151
|
Pham AQ, Dore K. Novel approaches to increase synaptic resilience as potential treatments for Alzheimer's disease. Semin Cell Dev Biol 2023; 139:84-92. [PMID: 35370089 DOI: 10.1016/j.semcdb.2022.03.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 12/31/2022]
Abstract
A significant proportion of brains with Alzheimer's disease pathology are obtained from patients that were cognitively normal, suggesting that differences within the brains of these individuals made them resilient to the disease. Here, we describe recent approaches that specifically increase synaptic resilience, as loss of synapses is considered to be the first change in the brains of Alzheimer's patients. We start by discussing studies showing benefit from increased expression of neurotrophic factors and protective genes. Methods that effectively make dendritic spines stronger, specifically by acting through actin network proteins, scaffolding proteins and inhibition of phosphatases are described next. Importantly, the therapeutic strategies presented in this review tackle Alzheimer's disease not by targeting plaques and tangles, but instead by making synapses resilient to the pathology associated with Alzheimer's disease, which has tremendous potential.
Collapse
Affiliation(s)
- Andrew Q Pham
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla 92093, United States
| | - Kim Dore
- Department of Neurosciences, Center for Neural Circuits and Behavior, UCSD, La Jolla 92093, United States.
| |
Collapse
|
152
|
Kasai H, Ucar H, Morimoto Y, Eto F, Okazaki H. Mechanical transmission at spine synapses: Short-term potentiation and working memory. Curr Opin Neurobiol 2023; 80:102706. [PMID: 36931116 DOI: 10.1016/j.conb.2023.102706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/17/2022] [Accepted: 02/15/2023] [Indexed: 03/17/2023]
Abstract
Do dendritic spines, which comprise the postsynaptic component of most excitatory synapses, exist only for their structural dynamics, receptor trafficking, and chemical and electrical compartmentation? The answer is no. Simultaneous investigation of both spine and presynaptic terminals has recently revealed a novel feature of spine synapses. Spine enlargement pushes the presynaptic terminals with muscle-like force and augments the evoked glutamate release for up to 20 min. We now summarize the evidence that such mechanical transmission shares critical features in common with short-term potentiation (STP) and may represent the cellular basis of short-term and working memory. Thus, spine synapses produce the force of learning to leave structural traces for both short and long-term memories.
Collapse
Affiliation(s)
- Haruo Kasai
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Hasan Ucar
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yuichi Morimoto
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Fumihiro Eto
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hitoshi Okazaki
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
153
|
Espadas I, Wingfield J, Grinman E, Ghosh I, Chanda K, Nakahata Y, Bauer K, Raveendra B, Kiebler M, Yasuda R, Rangaraju V, Puthanveettil S. SLAMR, a synaptically targeted lncRNA, facilitates the consolidation of contextual fear memory. RESEARCH SQUARE 2023:rs.3.rs-2489387. [PMID: 36993323 PMCID: PMC10055528 DOI: 10.21203/rs.3.rs-2489387/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
LncRNAs are involved in critical processes for cell homeostasis and function. However, it remains largely unknown whether and how the transcriptional regulation of long noncoding RNAs results in activity-dependent changes at the synapse and facilitate formation of long-term memories. Here, we report the identification of a novel lncRNA, SLAMR, that becomes enriched in CA1- but not in CA3-hippocampal neurons upon contextual fear conditioning. SLAMR is transported to dendrites via the molecular motor KIF5C and recruited to the synapse in response to stimulation. Loss of function of SLAMR reduced dendritic complexity and impaired activity dependent changes in spine structural plasticity. Interestingly, gain of function of SLAMR enhanced dendritic complexity, and spine density through enhanced translation. Analyses of the SLAMR interactome revealed its association with CaMKIIα protein through a 220-nucleotide element and its modulation of CaMKIIα activity. Furthermore, loss-of-function of SLAMR in CA1 selectively impairs consolidation but neither acquisition, recall, nor extinction of fear memory and spatial memory. Together, these results establish a new mechanism for activity dependent changes at the synapse and consolidation of contextual fear.
Collapse
Affiliation(s)
- Isabel Espadas
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Jenna Wingfield
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Eddie Grinman
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Ilika Ghosh
- Max Planck Florida Institute, Jupiter, FL, USA
| | - Kaushik Chanda
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | | | - Karl Bauer
- Biomedical Center (BMC), Department for Cell Biology, Medical Faculty, Ludwig-Maximilians-University of Munich, 82152 Planegg-Martinsried, Germany
| | - Bindu Raveendra
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Michael Kiebler
- Biomedical Center (BMC), Department for Cell Biology, Medical Faculty, Ludwig-Maximilians-University of Munich, 82152 Planegg-Martinsried, Germany
| | | | | | - Sathyanarayanan Puthanveettil
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| |
Collapse
|
154
|
Hyun JH, Hannan P, Iwamoto H, Blakely RD, Kwon HB. Serotonin in the orbitofrontal cortex enhances cognitive flexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531880. [PMID: 36945634 PMCID: PMC10028980 DOI: 10.1101/2023.03.09.531880] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Cognitive flexibility is a brain's ability to switch between different rules or action plans depending on the context. However, cellular level understanding of cognitive flexibility have been largely unexplored. We probed a specific serotonergic pathway from dorsal raphe nuclei (DRN) to the orbitofrontal cortex (OFC) while animals are performing reversal learning task. We found that serotonin release from DRN to the OFC promotes reversal learning. A long-range connection between these two brain regions was confirmed anatomically and functionally. We further show that spatiotemporally precise serotonergic action directly enhances the excitability of OFC neurons and offers enhanced spike probability of OFC network. Serotonergic action facilitated the induction of synaptic plasticity by enhancing Ca2+ influx at dendritic spines in the OFC. Thus, our findings suggest that a key signature of flexibility is the formation of choice specific ensembles via serotonin-dependent synaptic plasticity.
Collapse
Affiliation(s)
- Jung Ho Hyun
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
- Department of Brain Sciences, DGIST, Daegu, Republic of Korea
| | - Patrick Hannan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
| | - Hideki Iwamoto
- Department of Biomedical Science and Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Randy D. Blakely
- Department of Biomedical Science and Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
| |
Collapse
|
155
|
Yagishita S. Cellular bases for reward-related dopamine actions. Neurosci Res 2023; 188:1-9. [PMID: 36496085 DOI: 10.1016/j.neures.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 11/09/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022]
Abstract
Dopamine neurons exhibit transient increases and decreases in their firing rate upon reward and punishment for learning. This bidirectional modulation of dopamine dynamics occurs on the order of hundreds of milliseconds, and it is sensitively detected to reinforce the preceding sensorimotor events. These observations indicate that the mechanisms of dopamine detection at the projection sites are of remarkable precision, both in time and concentration. A major target of dopamine projection is the striatum, including the ventral region of the nucleus accumbens, which mainly comprises dopamine D1 and D2 receptor (D1R and D2R)-expressing spiny projection neurons. Although the involvement of D1R and D2R in dopamine-dependent learning has been suggested, the exact cellular bases for detecting transient dopamine signaling remain unclear. This review discusses recent cellular studies on the novel synaptic mechanisms for detecting dopamine transient signals associated with learning. Analyses of behavior based on these mechanisms have further revealed new behavioral aspects that are closely associated with these synaptic mechanisms. Thus, it is gradually possible to mechanistically explain behavioral learning via synaptic and cellular bases in rodents.
Collapse
Affiliation(s)
- Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan; International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
156
|
Montanez-Miranda C, Bramlett SN, Hepler JR. RGS14 expression in CA2 hippocampus, amygdala, and basal ganglia: Implications for human brain physiology and disease. Hippocampus 2023; 33:166-181. [PMID: 36541898 PMCID: PMC9974931 DOI: 10.1002/hipo.23492] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
RGS14 is a multifunctional scaffolding protein that is highly expressed within postsynaptic spines of pyramidal neurons in hippocampal area CA2. Known roles of RGS14 in CA2 include regulating G protein, H-Ras/ERK, and calcium signaling pathways to serve as a natural suppressor of synaptic plasticity and postsynaptic signaling. RGS14 also shows marked postsynaptic expression in major structures of the limbic system and basal ganglia, including the amygdala and both the ventral and dorsal subdivisions of the striatum. In this review, we discuss the signaling functions of RGS14 and its role in postsynaptic strength (long-term potentiation) and spine structural plasticity in CA2 hippocampal neurons, and how RGS14 suppression of plasticity impacts linked behaviors such as spatial learning, object memory, and fear conditioning. We also review RGS14 expression in the limbic system and basal ganglia and speculate on its possible roles in regulating plasticity in these regions, with a focus on behaviors related to emotion and motivation. Finally, we explore the functional implications of RGS14 in various brain circuits and speculate on its possible roles in certain disease states such as hippocampal seizures, addiction, and anxiety disorders.
Collapse
Affiliation(s)
| | | | - John R. Hepler
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322-3090
| |
Collapse
|
157
|
Rollenhagen A, Anstötz M, Zimmermann K, Kasugai Y, Sätzler K, Molnar E, Ferraguti F, Lübke JHR. Layer-specific distribution and expression pattern of AMPA- and NMDA-type glutamate receptors in the barrel field of the adult rat somatosensory cortex: a quantitative electron microscopic analysis. Cereb Cortex 2023; 33:2342-2360. [PMID: 35732315 PMCID: PMC9977369 DOI: 10.1093/cercor/bhac212] [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: 12/09/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/14/2022] Open
Abstract
AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) and NMDA (N-methyl-d-aspartate) glutamate receptors are driving forces for synaptic transmission and plasticity at neocortical synapses. However, their distribution pattern in the adult rat neocortex is largely unknown and was quantified using freeze fracture replication combined with postimmunogold-labeling. Both receptors were co-localized at layer (L)4 and L5 postsynaptic densities (PSDs). At L4 dendritic shaft and spine PSDs, the number of gold grains detecting AMPA was similar, whereas at L5 shaft PSDs AMPA-receptors outnumbered those on spine PSDs. Their number was significantly higher at L5 vs. L4 PSDs. At L4 and L5 dendritic shaft PSDs, the number of gold grains detecting GluN1 was ~2-fold higher than at spine PSDs. The number of gold grains detecting the GluN1-subunit was higher for both shaft and spine PSDs in L5 vs. L4. Both receptors showed a large variability in L4 and L5. A high correlation between the number of gold grains and PSD size for both receptors and targets was observed. Both receptors were distributed over the entire PSD but showed a layer- and target-specific distribution pattern. The layer- and target-specific distribution of AMPA and GluN1 glutamate receptors partially contribute to the observed functional differences in synaptic transmission and plasticity in the neocortex.
Collapse
Affiliation(s)
- Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany
| | - Max Anstötz
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany.,Institute of Anatomy II, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Universitätsstr. 1, Düsseldorf 40001, Germany
| | - Kerstin Zimmermann
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany
| | - Yu Kasugai
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1a, Innsbruck A-6020, Austria
| | - Kurt Sätzler
- School of Biomedical Sciences, University of Ulster, Cromore Rd., Londonderry BT52 1SA, United Kingdom
| | - Elek Molnar
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Francesco Ferraguti
- Department of Pharmacology, Medical University of Innsbruck, Peter Mayr Strasse 1a, Innsbruck A-6020, Austria
| | - Joachim H R Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Leo Brandt Str., Jülich 52425, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH/Medical University Aachen, Pauwelstr. 30, Aachen 52074, Germany.,JARA Translational Medicine Jülich/Aachen, Germany
| |
Collapse
|
158
|
Lukacs IP, Francavilla R, Field M, Hunter E, Howarth M, Horie S, Plaha P, Stacey R, Livermore L, Ansorge O, Tamas G, Somogyi P. Differential effects of group III metabotropic glutamate receptors on spontaneous inhibitory synaptic currents in spine-innervating double bouquet and parvalbumin-expressing dendrite-targeting GABAergic interneurons in human neocortex. Cereb Cortex 2023; 33:2101-2142. [PMID: 35667019 PMCID: PMC9977385 DOI: 10.1093/cercor/bhac195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/12/2022] Open
Abstract
Diverse neocortical GABAergic neurons specialize in synaptic targeting and their effects are modulated by presynaptic metabotropic glutamate receptors (mGluRs) suppressing neurotransmitter release in rodents, but their effects in human neocortex are unknown. We tested whether activation of group III mGluRs by L-AP4 changes GABAA receptor-mediated spontaneous inhibitory postsynaptic currents (sIPSCs) in 2 distinct dendritic spine-innervating GABAergic interneurons recorded in vitro in human neocortex. Calbindin-positive double bouquet cells (DBCs) had columnar "horsetail" axons descending through layers II-V innervating dendritic spines (48%) and shafts, but not somata of pyramidal and nonpyramidal neurons. Parvalbumin-expressing dendrite-targeting cell (PV-DTC) axons extended in all directions innervating dendritic spines (22%), shafts (65%), and somata (13%). As measured, 20% of GABAergic neuropil synapses innervate spines, hence DBCs, but not PV-DTCs, preferentially select spine targets. Group III mGluR activation paradoxically increased the frequency of sIPSCs in DBCs (to median 137% of baseline) but suppressed it in PV-DTCs (median 92%), leaving the amplitude unchanged. The facilitation of sIPSCs in DBCs may result from their unique GABAergic input being disinhibited via network effect. We conclude that dendritic spines receive specialized, diverse GABAergic inputs, and group III mGluRs differentially regulate GABAergic synaptic transmission to distinct GABAergic cell types in human cortex.
Collapse
Affiliation(s)
- Istvan P Lukacs
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | | | - Martin Field
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Emily Hunter
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Michael Howarth
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Sawa Horie
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Richard Stacey
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Laurent Livermore
- Department of Neurosurgery, John Radcliffe Hospital, OUH NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Olaf Ansorge
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Gabor Tamas
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, 6726 Szeged, Hungary
| | - Peter Somogyi
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| |
Collapse
|
159
|
Smilovic D, Rietsche M, Fellenz M, Drakew A, Vuksic M, Deller T. Loss of tumor necrosis factor (TNF)-receptor 1 and TNF-receptor 2 partially replicate effects of TNF deficiency on dendritic spines of granule cells in mouse dentate gyrus. J Comp Neurol 2023; 531:281-293. [PMID: 36221961 DOI: 10.1002/cne.25424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 12/24/2022]
Abstract
The cytokine tumor necrosis factor (TNF) is involved in the regulation of physiological and pathophysiological processes in the central nervous system. In previous work, we showed that mice lacking constitutive levels of TNF exhibit a reduction in spine density and changes in spine head size distribution of dentate granule cells. Here, we investigated which TNF-receptor pathway is responsible for this phenotype and analyzed granule cell spine morphology in TNF-R1-, TNF-R2-, and TNF-R1/R2-deficient mice. Single granule cells were filled with Alexa568 in fixed hippocampal brain slices and immunostained for the actin-modulating protein synaptopodin (SP), a marker for strong and stable spines. An investigator blind to genotype investigated dendritic spines using deconvolved confocal image stacks. Similar to TNF-deficient mice, TNF-R1 and TNF-R2 mutants showed a decrease in the size of small spines (SP-negative) with TNF-R1/R2-KO mice exhibiting an additive effect. TNF-R1 mutants also showed an increase in the size of large spines (SP-positive), mirroring the situation in TNF-deficient mice. Unlike the TNF-deficient mouse, none of the TNF-R mutants exhibited a reduction in their granule cell spine densities. Since TNF tunes the excitability of networks, lack of constitutive TNF reduces network excitation. This may explain why we observed alterations in spine head size distributions in TNF- and TNF-R-deficient granule cells. The changes in spine density observed in the TNF-deficient mouse could not be linked to canonical TNF-R-signaling. Instead, noncanonical pathways or unknown developmental functions of TNF may cause this phenomenon.
Collapse
Affiliation(s)
- Dinko Smilovic
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt, Germany.,Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Michael Rietsche
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt, Germany
| | - Meike Fellenz
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt, Germany
| | - Alexander Drakew
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt, Germany
| | - Mario Vuksic
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt, Germany.,Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
160
|
Kleinjan MS, Buchta WC, Ogelman R, Hwang IW, Kuwajima M, Hubbard DD, Kareemo DJ, Prikhodko O, Olah SL, Gomez Wulschner LE, Abraham WC, Franco SJ, Harris KM, Oh WC, Kennedy MJ. Dually innervated dendritic spines develop in the absence of excitatory activity and resist plasticity through tonic inhibitory crosstalk. Neuron 2023; 111:362-371.e6. [PMID: 36395772 PMCID: PMC9899020 DOI: 10.1016/j.neuron.2022.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 07/13/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
Dendritic spines can be directly connected to both inhibitory and excitatory presynaptic terminals, resulting in nanometer-scale proximity of opposing synaptic functions. While dually innervated spines (DiSs) are observed throughout the central nervous system, their developmental timeline and functional properties remain uncharacterized. Here we used a combination of serial section electron microscopy, live imaging, and local synapse activity manipulations to investigate DiS development and function in rodent hippocampus. Dual innervation occurred early in development, even on spines where the excitatory input was locally silenced. Synaptic NMDA receptor currents were selectively reduced at DiSs through tonic GABAB receptor signaling. Accordingly, spine enlargement normally associated with long-term potentiation on singly innervated spines (SiSs) was blocked at DiSs. Silencing somatostatin interneurons or pharmacologically blocking GABABRs restored NMDA receptor function and structural plasticity to levels comparable to neighboring SiSs. Thus, hippocampal DiSs are stable structures where function and plasticity are potently regulated by nanometer-scale GABAergic signaling.
Collapse
Affiliation(s)
- Mason S Kleinjan
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - William C Buchta
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Roberto Ogelman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - In-Wook Hwang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Masaaki Kuwajima
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Dusten D Hubbard
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Dean J Kareemo
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Olga Prikhodko
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Samantha L Olah
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Luis E Gomez Wulschner
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Santos J Franco
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Kristen M Harris
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Won Chan Oh
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Matthew J Kennedy
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
161
|
Zhang X, Yang S, Han S, Sun Y, Han M, Zheng X, Li F, Wei Y, Wang Y, Bi J. Differential methylation of circRNA m6A in an APP/PS1 Alzheimer's disease mouse model. Mol Med Rep 2023; 27:55. [PMID: 36660942 PMCID: PMC9879070 DOI: 10.3892/mmr.2023.12942] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 11/25/2022] [Indexed: 01/19/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurological disease characterized by memory loss and progressive cognitive impairment. The characteristic AD pathologies include extracellular senile plaques formed by β‑amyloid protein deposition, neurofibrillary tangles formed by hyper‑phosphorylation of τ protein and neuronal loss caused by glial cell proliferation. However, the pathogenesis of AD is still unclear. Dysregulation of RNA methylation is associated with biological processes, including neurodevelopment and neurodegenerative disease. N6‑methyladenosine (m6A) is the main modification in eukaryotic RNA and may be associated with the pathophysiology of AD. Circular RNA (circRNA) is a new type of evolutionarily conserved non‑coding RNA without 5'‑cap and 3'‑polyadenylic acid tail. circRNA undergoes m6A RNA methylation and may be involved in the pathogenesis of AD. In the present study, high‑throughput sequencing was performed to assess the degree of circRNA m6A methylation in APP/PS1 AD and C57BL/6 mice. These results suggested that circRNA m6A methylation in AD mice was markedly altered compared to the control group. Furthermore, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis was used to predict associated pathways; genes with different circRNA m6A methylation in AD mice were associated with 'axon guidance', 'long‑term potentiation', 'glutamatergic synapse', 'cholinergic synapse', 'GABAergic synapse' and 'long‑term depression'. Methylated RNA immunoprecipitation reverse transcription‑quantitative PCR demonstrated that among the eight selected circRNA m6A genes, there were five genes that demonstrated significantly increased methylation and three demonstrated significantly decreased methylation. In summary, the present study indicated that circRNA m6A methylation may be associated with pathogenesis of AD.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Suge Yang
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Song Han
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yuan Sun
- Department of Outpatients, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Min Han
- Department of Geriatric Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Xiaolei Zheng
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Fan Li
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yan Wei
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yun Wang
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China,Correspondence to: Dr Yun Wang, Department of Neurological Medicine, The Second Hospital of Shandong University, 247 Beiyuan Road, Jinan, Shandong 250033, P.R. China, E-mail:
| | - Jianzhong Bi
- Department of Neurological Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
162
|
Altered integration of excitatory inputs onto the basal dendrites of layer 5 pyramidal neurons in a mouse model of Fragile X syndrome. Proc Natl Acad Sci U S A 2023; 120:e2208963120. [PMID: 36595706 PMCID: PMC9926222 DOI: 10.1073/pnas.2208963120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Layer 5 (L5) pyramidal neurons receive predictive and sensory inputs in a compartmentalized manner at their apical and basal dendrites, respectively. To uncover how integration of sensory inputs is affected in autism spectrum disorders (ASD), we used two-photon glutamate uncaging to activate spines in the basal dendrites of L5 pyramidal neurons from a mouse model of Fragile X syndrome (FXS), the most common genetic cause of ASD. While subthreshold excitatory inputs integrate linearly in wild-type animals, surprisingly those with FXS summate sublinearly, contradicting what would be expected of sensory hypersensitivity classically associated with ASD. We next investigated the mechanism underlying this sublinearity by performing knockdown of the regulatory β4 subunit of BK channels, which rescued the synaptic integration, a result that was corroborated with numerical simulations. Taken together, these findings suggest that there is a differential impairment in the integration of feedforward sensory and feedback predictive inputs in L5 pyramidal neurons in FXS and potentially other forms of ASD, as a result of specifically localized subcellular channelopathies. These results challenge the traditional view that FXS and other ASD are characterized by sensory hypersensitivity, proposing instead a hyposensitivity of sensory inputs and hypersensitivity of predictive inputs onto cortical neurons.
Collapse
|
163
|
Mohibbullah M, Pringgo Wicaksono MY, Hannan MA, Dash R, Nur Meinita MD, Choi JS, Hong YK, Moon IS. The Edible Seaweed Gelidium amansii Promotes Structural Plasticity of Hippocampal Neurons and Improves Scopolamine-induced Learning and Memory Impairment in Mice. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1391-1402. [PMID: 36093815 DOI: 10.2174/1871527321666220909142158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/20/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Gelidium amansii has been gaining profound interest in East Asian countries due to its enormous commercial value for agar production and its extensive pharmacological properties. Previous studies have shown that the ethanol extract of Gelidium amansii (GAE) has promising neurotrophic effects in in vitro conditions. OBJECTIVES The present study aimed at investigating the protective effects of GAE against scopolamine-induced cognitive deficits and its modulatory effects on hippocampal plasticity in mice. METHODS For memory-related behavioral studies, the passive avoidance test and radial arm maze paradigm were conducted. The brain slices of the hippocampus CA1 neurons of experimental mice were then prepared to perform Golgi staining for analyzing spine density and its characteristic shape, and immunohistochemistry for assessing the expression of different pre- and postsynaptic proteins. RESULTS Following oral administration of GAE (0.5 mg/g body weight), mice with memory deficits exhibited a significant increase in the latency time on the passive avoidance test and a decrease in the number of working and reference memory errors and latency time on the radial arm maze test. Microscopic observations of Golgi-impregnated tissue sections and immunohistochemistry of hippocampal slices showed that neurons from GAE-treated mice displayed higher spine density and spine dynamics, increased synaptic contact, and the recruitment of memory-associated proteins such as N-methyl-D-aspartate receptors (NR2A and NR2B) and postsynaptic density-95 (PSD-95) when compared with the control group. CONCLUSION With these memory-protective functions and a modulatory role in underlying memory-related events, GAE could be a potential functional food and a promising source of pharmacological agents for the prevention and treatment of memory-related brain disorders.
Collapse
Affiliation(s)
- Md Mohibbullah
- Department of Fishing and Post Harvest Technology, Sher-e-Bangla Agricultural University, Sher-e-Bangla Nagar, Dhaka-1207, Bangladesh
- Seafood Research Center, Silla University, #605, Advanced Seafood Processing Complex, Wonyang-ro, Amnam-dong, Seo-gu, Busan 49277, Korea
- Department of Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958, Korea
- Department of Biotechnology, Pukyong National University, Namku, Busan 608-737, Korea
| | | | - Md Abdul Hannan
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh-2202, Bangladesh
- Department of Anatomy, College of Korean Medicine, Dongguk University, 38066, Korea
| | - Raju Dash
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Korea
| | - Maria Dyah Nur Meinita
- Faculty of Fisheries and Marine Science, Center for Maritime Biosciences Studies, Jenderal Soedirman University, Purwokerto 53123, Indonesia
| | - Jae-Suk Choi
- Seafood Research Center, Silla University, #605, Advanced Seafood Processing Complex, Wonyang-ro, Amnam-dong, Seo-gu, Busan 49277, Korea
- Department of Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958, Korea
| | - Yong-Ki Hong
- Department of Biotechnology, Pukyong National University, Namku, Busan 608-737, Korea
| | - Il Soo Moon
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Gyeongbuk 38066, Korea
| |
Collapse
|
164
|
Rasia-Filho AA, Calcagnotto ME, von Bohlen Und Halbach O. Glial Cell Modulation of Dendritic Spine Structure and Synaptic Function. ADVANCES IN NEUROBIOLOGY 2023; 34:255-310. [PMID: 37962798 DOI: 10.1007/978-3-031-36159-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Glia comprise a heterogeneous group of cells involved in the structure and function of the central and peripheral nervous system. Glial cells are found from invertebrates to humans with morphological specializations related to the neural circuits in which they are embedded. Glial cells modulate neuronal functions, brain wiring and myelination, and information processing. For example, astrocytes send processes to the synaptic cleft, actively participate in the metabolism of neurotransmitters, and release gliotransmitters, whose multiple effects depend on the targeting cells. Human astrocytes are larger and more complex than their mice and rats counterparts. Astrocytes and microglia participate in the development and plasticity of neural circuits by modulating dendritic spines. Spines enhance neuronal connectivity, integrate most postsynaptic excitatory potentials, and balance the strength of each input. Not all central synapses are engulfed by astrocytic processes. When that relationship occurs, a different pattern for thin and large spines reflects an activity-dependent remodeling of motile astrocytic processes around presynaptic and postsynaptic elements. Microglia are equally relevant for synaptic processing, and both glial cells modulate the switch of neuroendocrine secretion and behavioral display needed for reproduction. In this chapter, we provide an overview of the structure, function, and plasticity of glial cells and relate them to synaptic maturation and modulation, also involving neurotrophic factors. Together, neurons and glia coordinate synaptic transmission in both normal and abnormal conditions. Neglected over decades, this exciting research field can unravel the complexity of species-specific neural cytoarchitecture as well as the dynamic region-specific functional interactions between diverse neurons and glial subtypes.
Collapse
Affiliation(s)
- Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
165
|
KASAI H. Unraveling the mysteries of dendritic spine dynamics: Five key principles shaping memory and cognition. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2023; 99:254-305. [PMID: 37821392 PMCID: PMC10749395 DOI: 10.2183/pjab.99.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/11/2023] [Indexed: 10/13/2023]
Abstract
Recent research extends our understanding of brain processes beyond just action potentials and chemical transmissions within neural circuits, emphasizing the mechanical forces generated by excitatory synapses on dendritic spines to modulate presynaptic function. From in vivo and in vitro studies, we outline five central principles of synaptic mechanics in brain function: P1: Stability - Underpinning the integral relationship between the structure and function of the spine synapses. P2: Extrinsic dynamics - Highlighting synapse-selective structural plasticity which plays a crucial role in Hebbian associative learning, distinct from pathway-selective long-term potentiation (LTP) and depression (LTD). P3: Neuromodulation - Analyzing the role of G-protein-coupled receptors, particularly dopamine receptors, in time-sensitive modulation of associative learning frameworks such as Pavlovian classical conditioning and Thorndike's reinforcement learning (RL). P4: Instability - Addressing the intrinsic dynamics crucial to memory management during continual learning, spotlighting their role in "spine dysgenesis" associated with mental disorders. P5: Mechanics - Exploring how synaptic mechanics influence both sides of synapses to establish structural traces of short- and long-term memory, thereby aiding the integration of mental functions. We also delve into the historical background and foresee impending challenges.
Collapse
Affiliation(s)
- Haruo KASAI
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
166
|
Tan SY, Jiang JX, Huang HX, Mo XP, Feng JR, Chen Y, Yang L, Long C. Neural mechanism underlies CYLD modulation of morphology and synaptic function of medium spiny neurons in dorsolateral striatum. Front Mol Neurosci 2023; 16:1107355. [PMID: 36846565 PMCID: PMC9945542 DOI: 10.3389/fnmol.2023.1107355] [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: 11/24/2022] [Accepted: 01/17/2023] [Indexed: 02/11/2023] Open
Abstract
Although the deubiquitinase cylindromatosis (CYLD), an abundant protein in the postsynaptic density fraction, plays a crucial role in mediating the synaptic activity of the striatum, the precise molecular mechanism remains largely unclear. Here, using a Cyld-knockout mouse model, we demonstrate that CYLD regulates dorsolateral striatum (DLS) neuronal morphology, firing activity, excitatory synaptic transmission, and plasticity of striatal medium spiny neurons via, likely, interaction with glutamate receptor 1 (GluA1) and glutamate receptor 2 (GluA2), two key subunits of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs). CYLD deficiency reduces levels of GluA1 and GluA2 surface protein and increases K63-linked ubiquitination, resulting in functional impairments both in AMPAR-mediated excitatory postsynaptic currents and in AMPAR-dependent long-term depression. The results demonstrate a functional association of CYLD with AMPAR activity, which strengthens our understanding of the role of CYLD in striatal neuronal activity.
Collapse
Affiliation(s)
- Shu-Yi Tan
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jin-Xiang Jiang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Hui-Xian Huang
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiu-Ping Mo
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Jing-Ru Feng
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Yu Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China.,South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, China
| |
Collapse
|
167
|
Heck N, Santos MD. Dendritic Spines in Learning and Memory: From First Discoveries to Current Insights. ADVANCES IN NEUROBIOLOGY 2023; 34:311-348. [PMID: 37962799 DOI: 10.1007/978-3-031-36159-3_7] [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: 11/15/2023]
Abstract
The central nervous system is composed of neural ensembles, and their activity patterns are neural correlates of cognitive functions. Those ensembles are networks of neurons connected to each other by synapses. Most neurons integrate synaptic signal through a remarkable subcellular structure called spine. Dendritic spines are protrusions whose diverse shapes make them appear as a specific neuronal compartment, and they have been the focus of studies for more than a century. Soon after their first description by Ramón y Cajal, it has been hypothesized that spine morphological changes could modify neuronal connectivity and sustain cognitive abilities. Later studies demonstrated that changes in spine density and morphology occurred in experience-dependent plasticity during development, and in clinical cases of mental retardation. This gave ground for the assumption that dendritic spines are the particular locus of cerebral plasticity. With the discovery of synaptic long-term potentiation, a research program emerged with the aim to establish whether dendritic spine plasticity could explain learning and memory. The development of live imaging methods revealed on the one hand that dendritic spine remodeling is compatible with learning process and, on the other hand, that their long-term stability is compatible with lifelong memories. Furthermore, the study of the mechanisms of spine growth and maintenance shed new light on the rules of plasticity. In behavioral paradigms of memory, spine formation or elimination and morphological changes were found to correlate with learning. In a last critical step, recent experiments have provided evidence that dendritic spines play a causal role in learning and memory.
Collapse
Affiliation(s)
- Nicolas Heck
- Laboratory Neurosciences Paris Seine, Sorbonne Université, Paris, France.
| | - Marc Dos Santos
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
168
|
Righes Marafiga J, Calcagnotto ME. Electrophysiology of Dendritic Spines: Information Processing, Dynamic Compartmentalization, and Synaptic Plasticity. ADVANCES IN NEUROBIOLOGY 2023; 34:103-141. [PMID: 37962795 DOI: 10.1007/978-3-031-36159-3_3] [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: 11/15/2023]
Abstract
For many years, synaptic transmission was considered as information transfer between presynaptic neuron and postsynaptic cell. At the synaptic level, it was thought that dendritic arbors were only receiving and integrating all information flow sent along to the soma, while axons were primarily responsible for point-to-point information transfer. However, it is important to highlight that dendritic spines play a crucial role as postsynaptic components in central nervous system (CNS) synapses, not only integrating and filtering signals to the soma but also facilitating diverse connections with axons from many different sources. The majority of excitatory connections from presynaptic axonal terminals occurs on postsynaptic spines, although a subset of GABAergic synapses also targets spine heads. Several studies have shown the vast heterogeneous morphological, biochemical, and functional features of dendritic spines related to synaptic processing. In this chapter (adding to the relevant data on the biophysics of spines described in Chap. 1 of this book), we address the up-to-date functional dendritic characteristics assessed through electrophysiological approaches, including backpropagating action potentials (bAPs) and synaptic potentials mediated in dendritic and spine compartmentalization, as well as describing the temporal and spatial dynamics of glutamate receptors in the spines related to synaptic plasticity.
Collapse
Affiliation(s)
- Joseane Righes Marafiga
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Maria Elisa Calcagnotto
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
169
|
Rathour RK, Kaphzan H. Synergies between synaptic and HCN channel plasticity dictates firing rate homeostasis and mutual information transfer in hippocampal model neuron. Front Cell Neurosci 2023; 17:1096823. [PMID: 37020846 PMCID: PMC10067771 DOI: 10.3389/fncel.2023.1096823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/02/2023] [Indexed: 04/07/2023] Open
Abstract
Homeostasis is a precondition for any physiological system of any living organism. Nonetheless, models of learning and memory that are based on processes of synaptic plasticity are unstable by nature according to Hebbian rules, and it is not fully clear how homeostasis is maintained during these processes. This is where theoretical and computational frameworks can help in gaining a deeper understanding of the various cellular processes that enable homeostasis in the face of plasticity. A previous simplistic single compartmental model with a single synapse showed that maintaining input/output response homeostasis and stable synaptic learning could be enabled by introducing a linear relationship between synaptic plasticity and HCN conductance plasticity. In this study, we aimed to examine whether this approach could be extended to a more morphologically realistic model that entails multiple synapses and gradients of various VGICs. In doing so, we found that a linear relationship between synaptic plasticity and HCN conductance plasticity was able to maintain input/output response homeostasis in our morphologically realistic model, where the slope of the linear relationship was dependent on baseline HCN conductance and synaptic permeability values. An increase in either baseline HCN conductance or synaptic permeability value led to a decrease in the slope of the linear relationship. We further show that in striking contrast to the single compartment model, here linear relationship was insufficient in maintaining stable synaptic learning despite maintaining input/output response homeostasis. Additionally, we showed that homeostasis of input/output response profiles was at the expense of decreasing the mutual information transfer due to the increase in noise entropy, which could not be fully rescued by optimizing the linear relationship between synaptic and HCN conductance plasticity. Finally, we generated a place cell model based on theta oscillations and show that synaptic plasticity disrupts place cell activity. Whereas synaptic plasticity accompanied by HCN conductance plasticity through linear relationship maintains the stability of place cell activity. Our study establishes potential differences between a single compartmental model and a morphologically realistic model.
Collapse
|
170
|
Uchigashima M, Hayashi Y, Futai K. Regulation of Presynaptic Release Machinery by Cell Adhesion Molecules. ADVANCES IN NEUROBIOLOGY 2023; 33:333-356. [PMID: 37615873 DOI: 10.1007/978-3-031-34229-5_13] [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: 08/25/2023]
Abstract
The synapse is a highly specialized asymmetric structure that transmits and stores information in the brain. The size of pre- and postsynaptic structures and function is well coordinated at the individual synapse level. For example, large postsynaptic dendritic spines have a larger postsynaptic density with higher α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) number on their surface, while juxtaposing presynaptic terminals have a larger active zone and higher release probability. This indicates that pre- and postsynaptic domains bidirectionally communicate to coordinate assembly of specific molecules on both sides of the synaptic cleft. Cell adhesion molecules (CAMs) that localize at synapses form transsynaptic protein interactions across the synaptic cleft and play important roles in synapse formation and regulation. The extracellular domain of CAMs is essential for specific synapse formation and function. In contrast, the intracellular domain is necessary for binding with synaptic molecules and signal transduction. Therefore, CAMs play an essential role on synapse function and structure. In fact, ample evidence indicates that transsynaptic CAMs instruct and modulate functions at presynaptic sites. This chapter focuses on transsynaptic protein interactions that regulate presynaptic functions emphasizing the role of neuronal CAMs and the intracellular mechanism of their regulation.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kensuke Futai
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
171
|
Renner J, Rasia-Filho AA. Morphological Features of Human Dendritic Spines. ADVANCES IN NEUROBIOLOGY 2023; 34:367-496. [PMID: 37962801 DOI: 10.1007/978-3-031-36159-3_9] [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: 11/15/2023]
Abstract
Dendritic spine features in human neurons follow the up-to-date knowledge presented in the previous chapters of this book. Human dendrites are notable for their heterogeneity in branching patterns and spatial distribution. These data relate to circuits and specialized functions. Spines enhance neuronal connectivity, modulate and integrate synaptic inputs, and provide additional plastic functions to microcircuits and large-scale networks. Spines present a continuum of shapes and sizes, whose number and distribution along the dendritic length are diverse in neurons and different areas. Indeed, human neurons vary from aspiny or "relatively aspiny" cells to neurons covered with a high density of intermingled pleomorphic spines on very long dendrites. In this chapter, we discuss the phylogenetic and ontogenetic development of human spines and describe the heterogeneous features of human spiny neurons along the spinal cord, brainstem, cerebellum, thalamus, basal ganglia, amygdala, hippocampal regions, and neocortical areas. Three-dimensional reconstructions of Golgi-impregnated dendritic spines and data from fluorescence microscopy are reviewed with ultrastructural findings to address the complex possibilities for synaptic processing and integration in humans. Pathological changes are also presented, for example, in Alzheimer's disease and schizophrenia. Basic morphological data can be linked to current techniques, and perspectives in this research field include the characterization of spines in human neurons with specific transcriptome features, molecular classification of cellular diversity, and electrophysiological identification of coexisting subpopulations of cells. These data would enlighten how cellular attributes determine neuron type-specific connectivity and brain wiring for our diverse aptitudes and behavior.
Collapse
Affiliation(s)
- Josué Renner
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
172
|
Petanjek Z, Banovac I, Sedmak D, Hladnik A. Dendritic Spines: Synaptogenesis and Synaptic Pruning for the Developmental Organization of Brain Circuits. ADVANCES IN NEUROBIOLOGY 2023; 34:143-221. [PMID: 37962796 DOI: 10.1007/978-3-031-36159-3_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Synaptic overproduction and elimination is a regular developmental event in the mammalian brain. In the cerebral cortex, synaptic overproduction is almost exclusively correlated with glutamatergic synapses located on dendritic spines. Therefore, analysis of changes in spine density on different parts of the dendritic tree in identified classes of principal neurons could provide insight into developmental reorganization of specific microcircuits.The activity-dependent stabilization and selective elimination of the initially overproduced synapses is a major mechanism for generating diversity of neural connections beyond their genetic determination. The largest number of overproduced synapses was found in the monkey and human cerebral cortex. The highest (exceeding adult values by two- to threefold) and most protracted overproduction (up to third decade of life) was described for associative layer IIIC pyramidal neurons in the human dorsolateral prefrontal cortex.Therefore, the highest proportion and extraordinarily extended phase of synaptic spine overproduction is a hallmark of neural circuitry in human higher-order associative areas. This indicates that microcircuits processing the most complex human cognitive functions have the highest level of developmental plasticity. This finding is the backbone for understanding the effect of environmental impact on the development of the most complex, human-specific cognitive and emotional capacities, and on the late onset of human-specific neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Zdravko Petanjek
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia.
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia.
| | - Ivan Banovac
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dora Sedmak
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ana Hladnik
- Department of Anatomy and Clinical Anatomy, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Neuroscience, Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Center of Excellence for Basic, Clinical and Translational Neuroscience, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
173
|
Bonansco C, Cerpa W, Inestrosa NC. How Are Synapses Born? A Functional and Molecular View of the Role of the Wnt Signaling Pathway. Int J Mol Sci 2022; 24:ijms24010708. [PMID: 36614149 PMCID: PMC9821221 DOI: 10.3390/ijms24010708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 01/03/2023] Open
Abstract
Synaptic transmission is a dynamic process that requires precise regulation. Early in life, we must be able to forge appropriate connections (add and remove) to control our behavior. Neurons must recognize appropriate targets, and external soluble factors that activate specific signaling cascades provide the regulation needed to achieve this goal. Wnt signaling has been implicated in several forms of synaptic plasticity, including functional and structural changes associated with brain development. The analysis of synapses from an electrophysiological perspective allows us to characterize the functional role of cellular signaling pathways involved in brain development. The application of quantal theory to principles of developmental plasticity offers the possibility of dissecting the function of structural changes associated with the birth of new synapses as well as the maturation of immature silent synapses. Here, we focus on electrophysiological and molecular evidence that the Wnt signaling pathway regulates glutamatergic synaptic transmission, specifically N-methyl-d-aspartate receptors (NMDARs), to control the birth of new synapses. We also focus on the role of Wnts in the conversion of silent synapses into functional synapses.
Collapse
Affiliation(s)
- Christian Bonansco
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Correspondence: (C.B.); (N.C.I.)
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Nibaldo C. Inestrosa
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6200000, Chile
- Centro de Envejecimiento y Regeneración (CARE UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Correspondence: (C.B.); (N.C.I.)
| |
Collapse
|
174
|
Machado JP, Athie MC, Matos AH, Lopes-Cendes I, Vieira A. The transcriptome of rat hippocampal subfields. IBRO Neurosci Rep 2022; 13:322-329. [PMID: 36247526 PMCID: PMC9561749 DOI: 10.1016/j.ibneur.2022.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022] Open
Abstract
The hippocampus comprises several neuronal populations such as CA1, CA2, CA3, and the dentate gyrus (DG), which present different neuronal origins, morphologies, and molecular mechanisms. Laser capture microdissection (LCM) allows selectively collecting samples from target regions and eliminating unwanted cells to obtain more specific results. LCM of hippocampus neuronal populations coupĺed with RNA-seq analysis has the potential to allow the exploration of the molecular machinery unique to each of these subfields. Previous RNA-seq investigation has already provided a molecular blueprint of the hippocampus, however, there is no RNA-seq data specific for each of the rat hippocampal regions. Serial tissue sections covering the hippocampus were produced from frozen brains of adult male Wistar rats, and the hippocampal subfields CA1, CA2, CA3, and DG were identified and isolated by LCM. We found evident segregation of the transcriptomic profile from different regions of the hippocampus and the expression of known, as well as novel, specific marker genes for each region. Gene ontology enrichment analysis of CA1 subfield indicates an enrichment of actin regulation and postsynaptic membrane AMPA receptors genes indispensable for long-term potentiation. CA2 and CA3 transcripts were found associated with the increased metabolic processes. DG expression was enriched for ribosome and spliceosome, both required for protein synthesis and maintenance of cell life. The present findings contribute to a deeper understanding of the differences in the molecular machinery expressed by the rat hippocampal neuronal populations, further exploring underlying mechanisms responsible for each subflied specific functions.
Collapse
Affiliation(s)
- João P.D. Machado
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Maria C.P. Athie
- Department of Translational Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Alexandre H.B. Matos
- Department of Translational Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - Iscia Lopes-Cendes
- Department of Translational Medicine, School of Medical Sciences. University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| | - André.S. Vieira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- Brazilian Institute of Neuroscience and Neurotechnology (BRAINN), Campinas, São Paulo, Brazil
| |
Collapse
|
175
|
FOXG1 Contributes Adult Hippocampal Neurogenesis in Mice. Int J Mol Sci 2022; 23:ijms232314979. [PMID: 36499306 PMCID: PMC9735854 DOI: 10.3390/ijms232314979] [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: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Strategies to enhance hippocampal precursor cells efficiently differentiate into neurons could be crucial for structural repair after neurodegenerative damage. FOXG1 has been shown to play an important role in pattern formation, cell proliferation, and cell specification during embryonic and early postnatal neurogenesis. Thus far, the role of FOXG1 in adult hippocampal neurogenesis is largely unknown. Utilizing CAG-loxp-stop-loxp-Foxg1-IRES-EGFP (Foxg1fl/fl), a specific mouse line combined with CreAAV infusion, we successfully forced FOXG1 overexpressed in the hippocampal dentate gyrus (DG) of the genotype mice. Thereafter, we explored the function of FOXG1 on neuronal lineage progression and hippocampal neurogenesis in adult mice. By inhibiting p21cip1 expression, FOXG1-regulated activities enable the expansion of the precursor cell population. Besides, FOXG1 induced quiescent radial-glia like type I neural progenitor, giving rise to intermediate progenitor cells, neuroblasts in the hippocampal DG. Through increasing the length of G1 phase, FOXG1 promoted lineage-committed cells to exit the cell cycle and differentiate into mature neurons. The present results suggest that FOXG1 likely promotes neuronal lineage progression and thereby contributes to adult hippocampal neurogenesis. Elevating FOXG1 levels either pharmacologically or through other means could present a therapeutic strategy for disease related with neuronal loss.
Collapse
|
176
|
Dorkenwald S, Turner NL, Macrina T, Lee K, Lu R, Wu J, Bodor AL, Bleckert AA, Brittain D, Kemnitz N, Silversmith WM, Ih D, Zung J, Zlateski A, Tartavull I, Yu SC, Popovych S, Wong W, Castro M, Jordan CS, Wilson AM, Froudarakis E, Buchanan J, Takeno MM, Torres R, Mahalingam G, Collman F, Schneider-Mizell CM, Bumbarger DJ, Li Y, Becker L, Suckow S, Reimer J, Tolias AS, Macarico da Costa N, Reid RC, Seung HS. Binary and analog variation of synapses between cortical pyramidal neurons. eLife 2022; 11:e76120. [PMID: 36382887 PMCID: PMC9704804 DOI: 10.7554/elife.76120] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 11/15/2022] [Indexed: 11/17/2022] Open
Abstract
Learning from experience depends at least in part on changes in neuronal connections. We present the largest map of connectivity to date between cortical neurons of a defined type (layer 2/3 [L2/3] pyramidal cells in mouse primary visual cortex), which was enabled by automated analysis of serial section electron microscopy images with improved handling of image defects (250 × 140 × 90 μm3 volume). We used the map to identify constraints on the learning algorithms employed by the cortex. Previous cortical studies modeled a continuum of synapse sizes by a log-normal distribution. A continuum is consistent with most neural network models of learning, in which synaptic strength is a continuously graded analog variable. Here, we show that synapse size, when restricted to synapses between L2/3 pyramidal cells, is well modeled by the sum of a binary variable and an analog variable drawn from a log-normal distribution. Two synapses sharing the same presynaptic and postsynaptic cells are known to be correlated in size. We show that the binary variables of the two synapses are highly correlated, while the analog variables are not. Binary variation could be the outcome of a Hebbian or other synaptic plasticity rule depending on activity signals that are relatively uniform across neuronal arbors, while analog variation may be dominated by other influences such as spontaneous dynamical fluctuations. We discuss the implications for the longstanding hypothesis that activity-dependent plasticity switches synapses between bistable states.
Collapse
Affiliation(s)
- Sven Dorkenwald
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Computer Science Department, Princeton UniversityPrincetonUnited States
| | - Nicholas L Turner
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Computer Science Department, Princeton UniversityPrincetonUnited States
| | - Thomas Macrina
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Computer Science Department, Princeton UniversityPrincetonUnited States
| | - Kisuk Lee
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Brain & Cognitive Sciences Department, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Ran Lu
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Jingpeng Wu
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Agnes L Bodor
- Allen Institute for Brain ScienceSeattleUnited States
| | | | | | - Nico Kemnitz
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | | | - Dodam Ih
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Jonathan Zung
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Aleksandar Zlateski
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Ignacio Tartavull
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Szi-Chieh Yu
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Sergiy Popovych
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Computer Science Department, Princeton UniversityPrincetonUnited States
| | - William Wong
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Manuel Castro
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Chris S Jordan
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Alyssa M Wilson
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
| | - Emmanouil Froudarakis
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Center for Neuroscience and Artificial Intelligence, Baylor College of MedicineHoustonUnited States
| | | | - Marc M Takeno
- Allen Institute for Brain ScienceSeattleUnited States
| | - Russel Torres
- Allen Institute for Brain ScienceSeattleUnited States
| | | | | | | | | | - Yang Li
- Allen Institute for Brain ScienceSeattleUnited States
| | - Lynne Becker
- Allen Institute for Brain ScienceSeattleUnited States
| | - Shelby Suckow
- Allen Institute for Brain ScienceSeattleUnited States
| | - Jacob Reimer
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Center for Neuroscience and Artificial Intelligence, Baylor College of MedicineHoustonUnited States
| | - Andreas S Tolias
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Center for Neuroscience and Artificial Intelligence, Baylor College of MedicineHoustonUnited States
- Department of Electrical and Computer Engineering, Rice UniversityHoustonUnited States
| | | | - R Clay Reid
- Allen Institute for Brain ScienceSeattleUnited States
| | - H Sebastian Seung
- Princeton Neuroscience Institute, Princeton UniversityPrincetonUnited States
- Computer Science Department, Princeton UniversityPrincetonUnited States
| |
Collapse
|
177
|
Qin S, Zhang Z, Zhao Y, Liu J, Qiu J, Gong Y, Fan W, Guo Y, Guo Y, Xu Z, Guo Y. The impact of acupuncture on neuroplasticity after ischemic stroke: a literature review and perspectives. Front Cell Neurosci 2022; 16:817732. [PMID: 36439200 PMCID: PMC9685811 DOI: 10.3389/fncel.2022.817732] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 10/24/2022] [Indexed: 09/07/2023] Open
Abstract
Ischemic stroke is common in the elderly, and is one of the main causes of long-term disability worldwide. After ischemic stroke, spontaneous recovery and functional reconstruction take place. These processes are possible thanks to neuroplasticity, which involves neurogenesis, synaptogenesis, and angiogenesis. However, the repair of ischemic damage is not complete, and neurological deficits develop eventually. The WHO recommends acupuncture as an alternative and complementary method for the treatment of stroke. Moreover, clinical and experimental evidence has documented the potential of acupuncture to ameliorate ischemic stroke-induced neurological deficits, particularly sequelae such as dyskinesia, spasticity, cognitive impairment, and dysphagia. These effects are related to the ability of acupuncture to promote spontaneous neuroplasticity after ischemic stroke. Specifically, acupuncture can stimulate neurogenesis, activate axonal regeneration and sprouting, and improve the structure and function of synapses. These processes modify the neural network and function of the damaged brain area, producing the improvement of various skills and adaptability. Astrocytes and microglia may be involved in the regulation of neuroplasticity by acupuncture, such as by the production and release of a variety of neurotrophic factors, including brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF). Moreover, the evidence presented indicates that acupuncture promotes neuroplasticity by modulating the functional reconstruction of the whole brain after ischemia. Therefore, the promotion of neuroplasticity is expected to become a new target for acupuncture in the treatment of neurological deficits after ischemic stroke, and research into the mechanisms responsible for these actions will be of significant clinical value.
Collapse
Affiliation(s)
- Siru Qin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zichen Zhang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yadan Zhao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyi Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiwen Qiu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yinan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wen Fan
- Department of Rehabilitation Physical Therapy Course, Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Japan
| | - Yongming Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yi Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yang Guo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Acupuncture Department, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
178
|
Vallés AS, Barrantes FJ. The synaptic lipidome in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:184033. [PMID: 35964712 DOI: 10.1016/j.bbamem.2022.184033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
Adequate homeostasis of lipid, protein and carbohydrate metabolism is essential for cells to perform highly specific tasks in our organism, and the brain, with its uniquely high energetic requirements, posesses singular characteristics. Some of these are related to its extraordinary dotation of synapses, the specialized subcelluar structures where signal transmission between neurons occurs in the central nervous system. The post-synaptic compartment of excitatory synapses, the dendritic spine, harbors key molecules involved in neurotransmission tightly packed within a minute volume of a few femtoliters. The spine is further compartmentalized into nanodomains that facilitate the execution of temporo-spatially separate functions in the synapse. Lipids play important roles in this structural and functional compartmentalization and in mechanisms that impact on synaptic transmission. This review analyzes the structural and dynamic processes involving lipids at the synapse, highlighting the importance of their homeostatic balance for the physiology of this complex and highly specialized structure, and underscoring the pathologies associated with disbalances of lipid metabolism, particularly in the perinatal and late adulthood periods of life. Although small variations of the lipid profile in the brain take place throughout the adult lifespan, the pathophysiological consequences are clinically manifested mostly during late adulthood. Disturbances in lipid homeostasis in the perinatal period leads to alterations during nervous system development, while in late adulthood they favor the occurrence of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ana Sofia Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), 8000 Bahía Blanca, Argentina.
| | - Francisco J Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AAZ, Argentina.
| |
Collapse
|
179
|
Abstract
Activation of Ca2+/calmodulin-dependent kinase II (CaMKII) plays a critical role in long-term potentiation (LTP), a long accepted cellular model for learning and memory. However, how LTP and memories survive the turnover of synaptic proteins, particularly CaMKII, remains a mystery. Here, we take advantage of the finding that constitutive Ca2+-independent CaMKII activity, acquired prior to slice preparation, provides a lasting memory trace at synapses. In slice culture, this persistent CaMKII activity, in the absence of Ca2+ stimulation, remains stable over a 2-wk period, well beyond the turnover of CaMKII protein. We propose that the nascent CaMKII protein present at 2 wk acquired its activity from preexisting active CaMKII molecules, which transferred their activity to newly synthesized CaMKII molecules and thus maintain the memory in the face of protein turnover.
Collapse
|
180
|
Schneider MA, Malhotra D, Spritzer PM, Hatchard T, Minuzzi L, Frey BN, Haefner SA, Nicholson A, McKinnon M, Syan SK, Cardoso TDA, Schwarz K, Anés M, Santos-Díaz A, Lobato MIR. Estradiol Replacement as a Potential Enhancer of Working Memory and Neuroplasticity in Hypogonadal Trans Women. Neuroendocrinology 2022; 113:489-500. [PMID: 36130584 DOI: 10.1159/000527130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/25/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The cognitive effects of cross-sex hormone therapy (CSHT) are not well understood. In cisgender individuals, sex hormone therapy can impact neurotransmitter levels and structural anatomy. Similarly, in gender-diverse persons, CSHT has been associated with neural adaptations, such as growth in brain structures resembling those observed in cisgender individuals of the same sex. Hormone-related changes in learning and memory, as seen in menopause, are associated with physiological hypogonadism or a decline in hormones, such as estradiol. The present study examined the effect of estradiol administration in humans on glutamate concentration in brain regions involved in semantic and working memory (i.e., the dorsolateral prefrontal cortex [DLPFC], the posterior hippocampus, and the pregenual anterior cingulate cortex) and its relationship with memory. METHODS Eighteen trans women (male biological sex assigned at birth) ceased CSHT for 30 days for a washout phase (t1) upon study enrollment to reach a hypogonadal state. Working and semantic memory, cognition, hormonal assays, and brain imaging were assessed. Participants resumed CSHT for 60 days for a replacement phase (t2), after which the same evaluations from t1 were repeated. RESULTS Estradiol increased among trans women after 60 days of resumed CSHT with significant improvements in semantic memory compared to the hypogonadal phase. Working memory recall was significantly and positively correlated to glutamate in the DLPFC during the reinstatement phase, although the relationship was not moderated by levels of estradiol. DISCUSSION These results may have clinical implications for the therapeutic effects of estradiol replacement, serving as a protective factor against cognitive decline and impairment for trans women post-gonadectomy.
Collapse
Affiliation(s)
- Maiko A Schneider
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Devon Malhotra
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Poli M Spritzer
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Taylor Hatchard
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Luciano Minuzzi
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Benicio N Frey
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Sasha A Haefner
- Ontario Institute for Studies in Education, University of Toronto, Toronto, Ontario, Canada
| | - Andrew Nicholson
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Margaret McKinnon
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Sabrina K Syan
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Taiane de Azevedo Cardoso
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Karine Schwarz
- Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Maurício Anés
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | | | - Maria I R Lobato
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
181
|
The Genomic Architecture of Pregnancy-Associated Plasticity in the Maternal Mouse Hippocampus. eNeuro 2022; 9:ENEURO.0117-22.2022. [PMID: 36239981 PMCID: PMC9522463 DOI: 10.1523/eneuro.0117-22.2022] [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: 03/18/2022] [Revised: 08/10/2022] [Accepted: 08/25/2022] [Indexed: 12/15/2022] Open
Abstract
Pregnancy is associated with extraordinary plasticity in the maternal brain. Studies in humans and other mammals suggest extensive structural and functional remodeling of the female brain during and after pregnancy. However, we understand remarkably little about the molecular underpinnings of this natural phenomenon. To gain insight into pregnancy-associated hippocampal plasticity, we performed single nucleus RNA sequencing (snRNA-seq) and snATAC-seq from the mouse hippocampus before, during, and after pregnancy. We identified cell type-specific transcriptional and epigenetic signatures associated with pregnancy and postpartum adaptation. In addition, we analyzed receptor-ligand interactions and transcription factor (TF) motifs that inform hippocampal cell type identity and provide evidence of pregnancy-associated adaption. In total, these data provide a unique resource of coupled transcriptional and epigenetic data across a dynamic time period in the mouse hippocampus and suggest opportunities for functional interrogation of hormone-mediated plasticity.
Collapse
|
182
|
Yasuda R, Hayashi Y, Hell JW. CaMKII: a central molecular organizer of synaptic plasticity, learning and memory. Nat Rev Neurosci 2022; 23:666-682. [PMID: 36056211 DOI: 10.1038/s41583-022-00624-2] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 12/30/2022]
Abstract
Calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) is the most abundant protein in excitatory synapses and is central to synaptic plasticity, learning and memory. It is activated by intracellular increases in calcium ion levels and triggers molecular processes necessary for synaptic plasticity. CaMKII phosphorylates numerous synaptic proteins, thereby regulating their structure and functions. This leads to molecular events crucial for synaptic plasticity, such as receptor trafficking, localization and activity; actin cytoskeletal dynamics; translation; and even transcription through synapse-nucleus shuttling. Several new tools affording increasingly greater spatiotemporal resolution have revealed the link between CaMKII activity and downstream signalling processes in dendritic spines during synaptic and behavioural plasticity. These technologies have provided insights into the function of CaMKII in learning and memory.
Collapse
Affiliation(s)
- Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Johannes W Hell
- Department of Pharmacology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
183
|
Fujii H, Kidokoro H, Kondo Y, Kawaguchi M, Horigane SI, Natsume J, Takemoto-Kimura S, Bito H. Förster resonance energy transfer-based kinase mutation phenotyping reveals an aberrant facilitation of Ca2+/calmodulin-dependent CaMKIIα activity in de novo mutations related to intellectual disability. Front Mol Neurosci 2022; 15:970031. [PMID: 36117912 PMCID: PMC9474683 DOI: 10.3389/fnmol.2022.970031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
CaMKIIα plays a fundamental role in learning and memory and is a key determinant of synaptic plasticity. Its kinase activity is regulated by the binding of Ca2+/CaM and by autophosphorylation that operates in an activity-dependent manner. Though many mutations in CAMK2A were linked to a variety of neurological disorders, the multiplicity of its functional substrates renders the systematic molecular phenotyping challenging. In this study, we report a new case of CAMK2A P212L, a recurrent mutation, in a patient with an intellectual disability. To quantify the effect of this mutation, we developed a FRET-based kinase phenotyping strategy and measured aberrance in Ca2+/CaM-dependent activation dynamics in vitro and in synaptically connected neurons. CaMKIIα P212L revealed a significantly facilitated Ca2+/CaM-dependent activation in vitro. Consistently, this mutant showed faster activation and more delayed inactivation in neurons. More prolonged kinase activation was also accompanied by a leftward shift in the CaMKIIα input frequency tuning curve. In keeping with this, molecular phenotyping of other reported CAMK2A de novo mutations linked to intellectual disability revealed aberrant facilitation of Ca2+/CaM-dependent activation of CaMKIIα in most cases. Finally, the pharmacological reversal of CAMK2A P212L phenotype in neurons was demonstrated using an FDA-approved NMDA receptor antagonist memantine, providing a basis for targeted therapeutics in CAMK2A-linked intellectual disability. Taken together, FRET-based kinase mutation phenotyping sheds light on the biological impact of CAMK2A mutations and provides a selective, sensitive, quantitative, and scalable strategy for gaining novel insights into the molecular etiology of intellectual disability.
Collapse
Affiliation(s)
- Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- *Correspondence: Hajime Fujii
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yayoi Kondo
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiro Kawaguchi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shin-ichiro Horigane
- Department of Neuroscience I, Research Institute of Environmental Medicine (RIEM), Nagoya University, Nagoya, Japan
- Department of Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun Natsume
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Developmental Disability Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sayaka Takemoto-Kimura
- Department of Neuroscience I, Research Institute of Environmental Medicine (RIEM), Nagoya University, Nagoya, Japan
- Department of Molecular/Cellular Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Haruhiko Bito
| |
Collapse
|
184
|
Li DC, Dighe NM, Barbee BR, Pitts EG, Kochoian B, Blumenthal SA, Figueroa J, Leong T, Gourley SL. A molecularly integrated amygdalo-fronto-striatal network coordinates flexible learning and memory. Nat Neurosci 2022; 25:1213-1224. [PMID: 36042313 PMCID: PMC10614133 DOI: 10.1038/s41593-022-01148-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/18/2022] [Indexed: 01/28/2023]
Abstract
Behavioral flexibility-that is, the ability to deviate from established behavioral sequences-is critical for navigating dynamic environments and requires the durable encoding and retrieval of new memories to guide future choice. The orbitofrontal cortex (OFC) supports outcome-guided behaviors. However, the coordinated neural circuitry and cellular mechanisms by which OFC connections sustain flexible learning and memory remain elusive. Here we demonstrate in mice that basolateral amygdala (BLA)→OFC projections bidirectionally control memory formation when familiar behaviors are unexpectedly not rewarded, whereas OFC→dorsomedial striatum (DMS) projections facilitate memory retrieval. OFC neuronal ensembles store a memory trace for newly learned information, which appears to be facilitated by circuit-specific dendritic spine plasticity and neurotrophin signaling within defined BLA-OFC-DMS connections and obstructed by cocaine. Thus, we describe the directional transmission of information within an integrated amygdalo-fronto-striatal circuit across time, whereby novel memories are encoded by BLA→OFC inputs, represented within OFC ensembles and retrieved via OFC→DMS outputs during future choice.
Collapse
Affiliation(s)
- Dan C Li
- Medical Scientist Training Program, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Niharika M Dighe
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Britton R Barbee
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Elizabeth G Pitts
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Brik Kochoian
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Sarah A Blumenthal
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Janet Figueroa
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Traci Leong
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Shannon L Gourley
- Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA.
- Emory National Primate Research Center, Emory University, Atlanta, GA, USA.
| |
Collapse
|
185
|
Avalos MP, Guzman AS, Garcia-Keller C, Mongi-Bragato B, Esparza MA, Rigoni D, Sanchez MA, Calfa GD, Bollati FA, Cancela LM. Impairment of glutamate homeostasis in the nucleus accumbens core underpins cross-sensitization to cocaine following chronic restraint stress. Front Physiol 2022; 13:896268. [PMID: 36091376 PMCID: PMC9462460 DOI: 10.3389/fphys.2022.896268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022] Open
Abstract
Though the facilitating influence of stress on drug abuse is well documented, the mechanisms underlying this interaction have yet to be fully elucidated. The present study explores the neurobiological mechanisms underpinning the sensitized response to the psychomotor-stimulating effects of cocaine following chronic restraint stress (CRS), emphasizing the differential contribution of both subcompartments of the nucleus accumbens (NA), the core (NAcore) and shell (NAshell), to this phenomenon. Adult male Wistar rats were restrained for 2 h/day for 7 days and, 2 weeks after the last stress exposure (day 21), all animals were randomly assigned to behavioral, biochemical or neurochemical tests. Our results demonstrated that the enduring CRS-induced increase in psychostimulant response to cocaine was paralleled by an increase of extracellular dopamine levels in the NAcore, but not the NAshell, greater than that observed in the non-stress group. Furthermore, we found that CRS induced an impairment of glutamate homeostasis in the NAcore, but not the NAshell. Its hallmarks were increased basal extracellular glutamate concentrations driven by a CRS-induced downregulation of GLT-1, blunted glutamate levels in response to cocaine and postsynaptic structural remodeling in pre-stressed animals. In addition, ceftriaxone, a known GLT-1 enhancer, prevented the CRS-induced GLT-1 downregulation, increased basal extracellular glutamate concentrations and changes in structural plasticity in the NAcore as well as behavioral cross-sensitization to cocaine, emphasizing the biological importance of GLT-1 in the comorbidity between chronic stress exposure and drug abuse. A future perspective concerning the paramount relevance of the stress-induced disruption of glutamate homeostasis as a vulnerability factor to the development of stress and substance use disorders during early life or adulthood of descendants is provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Flavia A. Bollati
- Departamento de Farmacología Otto Orsingher, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Liliana M. Cancela
- Departamento de Farmacología Otto Orsingher, Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
186
|
Changes in Dendritic Spine Morphology and Density of Granule Cells in the Olfactory Bulb of Anguilla anguilla (L., 1758): A Possible Way to Understand Orientation and Migratory Behavior. BIOLOGY 2022; 11:biology11081244. [PMID: 36009870 PMCID: PMC9405168 DOI: 10.3390/biology11081244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022]
Abstract
Simple Summary The olfactory bulb can process odour cues through granular cells (GCs) and dendritic spines, changing their synaptic plasticity properties and their morphology. The GCs’ dendritic spines density and morphology were analysed in Anguilla anguilla, considering the olfaction as a driver involved in fish orientation and migration. For the head and neck morphology, spines were classified as mushroom, long thin, stubby, and filopodia. Spines’ density decreased from juvenile migrants to no-migrant stages and increased in the adult migrants. Spines’ density was comparable between glass and silver eels as an adaptation to migration, while at non-migrating phases, spines’ density decreased. For its phylogenetic Elopomorph attribution and its complex life cycle, A. anguilla could be recommended as a model species to study the development of dendritic spines in GCs of the olfactory bulb. Considering the role of olfaction in the orientation and migration of A. anguilla, the modification of environmental stimuli (ocean alterations and climate change) could represent contributing factors that threaten this critically endangered species. Abstract Olfaction could represent a pivotal process involved in fish orientation and migration. The olfactory bulb can manage olfactive signals at the granular cell (GC) and dendritic spine levels for their synaptic plasticity properties and changing their morphology and structural stability after environmental odour cues. The GCs’ dendritic spine density and morphology were analysed across the life stages of the catadromous Anguilla anguilla. According to the head and neck morphology, spines were classified as mushroom (M), long thin (LT), stubby (S), and filopodia (F). Total spines’ density decreased from juvenile migrants to no-migrant stages, to increase again in the adult migrant stage. Mean spines’ density was comparable between glass and silver eels as an adaptation to migration. At non-migrating phases, spines’ density decreased for M and LT, while M, LT, and S density increased in silver eels. A great dendritic spine development was found in the two migratory phases, regressing in trophic phases, but that could be recreated in adults, tracing the migratory memory of the routes travelled in juvenile phases. For its phylogenetic Elopomorph attribution and its complex life cycle, A. anguilla could be recommended as a model species to study the development of dendritic spines in GCs of the olfactory bulb as an index of synaptic plasticity involved in the modulation of olfactory stimuli. If olfaction is involved in the orientation and migration of A. anguilla and if eels possess a memory, these processes could be influenced by the modification of environmental stimuli (ocean alterations and rapid climate change) contributing to threatening this critically endangered species.
Collapse
|
187
|
Voglewede MM, Zhang H. Polarity proteins: Shaping dendritic spines and memory. Dev Biol 2022; 488:68-73. [PMID: 35580729 PMCID: PMC9953585 DOI: 10.1016/j.ydbio.2022.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023]
Abstract
The morphogenesis and plasticity of dendritic spines are associated with synaptic strength, learning, and memory. Dendritic spines are highly compartmentalized structures, which makes proteins involved in cellular polarization and membrane compartmentalization likely candidates regulating their formation and maintenance. Indeed, recent studies suggest polarity proteins help form and maintain dendritic spines by compartmentalizing the spine neck and head. Here, we review emerging evidence that polarity proteins regulate dendritic spine plasticity and stability through the cytoskeleton, scaffolding molecules, and signaling molecules. We specifically analyze various polarity complexes known to contribute to different forms of cell polarization processes and examine the essential conceptual context linking these groups of polarity proteins to dendritic spine morphogenesis, plasticity, and cognitive functions.
Collapse
Affiliation(s)
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
188
|
Ostroff LE, Cain CK. Persistent up-regulation of polyribosomes at synapses during long-term memory, reconsolidation, and extinction of associative memory. LEARNING & MEMORY (COLD SPRING HARBOR, N.Y.) 2022; 29:192-202. [PMID: 35882501 PMCID: PMC9374273 DOI: 10.1101/lm.053577.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/28/2022] [Indexed: 11/25/2022]
Abstract
Local protein synthesis at synapses can provide a rapid supply of proteins to support synaptic changes during consolidation of new memories, but its role in the maintenance or updating of established memories is unknown. Consolidation requires new protein synthesis in the period immediately following learning, whereas established memories are resistant to protein synthesis inhibitors. We have previously reported that polyribosomes are up-regulated in the lateral amygdala (LA) during consolidation of aversive-cued Pavlovian conditioning. In this study, we used serial section electron microscopy reconstructions to determine whether the distribution of dendritic polyribosomes returns to baseline during the long-term memory phase. Relative to control groups, long-term memory was associated with up-regulation of polyribosomes throughout dendrites, including in dendritic spines of all sizes. Retrieval of a consolidated memory by presentation of a small number of cues induces a new, transient requirement for protein synthesis to maintain the memory, while presentation of a large number of cues results in extinction learning, forming a new memory. One hour after retrieval or extinction training, the distribution of dendritic polyribosomes was similar except in the smallest spines, which had more polyribosomes in the extinction group. Our results demonstrate that the effects of learning on dendritic polyribosomes are not restricted to the transient translation-dependent phase of memory formation. Cued Pavlovian conditioning induces persistent synapse strengthening in the LA that is not reversed by retrieval or extinction, and dendritic polyribosomes may therefore correlate generally with synapse strength as opposed to recent activity or transient translational processes.
Collapse
Affiliation(s)
- Linnaea E Ostroff
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA.,Connecticut Institute for the Brain and Cognitive Science, University of Connecticut, Storrs, Connecticut 06269, USA.,Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Christopher K Cain
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, New York 10962, USA.,Child and Adolescent Psychiatry, New York University Langone Health, New York, New York 10016, USA
| |
Collapse
|
189
|
Ka M, Kim HG, Kim WY. WDR5-HOTTIP Histone Modifying Complex Regulates Neural Migration and Dendrite Polarity of Pyramidal Neurons via Reelin Signaling. Mol Neurobiol 2022; 59:5104-5120. [PMID: 35672601 PMCID: PMC9378496 DOI: 10.1007/s12035-022-02905-4] [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: 03/11/2022] [Accepted: 05/26/2022] [Indexed: 11/26/2022]
Abstract
WD-repeat domain 5 (WDR5), a core component of histone methyltransferase complexes, is associated with Kabuki syndrome and Kleefstra syndrome that feature intellectual disability and neurodevelopmental delay. Despite its critical status in gene regulation and neurological disorders, the role of WDR5 in neural development is unknown. Here we show that WDR5 is required for normal neuronal placement and dendrite polarization in the developing cerebral cortex. WDR5 knockdown led to defects in both entry into the bipolar transition of pyramidal neurons within the intermediate zone and radial migration into cortical layers. Moreover, WDR5 deficiency disrupted apical and basal polarity of cortical dendrites. Aberrant dendritic spines and synapses accompanied the dendrite polarity phenotype. WDR5 deficiency reduced expression of reelin signaling receptors, ApoER and VdldR, which were associated with abnormal H3K4 methylation and H4 acetylation on their promoter regions. Finally, an lncRNA, HOTTIP, was found to be a partner of WDR5 to regulate dendritic polarity and reelin signaling via histone modification. Our results demonstrate a novel role for WDR5 in neuronal development and provide mechanistic insights into the neuropathology associated with histone methyltransferase dysfunction.
Collapse
Affiliation(s)
- Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, KRICT, Daejeon, 34114, Republic of Korea
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamid Bin Khalifa University, Doha, Qatar
| | - Woo-Yang Kim
- Department of Biological Sciences, Kent State University, Kent, OH, 44242, USA.
| |
Collapse
|
190
|
Sohn J, Suzuki M, Youssef M, Hatada S, Larkum ME, Kawaguchi Y, Kubota Y. Presynaptic supervision of cortical spine dynamics in motor learning. SCIENCE ADVANCES 2022; 8:eabm0531. [PMID: 35895812 PMCID: PMC9328689 DOI: 10.1126/sciadv.abm0531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
In mammalian neocortex, learning triggers the formation and turnover of new postsynaptic spines on pyramidal cell dendrites. However, the biological principles of spine reorganization during learning remain elusive because the identity of their presynaptic neuronal partners is unknown. Here, we show that two presynaptic neural circuits supervise distinct programs of spine dynamics to execute learning. We imaged spine dynamics in motor cortex during learning and performed post hoc identification of their afferent presynaptic neurons. New spines that appeared during learning formed small transient contacts with corticocortical neurons that were eliminated on skill acquisition. In contrast, persistent spines with axons from thalamic neurons were formed and enlarged. These results suggest that pyramidal cell dendrites in motor cortex use a neural circuit division of labor during skill learning, with dynamic teaching contacts from top-down intracortical axons followed by synaptic memory formation driven by thalamic axons. Dual spine supervision may govern diverse skill learning in the neocortex.
Collapse
Affiliation(s)
- Jaerin Sohn
- Division of Cerebral Circuitry, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
| | - Mototaka Suzuki
- Neurocure Center for Excellence, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Mohammed Youssef
- Division of Cerebral Circuitry, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
- Department of Animal Physiology, Faculty of Veterinary Medicine, South Valley University, Qena 83523, Egypt
| | - Sayuri Hatada
- Division of Cerebral Circuitry, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
| | - Matthew E. Larkum
- Neurocure Center for Excellence, Charité Universitätsmedizin Berlin, 10117 Berlin, Germany
- Institute of Biology, Humboldt University of Berlin, 10117 Berlin, Germany
| | - Yasuo Kawaguchi
- Division of Cerebral Circuitry, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
- Brain Science Institute, Tamagawa University, Machida, Tokyo 194-8610, Japan
| | - Yoshiyuki Kubota
- Division of Cerebral Circuitry, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences (NIPS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
- Support Unit for Electron Microscopy Techniques, Research Resources Division, RIKEN Center for Brain Science, Wako 351-0198, Japan
| |
Collapse
|
191
|
The Role of Acetylcholine on the Effects of Different Doses of Sulfite in Learning and Memory. Neurochem Res 2022; 47:3331-3343. [PMID: 35895153 DOI: 10.1007/s11064-022-03684-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 10/16/2022]
Abstract
In this study, the effects of different doses of sulfite on learning, memory, and long term potentiation as well as the relationship of these effects with acetylcholine pathways, Arc and synapsin 1 levels were investigated. Sixty male Wistar albino rats were randomly divided into three groups as control, S100, and S260. Sodiummetabisulfite (S100;100 mg/kg/day, S260;260 mg/kg/day) was given by oral administration. Behavioral changes were evaluated. After long term potentiation recordings from the perforant pathway-dentate gyrus synapses, animals were sacrificed. Acetylcholinesterase activity, choline acetyltransferase activity, acetylcholine level as well as Arc and Synapsin 1 expressions were analyzed on the hippocampi. The total distance and average velocity values in the open field and Morris water maze tests increased in the sulfite groups, while the discrimination index in the novel object recognition test decreased compared to controls. Acetylcholine levels and choline acetyltransferase activity were also increased in the sulfite groups, while acetylcholinesterase activity was decreased compared to controls. Sulfite intake attenuated long term potentiation in the hippocampus. It has been observed that the excitatory postsynaptic potential slope and population spike amplitude of the field potentials obtained in sulfite groups decreased. This impairment was accompanied by a decrease in Arc and synapsin 1 expressions. In conclusion, it has been shown that sulfite intake in adults impairs learning and memory, possibly mediated by the cholinergic pathway. It is considered that the decrement in Arc and synapsin expressions may play a role in the mechanism underlying the impairment in long term potentiation caused by toxicity.
Collapse
|
192
|
Blume T, Filser S, Sgobio C, Peters F, Neumann U, Shimshek D, Saito T, Saido TC, Brendel M, Herms J. β-secretase inhibition prevents structural spine plasticity deficits in AppNL-G-F mice. Front Aging Neurosci 2022; 14:909586. [PMID: 35936777 PMCID: PMC9354544 DOI: 10.3389/fnagi.2022.909586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
All clinical BACE1-inhibitor trials for the treatment of Alzheimer's Disease (AD) have failed due to insufficient efficacy or side effects like worsening of cognitive symptoms. However, the scientific evidence to date suggests that BACE1-inhibition could be an effective preventative measure if applied prior to the accumulation of amyloid-beta (Aβ)-peptide and resultant impairment of synaptic function. Preclinical studies have associated BACE1-inhibition-induced cognitive deficits with decreased dendritic spine density. Therefore, we investigated dose-dependent effects of BACE1-inhibition on hippocampal dendritic spine dynamics in an APP knock-in mouse line for the first time. We conducted in vivo two-photon microscopy in the stratum oriens layer of hippocampal CA1 neurons in 3.5-month-old AppNL-G-FGFP-M mice over 6 weeks to monitor the effect of potential preventive treatment with a high and low dose of the BACE1-inhibitor NB-360 on dendritic spine dynamics. Structural spine plasticity was severely impaired in untreated AppNL-G-FGFP-M mice, although spines were not yet showing signs of degeneration. Prolonged high-dose BACE1-inhibition significantly enhanced spine formation, improving spine dynamics in the AD mouse model. We conclude that in an early AD stage characterized by low Aβ-accumulation and no irreversible spine loss, BACE1-inhibition could hold the progressive synapse loss and cognitive decline by improving structural spine dynamics.
Collapse
Affiliation(s)
- Tanja Blume
- German Center for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Severin Filser
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Carmelo Sgobio
- German Center for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
| | | | - Ulf Neumann
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Derya Shimshek
- Department of Neuroscience, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Matthias Brendel
- Munich Cluster for Systems Neurology, Munich, Germany
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases, Munich, Germany
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology, Munich, Germany
- *Correspondence: Jochen Herms
| |
Collapse
|
193
|
Argunşah AÖ, Erdil E, Ghani MU, Ramiro-Cortés Y, Hobbiss AF, Karayannis T, Çetin M, Israely I, Ünay D. An interactive time series image analysis software for dendritic spines. Sci Rep 2022; 12:12405. [PMID: 35859092 PMCID: PMC9300710 DOI: 10.1038/s41598-022-16137-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/05/2022] [Indexed: 11/09/2022] Open
Abstract
Live fluorescence imaging has demonstrated the dynamic nature of dendritic spines, with changes in shape occurring both during development and in response to activity. The structure of a dendritic spine correlates with its functional efficacy. Learning and memory studies have shown that a great deal of the information stored by a neuron is contained in the synapses. High precision tracking of synaptic structures can give hints about the dynamic nature of memory and help us understand how memories evolve both in biological and artificial neural networks. Experiments that aim to investigate the dynamics behind the structural changes of dendritic spines require the collection and analysis of large time-series datasets. In this paper, we present an open-source software called SpineS for automatic longitudinal structural analysis of dendritic spines with additional features for manual intervention to ensure optimal analysis. We have tested the algorithm on in-vitro, in-vivo, and simulated datasets to demonstrate its performance in a wide range of possible experimental scenarios.
Collapse
Affiliation(s)
- Ali Özgür Argunşah
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, 1400-038, Portugal. .,Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zürich, Zürich, Switzerland. .,UZH/ETH Zürich, Neuroscience Center Zurich (ZNZ), Zürich, Switzerland.
| | - Ertunç Erdil
- ETH Zürich, Computer Vision Laboratory, Zürich, Switzerland
| | - Muhammad Usman Ghani
- Department of Electrical and Computer Engineering, Boston University, Boston, 02215, MA, USA
| | - Yazmín Ramiro-Cortés
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, 1400-038, Portugal.,Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, C.P. 04510, Mexico
| | - Anna F Hobbiss
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, 1400-038, Portugal
| | - Theofanis Karayannis
- Laboratory of Neural Circuit Assembly, Brain Research Institute (HiFo), University of Zürich, Zürich, Switzerland.,UZH/ETH Zürich, Neuroscience Center Zurich (ZNZ), Zürich, Switzerland
| | - Müjdat Çetin
- Department of Electrical and Computer Engineering, Goergen Institute for Data Science, University of Rochester, Rochester, 14627, NY, USA
| | - Inbal Israely
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, 1400-038, Portugal.,Department of Pathology and Cell Biology, Columbia University, New York, 10032, NY, USA
| | - Devrim Ünay
- Department of Biomedical Engineering, İzmir University of Economics, İzmir, Turkey. .,Department of Electrical and Electronics Engineering, İzmir Democracy University, İzmir, Turkey.
| |
Collapse
|
194
|
Feng Z, Lee S, Jia B, Jian T, Kim E, Zhang M. IRSp53 promotes postsynaptic density formation and actin filament bundling. J Cell Biol 2022; 221:213346. [PMID: 35819332 PMCID: PMC9280192 DOI: 10.1083/jcb.202105035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/04/2021] [Accepted: 06/13/2022] [Indexed: 01/14/2023] Open
Abstract
IRSp53 (aka BAIAP2) is a scaffold protein that couples membranes with the cytoskeleton in actin-filled protrusions such as filopodia and lamellipodia. The protein is abundantly expressed in excitatory synapses and is essential for synapse development and synaptic plasticity, although with poorly understood mechanisms. Here we show that specific multivalent interactions between IRSp53 and its binding partners PSD-95 or Shank3 drive phase separation of the complexes in solution. IRSp53 can be enriched to the reconstituted excitatory PSD (ePSD) condensates via bridging to the core and deeper layers of ePSD. Overexpression of a mutant defective in the IRSp53/PSD-95 interaction perturbs synaptic enrichment of IRSp53 in mouse cortical neurons. The reconstituted PSD condensates promote bundled actin filament formation both in solution and on membranes, via IRSp53-mediated actin binding and bundling. Overexpression of mutants that perturb IRSp53-actin interaction leads to defects in synaptic maturation of cortical neurons. Together, our studies provide potential mechanistic insights into the physiological roles of IRSp53 in synapse formation and function.
Collapse
Affiliation(s)
- Zhe Feng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China,State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Suho Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Bowen Jia
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Tao Jian
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China
| | - Eunjoon Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea,Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea,Correspondence to Eunjoon Kim:
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Hong Kong, China,School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
195
|
Yang L, Hao JR, Gao Y, Yang X, Shen XR, Wang HY, Sun N, Gao C. HDAC3 of dorsal hippocampus induces postoperative cognitive dysfunction in aged mice. Behav Brain Res 2022; 433:114002. [PMID: 35810999 DOI: 10.1016/j.bbr.2022.114002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/16/2022] [Accepted: 07/05/2022] [Indexed: 12/18/2022]
Abstract
Postoperative cognitive dysfunction (POCD) affects a substantial number of aged individuals. Although advanced age has been regarded as the only independent risk factor for cognitive decline following anesthesia and surgery, the exact cellular and molecular mechanisms remain poorly understood. Histone deacetylase 3 (HDAC3), an epigenetic regulator of memory plays an important role in age-dependent disease. In this study, we investigated the role of HDAC3 in POCD using a laparotomy mouse model. The results showed that the level of HDAC3 in the dorsal hippocampus (DH) was elevated in aged mice compared with young mice. The surgery impaired the spatial-temporal memory in aged mice, as indicated in the object location memory (OLM) and temporal order memory (TOM) tests. Model mice also exhibited increased expression of HDAC3 protein and decreased levels of dendritic spine density and synaptic plasticity-related proteins in the DH. Selectively blocking HDAC3 in the DH of aged mice reversed spatial-temporal memory impairment induced by surgery and restored dendritic spine density and synaptic plasticity-related proteins in the DH. Overexpression of HDAC3 by adeno-associated virus in the DH of young mice mimicked the behavioral deficits induced by anesthesia and surgery. Our results indicated that HDAC3 negatively regulates spatial-temporal memory in aged mice after anesthesia and surgery. Targeting HDAC3 might represent a potential therapy to avoid POCD.
Collapse
Affiliation(s)
- Li Yang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China; The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.
| | - Jing-Ru Hao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China.
| | - Yin Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China; The Affiliated Nanjing Drum Tower Hospital of Xuzhou Medical University, Nanjing, Jiangsu 210008, China.
| | - Xiu Yang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China.
| | - Xiao-Ran Shen
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China.
| | - Hu-Yi Wang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China.
| | - Nan Sun
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China.
| | - Can Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004 Xuzhou, Jiangsu, China.
| |
Collapse
|
196
|
Ueda Y, Sugimoto N, Ozawa T. Increased spine PIP3 is sequestered from dendritic shafts. Mol Brain 2022; 15:59. [PMID: 35787719 PMCID: PMC9254409 DOI: 10.1186/s13041-022-00944-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/26/2022] [Indexed: 11/30/2022] Open
Abstract
Phosphatidylinositol 3,4,5-trisphosphate (PIP3) is a lipid second messenger that is crucial for the synaptic plasticity underlying learning and memory in pyramidal neurons in the brain. Our previous study uncovered PIP3 enrichment in the dendritic spines of hippocampal pyramidal neurons in the static state using a fluorescence lifetime-based PIP3 probe. However, the extent to which PIP3 enrichment is preserved in different states has not been fully investigated. Here, we revealed that PIP3 accumulation in dendritic spines is strictly controlled even in an active state in which PIP3 is increased by glutamate stimulation and high potassium-induced membrane depolarization. Time-course PIP3 analysis clarified the gradual PIP3 accumulation in dendritic spines over days during neuronal development. Collectively, these results deepen our understanding of PIP3 dynamics in dendritic spines, and the dysregulation of the PIP3 gradient between dendritic spines and shafts could cause neuronal diseases and mental disorders, such as autism spectrum disorder.
Collapse
Affiliation(s)
- Yoshibumi Ueda
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan.
| | - Naotoshi Sugimoto
- Department of Physiology, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
197
|
Identification of sulfonamide-based butyrylcholinesterase inhibitors using machine learning. Future Med Chem 2022; 14:1049-1070. [PMID: 35707942 DOI: 10.4155/fmc-2021-0325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: This study reports the designing of BChE inhibitors through machine learning (ML), followed by in silico and in vitro evaluations. Methodology: ML technique was used to predict the virtual hit, and its derivatives were synthesized and characterized. The compounds were evaluated by using various in vitro tests and in silico methods. Results: The gradient boosting classifier predicted N-phenyl-4-(phenylsulfonamido) benzamide as an active BChE inhibitor. The derivatives of the inhibitor, i.e., compounds 34, 37 and 54 were potent BChE inhibitors and displayed blood-brain barrier permeability with no significant AChE inhibition. Conclusion: The ML prediction was effective, and the synthesized compounds showed the BChE inhibitory activity, which was also supported by the in silico studies.
Collapse
|
198
|
3dSpAn: An interactive software for 3D segmentation and analysis of dendritic spines. Neuroinformatics 2022; 20:679-698. [PMID: 34743262 DOI: 10.1007/s12021-021-09549-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2021] [Indexed: 12/31/2022]
Abstract
Three-dimensional segmentation and analysis of dendritic spine morphology involve two major challenges: 1) how to segment individual spines from the dendrites and 2) how to quantitatively assess the morphology of individual spines. To address these two issues, we developed software called 3dSpAn (3-dimensional Spine Analysis), based on implementing a previously published method, 3D multi-scale opening algorithm in shared intensity space. 3dSpAn consists of four modules: a) Preprocessing and Region of Interest (ROI) selection, b) Intensity thresholding and seed selection, c) Multi-scale segmentation, and d) Quantitative morphological feature extraction. In this article, we present the results of segmentation and morphological analysis for different observation methods and conditions, including in vitro and ex vivo imaging with confocal microscopy, and in vivo observations using high-resolution two-photon microscopy. In particular, we focus on software usage, the influence of adjustable parameters on the obtained results, user reproducibility, accuracy analysis, and also include a qualitative comparison with a commercial benchmark. 3dSpAn software is freely available for non-commercial use at www.3dSpAn.org .
Collapse
|
199
|
Contreras D, Piña R, Carvallo C, Godoy F, Ugarte G, Zeise M, Rozas C, Morales B. Methylphenidate Restores Behavioral and Neuroplasticity Impairments in the Prenatal Nicotine Exposure Mouse Model of ADHD: Evidence for Involvement of AMPA Receptor Subunit Composition and Synaptic Spine Morphology in the Hippocampus. Int J Mol Sci 2022; 23:ijms23137099. [PMID: 35806103 PMCID: PMC9266648 DOI: 10.3390/ijms23137099] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
In ADHD treatment, methylphenidate (MPH) is the most frequently used medication. The present work provides evidence that MPH restored behavioral impairments and neuroplasticity due to changes in AMPAR subunit composition and distribution, as well as maturation of dendritic spines, in a prenatal nicotine exposure (PNE) ADHD mouse model. PNE animals and controls were given a single oral dose of MPH (1 mg/kg), and their behavior was tested for attention, hyperactivity, and working memory. Long-term potentiation (LTP) was induced and analyzed at the CA3/CA1 synapse in hippocampal slices taken from the same animals tested behaviorally, measuring fEPSPs and whole-cell patch-clamp EPSCs. By applying crosslinking and Western blots, we estimated the LTP effects on AMPAR subunit composition and distribution. The density and types of dendritic spines were quantified by using the Golgi staining method. MPH completely restored the behavioral impairments of PNE mice. Reduced LTP and AMPA-receptor-mediated EPSCs were also restored. EPSC amplitudes were tightly correlated with numbers of GluA1/GluA1 AMPA receptors at the cell surface. Finally, we found a lower density of dendritic spines in hippocampal pyramidal neurons in PNE mice, with a higher fraction of thin-type immature spines and a lower fraction of mushroom mature spines; the latter effect was also reversed by MPH.
Collapse
Affiliation(s)
- Darwin Contreras
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Ricardo Piña
- Departamento de Biología, Facultad de Ciencias Básicas, Universidad Metropolitana de Ciencias de la Educación, Santiago 7760197, Chile;
- Departamento de Ciencias Pedagógicas, Facultad de Educación, Universidad Bernardo O’Higgins, Santiago 8370993, Chile
| | - Claudia Carvallo
- Centro de investigación e innovación en Gerontología Aplicada (CIGAP), Facultad de Salud, Universidad Santo Tomás, Santiago 8370003, Chile;
| | - Felipe Godoy
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Gonzalo Ugarte
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
| | - Marc Zeise
- School of Psychology, Faculty of Humanities, University of Santiago de Chile, Santiago 9170022, Chile;
| | - Carlos Rozas
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
- Correspondence: (C.R.); (B.M.)
| | - Bernardo Morales
- Laboratory of Neuroscience, Faculty of Chemistry and Biology, University of Santiago de Chile, Alameda 3363, Santiago 9170022, Chile; (D.C.); (F.G.); (G.U.)
- Correspondence: (C.R.); (B.M.)
| |
Collapse
|
200
|
Mohanan AG, Gunasekaran S, Jacob RS, Omkumar RV. Role of Ca2+/Calmodulin-Dependent Protein Kinase Type II in Mediating Function and Dysfunction at Glutamatergic Synapses. Front Mol Neurosci 2022; 15:855752. [PMID: 35795689 PMCID: PMC9252440 DOI: 10.3389/fnmol.2022.855752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/21/2022] [Indexed: 01/25/2023] Open
Abstract
Glutamatergic synapses harbor abundant amounts of the multifunctional Ca2+/calmodulin-dependent protein kinase type II (CaMKII). Both in the postsynaptic density as well as in the cytosolic compartment of postsynaptic terminals, CaMKII plays major roles. In addition to its Ca2+-stimulated kinase activity, it can also bind to a variety of membrane proteins at the synapse and thus exert spatially restricted activity. The abundance of CaMKII in glutamatergic synapse is akin to scaffolding proteins although its prominent function still appears to be that of a kinase. The multimeric structure of CaMKII also confers several functional capabilities on the enzyme. The versatility of the enzyme has prompted hypotheses proposing several roles for the enzyme such as Ca2+ signal transduction, memory molecule function and scaffolding. The article will review the multiple roles played by CaMKII in glutamatergic synapses and how they are affected in disease conditions.
Collapse
Affiliation(s)
- Archana G. Mohanan
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sowmya Gunasekaran
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - Reena Sarah Jacob
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Research Scholar, Manipal Academy of Higher Education, Manipal, India
| | - R. V. Omkumar
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- *Correspondence: R. V. Omkumar,
| |
Collapse
|