corticostriatal Transmission and Action Control. J Neurosci 2018; 39:1457-1470. [PMID: 30559150 DOI: 10.1523/jneurosci.1940-18.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/13/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
Dynamic regulation of synaptic transmission at cortical inputs to the dorsal striatum is considered critical for flexible and efficient action learning and control. Presynaptic mechanisms governing the properties and plasticity of glutamate release from these inputs are not fully understood, and the corticostriatal synaptic processes that support normal action learning and control remain unclear. Here we show in male and female mice that conditional deletion of presynaptic proteins RIM1αβ (RIM1) from excitatory cortical neurons impairs corticostriatal synaptic transmission in the dorsolateral striatum. Key forms of presynaptic G-protein-coupled receptor-mediated short- and long-term striatal plasticity are spared following RIM1 deletion. Conditional RIM1 KO mice show heightened novelty-induced locomotion and impaired motor learning on the accelerating rotarod. They further show heightened self-paced instrumental responding for food and impaired learning of a habitual instrumental response strategy. Together, these findings reveal a selective role for presynaptic RIM1 in neurotransmitter release at prominent basal ganglia synapses, and provide evidence that RIM1-dependent processes help to promote the refinement of skilled actions, constrain goal-directed behaviors, and support the learning and use of habits.SIGNIFICANCE STATEMENT Our daily functioning hinges on the ability to flexibly and efficiently learn and control our actions. How the brain encodes these capacities is unclear. Here we identified a selective role for presynaptic proteins RIM1αβ in controlling glutamate release from cortical inputs to the dorsolateral striatum, a brain structure critical for action learning and control. Behavioral analysis of mice with restricted genetic deletion of RIM1αβ further revealed roles for RIM1αβ-dependent processes in the learning and refinement of motor skills and the balanced expression of goal-directed and habitual actions.
Collapse
|
29
|
Korteling JE, Brouwer AM, Toet A. A Neural Network Framework for Cognitive Bias. Front Psychol 2018; 9:1561. [PMID: 30233451 PMCID: PMC6129743 DOI: 10.3389/fpsyg.2018.01561] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/06/2018] [Indexed: 11/13/2022] Open
Abstract
Human decision-making shows systematic simplifications and deviations from the tenets of rationality ('heuristics') that may lead to suboptimal decisional outcomes ('cognitive biases'). There are currently three prevailing theoretical perspectives on the origin of heuristics and cognitive biases: a cognitive-psychological, an ecological and an evolutionary perspective. However, these perspectives are mainly descriptive and none of them provides an overall explanatory framework for the underlying mechanisms of cognitive biases. To enhance our understanding of cognitive heuristics and biases we propose a neural network framework for cognitive biases, which explains why our brain systematically tends to default to heuristic ('Type 1') decision making. We argue that many cognitive biases arise from intrinsic brain mechanisms that are fundamental for the working of biological neural networks. To substantiate our viewpoint, we discern and explain four basic neural network principles: (1) Association, (2) Compatibility, (3) Retainment, and (4) Focus. These principles are inherent to (all) neural networks which were originally optimized to perform concrete biological, perceptual, and motor functions. They form the basis for our inclinations to associate and combine (unrelated) information, to prioritize information that is compatible with our present state (such as knowledge, opinions, and expectations), to retain given information that sometimes could better be ignored, and to focus on dominant information while ignoring relevant information that is not directly activated. The supposed mechanisms are complementary and not mutually exclusive. For different cognitive biases they may all contribute in varying degrees to distortion of information. The present viewpoint not only complements the earlier three viewpoints, but also provides a unifying and binding framework for many cognitive bias phenomena.
Collapse
|
30
|
Chaussenot R, Amar M, Fossier P, Vaillend C. Dp71-Dystrophin Deficiency Alters Prefrontal Cortex Excitation-Inhibition Balance and Executive Functions. Mol Neurobiol 2018; 56:2670-2684. [PMID: 30051354 DOI: 10.1007/s12035-018-1259-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/17/2018] [Indexed: 01/19/2023]
Abstract
In the Duchenne muscular dystrophy (DMD) syndrome, mutations affecting expression of Dp71, the main dystrophin isoform of the multipromoter dmd gene in brain, have been associated with intellectual disability and neuropsychiatric disturbances. Patients' profile suggests alterations in prefrontal cortex-dependent executive processes, but the specific dysfunctions due to Dp71 deficiency are unclear. Dp71 is involved in brain ion homeostasis, and its deficiency is expected to increase neuronal excitability, which might compromise the integrity of neuronal networks undertaking high-order cognitive functions. Here, we used electrophysiological (patch clamp) and behavioral techniques in a transgenic mouse that display a selective loss of Dp71 and no muscular dystrophy, to identify changes in prefrontal cortex excitatory/inhibitory (E/I) balance and putative executive dysfunctions. We found prefrontal cortex E/I balance is shifted toward enhanced excitation in Dp71-null mice. This is associated with a selective alteration of AMPA receptor-mediated glutamatergic transmission and reduced synaptic plasticity, while inhibitory transmission is unaffected. Moreover, Dp71-null mice display deficits in cognitive processes that depend on prefrontal cortex integrity, such as cognitive flexibility and sensitivity of spatial working memory to proactive interference. Our data suggest that impaired cortical E/I balance and executive dysfunctions contribute to the intellectual and behavioral disturbances associated with Dp71 deficiency in DMD, in line with current neurobehavioral models considering these functions as key pathophysiological factors in various neurodevelopmental disorders. These new insights in DMD neurobiology also suggest new directions for therapeutic developments targeting excitatory neurotransmission, as well as for guidance of academic environment in severely affected DMD children.
Collapse
Affiliation(s)
- Rémi Chaussenot
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, Orsay, France
| | - Muriel Amar
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, Orsay, France.,Laboratoire de Toxinologie moléculaire et Biotechnologies, Institut des Sciences du Vivant Frédéric Joliot, CEA de Saclay, 91191, Gif-sur-Yvette, France
| | - Philippe Fossier
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, Orsay, France
| | - Cyrille Vaillend
- Neuroscience Paris-Saclay Institute (Neuro-PSI), UMR 9197, Université Paris Sud, CNRS, Université Paris Saclay, Orsay, France.
| |
Collapse
|
31
|
Courtney CD, Christian CA. Subregion-Specific Impacts of Genetic Loss of Diazepam Binding Inhibitor on Synaptic Inhibition in the Murine Hippocampus. Neuroscience 2018; 388:128-138. [PMID: 30031126 DOI: 10.1016/j.neuroscience.2018.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/27/2018] [Accepted: 07/09/2018] [Indexed: 11/16/2022]
Abstract
Benzodiazepines are commonly prescribed to treat neurological conditions including epilepsy, insomnia, and anxiety. The discovery of benzodiazepine-specific binding sites on γ-aminobutyric acid type-A receptors (GABAARs) led to the hypothesis that the brain may produce endogenous benzodiazepine-binding site ligands. An endogenous peptide, diazepam binding inhibitor (DBI), which can bind these sites, is thought to be capable of both enhancing and attenuating GABAergic transmission in different brain regions. However, the role that DBI plays in modulating GABAARs in the hippocampus remains unclear. Here, we investigated the role of DBI in modulating synaptic inhibition in the hippocampus using a constitutive DBI knockout mouse. Miniature and evoked inhibitory postsynaptic currents (mIPSCs, eIPSCs) were recorded from CA1 pyramidal cells and dentate gyrus (DG) granule cells. Loss of DBI signaling increased mIPSC frequency and amplitude in CA1 pyramidal cells from DBI knockout mice compared to wild-types. In DG granule cells, conversely, the loss of DBI decreased mIPSC amplitude and increased mIPSC decay time, indicating bidirectional modulation of GABAAR-mediated transmission in specific subregions of the hippocampus. eIPSC paired-pulse ratios were consistent across genotypes, suggesting that alterations in mIPSC frequency were not due to changes in presynaptic release probability. Furthermore, cells from DBI knockout mice did not display altered responsiveness to pharmacological applications of diazepam, a benzodiazepine, nor flumazenil, a benzodiazepine-binding site antagonist. These results provide evidence that genetic loss of DBI alters synaptic inhibition in the adult hippocampus, and that the direction of DBI-mediated modulation can vary discretely between specific subregions of the same brain structure.
Collapse
Affiliation(s)
- Connor D Courtney
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Catherine A Christian
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
32
|
Abstract
Various forms of synaptic plasticity underlie aspects of learning and memory. Synaptic augmentation is a form of short-term plasticity characterized by synaptic enhancement that persists for seconds following specific patterns of stimulation. The mechanisms underlying this form of plasticity are unclear but are thought to involve residual presynaptic Ca2+ Here, we report that augmentation was reduced in cultured mouse hippocampal neurons lacking the Ca2+ sensor, Doc2; other forms of short-term enhancement were unaffected. Doc2 binds Ca2+ and munc13 and translocates to the plasma membrane to drive augmentation. The underlying mechanism was not associated with changes in readily releasable pool size or Ca2+ dynamics, but rather resulted from superpriming a subset of synaptic vesicles. Hence, Doc2 forms part of the Ca2+-sensing apparatus for synaptic augmentation via a mechanism that is molecularly distinct from other forms of short-term plasticity.
Collapse
|
33
|
AD-Related N-Terminal Truncated Tau Is Sufficient to Recapitulate In Vivo the Early Perturbations of Human Neuropathology: Implications for Immunotherapy. Mol Neurobiol 2018; 55:8124-8153. [PMID: 29508283 DOI: 10.1007/s12035-018-0974-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/19/2018] [Indexed: 01/08/2023]
Abstract
The NH2tau 26-44 aa (i.e., NH2htau) is the minimal biologically active moiety of longer 20-22-kDa NH2-truncated form of human tau-a neurotoxic fragment mapping between 26 and 230 amino acids of full-length protein (htau40)-which is detectable in presynaptic terminals and peripheral CSF from patients suffering from AD and other non-AD neurodegenerative diseases. Nevertheless, whether its exogenous administration in healthy nontransgenic mice is able to elicit a neuropathological phenotype resembling human tauopathies has not been yet investigated. We explored the in vivo effects evoked by subchronic intracerebroventricular (i.c.v.) infusion of NH2htau or its reverse counterpart into two lines of young (2-month-old) wild-type mice (C57BL/6 and B6SJL). Six days after its accumulation into hippocampal parenchyma, significant impairment in memory/learning performance was detected in NH2htau-treated group in association with reduced synaptic connectivity and neuroinflammatory response. Compromised short-term plasticity in paired-pulse facilitation paradigm (PPF) was detected in the CA3/CA1 synapses from NH2htau-impaired animals along with downregulation in calcineurin (CaN)-stimulated pCREB/c-Fos pathway(s). Importantly, these behavioral, synaptotoxic, and neuropathological effects were independent from the genetic background, occurred prior to frank neuronal loss, and were specific because no alterations were detected in the control group infused with its reverse counterpart. Finally, a 2.0-kDa peptide which biochemically and immunologically resembles the injected NH2htau was endogenously detected in vivo, being present in hippocampal synaptosomal preparations from AD subjects. Given that the identification of the neurotoxic tau species is mandatory to develop a more effective tau-based immunological approach, our evidence can have important translational implications for cure of human tauopathies.
Collapse
|
34
|
Nouhi M, Zhang X, Yao N, Chergui K. CIQ, a positive allosteric modulator of GluN2C/D-containing N-methyl-d-aspartate receptors, rescues striatal synaptic plasticity deficit in a mouse model of Parkinson's disease. CNS Neurosci Ther 2017; 24:144-153. [PMID: 29230960 DOI: 10.1111/cns.12784] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/22/2017] [Accepted: 11/23/2017] [Indexed: 12/15/2022] Open
Abstract
AIMS To investigate if CIQ, a positive allosteric modulator of N-methyl-d-aspartate receptors (NMDARs) containing GluN2C/D subunits, rescues the loss of long-term potentiation (LTP) and forelimb-use asymmetry in a mouse model of Parkinson's disease (PD). METHODS We have used electrophysiology in brain slices and the cylinder test to examine the effect of CIQ on glutamatergic synaptic transmission, synaptic plasticity, and forelimb-use in the unilateral 6-hydroxydopamine-lesion mouse model of PD. RESULTS CIQ, applied in the perfusion solution, reversibly reduced glutamatergic synaptic transmission in the dopamine-depleted striatum and had no effect in the dopamine-intact striatum. LTP, a dopamine- and NMDAR-dependent form of synaptic plasticity, was induced in the dopamine-intact striatum but was lost in the dopamine-depleted striatum. This impaired LTP was restored in the presence of CIQ applied in the perfusion solution. This treatment, however, prevented LTP induction in control slices. In brain slices from mice which received single and chronic intraperitoneal injections of CIQ, LTP was restored in the dopamine-depleted striatum and unaffected in the dopamine-intact striatum. Forelimb-use asymmetry, a test which assesses deficits in paw usage in the unilateral lesion model of PD, was reversed by systemic chronic treatment with CIQ. CONCLUSION A positive allosteric modulator of GluN2C/D-containing NMDARs rescues LTP and forelimb-use asymmetry in a mouse model of PD. This study proposes GluN2D as a potential candidate for therapeutic intervention in PD.
Collapse
Affiliation(s)
- Mona Nouhi
- Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, The Karolinska Institute, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, The Karolinska Institute, Stockholm, Sweden
| | - Ning Yao
- Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, The Karolinska Institute, Stockholm, Sweden
| | - Karima Chergui
- Department of Physiology and Pharmacology, Section of Molecular Neurophysiology, The Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
35
|
Campbell SL, van Groen T, Kadish I, Smoot LHM, Bolger GB. Altered phosphorylation, electrophysiology, and behavior on attenuation of PDE4B action in hippocampus. BMC Neurosci 2017; 18:77. [PMID: 29197324 PMCID: PMC5712142 DOI: 10.1186/s12868-017-0396-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 11/28/2017] [Indexed: 01/19/2023] Open
Abstract
Background PDE4 cyclic nucleotide phosphodiesterases regulate 3′, 5′ cAMP abundance in the CNS and thereby regulate PKA activity and phosphorylation of CREB, which has been implicated in learning and memory, depression and other functions. The PDE4 isoform PDE4B1 also interacts with the DISC1 protein, implicated in neural development and behavioral disorders. The cellular functions of PDE4B1 have been investigated extensively, but its function(s) in the intact organism remained unexplored. Results To specifically disrupt PDE4B1, we developed mice that express a PDE4B1-D564A transgene in the hippocampus and forebrain. The transgenic mice showed enhanced phosphorylation of CREB and ERK1/2 in hippocampus. Hippocampal neurogenesis was increased in the transgenic mice. Hippocampal electrophysiological studies showed increased baseline synaptic transmission and enhanced LTP in male transgenic mice. Behaviorally, male transgenic mice showed increased activity in prolonged open field testing, but neither male nor female transgenic mice showed detectable anxiety-like behavior or antidepressant effects in the elevated plus-maze, tail-suspension or forced-swim tests. Neither sex showed any significant differences in associative fear conditioning or showed any demonstrable abnormalities in pre-pulse inhibition. Conclusions These data support the use of an isoform-selective approach to the study of PDE4B1 function in the CNS and suggest a probable role of PDE4B1 in synaptic plasticity and behavior. They also provide additional rationale and a refined approach to the development of small-molecule PDE4B1-selective inhibitors, which have potential functions in disorders of cognition, memory, mood and affect.
Collapse
Affiliation(s)
- Susan L Campbell
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA
| | - Thomas van Groen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Inga Kadish
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Lisa High Mitchell Smoot
- Department of Medicine, University of Alabama at Birmingham, NP 2501, 1720 2nd Ave S, Birmingham, AL, 35294-3300, USA
| | - Graeme B Bolger
- Department of Pharmacology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA. .,Department of Medicine, University of Alabama at Birmingham, NP 2501, 1720 2nd Ave S, Birmingham, AL, 35294-3300, USA. .,Center for Glial Biology in Health, Disease, and Cancer, Virginia Tech Carilion Research Institute, 2 Riverside Circle, Roanoke, VA, 24016, USA.
| |
Collapse
|
36
|
Roy N, Sanz-Leon P, Robinson PA. Spectral signatures of activity-dependent neural feedback in the corticothalamic system. Phys Rev E 2017; 96:052310. [PMID: 29347805 DOI: 10.1103/physreve.96.052310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Indexed: 11/07/2022]
Abstract
The modulation of neural quantities by presynaptic and postsynaptic activities via local feedback processes is investigated by incorporating nonlinear phenomena such as relative refractory period, synaptic enhancement, synaptic depression, and habituation. This is done by introducing susceptibilities, which quantify the response in either firing threshold or synaptic strength to unit change in either presynaptic or postsynaptic activity. Effects on the power spectra are then analyzed for a realistic corticothalamic model to determine the spectral signatures of various nonlinear processes and to what extent these are distinct. Depending on the feedback processes, there can be enhancements or reductions in low-frequency and/or alpha power, splitting of the alpha resonance, and/or appearance of new resonances at high frequencies. These features in the power spectra allow processes to be fully distinguished where they are unique, or partly distinguished if they are common to only a subset of feedbacks, and can potentially be used to constrain the types, strengths, and dynamics of feedbacks present.
Collapse
Affiliation(s)
- N Roy
- School of Physics, University of Sydney, New South Wales 2006, Australia and Center for Integrative Brain Function, University of Sydney, New South Wales 2006, Australia
| | - P Sanz-Leon
- School of Physics, University of Sydney, New South Wales 2006, Australia and Center for Integrative Brain Function, University of Sydney, New South Wales 2006, Australia
| | - P A Robinson
- School of Physics, University of Sydney, New South Wales 2006, Australia and Center for Integrative Brain Function, University of Sydney, New South Wales 2006, Australia
| |
Collapse
|
37
|
Sánchez-Rodríguez I, Temprano-Carazo S, Nájera A, Djebari S, Yajeya J, Gruart A, Delgado-García JM, Jiménez-Díaz L, Navarro-López JD. Activation of G-protein-gated inwardly rectifying potassium (Kir3/GirK) channels rescues hippocampal functions in a mouse model of early amyloid-β pathology. Sci Rep 2017; 7:14658. [PMID: 29116174 PMCID: PMC5676742 DOI: 10.1038/s41598-017-15306-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/25/2017] [Indexed: 12/15/2022] Open
Abstract
The hippocampus plays a critical role in learning and memory. Its correct performance relies on excitatory/inhibitory synaptic transmission balance. In early stages of Alzheimer’s disease (AD), neuronal hyperexcitability leads to network dysfunction observed in cortical regions such as the hippocampus. G-protein-gated potassium (GirK) channels induce neurons to hyperpolarize, contribute to the resting membrane potential and could compensate any excesses of excitation. Here, we have studied the relationship between GirK channels and hippocampal function in a mouse model of early AD pathology. Intracerebroventricular injections of amyloid-β (Aβ1-42) peptide—which have a causal role in AD pathogenesis—were performed to evaluate CA3–CA1 hippocampal synapse functionality in behaving mice. Aβ increased the excitability of the CA3–CA1 synapse, impaired long-term potentiation (LTP) and hippocampal oscillatory activity, and induced deficits in novel object recognition (NOR) tests. Injection of ML297 alone, a selective GirK activator, was also translated in LTP and NOR deficits. However, increasing GirK activity rescued all hippocampal deficits induced by Aβ due to the restoration of excitability values in the CA3–CA1 synapse. Our results show a synaptic mechanism, through GirK channel modulation, for the prevention of the hyperexcitability that causally contributes to synaptic, network, and cognitive deficits found in early AD pathogenesis.
Collapse
Affiliation(s)
- Irene Sánchez-Rodríguez
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, Ciudad Real, Spain
| | - Sara Temprano-Carazo
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, Ciudad Real, Spain
| | - Alberto Nájera
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, Ciudad Real, Spain
| | - Souhail Djebari
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, Ciudad Real, Spain
| | - Javier Yajeya
- University of Salamanca, Instituto de Neurociencias de Castilla y León, Salamanca, Spain
| | - Agnès Gruart
- Pablo de Olavide University, Division of Neurosciences, Seville, Spain
| | | | - Lydia Jiménez-Díaz
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, Ciudad Real, Spain.
| | - Juan D Navarro-López
- University of Castilla-La Mancha, NeuroPhysiology & Behavior Laboratory, Centro Regional de Investigaciones Biomédicas, School of Medicine of Ciudad Real, Ciudad Real, Spain.
| |
Collapse
|
38
|
Yuan K, Sheng H, Song J, Yang L, Cui D, Ma Q, Zhang W, Lai B, Chen M, Zheng P. Morphine treatment enhances glutamatergic input onto neurons of the nucleus accumbens via both disinhibitory and stimulating effect. Addict Biol 2017; 22:1756-1767. [PMID: 27549902 DOI: 10.1111/adb.12438] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/14/2016] [Accepted: 07/22/2016] [Indexed: 11/28/2022]
Abstract
Drug addiction is a chronic brain disorder characterized by the compulsive repeated use of drugs. The reinforcing effect of repeated use of drugs on reward plays an important role in morphine-induced addictive behaviors. The nucleus accumbens (NAc) is an important site where morphine treatment produces its reinforcing effect on reward. However, how morphine treatment produces its reinforcing effect on reward in the NAc remains to be clarified. In the present study, we studied the influence of morphine treatment on the effects of DA and observed whether morphine treatment could directly change glutamatergic synaptic transmission in the NAc. We also explored the functional significance of morphine-induced potentiation of glutamatergic synaptic transmission in the NAc at behavioral level. Our results show that (1) morphine treatment removes the inhibitory effect of DA on glutamatergic input onto NAc neurons; (2) morphine treatment potentiates glutamatergic input onto NAc neurons, especially the one from the basolateral amygdala (BLA) to the NAc; (3) blockade of glutamatergic synaptic transmission in the NAc or ablation of projection neurons from BLA to NAc significantly decreases morphine treatment-induced increase in locomotor activity. These results suggest that morphine treatment enhances glutamatergic input onto neurons of the NAc via both disinhibitory and stimulating effect and therefore increases locomotor activity.
Collapse
Affiliation(s)
- Kejing Yuan
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Huan Sheng
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Jiaojiao Song
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Li Yang
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Dongyang Cui
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Qianqian Ma
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Wen Zhang
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Bin Lai
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Ming Chen
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| | - Ping Zheng
- State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences and Institutes of Brain Science; Fudan University; China
| |
Collapse
|
39
|
Mercer A, Thomson AM. Cornu Ammonis Regions-Antecedents of Cortical Layers? Front Neuroanat 2017; 11:83. [PMID: 29018334 PMCID: PMC5622992 DOI: 10.3389/fnana.2017.00083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022] Open
Abstract
Studying neocortex and hippocampus in parallel, we are struck by the similarities. All three to four layered allocortices and the six layered mammalian neocortex arise in the pallium. All receive and integrate multiple cortical and subcortical inputs, provide multiple outputs and include an array of neuronal classes. During development, each cell positions itself to sample appropriate local and distant inputs and to innervate appropriate targets. Simpler cortices had already solved the need to transform multiple coincident inputs into serviceable outputs before neocortex appeared in mammals. Why then do phylogenetically more recent cortices need multiple pyramidal cell layers? A simple answer is that more neurones can compute more complex functions. The dentate gyrus and hippocampal CA regions-which might be seen as hippocampal antecedents of neocortical layers-lie side by side, albeit around a tight bend. Were the millions of cells of rat neocortex arranged in like fashion, the surface area of the CA pyramidal cell layers would be some 40 times larger. Even if evolution had managed to fold this immense sheet into the space available, the distances between neurones that needed to be synaptically connected would be huge and to maintain the speed of information transfer, massive, myelinated fiber tracts would be needed. How much more practical to stack the "cells that fire and wire together" into narrow columns, while retaining the mechanisms underlying the extraordinary precision with which circuits form. This demonstrably efficient arrangement presents us with challenges, however, not the least being to categorize the baffling array of neuronal subtypes in each of five "pyramidal layers." If we imagine the puzzle posed by this bewildering jumble of apical dendrites, basal dendrites and axons, from many different pyramidal and interneuronal classes, that is encountered by a late-arriving interneurone insinuating itself into a functional circuit, we can perhaps begin to understand why definitive classification, covering every aspect of each neurone's structure and function, is such a challenge. Here, we summarize and compare the development of these two cortices, the properties of their neurones, the circuits they form and the ordered, unidirectional flow of information from one hippocampal region, or one neocortical layer, to another.
Collapse
Affiliation(s)
- Audrey Mercer
- Department of Pharmacology, School of Pharmacy, University College London, London, United Kingdom
| | - Alex M. Thomson
- Department of Pharmacology, School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
40
|
Thompson JL, Drysdale M, Baimel C, Kaur M, MacGowan T, Pitman KA, Borgland SL. Obesity-Induced Structural and Neuronal Plasticity in the Lateral Orbitofrontal Cortex. Neuropsychopharmacology 2017; 42:1480-1490. [PMID: 28042870 PMCID: PMC5398895 DOI: 10.1038/npp.2016.284] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/22/2016] [Accepted: 12/18/2016] [Indexed: 12/19/2022]
Abstract
The orbitofrontal cortex (OFC) integrates sensory information with the current value of foods and updates actions based on this information. Obese humans and rats fed a cafeteria diet have impaired devaluation of food rewards, implicating a potential obesity-induced dysfunction of the OFC. We hypothesized that obesity alters OFC pyramidal neuronal structure and function and reduces conditioned suppression of feeding. Rats were given restricted (1 h/day), extended (23 h/day) or no (chow only) access to a cafeteria diet and tested for a conditioned suppression of feeding. Golgi-cox impregnation and whole-cell patch clamp experiments were performed in lateral OFC pyramidal neurons of rats from the 3 feeding groups. Rats with 40 days of extended, but not restricted, access to a cafeteria diet became obese and continued to feed during foot shock-predicting cues. Access to a cafeteria diet induced morphological changes in basilar dendrites of lateral OFC pyramidal neurons. While there were no alterations in excitatory synaptic transmission underlying altered spine density, we observed a more depolarized resting membrane potential. This was accompanied by decreased inhibitory synaptic transmission onto lateral OFC pyramidal neurons due to decreased release probability at GABAergic inputs. These changes could underlie the inability of the OFC to encode changes in the motivation value of food that is observed in obese rodents and humans.
Collapse
Affiliation(s)
- Jennifer L Thompson
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Michael Drysdale
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Corey Baimel
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Manpreet Kaur
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Taigan MacGowan
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Kimberley A Pitman
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Stephanie L Borgland
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada,Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW Calgary, Alberta T2N 4N1, Canada, Tel: +1 403 220 6967, Fax: +4032832700, E-mail:
| |
Collapse
|
41
|
Sase T, Katori Y, Komuro M, Aihara K. Bifurcation Analysis on Phase-Amplitude Cross-Frequency Coupling in Neural Networks with Dynamic Synapses. Front Comput Neurosci 2017; 11:18. [PMID: 28424606 PMCID: PMC5371682 DOI: 10.3389/fncom.2017.00018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/13/2017] [Indexed: 11/13/2022] Open
Abstract
We investigate a discrete-time network model composed of excitatory and inhibitory neurons and dynamic synapses with the aim at revealing dynamical properties behind oscillatory phenomena possibly related to brain functions. We use a stochastic neural network model to derive the corresponding macroscopic mean field dynamics, and subsequently analyze the dynamical properties of the network. In addition to slow and fast oscillations arising from excitatory and inhibitory networks, respectively, we show that the interaction between these two networks generates phase-amplitude cross-frequency coupling (CFC), in which multiple different frequency components coexist and the amplitude of the fast oscillation is modulated by the phase of the slow oscillation. Furthermore, we clarify the detailed properties of the oscillatory phenomena by applying the bifurcation analysis to the mean field model, and accordingly show that the intermittent and the continuous CFCs can be characterized by an aperiodic orbit on a closed curve and one on a torus, respectively. These two CFC modes switch depending on the coupling strength from the excitatory to inhibitory networks, via the saddle-node cycle bifurcation of a one-dimensional torus in map (MT1SNC), and may be associated with the function of multi-item representation. We believe that the present model might have potential for studying possible functional roles of phase-amplitude CFC in the cerebral cortex.
Collapse
Affiliation(s)
- Takumi Sase
- Graduate School of Information Science and Technology, The University of TokyoTokyo, Japan
| | - Yuichi Katori
- Institute of Industrial Science, The University of TokyoTokyo, Japan.,The School of Systems Information Science, Future University HakodateHokkaido, Japan
| | - Motomasa Komuro
- Center for Fundamental Education, Teikyo University of ScienceYamanashi, Japan
| | - Kazuyuki Aihara
- Graduate School of Information Science and Technology, The University of TokyoTokyo, Japan.,Institute of Industrial Science, The University of TokyoTokyo, Japan
| |
Collapse
|
42
|
Blitz DM, Pritchard AE, Latimer JK, Wakefield AT. Muscles innervated by a single motor neuron exhibit divergent synaptic properties on multiple time scales. ACTA ACUST UNITED AC 2017; 220:1233-1244. [PMID: 28104799 DOI: 10.1242/jeb.148908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/10/2017] [Indexed: 11/20/2022]
Abstract
Adaptive changes in the output of neural circuits underlying rhythmic behaviors are relayed to muscles via motor neuron activity. Presynaptic and postsynaptic properties of neuromuscular junctions can impact the transformation from motor neuron activity to muscle response. Further, synaptic plasticity occurring on the time scale of inter-spike intervals can differ between multiple muscles innervated by the same motor neuron. In rhythmic behaviors, motor neuron bursts can elicit additional synaptic plasticity. However, it is unknown whether plasticity regulated by the longer time scale of inter-burst intervals also differs between synapses from the same neuron, and whether any such distinctions occur across a physiological activity range. To address these issues, we measured electrical responses in muscles innervated by a chewing circuit neuron, the lateral gastric (LG) motor neuron, in a well-characterized small motor system, the stomatogastric nervous system (STNS) of the Jonah crab, Cancer borealisIn vitro and in vivo, sensory, hormonal and modulatory inputs elicit LG bursting consisting of inter-spike intervals of 50-250 ms and inter-burst intervals of 2-24 s. Muscles expressed similar facilitation measured with paired stimuli except at the shortest inter-spike interval. However, distinct decay time constants resulted in differences in temporal summation. In response to bursting activity, augmentation occurred to different extents and saturated at different inter-burst intervals. Further, augmentation interacted with facilitation, resulting in distinct intra-burst facilitation between muscles. Thus, responses of multiple target muscles diverge across a physiological activity range as a result of distinct synaptic properties sensitive to multiple time scales.
Collapse
Affiliation(s)
- Dawn M Blitz
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - Amy E Pritchard
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | - John K Latimer
- Department of Biology, Miami University, Oxford, OH 45056, USA
| | | |
Collapse
|
43
|
Functional Organization of Cutaneous and Muscle Afferent Synapses onto Immature Spinal Lamina I Projection Neurons. J Neurosci 2017; 37:1505-1517. [PMID: 28069928 DOI: 10.1523/jneurosci.3164-16.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/13/2016] [Accepted: 12/24/2016] [Indexed: 11/21/2022] Open
Abstract
It is well established that sensory afferents innervating muscle are more effective at inducing hyperexcitability within spinal cord circuits compared with skin afferents, which likely contributes to the higher prevalence of chronic musculoskeletal pain compared with pain of cutaneous origin. However, the mechanisms underlying these differences in central nociceptive signaling remain incompletely understood, as nothing is known about how superficial dorsal horn neurons process sensory input from muscle versus skin at the synaptic level. Using a novel ex vivo spinal cord preparation, here we identify the functional organization of muscle and cutaneous afferent synapses onto immature rat lamina I spino-parabrachial neurons, which serve as a major source of nociceptive transmission to the brain. Stimulation of the gastrocnemius nerve and sural nerve revealed significant convergence of muscle and cutaneous afferent synaptic input onto individual projection neurons. Muscle afferents displayed a higher probability of glutamate release, although short-term synaptic plasticity was similar between the groups. Importantly, muscle afferent synapses exhibited greater relative expression of Ca2+-permeable AMPARs compared with cutaneous inputs. In addition, the prevalence and magnitude of spike timing-dependent long-term potentiation were significantly higher at muscle afferent synapses, where it required Ca2+-permeable AMPAR activation. Collectively, these results provide the first evidence for afferent-specific properties of glutamatergic transmission within the superficial dorsal horn. A larger propensity for activity-dependent strengthening at muscle afferent synapses onto developing spinal projection neurons could contribute to the enhanced ability of these sensory inputs to sensitize central nociceptive networks and thereby evoke persistent pain in children following injury.SIGNIFICANCE STATEMENT The neurobiological mechanisms underlying the high prevalence of chronic musculoskeletal pain remain poorly understood, in part because little is known about why sensory neurons innervating muscle appear more capable of sensitizing nociceptive pathways in the CNS compared with skin afferents. The present study identifies, for the first time, the functional properties of muscle and cutaneous afferent synapses onto immature lamina I projection neurons, which convey nociceptive information to the brain. Despite many similarities, an enhanced relative expression of Ca2+-permeable AMPA receptors at muscle afferent synapses drives greater LTP following repetitive stimulation. A preferential ability of the dorsal horn synaptic network to amplify nociceptive input arising from muscle is predicted to favor the generation of musculoskeletal pain following injury.
Collapse
|
44
|
Zhang YF, Li QQ, Qu J, Sun CM, Wang Y. Alterations of motor cortical microcircuit in a depressive-like mouse model produced by light deprivation. Neuroscience 2017; 341:79-94. [DOI: 10.1016/j.neuroscience.2016.11.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 01/01/2023]
|
45
|
Pecoraro V, Sardone LM, Chisari M, Licata F, Li Volsi G, Perciavalle V, Ciranna L, Costa L. A subnanomolar concentration of Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) pre-synaptically modulates glutamatergic transmission in the rat hippocampus acting through acetylcholine. Neuroscience 2016; 340:551-562. [PMID: 27816700 DOI: 10.1016/j.neuroscience.2016.10.061] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 10/16/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022]
Abstract
The neuropeptide PACAP modulates synaptic transmission in the hippocampus exerting multiple effects through different receptor subtypes: the underlying mechanisms have not yet been completely elucidated. The neurotransmitter acetylcholine (ACh) also exerts a well-documented modulation of hippocampal synaptic transmission and plasticity. Since PACAP was shown to stimulate ACh release in the hippocampus, we tested whether PACAP acting through ACh might indirectly modulate glutamate-mediated synaptic transmission at a pre- and/or at a post-synaptic level. Using patch clamp on rat hippocampal slices, we tested PACAP effects on stimulation-evoked AMPA receptor-mediated excitatory post-synaptic currents (EPSCsAMPA) in the CA3-CA1 synapse and on spontaneous miniature EPSCs (mEPSCs) in CA1 pyramidal neurons. A subnanomolar dose of PACAP (0.5nM) decreased EPSCsAMPA amplitude, enhanced EPSC paired-pulse facilitation (PPF) and reduced mEPSC frequency, indicating a pre-synaptic decrease of glutamate release probability: these effects were abolished by simultaneous blockade of muscarinic and nicotinic ACh receptors, indicating the involvement of endogenous ACh. The effect of subnanomolar PACAP was abolished by a PAC1 receptor antagonist but not by a VPAC receptor blocker. At a higher concentration (10nM), PACAP inhibited EPSCsAMPA: this effect persisted in the presence of ACh receptor antagonists and did not involve any change in PPF or in mEPSC frequency, thus was not mediated by ACh and was exerted post- synaptically on CA1 pyramidal neurons. We suggest that a high-affinity PAC1 receptor pre-synaptically modulates hippocampal glutamatergic transmission acting through ACh. Therefore, administration of PACAP at very low doses might be envisaged in cognitive diseases with reduced cholinergic transmission.
Collapse
Affiliation(s)
- Valeria Pecoraro
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández de Elche, San Juan de Alicante, Spain
| | - Lara Maria Sardone
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Italy
| | - Mariangela Chisari
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Italy
| | - Flora Licata
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Italy
| | - Guido Li Volsi
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Italy
| | - Vincenzo Perciavalle
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Italy
| | - Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Italy.
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, Italy
| |
Collapse
|
46
|
Stanley EF. Single calcium channel domain gating of synaptic vesicle fusion at fast synapses; analysis by graphic modeling. Channels (Austin) 2016; 9:324-33. [PMID: 26457441 PMCID: PMC4826128 DOI: 10.1080/19336950.2015.1098793] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
At fast-transmitting presynaptic terminals Ca2+ enter through voltage gated calcium channels (CaVs) and bind to a synaptic vesicle (SV) -associated calcium sensor (SV-sensor) to gate fusion and discharge. An open CaV generates a high-concentration plume, or nanodomain of Ca2+ that dissipates precipitously with distance from the pore. At most fast synapses, such as the frog neuromuscular junction (NMJ), the SV sensors are located sufficiently close to individual CaVs to be gated by single nanodomains. However, at others, such as the mature rodent calyx of Held (calyx of Held), the physiology is more complex with evidence that CaVs that are both close and distant from the SV sensor and it is argued that release is gated primarily by the overlapping Ca2+ nanodomains from many CaVs. We devised a 'graphic modeling' method to sum Ca2+ from individual CaVs located at varying distances from the SV-sensor to determine the SV release probability and also the fraction of that probability that can be attributed to single domain gating. This method was applied first to simplified, low and high CaV density model release sites and then to published data on the contrasting frog NMJ and the rodent calyx of Held native synapses. We report 3 main predictions: the SV-sensor is positioned very close to the point at which the SV fuses with the membrane; single domain-release gating predominates even at synapses where the SV abuts a large cluster of CaVs, and even relatively remote CaVs can contribute significantly to single domain-based gating.
Collapse
Affiliation(s)
- Elise F Stanley
- a Toronto Western Research Institute ; Toronto , Ontario Canada
| |
Collapse
|
47
|
Felix RA, Magnusson AK. Development of excitatory synaptic transmission to the superior paraolivary and lateral superior olivary nuclei optimizes differential decoding strategies. Neuroscience 2016; 334:1-12. [PMID: 27476438 DOI: 10.1016/j.neuroscience.2016.07.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/08/2016] [Accepted: 07/23/2016] [Indexed: 11/17/2022]
Abstract
The superior paraolivary nucleus (SPON) is a prominent structure in the mammalian auditory brainstem with a proposed role in encoding transient broadband sounds such as vocalized utterances. Currently, the source of excitatory pathways that project to the SPON and how these inputs contribute to SPON function are poorly understood. To shed light on the nature of these inputs, we measured evoked excitatory postsynaptic currents (EPSCs) in the SPON originating from the intermediate acoustic stria and compared them with the properties of EPSCs in the lateral superior olive (LSO) originating from the ventral acoustic stria during auditory development from postnatal day 5 to 22 in mice. Before hearing onset, EPSCs in the SPON and LSO are very similar in size and kinetics. After the onset of hearing, SPON excitation is refined to extremely few (2:1) fibers, with each strengthened by an increase in release probability, yielding fast and strong EPSCs. LSO excitation is recruited from more fibers (5:1), resulting in strong EPSCs with a comparatively broader stimulus-response range after hearing onset. Evoked SPON excitation is comparatively weaker than evoked LSO excitation, likely due to a larger fraction of postsynaptic GluR2-containing Ca2+-impermeable AMPA receptors after hearing onset. Taken together, SPON excitation develops synaptic properties that are suited for transmitting single events with high temporal reliability and the strong, dynamic LSO excitation is compatible with high rate-level sensitivity. Thus, the excitatory input pathways to the SPON and LSO mature to support different decoding strategies of respective coarse temporal and sound intensity information at the brainstem level.
Collapse
Affiliation(s)
- Richard A Felix
- Unit of Audiology, Department of Clinical Science Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Anna K Magnusson
- Unit of Audiology, Department of Clinical Science Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
48
|
Liu H, Lan Y, Bing YH, Chu CP, Qiu DL. N-methyl-D-Aspartate Receptors Contribute to Complex Spike Signaling in Cerebellar Purkinje Cells: An In vivo Study in Mice. Front Cell Neurosci 2016; 10:172. [PMID: 27445699 PMCID: PMC4928496 DOI: 10.3389/fncel.2016.00172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/16/2016] [Indexed: 11/13/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are post-synaptically expressed at climbing fiber-Purkinje cell (CF-PC) synapses in cerebellar cortex in adult mice and contributed to CF-PC synaptic transmission under in vitro conditions. In this study, we investigated the role of NMDARs at CF-PC synapses during the spontaneous complex spike (CS) activity in cerebellar cortex in urethane-anesthetized mice, by in vivo whole-cell recording technique and pharmacological methods. Under current-clamp conditions, cerebellar surface application of NMDA (50 μM) induced an increase in the CS-evoked pause of simple spike (SS) firing accompanied with a decrease in the SS firing rate. Under voltage-clamp conditions, application of NMDA enhanced the waveform of CS-evoked inward currents, which expressed increases in the area under curve (AUC) and spikelet number of spontaneous CS. NMDA increased the AUC of spontaneous CS in a concentration-dependent manner. The EC50 of NMDA for increasing AUC of spontaneous CS was 33.4 μM. Moreover, NMDA significantly increased the amplitude, half-width and decay time of CS-evoked after-hyperpolarization (AHP) currents. Blockade of NMDARs with D-(-)-2-amino-5-phosphonopentanoic acid (D-APV, 250 μM) decreased the AUC, spikelet number, and amplitude of AHP currents. In addition, the NMDA-induced enhancement of CS activity could not be observed after α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors were blocked. The results indicated that NMDARs of CF-PC synapses contributed to the spontaneous CS activity by enhancing CS-evoked inward currents and AHP currents.
Collapse
Affiliation(s)
- Heng Liu
- Cellular Function Research Center, Yanbian UniversityYanji, China; Department of Physiology and Pathophysiology, College of Medicine, Yanbian UniversityYanji, China
| | - Yan Lan
- Cellular Function Research Center, Yanbian UniversityYanji, China; Department of Physiology and Pathophysiology, College of Medicine, Yanbian UniversityYanji, China
| | - Yan-Hua Bing
- Cellular Function Research Center, Yanbian UniversityYanji, China; Department of Physiology and Pathophysiology, College of Medicine, Yanbian UniversityYanji, China
| | - Chun-Ping Chu
- Cellular Function Research Center, Yanbian University Yanji, China
| | - De-Lai Qiu
- Cellular Function Research Center, Yanbian UniversityYanji, China; Department of Physiology and Pathophysiology, College of Medicine, Yanbian UniversityYanji, China; Key Laboratory of Natural Resource of the Changbai Mountain and Functional Molecular of the Ministry of Education, Yanbian UniversityYanji, China
| |
Collapse
|
49
|
Effects of pre-natal alcohol exposure on hippocampal synaptic plasticity: Sex, age and methodological considerations. Neurosci Biobehav Rev 2016; 64:12-34. [PMID: 26906760 DOI: 10.1016/j.neubiorev.2016.02.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 01/14/2016] [Accepted: 02/18/2016] [Indexed: 12/28/2022]
Abstract
The consumption of alcohol during gestation is detrimental to the developing central nervous system (CNS). The severity of structural and functional brain alterations associated with alcohol intake depends on many factors including the timing and duration of alcohol consumption. The hippocampal formation, a brain region implicated in learning and memory, is highly susceptible to the effects of developmental alcohol exposure. Some of the observed effects of alcohol on learning and memory may be due to changes at the synaptic level, as this teratogen has been repeatedly shown to interfere with hippocampal synaptic plasticity. At the molecular level alcohol interferes with receptor proteins and can disrupt hormones that are important for neuronal signaling and synaptic plasticity. In this review we examine the consequences of prenatal and early postnatal alcohol exposure on hippocampal synaptic plasticity and highlight the numerous factors that can modulate the effects of alcohol. We also discuss some potential mechanisms responsible for these changes as well as emerging therapeutic avenues that are beginning to be explored.
Collapse
|
50
|
Jia Y, Parker D. Short-Term Synaptic Plasticity at Interneuronal Synapses Could Sculpt Rhythmic Motor Patterns. Front Neural Circuits 2016; 10:4. [PMID: 26869889 PMCID: PMC4738240 DOI: 10.3389/fncir.2016.00004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 01/15/2016] [Indexed: 01/19/2023] Open
Abstract
The output of a neuronal network depends on the organization and functional properties of its component cells and synapses. While the characterization of synaptic properties has lagged cellular analyses, a potentially important aspect in rhythmically active networks is how network synapses affect, and are in turn affected by, network activity. This could lead to a potential circular interaction where short-term activity-dependent synaptic plasticity is both influenced by and influences the network output. The analysis of synaptic plasticity in the lamprey locomotor network was extended here to characterize the short-term plasticity of connections between network interneurons and to try and address its potential network role. Paired recordings from identified interneurons in quiescent networks showed synapse-specific synaptic properties and plasticity that supported the presence of two hemisegmental groups that could influence bursting: depression in an excitatory interneuron group, and facilitation in an inhibitory feedback circuit. The influence of activity-dependent synaptic plasticity on network activity was investigated experimentally by changing Ringer Ca(2+) levels, and in a simple computer model. A potential caveat of the experimental analyses was that changes in Ringer Ca(2+) (and compensatory adjustments in Mg(2+) in some cases) could alter several other cellular and synaptic properties. Several of these properties were tested, and while there was some variability, these were not usually significantly affected by the Ringer changes. The experimental analyses suggested that depression of excitatory inputs had the strongest influence on the patterning of network activity. The simulation supported a role for this effect, and also suggested that the inhibitory facilitating group could modulate the influence of the excitatory synaptic depression. Short-term activity-dependent synaptic plasticity has not generally been considered in spinal cord models. These results provide further evidence for short-term plasticity between locomotor network interneurons. As this plasticity could influence the patterning of the network output it should be considered as a potential functional component of spinal cord networks.
Collapse
Affiliation(s)
| | - David Parker
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, UK
| |
Collapse
|