1
|
Wardaszka-Pianka P, Kuzniewska B, GumiNska N, Hojka-Osinska A, Puchalska M, Milek J, Stawikowska A, Krawczyk P, Pauzin FP, Wojtowicz T, Radwanska K, Bramham CR, Dziembowski A, Dziembowska M. Terminal nucleotidyltransferase Tent2 microRNA A-tailing enzyme regulates excitatory/inhibitory balance in the hippocampus. RNA (NEW YORK, N.Y.) 2025; 31:756-771. [PMID: 40101932 DOI: 10.1261/rna.080240.124] [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: 08/22/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
One of the posttranscriptional mechanisms regulating the stability of RNA molecules involves the addition of nontemplated nucleotides to their 3' ends, a process known as RNA tailing. To systematically investigate the physiological consequences of terminal nucleotidyltransferase TENT2 absence on RNA 3' end modifications in the mouse hippocampus, we developed a new Tent2 knockout mouse. Electrophysiological measurements revealed increased excitability in Tent2 KO hippocampal neurons, and behavioral analyses showed decreased anxiety and improved fear extinction in these mice. At the molecular level, we observed changes in miRNAs' monoadenylation in Tent2 KO mouse hippocampus, but found no effect of the TENT2 loss on the mRNAs' total poly(A) tail length, as measured by direct nanopore RNA sequencing. Moreover, differential expression analysis revealed transcripts related to synaptic transmission to be downregulated in the hippocampus of Tent2 knockout mice. These changes may explain the observed behavioral and electrophysiological alterations. Our data thus establish a link between TENT2-dependent miRNA tailing and the balance of inhibitory and excitatory neurotransmission.
Collapse
Affiliation(s)
| | - Bozena Kuzniewska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Natalia GumiNska
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Anna Hojka-Osinska
- Bioinformatics Facility, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Monika Puchalska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Jacek Milek
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Aleksandra Stawikowska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| | - Pawel Krawczyk
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Francois P Pauzin
- Department of Biomedicine, University of Bergen, 5007 Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, 5007 Bergen, Norway
| | - Tomasz Wojtowicz
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, 5007 Bergen, Norway
- Mohn Research Center for the Brain, University of Bergen, 5007 Bergen, Norway
| | - Andrzej Dziembowski
- Laboratory of RNA Biology, International Institute of Molecular and Cell Biology, 02-109 Warsaw, Poland
| | - Magdalena Dziembowska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland
| |
Collapse
|
2
|
Hwang EK, Wunsch AM, Wolf ME. Retinoic acid-mediated homeostatic plasticity drives cell type-specific CP-AMPAR accumulation in nucleus accumbens core and incubation of cocaine craving. Mol Psychiatry 2025:10.1038/s41380-025-03026-9. [PMID: 40316677 DOI: 10.1038/s41380-025-03026-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 03/06/2025] [Accepted: 04/08/2025] [Indexed: 05/04/2025]
Abstract
Incubation of cocaine craving, a translationally relevant model for the persistence of drug craving during abstinence, ultimately depends on strengthening of nucleus accumbens core (NAcc) synapses through synaptic insertion of homomeric GluA1 Ca2+-permeable AMPA receptors (CP-AMPARs). Here we tested the hypothesis that CP-AMPAR upregulation results from a form of homeostatic plasticity, previously characterized in vitro and in other brain regions, that depends on retinoic acid (RA) signaling in dendrites. Under normal conditions, ongoing synaptic transmission maintains intracellular Ca2+ at levels sufficient to suppress RA synthesis. Prolonged blockade of neuronal activity results in disinhibition of RA synthesis, leading to increased GluA1 translation and synaptic insertion of homomeric GluA1 CP-AMPARs. Using slice recordings, we found that increasing RA signaling in NAcc medium spiny neurons (MSN) from drug-naïve rats rapidly upregulates CP-AMPARs. This is observed only in MSN expressing the D1 dopamine receptor. In MSN recorded from rats that have undergone incubation of craving, we observe CP-AMPAR upregulation in D1 MSN (but not D2 MSN) and the effect of exogenous RA application is occluded in these D1 MSN. Instead, interruption of RA signaling in the slice normalizes the incubation-associated elevation of synaptic CP-AMPARs. Paralleling this in vitro finding, interruption of RA signaling in the NAcc of 'incubated rats' normalizes elevated cue-induced cocaine seeking back to non-incubated levels. These results suggest that RA signaling becomes tonically active in the NAcc during cocaine withdrawal and, by maintaining elevated CP-AMPAR levels, contributes to the incubation of cocaine craving.
Collapse
Affiliation(s)
- Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- National Center for Wellness and Recovery, Oklahoma State University Center for Health Sciences, Tulsa, OK, 74107, USA
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA.
| |
Collapse
|
3
|
Wong RK, Spencer GE. Memory formation following appetitive conditioning is variably dependent on retinoid signaling. Neurobiol Learn Mem 2025; 219:108048. [PMID: 40180148 DOI: 10.1016/j.nlm.2025.108048] [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: 11/28/2024] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025]
Abstract
Retinoic acid (RA), the metabolite of Vitamin A, plays an important role in central nervous system development and regeneration, as well as learning and memory in vertebrates. We have previously shown that RA signaling is also important for consolidation of long-term memory (LTM) in the invertebrate mollusc, Lymnaea stagnalis, following operant conditioning of the aerial respiratory behaviour. Here, we examine whether retinoids also play a role in classical reward conditioning in this mollusc. A single-trial appetitive conditioning paradigm was used, with amyl acetate as the conditioned stimulus (CS) and sucrose as the unconditioned stimulus (US). This produced an acquired conditioned response whereby the animal exhibited a feeding response to amyl acetate. A single-pairing of CS with US produced long-term memory at both 1d and 6d after training. Pharmacological treatments that disrupt RA signaling did not block the formation of long term memory when a 6-day food deprivation period was implemented before training. However, two different paradigms induced susceptibility of the conditioned response (memory) to retinoid signaling inhibitors. The first paradigm change involved using a shorter, 3-day food deprivation period in order to reduce motivational drive to feed, whereas the second paradigm manipulation reduced the strength of the unconditioned stimulus (sucrose). These findings suggest different susceptibility of memories to retinoid inhibition, depending on shifts in both external parameters of the experiment, as well as internal motivational states of the animal.
Collapse
Affiliation(s)
- Raymond K Wong
- Centre for Neuroscience, Brock University, St. Catharines, ON L2S 3A1, Canada; Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Gaynor E Spencer
- Centre for Neuroscience, Brock University, St. Catharines, ON L2S 3A1, Canada; Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada.
| |
Collapse
|
4
|
Dunham TL, Wilkerson JR, Johnson RC, Huganir RL, Volk LJ. WWC2 modulates GABA A-receptor-mediated synaptic transmission, revealing class-specific mechanisms of synapse regulation by WWC family proteins. Cell Rep 2024; 43:114841. [PMID: 39388350 PMCID: PMC11913214 DOI: 10.1016/j.celrep.2024.114841] [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: 08/03/2023] [Revised: 07/22/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
The WW and C2 domain-containing protein (WWC2) is implicated in several neurological disorders. Here, we demonstrate that WWC2 interacts with inhibitory, but not excitatory, postsynaptic scaffolds, consistent with prior proteomic identification of WWC2 as a putative component of the inhibitory postsynaptic density. Using mice lacking WWC2 expression in excitatory forebrain neurons, we show that WWC2 suppresses γ-aminobutyric acid type-A receptor (GABAAR) incorporation into the plasma membrane and regulates HAP1 and GRIP1, which form a complex promoting GABAAR recycling to the membrane. Inhibitory synaptic transmission is increased in CA1 pyramidal cells lacking WWC2. Furthermore, unlike the WWC2 homolog KIBRA (kidney/brain protein; WWC1), a key regulator of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking at excitatory synapses, the deletion of WWC2 does not affect synaptic AMPAR expression. In contrast, loss of KIBRA does not affect GABAAR membrane expression. These data reveal synapse class-selective functions for WWC proteins as regulators of ionotropic neurotransmitter receptors and provide insight into mechanisms regulating GABAAR membrane expression.
Collapse
Affiliation(s)
- Thomas L Dunham
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Julia R Wilkerson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lenora J Volk
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Psychiatry UT Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
5
|
Wunsch AM, Hwang EK, Funke JR, Baker R, Moutier A, Milovanovic M, Green TA, Wolf ME. Retinoic acid-mediated homeostatic plasticity in the nucleus accumbens core contributes to incubation of cocaine craving. Psychopharmacology (Berl) 2024; 241:1983-2001. [PMID: 38935096 DOI: 10.1007/s00213-024-06612-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/10/2024] [Indexed: 06/28/2024]
Abstract
RATIONALE Incubation of cocaine craving refers to the progressive intensification of cue-induced craving during abstinence from cocaine self-administration. We showed previously that homomeric GluA1 Ca2+-permeable AMPARs (CP-AMPAR) accumulate in excitatory synapses of nucleus accumbens core (NAcc) medium spiny neurons (MSN) after ∼1 month of abstinence and thereafter their activation is required for expression of incubation. Therefore, it is important to understand mechanisms underlying CP-AMPAR plasticity. OBJECTIVES We hypothesize that CP-AMPAR upregulation represents a retinoic acid (RA)-dependent form of homeostatic plasticity, previously described in other brain regions, in which a reduction in neuronal activity disinhibits RA synthesis, leading to GluA1 translation and CP-AMPAR synaptic insertion. We tested this using viral vectors to bidirectionally manipulate RA signaling in NAcc during abstinence following extended-access cocaine self-administration. RESULTS We used shRNA targeted to the RA degradative enzyme Cyp26b1 to increase RA signaling. This treatment accelerated incubation; rats expressed incubation on abstinence day (AD) 15, when it is not yet detected in control rats. It also accelerated CP-AMPAR synaptic insertion measured with slice physiology. CP-AMPARs were detected in Cyp26b1 shRNA-expressing MSN, but not control MSN, on AD15-18. Next, we used shRNA targeted to the major RA synthetic enzyme Aldh1a1 to reduce RA signaling. In MSN expressing Aldh1a1 shRNA, synaptic CP-AMPARs were reduced in late withdrawal (AD42-60) compared to controls. However, we did not detect an effect of this manipulation on incubated cocaine seeking (AD40). CONCLUSIONS These findings support the hypothesis that increased RA signaling during abstinence contributes to CP-AMPAR accumulation and incubation of cocaine craving.
Collapse
Affiliation(s)
- Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
| | - Jonathan R Funke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA, 02111, USA
| | - Raines Baker
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Alana Moutier
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA
- Yecuris Corporation, Tualatin, OR, 97062, USA
| | - Mike Milovanovic
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97212, USA.
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA.
| |
Collapse
|
6
|
Hwang EK, Wunsch AM, Wolf ME. Retinoic acid-mediated homeostatic plasticity drives cell type-specific CP-AMPAR accumulation in nucleus accumbens core and incubation of cocaine craving. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.611703. [PMID: 39314388 PMCID: PMC11419102 DOI: 10.1101/2024.09.12.611703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Incubation of cocaine craving, a translationally relevant model for the persistence of drug craving during abstinence, ultimately depends on strengthening of nucleus accumbens core (NAcc) synapses through synaptic insertion of homomeric GluA1 Ca2+-permeable AMPA receptors (CP-AMPARs). Here we tested the hypothesis that CP-AMPAR upregulation results from a form of homeostatic plasticity, previously characterized in vitro and in other brain regions, that depends on retinoic acid (RA) signaling in dendrites. Under normal conditions, ongoing synaptic transmission maintains intracellular Ca2+ at levels sufficient to suppress RA synthesis. Prolonged blockade of neuronal activity results in disinhibition of RA synthesis, leading to increased GluA1 translation and synaptic insertion of homomeric GluA1 CP-AMPARs. Using slice recordings, we found that increasing RA signaling in NAcc medium spiny neurons (MSN) from drug-naïve rats rapidly upregulates CP-AMPARs, and that this pathway is operative only in MSN expressing the D1 dopamine receptor. In MSN recorded from rats that have undergone incubation of craving, this effect of RA is occluded; instead, interruption of RA signaling in the slice normalizes the incubation-associated elevation of synaptic CP-AMPARs. Paralleling this in vitro finding, interruption of RA signaling in the NAcc of 'incubated rats' normalizes the incubation-associated elevation of cue-induced cocaine seeking. These results suggest that RA signaling becomes tonically active in the NAcc during cocaine withdrawal and, by maintaining elevated CP-AMPAR levels, contributes to the incubation of cocaine craving.
Collapse
Affiliation(s)
- Eun-Kyung Hwang
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| | - Amanda M Wunsch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| | - Marina E Wolf
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, U.S.A. 97212
| |
Collapse
|
7
|
Carrazana R, Espinoza F, Ávila A. Mechanistic perspective on the actions of vitamin a in autism spectrum disorder etiology. Neuroscience 2024; 554:72-82. [PMID: 39002756 DOI: 10.1016/j.neuroscience.2024.07.012] [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: 09/27/2023] [Revised: 05/07/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Vitamin A (VA) has many functions in the body, some of which are key for the development and functioning of the nervous system, while some others might indirectly influence neural function. Both hypovitaminosis and hypervitaminosis A can lead to clinical manifestations of concern for individuals and for general global health. Scientific evidence on the link between VA and autism spectrum disorder (ASD) is growing, with some clinical studies and accumulating results obtained from basic research using cellular and animal models. Remarkably, it has been shown that VA deficiency can exacerbate autistic symptomatology. In turn, VA supplementation has been shown to be able to improve autistic symptomatology in selected groups of individuals with ASD. However, it is important to recognize that ASD is a highly heterogeneous condition. Therefore, it is important to clarify how and when VA supplementation can be of benefit for affected individuals. Here we delve into the relationship between VA and ASD, discussing clinical observations and mechanistic insights obtained from research on selected autistic syndromes and laboratory models to advance in defining how the VA signaling pathway can be exploited for treatment of ASD.
Collapse
Affiliation(s)
- Ramón Carrazana
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisca Espinoza
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Ariel Ávila
- Neurodevelopmental Biology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile.
| |
Collapse
|
8
|
Dunham TL, Wilkerson JR, Johnson RC, Huganir RL, Volk LJ. Modulation of GABA A receptor trafficking by WWC2 reveals class-specific mechanisms of synapse regulation by WWC family proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584487. [PMID: 38559047 PMCID: PMC10979870 DOI: 10.1101/2024.03.11.584487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
WWC2 (WW and C2 domain-containing protein) is implicated in several neurological disorders, however its function in the brain has yet to be determined. Here, we demonstrate that WWC2 interacts with inhibitory but not excitatory postsynaptic scaffolds, consistent with prior proteomic identification of WWC2 as a putative component of the inhibitory postsynaptic density. Using mice lacking WWC2 expression in excitatory forebrain neurons, we show that WWC2 suppresses GABA A R incorporation into the plasma membrane and regulates HAP1 and GRIP1, which form a complex promoting GABA A R recycling to the membrane. Inhibitory synaptic transmission is dysregulated in CA1 pyramidal cells lacking WWC2. Furthermore, unlike the WWC2 homolog KIBRA (WWC1), a key regulator of AMPA receptor trafficking at excitatory synapses, deletion of WWC2 does not affect synaptic AMPAR expression. In contrast, loss of KIBRA does not affect GABA A R membrane expression. These data reveal unique, synapse class-selective functions for WWC proteins as regulators of ionotropic neurotransmitter receptors and provide insight into mechanisms regulating GABA A R membrane expression.
Collapse
|
9
|
Piazza A, Carlone R, Spencer GE. Non-canonical retinoid signaling in neural development, regeneration and synaptic function. Front Mol Neurosci 2024; 17:1371135. [PMID: 38516042 PMCID: PMC10954794 DOI: 10.3389/fnmol.2024.1371135] [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: 01/15/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Canonical retinoid signaling via nuclear receptors and gene regulation is critical for the initiation of developmental processes such as cellular differentiation, patterning and neurite outgrowth, but also mediates nerve regeneration and synaptic functions in adult nervous systems. In addition to canonical transcriptional regulation, retinoids also exert rapid effects, and there are now multiple lines of evidence supporting non-canonical retinoid actions outside of the nucleus, including in dendrites and axons. Together, canonical and non-canonical retinoid signaling provide the precise temporal and spatial control necessary to achieve the fine cellular coordination required for proper nervous system function. Here, we examine and discuss the evidence supporting non-canonical actions of retinoids in neural development and regeneration as well as synaptic function, including a review of the proposed molecular mechanisms involved.
Collapse
Affiliation(s)
| | | | - Gaynor E. Spencer
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
10
|
Boerma T, Ter Haar S, Ganga R, Wijnen F, Blom E, Wierenga CJ. What risk factors for Developmental Language Disorder can tell us about the neurobiological mechanisms of language development. Neurosci Biobehav Rev 2023; 154:105398. [PMID: 37741516 DOI: 10.1016/j.neubiorev.2023.105398] [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: 04/21/2023] [Revised: 07/03/2023] [Accepted: 09/17/2023] [Indexed: 09/25/2023]
Abstract
Language is a complex multidimensional cognitive system that is connected to many neurocognitive capacities. The development of language is therefore strongly intertwined with the development of these capacities and their neurobiological substrates. Consequently, language problems, for example those of children with Developmental Language Disorder (DLD), are explained by a variety of etiological pathways and each of these pathways will be associated with specific risk factors. In this review, we attempt to link previously described factors that may interfere with language development to putative underlying neurobiological mechanisms of language development, hoping to uncover openings for future therapeutical approaches or interventions that can help children to optimally develop their language skills.
Collapse
Affiliation(s)
- Tessel Boerma
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Sita Ter Haar
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands; Cognitive Neurobiology and Helmholtz Institute, Department of Psychology, Utrecht University/Translational Neuroscience, University Medical Center Utrecht, the Netherlands
| | - Rachida Ganga
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Frank Wijnen
- Institute for Language Sciences, Department of Languages, Literature and Communication, Utrecht University, Utrecht, the Netherlands
| | - Elma Blom
- Department of Development and Education of youth in Diverse Societies (DEEDS), Utrecht University, Utrecht, the Netherlands; Department of Language and Culture, The Arctic University of Norway UiT, Tromsø, Norway.
| | - Corette J Wierenga
- Biology Department, Faculty of Science, Utrecht University, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
11
|
Monday HR, Wang HC, Feldman DE. Circuit-level theories for sensory dysfunction in autism: convergence across mouse models. Front Neurol 2023; 14:1254297. [PMID: 37745660 PMCID: PMC10513044 DOI: 10.3389/fneur.2023.1254297] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Individuals with autism spectrum disorder (ASD) exhibit a diverse range of behavioral features and genetic backgrounds, but whether different genetic forms of autism involve convergent pathophysiology of brain function is unknown. Here, we analyze evidence for convergent deficits in neural circuit function across multiple transgenic mouse models of ASD. We focus on sensory areas of neocortex, where circuit differences may underlie atypical sensory processing, a central feature of autism. Many distinct circuit-level theories for ASD have been proposed, including increased excitation-inhibition (E-I) ratio and hyperexcitability, hypofunction of parvalbumin (PV) interneuron circuits, impaired homeostatic plasticity, degraded sensory coding, and others. We review these theories and assess the degree of convergence across ASD mouse models for each. Behaviorally, our analysis reveals that innate sensory detection behavior is heightened and sensory discrimination behavior is impaired across many ASD models. Neurophysiologically, PV hypofunction and increased E-I ratio are prevalent but only rarely generate hyperexcitability and excess spiking. Instead, sensory tuning and other aspects of neural coding are commonly degraded and may explain impaired discrimination behavior. Two distinct phenotypic clusters with opposing neural circuit signatures are evident across mouse models. Such clustering could suggest physiological subtypes of autism, which may facilitate the development of tailored therapeutic approaches.
Collapse
Affiliation(s)
- Hannah R. Monday
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| | | | - Daniel E. Feldman
- Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
12
|
Wingrove J, de Hoog E, Spencer GE. Disruptions in network plasticity precede deficits in memory following inhibition of retinoid signaling. J Neurophysiol 2023; 129:41-55. [PMID: 36448682 DOI: 10.1152/jn.00270.2022] [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: 12/02/2022] Open
Abstract
Retinoic acid, the active metabolite of vitamin A, is important for vertebrate cognition and hippocampal plasticity, but few studies have examined its role in invertebrate learning and memory, and its actions in the invertebrate central nervous system are currently unknown. Using the mollusc Lymnaea stagnalis, we examined operant conditioning of the respiratory behavior, controlled by a well-defined central pattern generator (CPG), and used citral to inhibit retinoic acid signaling. Both citral- and vehicle-treated animals showed normal learning, but citral-treated animals failed to exhibit long-term memory at 24 h. Cohorts of citral- or vehicle-treated animals were dissected into semi-intact preparations, either 1 h after training, or after the memory test 24 h later. Simultaneous electrophysiological recordings from the CPG pacemaker cell (right pedal dorsal 1; RPeD1) and an identified motorneuron (VI) were made while monitoring respiratory activity (pneumostome opening). Activity of the CPG pneumostome opener interneuron (input 3 interneuron; IP3) was also monitored indirectly. Vehicle-treated conditioned preparations showed significant changes in network parameters immediately after learning, such as reduced motorneuron bursting activity (from IP3 input), delayed pneumostome opening, and decoupling of coincident IP3 input within the network. However, citral-treated preparations failed to exhibit these network changes and more closely resembled naïve preparations. Importantly, these citral-induced differences were manifested immediately after training and before any overt changes in the behavioral response (memory impairment). These studies shed light on where and when retinoid signaling might affect a central pattern-generating network to promote memory formation during conditioning of a homeostatic behavior.NEW & NOTEWORTHY We provide novel evidence for how conditioning-induced changes in a CPG network are disrupted when retinoid signaling is inhibited. Inhibition of retinoic acid signaling prevents long-term memory formation following operant conditioning, but has no effect on learning. Simultaneous electrophysiological and behavioral analyses indicate network changes immediately following learning, but these changes are prevented with inhibition of retinoid signaling, before any overt changes in behavior. These data suggest sites for retinoid actions during memory formation.
Collapse
Affiliation(s)
- Joel Wingrove
- Department Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Eric de Hoog
- Department Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Gaynor E Spencer
- Department Biological Sciences, Brock University, St Catharines, Ontario, Canada
| |
Collapse
|
13
|
Cao B, Scherrer G, Chen L. Spinal cord retinoic acid receptor signaling gates mechanical hypersensitivity in neuropathic pain. Neuron 2022; 110:4108-4124.e6. [PMID: 36223767 PMCID: PMC9789181 DOI: 10.1016/j.neuron.2022.09.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/27/2022] [Accepted: 09/22/2022] [Indexed: 02/08/2023]
Abstract
Central sensitization caused by spinal disinhibition is a key mechanism of mechanical allodynia in neuropathic pain. However, the molecular mechanisms underlying spinal disinhibition after nerve injury remain unclear. Here, we show in mice that spared nerve injury (SNI), which induces mechanical hypersensitivity and neuropathic pain, triggers homeostatic reduction of inhibitory outputs from dorsal horn parvalbumin-positive (PV+) interneurons onto both primary afferent terminals and excitatory interneurons. The reduction in inhibitory outputs drives hyperactivation of the spinal cord nociceptive pathway, causing mechanical hypersensitivity. We identified the retinoic acid receptor RARα, a central regulator of homeostatic plasticity, as the key molecular mediator for this synaptic disinhibition. Deletion of RARα in spinal PV+ neurons or application of an RARα antagonist in the spinal cord prevented the development of SNI-induced mechanical hypersensitivity. Our results identify RARα as a crucial molecular effector for neuropathic pain and a potential target for its treatment.
Collapse
Affiliation(s)
- Bing Cao
- Department of Neurosurgery, Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gregory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lu Chen
- Department of Neurosurgery, Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
14
|
Thapliyal S, Arendt KL, Lau AG, Chen L. Retinoic acid-gated BDNF synthesis in neuronal dendrites drives presynaptic homeostatic plasticity. eLife 2022; 11:e79863. [PMID: 36515276 PMCID: PMC9797192 DOI: 10.7554/elife.79863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Homeostatic synaptic plasticity is a non-Hebbian synaptic mechanism that adjusts synaptic strength to maintain network stability while achieving optimal information processing. Among the molecular mediators shown to regulate this form of plasticity, synaptic signaling through retinoic acid (RA) and its receptor, RARα, has been shown to be critically involved in the homeostatic adjustment of synaptic transmission in both hippocampus and sensory cortices. In this study, we explore the molecular mechanism through which postsynaptic RA and RARα regulates presynaptic neurotransmitter release during prolonged synaptic inactivity at mouse glutamatertic synapses. We show that RARα binds to a subset of dendritically sorted brain-derived neurotrophic factor (Bdnf) mRNA splice isoforms and represses their translation. The RA-mediated translational de-repression of postsynaptic BDNF results in the retrograde activation of presynaptic tropomyosin receptor kinase B (TrkB) receptors, facilitating presynaptic homeostatic compensation through enhanced presynaptic release. Together, our study illustrates an RA-mediated retrograde synaptic signaling pathway through which postsynaptic protein synthesis during synaptic inactivity drives compensatory changes at the presynaptic site.
Collapse
Affiliation(s)
- Shruti Thapliyal
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| | - Kristin L Arendt
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| | - Anthony G Lau
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| | - Lu Chen
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
15
|
Abstract
Vitamin A (retinol) is a critical micronutrient required for the control of stem cell functions, cell differentiation, and cell metabolism in many different cell types, both during embryogenesis and in the adult organism. However, we must obtain vitamin A from food sources. Thus, the uptake and metabolism of vitamin A by intestinal epithelial cells, the storage of vitamin A in the liver, and the metabolism of vitamin A in target cells to more biologically active metabolites, such as retinoic acid (RA) and 4-oxo-RA, must be precisely regulated. Here, I will discuss the enzymes that metabolize vitamin A to RA and the cytochrome P450 Cyp26 family of enzymes that further oxidize RA. Because much progress has been made in understanding the regulation of ALDH1a2 (RALDH2) actions in the intestine, one focus of this review is on the metabolism of vitamin A in intestinal epithelial cells and dendritic cells. Another focus is on recent data that 4-oxo-RA is a ligand required for the maintenance of hematopoietic stem cell dormancy and the important role of RARβ (RARB) in these stem cells. Despite this progress, many questions remain in this research area, which links vitamin A metabolism to nutrition, immune functions, developmental biology, and nuclear receptor pharmacology.
Collapse
Affiliation(s)
- Lorraine J Gudas
- Department of Pharmacology, and Revlon Pharmaceutical Professor of Pharmacology and Toxicology, Pharmacology Department, and the Meyer Cancer Center of Weill Cornell Medicine of Cornell University, 1300 York Ave, New York, NY 10065
| |
Collapse
|
16
|
Yang L, Xia Z, Feng J, Zhang M, Miao P, Nie Y, Zhang X, Hao Z, Hu R. Retinoic Acid Supplementation Rescues the Social Deficits in Fmr1 Knockout Mice. Front Genet 2022; 13:928393. [PMID: 35783275 PMCID: PMC9247356 DOI: 10.3389/fgene.2022.928393] [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: 04/25/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a heritable neurodevelopmental disorder with the underlying etiology yet incompletely understood and no cure treatment. Patients of fragile X syndrome (FXS) also manifest symptoms, e.g. deficits in social behaviors, that are core traits with ASD. Several studies demonstrated that a mutual defect in retinoic acid (RA) signaling was observed in FXS and ASD. However, it is still unknown whether RA replenishment could pose a positive effect on autistic-like behaviors in FXS. Herein, we found that RA signaling was indeed down-regulated when the expression of FMR1 was impaired in SH-SY5Y cells. Furthermore, RA supplementation rescued the atypical social novelty behavior, but failed to alleviate the defects in sociability behavior or hyperactivity, in Fmr1 knock-out (KO) mouse model. The repetitive behavior and motor coordination appeared to be normal. The RNA sequencing results of the prefrontal cortex in Fmr1 KO mice indicated that deregulated expression of Foxp2, Tnfsf10, Lepr and other neuronal genes was restored to normal after RA treatment. Gene ontology terms of metabolic processes, extracellular matrix organization and behavioral pathways were enriched. Our findings provided a potential therapeutic intervention for social novelty defects in FXS.
Collapse
Affiliation(s)
- Liqin Yang
- School of Medicine, Guizhou University, Guiyang, China
| | - Zhixiong Xia
- School of Life and Health Sciences, Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences, Hangzhou, China
| | - Jianhua Feng
- Department of Pediatrics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Menghuan Zhang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Pu Miao
- Department of Pediatrics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yingjie Nie
- School of Medicine, Guizhou University, Guiyang, China
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiangyan Zhang
- School of Medicine, Guizhou University, Guiyang, China
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People’s Hospital, Guiyang, China
- *Correspondence: Xiangyan Zhang, ; Zijian Hao, ; Ronggui Hu,
| | - Zijian Hao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- *Correspondence: Xiangyan Zhang, ; Zijian Hao, ; Ronggui Hu,
| | - Ronggui Hu
- School of Medicine, Guizhou University, Guiyang, China
- School of Life and Health Sciences, Hangzhou Institute for Advanced Study University of Chinese Academy of Sciences, Hangzhou, China
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Xiangyan Zhang, ; Zijian Hao, ; Ronggui Hu,
| |
Collapse
|
17
|
Bülow P, Segal M, Bassell GJ. Mechanisms Driving the Emergence of Neuronal Hyperexcitability in Fragile X Syndrome. Int J Mol Sci 2022; 23:ijms23116315. [PMID: 35682993 PMCID: PMC9181819 DOI: 10.3390/ijms23116315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperexcitability is a shared neurophysiological phenotype across various genetic neurodevelopmental disorders, including Fragile X syndrome (FXS). Several patient symptoms are associated with hyperexcitability, but a puzzling feature is that their onset is often delayed until their second and third year of life. It remains unclear how and why hyperexcitability emerges in neurodevelopmental disorders. FXS is caused by the loss of FMRP, an RNA-binding protein which has many critical roles including protein synthesis-dependent and independent regulation of ion channels and receptors, as well as global regulation of protein synthesis. Here, we discussed recent literature uncovering novel mechanisms that may drive the progressive onset of hyperexcitability in the FXS brain. We discussed in detail how recent publications have highlighted defects in homeostatic plasticity, providing new insight on the FXS brain and suggest pharmacotherapeutic strategies in FXS and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: (P.B.); (G.J.B.)
| | - Menahem Segal
- Department of Brain Science, Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Gary J. Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Correspondence: (P.B.); (G.J.B.)
| |
Collapse
|
18
|
Homeostatic plasticity and excitation-inhibition balance: The good, the bad, and the ugly. Curr Opin Neurobiol 2022; 75:102553. [PMID: 35594578 DOI: 10.1016/j.conb.2022.102553] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/15/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
Abstract
In this review, we discuss the significance of the synaptic excitation/inhibition (E/I) balance in the context of homeostatic plasticity, whose primary goal is thought to maintain neuronal firing rates at a set point. We first provide an overview of the processes through which patterned input activity drives synaptic E/I tuning and maturation of circuits during development. Next, we emphasize the importance of the E/I balance at the synaptic level (homeostatic control of message reception) as a means to achieve the goal (homeostatic control of information transmission) at the network level and consider how compromised homeostatic plasticity associated with neurological diseases leads to hyperactivity, network instability, and ultimately improper information processing. Lastly, we highlight several pathological conditions related to sensory deafferentation and describe how, in some cases, homeostatic compensation without appropriate sensory inputs can result in phantom perceptions.
Collapse
|
19
|
de Hoog E, Spencer GE. Activity-dependent modulation of neuronal K V channels by retinoic acid enhances Ca V channel activity. J Biol Chem 2022; 298:101959. [PMID: 35452677 PMCID: PMC9127218 DOI: 10.1016/j.jbc.2022.101959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/14/2022] [Accepted: 04/17/2022] [Indexed: 12/18/2022] Open
Abstract
The metabolite of vitamin A, retinoic acid (RA), is known to affect synaptic plasticity in the nervous system and to play an important role in learning and memory. A ubiquitous mechanism by which neuronal plasticity develops in the nervous system is through modulation of voltage-gated Ca2+ (CaV) and voltage-gated K+ channels. However, how retinoids might regulate the activity of these channels has not been determined. Here, we show that RA modulates neuronal firing by inducing spike broadening and complex spiking in a dose-dependent manner in peptidergic and dopaminergic cell types. Using patch-clamp electrophysiology, we show that RA-induced complex spiking is activity dependent and involves enhanced inactivation of delayed rectifier voltage-gated K+ channels. The prolonged depolarizations observed during RA-modulated spiking lead to an increase in Ca2+ influx through CaV channels, though we also show an opposing effect of RA on the same neurons to inhibit Ca2+ influx. At physiological levels of Ca2+, this inhibition is specific to CaV2 (not CaV1) channels. Examining the interaction between the spike-modulating effects of RA and its inhibition of CaV channels, we found that inhibition of CaV2 channels limits the Ca2+ influx resulting from spike modulation. Our data thus provide novel evidence to suggest that retinoid signaling affects both delayed rectifier K+ channels and CaV channels to fine-tune Ca2+ influx through CaV2 channels. As these channels play important roles in synaptic function, we propose that these modulatory effects of retinoids likely contribute to synaptic plasticity in the nervous system.
Collapse
Affiliation(s)
- Eric de Hoog
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock's Way, St Catharines, Ontario. Canada. L2S 3A1
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, 1812 Sir Isaac Brock's Way, St Catharines, Ontario. Canada. L2S 3A1.
| |
Collapse
|
20
|
Lenz M, Eichler A, Kruse P, Muellerleile J, Deller T, Jedlicka P, Vlachos A. All-trans retinoic acid induces synaptopodin-dependent metaplasticity in mouse dentate granule cells. eLife 2021; 10:71983. [PMID: 34723795 PMCID: PMC8560091 DOI: 10.7554/elife.71983] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/15/2021] [Indexed: 12/27/2022] Open
Abstract
Previously we showed that the vitamin A metabolite all-trans retinoic acid (atRA) induces synaptic plasticity in acute brain slices prepared from the mouse and human neocortex (Lenz et al., 2021). Depending on the brain region studied, distinct effects of atRA on excitatory and inhibitory neurotransmission have been reported. Here, we used intraperitoneal injections of atRA (10 mg/kg) in adult C57BL/6J mice to study the effects of atRA on excitatory and inhibitory neurotransmission in the mouse fascia dentata—a brain region implicated in memory acquisition. No major changes in synaptic transmission were observed in the ventral hippocampus while a significant increase in both spontaneous excitatory postsynaptic current frequencies and synapse numbers were evident in the dorsal hippocampus 6 hr after atRA administration. The intrinsic properties of hippocampal dentate granule cells were not significantly different and hippocampal transcriptome analysis revealed no essential neuronal changes upon atRA treatment. In light of these findings, we tested for the metaplastic effects of atRA, that is, for its ability to modulate synaptic plasticity expression in the absence of major changes in baseline synaptic strength. Indeed, in vivo long-term potentiation (LTP) experiments demonstrated that systemic atRA treatment improves the ability of dentate granule cells to express LTP. The plasticity-promoting effects of atRA were not observed in synaptopodin-deficient mice, therefore, extending our previous results regarding the relevance of synaptopodin in atRA-mediated synaptic strengthening in the mouse prefrontal cortex. Taken together, our data show that atRA mediates synaptopodin-dependent metaplasticity in mouse dentate granule cells.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Muellerleile
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany.,ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
Wang N, Zhao Y, Gao J. Association Between Peripheral Blood Levels of Vitamin A and Autism Spectrum Disorder in Children: A Meta-Analysis. Front Psychiatry 2021; 12:742937. [PMID: 34658977 PMCID: PMC8515042 DOI: 10.3389/fpsyt.2021.742937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022] Open
Abstract
Vitamin A is an essential fat-soluble micronutrient that plays important roles in a surprisingly wide variety of biological processes from early growth and development to brain maintenance. Numerous clinical studies have been conducted to explore the relationship between peripheral vitamin A levels and autism spectrum disorder (ASD), but the results of these studies are controversial. Therefore, we assessed the association between peripheral vitamin A levels and ASD in the present meta-analysis. Relevant records were retrieved through the Embase, Web of Knowledge and PubMed databases up to 13 November 2020. Reference lists were also searched and analyzed. Hedges' g with its corresponding 95% confidence interval (CI) was used to assess the association between peripheral vitamin A levels and ASD. A fixed or random effects model was selected according to a heterogeneity test in overall and subgroup analyses. Five records (six studies) with 935 ASD children and 516 healthy children were included in the present study. Significantly decreased peripheral vitamin A concentrations were observed in ASD children compared with healthy children (Hedges' g = -0.600, 95% CI -1.153 to -0.048, P = 0.033). A similar result was also obtained after removing the studies identified by Galbraith plots. In addition, no obvious publication bias was found in the meta-analysis. The findings of our meta-analysis suggested decreased peripheral vitamin A levels in ASD children compared with healthy children. Further investigations into the effects of vitamin A on the development of ASD are warranted.
Collapse
Affiliation(s)
- Ni Wang
- Nursing Office of Beijing Road Medical District, General Hospital of Xinjiang Military Region, Wulumuqi, China
| | | | - Junwei Gao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
22
|
Differential Retinoic Acid Signaling in the Hippocampus of Aged Rats with and without Memory Impairment. eNeuro 2021; 8:ENEURO.0120-21.2021. [PMID: 34417282 PMCID: PMC8442538 DOI: 10.1523/eneuro.0120-21.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 12/21/2022] Open
Abstract
Retinoic acid (RA), a metabolite of vitamin A, has many physiological functions, and mounting evidence points to important roles in cognition. In vitro experiments indicate that RA is involved in homeostatic synaptic scaling in the hippocampus, which supports overall network stability during learning. It has been previously determined that disrupted RA signaling in the hippocampus causes deterioration of memory, that RA signaling declines with age in brain, and that application of RA reverses this decline. Here, we explore whether RA signaling is altered in an animal model of neurocognitive aging. We used a Morris water maze protocol to study cognitive decline in aged rats, which assesses hippocampus-dependent spatial memory and reveals substantial interindividual differences in aged animals. Aged unimpaired (AU) rats perform on par with young (Y), while aged impaired (AI) animals exhibit spatial memory deficits. We show that the major substrate for RA, retinol binding protein 4 (RBP4), is decreased in AU rats, and retinol cell surface receptor declines with chronological age. Other affected components of RA signaling include selective increases in AI animals in hippocampal synthesis (RALDH1) and catabolism of RA (CYP26B1), RA receptor α, the RA regulated ionotropic glutamate receptor (GluR1), as well as fragile X mental retardation protein (FMRP). The results support the conclusion that, surprisingly, increased RA signaling in the aged hippocampus is associated with poor cognitive outcome.
Collapse
|
23
|
Pfaff J, Reinwald H, Ayobahan SU, Alvincz J, Göckener B, Shomroni O, Salinas G, Düring RA, Schäfers C, Eilebrecht S. Toxicogenomic differentiation of functional responses to fipronil and imidacloprid in Daphnia magna. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 238:105927. [PMID: 34340001 DOI: 10.1016/j.aquatox.2021.105927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/10/2021] [Accepted: 07/19/2021] [Indexed: 06/13/2023]
Abstract
Active substances of pesticides, biocides or pharmaceuticals can induce adverse side effects in the aquatic ecosystem, necessitating environmental hazard and risk assessment prior to substance registration. The freshwater crustacean Daphnia magna is a model organism for acute and chronic toxicity assessment representing aquatic invertebrates. However, standardized tests involving daphnia are restricted to the endpoints immobility and reproduction and thus provide only limited insights into the underlying modes-of-action. Here, we applied transcriptome profiling to a modified D. magna Acute Immobilization test to analyze and compare gene expression profiles induced by the GABA-gated chloride channel blocker fipronil and the nicotinic acetylcholine receptor (nAChR) agonist imidacloprid. Daphnids were expose to two low effect concentrations of each substance followed by RNA sequencing and functional classification of affected gene ontologies and pathways. For both insecticides, we observed a concentration-dependent increase in the number of differentially expressed genes, whose expression changes were highly significantly positively correlated when comparing both test concentrations. These gene expression fingerprints showed virtually no overlap between the test substances and they related well to previous data of diazepam and carbaryl, two substances targeting similar molecular key events. While, based on our results, fipronil predominantly interfered with molecular functions involved in ATPase-coupled transmembrane transport and transcription regulation, imidacloprid primarily affected oxidase and oxidoreductase activity. These findings provide evidence that systems biology approaches can be utilized to identify and differentiate modes-of-action of chemical stressors in D. magna as an invertebrate aquatic non-target organism. The mechanistic knowledge extracted from such data will in future contribute to the development of Adverse Outcome Pathways (AOPs) for read-across and prediction of population effects.
Collapse
Affiliation(s)
- Julia Pfaff
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany; Institute of Soil Science and Soil Conservation, Research Centre for BioSystems, Land Use and Nutrition (iFZ), Justus Liebig University Giessen, Giessen, Germany
| | - Hannes Reinwald
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany; Department Evolutionary Ecology and Environmental Toxicology, Faculty Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Steve U Ayobahan
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Julia Alvincz
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Bernd Göckener
- Department Environmental and Food Analysis, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Orr Shomroni
- NGS-Services for Integrative Genomics, University of Göttingen, Göttingen, Germany
| | - Gabriela Salinas
- NGS-Services for Integrative Genomics, University of Göttingen, Göttingen, Germany
| | - Rolf-Alexander Düring
- Institute of Soil Science and Soil Conservation, Research Centre for BioSystems, Land Use and Nutrition (iFZ), Justus Liebig University Giessen, Giessen, Germany
| | - Christoph Schäfers
- Department of Ecotoxicology, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany
| | - Sebastian Eilebrecht
- Fraunhofer Attract Eco'n'OMICs, Fraunhofer Institute for Molecular Biology and Applied Ecology, Schmallenberg, Germany.
| |
Collapse
|
24
|
Booker SA, Kind PC. Mechanisms regulating input-output function and plasticity of neurons in the absence of FMRP. Brain Res Bull 2021; 175:69-80. [PMID: 34245842 DOI: 10.1016/j.brainresbull.2021.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/13/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
The function of brain circuits relies on high-fidelity information transfer within neurons. Synaptic inputs arrive primarily at dendrites, where they undergo integration and summation throughout the somatodendritic domain, ultimately leading to the generation of precise patterns of action potentials. Emerging evidence suggests that the ability of neurons to transfer synaptic information and modulate their output is impaired in a number of neurodevelopmental disorders including Fragile X Syndrome. In this review we summarise recent findings that have revealed the pathophysiological and plasticity mechanisms that alter the ability of neurons in sensory and limbic circuits to reliably code information in the absence of FMRP. We examine which aspects of this transform may result directly from the loss of FMRP and those that a result from compensatory or homeostatic alterations to neuronal function. Dissection of the mechanisms leading to altered input-output function of neurons in the absence of FMRP and their effects on regulating neuronal plasticity throughout development could have important implications for potential therapies for Fragile X Syndrome, including directing the timing and duration of different treatment options.
Collapse
Affiliation(s)
- Sam A Booker
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK; Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Peter C Kind
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh, UK; Patrick Wild Centre for Autism Research, University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK; National Centre for Biological Sciences (NCBS), Bangalore, India.
| |
Collapse
|
25
|
Park E, Lau AG, Arendt KL, Chen L. FMRP Interacts with RARα in Synaptic Retinoic Acid Signaling and Homeostatic Synaptic Plasticity. Int J Mol Sci 2021; 22:ijms22126579. [PMID: 34205274 PMCID: PMC8235556 DOI: 10.3390/ijms22126579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 01/08/2023] Open
Abstract
The fragile X syndrome (FXS) is an X-chromosome-linked neurodevelopmental disorder with severe intellectual disability caused by inactivation of the fragile X mental retardation 1 (FMR1) gene and subsequent loss of the fragile X mental retardation protein (FMRP). Among the various types of abnormal synaptic function and synaptic plasticity phenotypes reported in FXS animal models, defective synaptic retinoic acid (RA) signaling and subsequent defective homeostatic plasticity have emerged as a major synaptic dysfunction. However, the mechanism underlying the defective synaptic RA signaling in the absence of FMRP is unknown. Here, we show that RARα, the RA receptor critically involved in synaptic RA signaling, directly interacts with FMRP. This interaction is enhanced in the presence of RA. Blocking the interaction between FMRP and RARα with a small peptide corresponding to the critical binding site in RARα abolishes RA-induced increases in excitatory synaptic transmission, recapitulating the phenotype seen in the Fmr1 knockout mouse. Taken together, these data suggest that not only are functional FMRP and RARα necessary for RA-dependent homeostatic synaptic plasticity, but that the interaction between these two proteins is essential for proper transcription-independent RA signaling. Our results may provide further mechanistic understanding into FXS synaptic pathophysiology.
Collapse
|
26
|
Salcedo-Arellano MJ, Cabal-Herrera AM, Punatar RH, Clark CJ, Romney CA, Hagerman RJ. Overlapping Molecular Pathways Leading to Autism Spectrum Disorders, Fragile X Syndrome, and Targeted Treatments. Neurotherapeutics 2021; 18:265-283. [PMID: 33215285 PMCID: PMC8116395 DOI: 10.1007/s13311-020-00968-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Autism spectrum disorders (ASD) are subdivided into idiopathic (unknown) etiology and secondary, based on known etiology. There are hundreds of causes of ASD and most of them are genetic in origin or related to the interplay of genetic etiology and environmental toxicology. Approximately 30 to 50% of the etiologies can be identified when using a combination of available genetic testing. Many of these gene mutations are either core components of the Wnt signaling pathway or their modulators. The full mutation of the fragile X mental retardation 1 (FMR1) gene leads to fragile X syndrome (FXS), the most common cause of monogenic origin of ASD, accounting for ~ 2% of the cases. There is an overlap of molecular mechanisms in those with idiopathic ASD and those with FXS, an interaction between various signaling pathways is suggested during the development of the autistic brain. This review summarizes the cross talk between neurobiological pathways found in ASD and FXS. These signaling pathways are currently under evaluation to target specific treatments in search of the reversal of the molecular abnormalities found in both idiopathic ASD and FXS.
Collapse
Affiliation(s)
- Maria Jimena Salcedo-Arellano
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Department of Pathology and Laboratory Medicine, UC Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| | - Ana Maria Cabal-Herrera
- Group on Congenital Malformations and Dysmorphology, Faculty of Health, Universidad del Valle, Cali, 00000, Colombia
| | - Ruchi Harendra Punatar
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Courtney Jessica Clark
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Christopher Allen Romney
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA
| | - Randi J Hagerman
- Department of Pediatrics, University of California Davis School of Medicine, Sacramento, CA, 95817, USA.
- Medical Investigation of Neurodevelopmental Disorders (MIND) Institute UCDHS, University of California Davis, 2825 50th Street, Sacramento, CA, 95817, USA.
| |
Collapse
|
27
|
Rosiles-Abonce A, Rubio C, Taddei E, Rosiles D, Rubio-Osornio M. Antiepileptogenic Effect of Retinoic Acid. Curr Neuropharmacol 2021; 19:383-391. [PMID: 32351181 PMCID: PMC8033965 DOI: 10.2174/1570159x18666200429232104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/17/2020] [Accepted: 04/24/2020] [Indexed: 11/24/2022] Open
Abstract
Retinoic acid, a metabolite of vitamin A, acts through either genomic or nongenomic actions. The genomic action of retinoids exerts effects on gene transcription through interaction with retinoid receptors such as retinoic acid receptors (RARα, β, and γ) and retinoid X receptors (RXRα, β, and γ) that are primarily concentrated in the amygdala, pre-frontal cortex, and hippocampal areas in the brain. In response to retinoid binding, RAR/RXR heterodimers undergo major conformational changes and orchestrate the transcription of specific gene networks. Previous experimental studies have reported that retinoic acid exerts an antiepileptogenic effect through diverse mechanisms, including the modulation of gap junctions, neurotransmitters, long-term potentiation, calcium channels and some genes. To our knowledge, there are no previous or current clinical trials evaluating the use of retinoic acid for seizure control.
Collapse
Affiliation(s)
| | | | | | | | - Moisés Rubio-Osornio
- Address correspondence to this author at the Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Ciudad de Mexico; E-mail:
| |
Collapse
|
28
|
Marie A, Darricau M, Touyarot K, Parr-Brownlie LC, Bosch-Bouju C. Role and Mechanism of Vitamin A Metabolism in the Pathophysiology of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2021; 11:949-970. [PMID: 34120916 PMCID: PMC8461657 DOI: 10.3233/jpd-212671] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/12/2021] [Indexed: 01/09/2023]
Abstract
Evidence shows that altered retinoic acid signaling may contribute to the pathogenesis and pathophysiology of Parkinson's disease (PD). Retinoic acid is the bioactive derivative of the lipophilic vitamin A. Vitamin A is involved in several important homeostatic processes, such as cell differentiation, antioxidant activity, inflammation and neuronal plasticity. The role of vitamin A and its derivatives in the pathogenesis and pathophysiology of neurodegenerative diseases, and their potential as therapeutics, has drawn attention for more than 10 years. However, the literature sits in disparate fields. Vitamin A could act at the crossroad of multiple environmental and genetic factors of PD. The purpose of this review is to outline what is known about the role of vitamin A metabolism in the pathogenesis and pathophysiology of PD. We examine key biological systems and mechanisms that are under the control of vitamin A and its derivatives, which are (or could be) exploited for therapeutic potential in PD: the survival of dopaminergic neurons, oxidative stress, neuroinflammation, circadian rhythms, homeostasis of the enteric nervous system, and hormonal systems. We focus on the pivotal role of ALDH1A1, an enzyme expressed by dopaminergic neurons for the detoxification of these neurons, which is under the control of retinoic acid. By providing an integrated summary, this review will guide future studies on the potential role of vitamin A in the management of symptoms, health and wellbeing for PD patients.
Collapse
Affiliation(s)
- Anaıs Marie
- University Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Morgane Darricau
- University Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
- University Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Katia Touyarot
- University Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Louise C. Parr-Brownlie
- Department of Anatomy, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand (Center of Research Excellence), Dunedin, New Zealand
| | | |
Collapse
|
29
|
Li J, Jiang RY, Arendt KL, Hsu YT, Zhai SR, Chen L. Defective memory engram reactivation underlies impaired fear memory recall in Fragile X syndrome. eLife 2020; 9:61882. [PMID: 33215988 PMCID: PMC7679137 DOI: 10.7554/elife.61882] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/13/2020] [Indexed: 11/25/2022] Open
Abstract
Fragile X syndrome (FXS) is an X chromosome-linked disease associated with severe intellectual disabilities. Previous studies using the Fmr1 knockout (KO) mouse, an FXS mouse model, have attributed behavioral deficits to synaptic dysfunctions. However, how functional deficits at neural network level lead to abnormal behavioral learning remains unexplored. Here, we show that the efficacy of hippocampal engram reactivation is reduced in Fmr1 KO mice performing contextual fear memory recall. Experiencing an enriched environment (EE) prior to learning improved the engram reactivation efficacy and rescued memory recall in the Fmr1 KO mice. In addition, chemogenetically inhibiting EE-engaged neurons in CA1 reverses the rescue effect of EE on memory recall. Thus, our results suggest that inappropriate engram reactivation underlies cognitive deficits in FXS, and enriched environment may rescue cognitive deficits by improving network activation accuracy.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurosurgery, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Rena Y Jiang
- Department of Neurosurgery, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Kristin L Arendt
- Department of Neurosurgery, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Yu-Tien Hsu
- Department of Neurosurgery, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Sophia R Zhai
- Department of Neurosurgery, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| | - Lu Chen
- Department of Neurosurgery, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, United States
| |
Collapse
|
30
|
Wołoszynowska-Fraser MU, Kouchmeshky A, McCaffery P. Vitamin A and Retinoic Acid in Cognition and Cognitive Disease. Annu Rev Nutr 2020; 40:247-272. [PMID: 32966186 DOI: 10.1146/annurev-nutr-122319-034227] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The history of vitamin A goes back over one hundred years, but our realization of its importance for the brain and cognition is much more recent. The brain is more efficient than other target tissues at converting vitamin A to retinoic acid (RA), which activates retinoic acid receptors (RARs). RARs regulate transcription, but their function in the cytoplasm to control nongenomic actions is also crucial. Controlled synthesis of RA is essential for regulating synaptic plasticity in regions of the brain involved in learning and memory, such as the hippocampus. Vitamin A deficiency results in a deterioration of these functions, and failure of RA signaling is perhaps associated with normal cognitive decline with age as well as with Alzheimer's disease. Further, several psychiatric and developmental disorders that disrupt cognition are also linked with vitamin A and point to their possible treatment with vitamin A or RA.
Collapse
Affiliation(s)
| | - Azita Kouchmeshky
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, United Kingdom;
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, United Kingdom;
| |
Collapse
|
31
|
Qin XY, Fang H, Shan QH, Qi CC, Zhou JN. All-trans Retinoic Acid-induced Abnormal Hippocampal Expression of Synaptic Genes SynDIG1 and DLG2 is Correlated with Anxiety or Depression-Like Behavior in Mice. Int J Mol Sci 2020; 21:ijms21082677. [PMID: 32290523 PMCID: PMC7215843 DOI: 10.3390/ijms21082677] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/02/2020] [Accepted: 04/10/2020] [Indexed: 11/16/2022] Open
Abstract
Clinical reports suggest a potential link between excess retinoids and development of depression. Although it has been shown that all-trans retinoic acid (ATRA) administration induces behavioral changes, further insight into how ATRA is involved is lacking. The hippocampus seems to be a major target of retinoids, and abnormal synaptic plasticity of the hippocampus is involved in depression. We examined two genes associated with synaptic function, discs large homolog 2 (DLG2), and synapse differentiation-inducing gene protein 1 (SynDIG1) in terms of hippocampal expression and correlation with behavior. Three different doses of ATRA were injected into young mice and 10 mg/kg ATRA was found to induce depression-like behavior. In the hippocampus, DLG2 mRNA was significantly decreased by ATRA. mRNA levels were positively correlated with central area duration and distance in the open-field test. Increased SynDIG1 mRNA levels were observed. There was a negative correlation between SynDIG1 mRNA levels and mobility time in the forced swimming test. Retinoic acid receptor γ mRNA was significantly positively correlated with DLG2 and negatively correlated with SynDIG1. To summarize, ATRA administration induced anxiety- and depression-like behavior accompanied by a decreased expression of DLG2 and an increased expression of SynDIG1. Moreover, DLG2 was correlated with anxiety-like behavior and SynDIG1 was correlated with depression-like behavior. These results might constitute a novel target underlying ATRA-induced anxiety- and depression-like behavior.
Collapse
Affiliation(s)
- Xin-Ya Qin
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; (X.-Y.Q.); (H.F.); (Q.-H.S.)
| | - Hui Fang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; (X.-Y.Q.); (H.F.); (Q.-H.S.)
| | - Qing-Hong Shan
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; (X.-Y.Q.); (H.F.); (Q.-H.S.)
| | - Cong-Cong Qi
- Department of Laboratory Animal Science, Fudan University, Shanghai 200000, China;
| | - Jiang-Ning Zhou
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; (X.-Y.Q.); (H.F.); (Q.-H.S.)
- Correspondence:
| |
Collapse
|
32
|
Bülow P, Murphy TJ, Bassell GJ, Wenner P. Homeostatic Intrinsic Plasticity Is Functionally Altered in Fmr1 KO Cortical Neurons. Cell Rep 2020; 26:1378-1388.e3. [PMID: 30726724 PMCID: PMC6443253 DOI: 10.1016/j.celrep.2019.01.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/20/2018] [Accepted: 01/09/2019] [Indexed: 12/12/2022] Open
Abstract
Cortical hyperexcitability is a hallmark of fragile X syndrome (FXS). In the Fmr1 knockout (KO) mouse model of FXS,
cortical hyperexcitability is linked to sensory hypersensitivity and seizure susceptibility. It remains unclear why homeostatic
mechanisms fail to prevent such activity. Homeostatic intrinsic plasticity (HIP) adjusts membrane excitability through regulation
of ion channels to maintain activity levels following activity perturbation. Despite the critical role of HIP in the maturation of
excitability, it has not been examined in FXS. Here, we demonstrate that HIP does not operate normally in a disease model, FXS.
HIP was either lost or exaggerated in two distinct neuronal populations from Fmr1 KO cortical cultures. In addition, we have
identified a mechanism for homeostatic intrinsic plasticity. Compromising HIP function during development could leave cortical
neurons in the FXS nervous system vulnerable to hyperexcitability. Fragile X syndrome (FXS) is characterized by cortical hyperexcitability, but the mechanisms driving hyperexcitability are
poorly understood. Homeostatic intrinsic plasticity (HIP) regulates ion channel function to maintain appropriate activity levels.
Bülow et al. show that HIP is functionally altered in FXS neurons, which may leave cortical neurons vulnerable to
hyperexcitability.
Collapse
Affiliation(s)
- Pernille Bülow
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - T J Murphy
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA.
| | - Peter Wenner
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
33
|
Vitamin A: its many roles-from vision and synaptic plasticity to infant mortality. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2020; 206:389-399. [PMID: 32034476 DOI: 10.1007/s00359-020-01403-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/06/2019] [Accepted: 01/15/2020] [Indexed: 10/25/2022]
Abstract
The recognition that a dietary factor is essential to maintain good and sensitive vision as well as overall health goes back over 3,000 years to the ancient Egyptians. With the discovery of the vitamins at the turn of the twentieth century, fat-soluble vitamin A was soon shown to be the essential factor. In the first half of the twentieth century, the role vitamin A plays in vision, as precursor to the light-sensitive visual pigment molecules in the photoreceptors was elegantly worked out, especially by George Wald and his colleagues. Beginning in the 1960s, with the recognition of the active metabolite of vitamin A, its acid form now called retinoic acid, the roles of vitamin A in maintaining overall health of an organism began to be explored, and this research continues to this day. Receptors activated by retinoic acid, the RARs and RXRs have been shown to regulate gene transcription in a surprisingly wide variety of biological processes from early growth and development to the maintenance of epithelial tissues in many organs, the regulation of the immune system, and even the modulation of synaptic function in the brain involved in mechanisms underlying memory and learning. Therapeutic uses for retinoic acid have been developed, including one for a specific form of leukemia. The story is by no means complete and it is likely more surprises await with regard to this remarkable molecule.
Collapse
|
34
|
Zou RX, Gu X, Ding JJ, Wang T, Bi N, Niu K, Ge M, Chen XT, Wang HL. Pb exposure induces an imbalance of excitatory and inhibitory synaptic transmission in cultured rat hippocampal neurons. Toxicol In Vitro 2019; 63:104742. [PMID: 31785328 DOI: 10.1016/j.tiv.2019.104742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 11/12/2019] [Accepted: 11/24/2019] [Indexed: 12/16/2022]
Abstract
An appropriate balance of excitatory and inhibitory synapse maintains the network stability of the central nervous system. Our recent work showed lead (Pb) exposure can inhibit synaptic transmission in cultured hippocampal neurons. However, it is not clear whether Pb exposure disrupt the balance of excitatory and inhibitory synaptic transmission. Here, primary cultured hippocampal neurons from Sprague-Dawley (SD) rats were exposed to Pb (0.2 μM, 1 μM, 5 μM, respectively) from Days in Vitro (DIV) 7 to DIV 12 for 5 days and the excitatory and inhibitory synaptic transmission was examined. Patch clamp recording results showed that distinct from exposures of 0.2 μM and 5 μM, 1 μM Pb exposure significantly increased the mIPSC frequency and decreased the mEPSC frequency, leading to a uniform inhibitory outcome. Further, the number of inhibitory presynaptic puncta was significantly increased after 1 μM Pb exposure, while the number of excitatory presynaptic terminals was decreased. In addition 1 μM Pb increased the glutamic acid decarboxylase (GAD65) expression and the surface GABAA receptor (GABAAR) clusters. This shift might potentiate the synthesis of GABA and enhance the surface distribution of postsynaptic GABAAR clusters in hippocampus neurons. Together, these data showed that Pb exposure disrupted the balance of excitatory and inhibitory synaptic transmission via abnormal GABAergic neurotransmission.
Collapse
Affiliation(s)
- Rong-Xin Zou
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xiaozhen Gu
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Jin-Jun Ding
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Tiandong Wang
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Nanxi Bi
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Kang Niu
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Mengmeng Ge
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xiang-Tao Chen
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230031, PR China.
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|
35
|
Synaptic retinoic acid receptor signaling mediates mTOR-dependent metaplasticity that controls hippocampal learning. Proc Natl Acad Sci U S A 2019; 116:7113-7122. [PMID: 30782829 DOI: 10.1073/pnas.1820690116] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Homeostatic synaptic plasticity is a stabilizing mechanism engaged by neural circuits in response to prolonged perturbation of network activity. The non-Hebbian nature of homeostatic synaptic plasticity is thought to contribute to network stability by preventing "runaway" Hebbian plasticity at individual synapses. However, whether blocking homeostatic synaptic plasticity indeed induces runaway Hebbian plasticity in an intact neural circuit has not been explored. Furthermore, how compromised homeostatic synaptic plasticity impacts animal learning remains unclear. Here, we show in mice that the experience of an enriched environment (EE) engaged homeostatic synaptic plasticity in hippocampal circuits, thereby reducing excitatory synaptic transmission. This process required RARα, a nuclear retinoic acid receptor that doubles as a cytoplasmic retinoic acid-induced postsynaptic regulator of protein synthesis. Blocking RARα-dependent homeostatic synaptic plasticity during an EE experience by ablating RARα signaling induced runaway Hebbian plasticity, as evidenced by greatly enhanced long-term potentiation (LTP). As a consequence, RARα deletion in hippocampal circuits during an EE experience resulted in enhanced spatial learning but suppressed learning flexibility. In the absence of RARα, moreover, EE experience superactivated mammalian target of rapamycin (mTOR) signaling, causing a shift in protein translation that enhanced the expression levels of AMPA-type glutamate receptors. Treatment of mice with the mTOR inhibitor rapamycin during an EE experience not only restored normal AMPA-receptor expression levels but also reversed the increases in runaway Hebbian plasticity and learning after hippocampal RARα deletion. Thus, our findings reveal an RARα- and mTOR-dependent mechanism by which homeostatic plasticity controls Hebbian plasticity and learning.
Collapse
|
36
|
Li J, Park E, Zhong LR, Chen L. Homeostatic synaptic plasticity as a metaplasticity mechanism - a molecular and cellular perspective. Curr Opin Neurobiol 2019; 54:44-53. [PMID: 30212714 PMCID: PMC6361678 DOI: 10.1016/j.conb.2018.08.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/16/2018] [Accepted: 08/20/2018] [Indexed: 01/08/2023]
Abstract
The molecular mechanisms underlying various types of synaptic plasticity are historically regarded as separate processes involved in independent cellular events. However, recent progress in our molecular understanding of Hebbian and homeostatic synaptic plasticity supports the observation that these two types of plasticity share common cellular events, and are often altered together in neurological diseases. Here, we discuss the emerging concept of homeostatic synaptic plasticity as a metaplasticity mechanism with a focus on cellular signaling processes that enable a direct interaction between Hebbian and homeostatic plasticity. We also identify distinct and shared molecular players involved in these cellular processes that may be explored experimentally in future studies to test the hypothesis that homeostatic synaptic plasticity serves as a metaplasticity mechanism to integrate changes in neuronal activity and support optimal Hebbian learning.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Esther Park
- Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Lei R Zhong
- Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Lu Chen
- Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA.
| |
Collapse
|
37
|
Neurobehavioral and oxidative stress alterations following methylmercury and retinyl palmitate co-administration in pregnant and lactating rats and their offspring. Neurotoxicology 2018; 69:164-180. [DOI: 10.1016/j.neuro.2018.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/26/2022]
|
38
|
Retinoic Acid Receptor RARα-Dependent Synaptic Signaling Mediates Homeostatic Synaptic Plasticity at the Inhibitory Synapses of Mouse Visual Cortex. J Neurosci 2018; 38:10454-10466. [PMID: 30355624 DOI: 10.1523/jneurosci.1133-18.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 09/17/2018] [Accepted: 10/10/2018] [Indexed: 11/21/2022] Open
Abstract
Homeostatic synaptic plasticity is a synaptic mechanism through which the nervous system adjusts synaptic excitation and inhibition to maintain network stability. Retinoic acid (RA) and its receptor RARα have been established as critical mediators of homeostatic synaptic plasticity. In vitro studies reveal that RA signaling enhances excitatory synaptic strength and decreases inhibitory synaptic strength. However, it is unclear whether RA-mediated homeostatic synaptic plasticity occurs in vivo, and if so, whether it operates at specific types of synapses. Here, we examine the impact of RA/RARα signaling in the monocular zone of primary visual cortex (V1m) in mice of either sex. Exogenous RA treatment in acute cortical slices resulted in a reduction in mIPSCs of layer 2/3 pyramidal neurons, an effect mimicked by visual deprivation induced by binocular enucleation in postcritical period animals. Postnatal deletion of RARα blocked RA's effect on mIPSCs. Cell type-specific deletion of RARα revealed that RA acted specifically on parvalbumin (PV)-expressing interneurons. RARα deletion in PV+ interneurons blocked visual deprivation-induced changes in mIPSCs, demonstrating the critical involvement of RA signaling in PV+ interneurons in vivo Moreover, visual deprivation- or RA-induced downregulation of synaptic inhibition was absent in the visual cortical circuit of constitutive and PV-specific Fmr1 KO mice, strongly suggesting a functional interaction between fragile X mental retardation protein and RA signaling pathways. Together, our results demonstrate that RA/RARα signaling acts as a key component for homeostatic regulation of synaptic transmission at the inhibitory synapses of the visual cortex.SIGNIFICANCE STATEMENT In vitro studies established that retinoic acid (RA) and its receptor RARα play key roles in homeostatic synaptic plasticity, a mechanism by which synaptic excitation/inhibition balance and network stability are maintained. However, whether synaptic RA signaling operates in vivo remains undetermined. Here, using a conditional RARα KO mouse and cell type-specific Cre-driver lines, we showed that RARα signaling in parvalbumin-expressing interneurons is crucial for visual deprivation-induced homeostatic synaptic plasticity at inhibitory synapses in visual cortical circuits. Importantly, this form of synaptic plasticity is absent when fragile X mental retardation protein is selectively deleted in parvalbumin-expressing interneurons, suggesting a functional connection between RARα and fragile X mental retardation protein signaling pathways in vivo Thus, dysfunction of RA-dependent homeostatic plasticity may contribute to cortical circuit abnormalities in fragile X syndrome.
Collapse
|
39
|
Zhang Z, Marro SG, Zhang Y, Arendt KL, Patzke C, Zhou B, Fair T, Yang N, Südhof TC, Wernig M, Chen L. The fragile X mutation impairs homeostatic plasticity in human neurons by blocking synaptic retinoic acid signaling. Sci Transl Med 2018; 10:eaar4338. [PMID: 30068571 PMCID: PMC6317709 DOI: 10.1126/scitranslmed.aar4338] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 07/12/2018] [Indexed: 11/02/2022]
Abstract
Fragile X syndrome (FXS) is an X chromosome-linked disease leading to severe intellectual disabilities. FXS is caused by inactivation of the fragile X mental retardation 1 (FMR1) gene, but how FMR1 inactivation induces FXS remains unclear. Using human neurons generated from control and FXS patient-derived induced pluripotent stem (iPS) cells or from embryonic stem cells carrying conditional FMR1 mutations, we show here that loss of FMR1 function specifically abolished homeostatic synaptic plasticity without affecting basal synaptic transmission. We demonstrated that, in human neurons, homeostatic plasticity induced by synaptic silencing was mediated by retinoic acid, which regulated both excitatory and inhibitory synaptic strength. FMR1 inactivation impaired homeostatic plasticity by blocking retinoic acid-mediated regulation of synaptic strength. Repairing the genetic mutation in the FMR1 gene in an FXS patient cell line restored fragile X mental retardation protein (FMRP) expression and fully rescued synaptic retinoic acid signaling. Thus, our study reveals a robust functional impairment caused by FMR1 mutations that might contribute to neuronal dysfunction in FXS. In addition, our results suggest that FXS patient iPS cell-derived neurons might be useful for studying the mechanisms mediating functional abnormalities in FXS.
Collapse
Affiliation(s)
- Zhenjie Zhang
- Departments of Neurosurgery, and Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Samuele G Marro
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Yingsha Zhang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Kristin L Arendt
- Departments of Neurosurgery, and Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Christopher Patzke
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Bo Zhou
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Tyler Fair
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Nan Yang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5453, USA.
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305-5453, USA
| | - Lu Chen
- Departments of Neurosurgery, and Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA.
| |
Collapse
|
40
|
Postnatal Ablation of Synaptic Retinoic Acid Signaling Impairs Cortical Information Processing and Sensory Discrimination in Mice. J Neurosci 2018; 38:5277-5288. [PMID: 29760176 DOI: 10.1523/jneurosci.3028-17.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 04/28/2018] [Accepted: 05/03/2018] [Indexed: 12/28/2022] Open
Abstract
Retinoic acid (RA) and its receptors (RARs) are well established essential transcriptional regulators during embryonic development. Recent findings in cultured neurons identified an independent and critical post-transcriptional role of RA and RARα in the homeostatic regulation of excitatory and inhibitory synaptic transmission in mature neurons. However, the functional relevance of synaptic RA signaling in vivo has not been established. Here, using somatosensory cortex as a model system and the RARα conditional knock-out mouse as a tool, we applied multiple genetic manipulations to delete RARα postnatally in specific populations of cortical neurons, and asked whether synaptic RA signaling observed in cultured neurons is involved in cortical information processing in vivo Indeed, conditional ablation of RARα in mice via a CaMKIIα-Cre or a layer 5-Cre driver line or via somatosensory cortex-specific viral expression of Cre-recombinase impaired whisker-dependent texture discrimination, suggesting a critical requirement of RARα expression in L5 pyramidal neurons of somatosensory cortex for normal tactile sensory processing. Transcranial two-photon imaging revealed a significant increase in dendritic spine elimination on apical dendrites of somatosensory cortical layer 5 pyramidal neurons in these mice. Interestingly, the enhancement of spine elimination is whisker experience-dependent as whisker trimming rescued the spine elimination phenotype. Additionally, experiencing an enriched environment improved texture discrimination in RARα-deficient mice and reduced excessive spine pruning. Thus, RA signaling is essential for normal experience-dependent cortical circuit remodeling and sensory processing.SIGNIFICANCE STATEMENT The importance of synaptic RA signaling has been demonstrated in in vitro studies. However, whether RA signaling mediated by RARα contributes to neural circuit functions in vivo remains largely unknown. In this study, using a RARα conditional knock-out mouse, we performed multiple regional/cell-type-specific manipulation of RARα expression in the postnatal brain, and show that RARα signaling contributes to normal whisker-dependent texture discrimination as well as regulating spine dynamics of apical dendrites from layer (L5) pyramidal neurons in S1. Deletion of RARα in excitatory neurons in the forebrain induces elevated spine elimination and impaired sensory discrimination. Our study provides novel insights into the role of RARα signaling in cortical processing and experience-dependent spine maturation.
Collapse
|
41
|
Gao M, Whitt JL, Huang S, Lee A, Mihalas S, Kirkwood A, Lee HK. Experience-dependent homeostasis of 'noise' at inhibitory synapses preserves information coding in adult visual cortex. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0156. [PMID: 28093550 DOI: 10.1098/rstb.2016.0156] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2016] [Indexed: 02/05/2023] Open
Abstract
Synapses are intrinsically 'noisy' in that neurotransmitter is occasionally released in the absence of an action potential. At inhibitory synapses, the frequency of action potential-independent release is orders of magnitude higher than that at excitatory synapses raising speculations that it may serve a function. Here we report that the frequency of action potential-independent inhibitory synaptic 'noise' (i.e. miniature inhibitory postsynaptic currents, mIPSCs) is highly regulated by sensory experience in visual cortex. Importantly, regulation of mIPSC frequency is so far the predominant form of functional plasticity at inhibitory synapses in adults during the refractory period for plasticity and is a locus of rapid non-genomic actions of oestrogen. Models predict that regulating the frequency of mIPSCs, together with the previously characterized synaptic scaling of miniature excitatory PSCs, allows homeostatic maintenance of both the mean and variance of inputs to a neuron, a necessary feature of probabilistic population codes. Furthermore, mIPSC frequency regulation allows preservation of the temporal profile of neural responses while homeostatically regulating the overall firing rate. Our results suggest that the control of inhibitory 'noise' allows adaptive maintenance of adult cortical function in tune with the sensory environment.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
Affiliation(s)
- Ming Gao
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jessica L Whitt
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Shiyong Huang
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Angela Lee
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Stefan Mihalas
- Allen Institute for Brain Science, Seattle, WA 98103, USA
| | - Alfredo Kirkwood
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hey-Kyoung Lee
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, USA .,Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
42
|
Yee AX, Hsu YT, Chen L. A metaplasticity view of the interaction between homeostatic and Hebbian plasticity. Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0155. [PMID: 28093549 DOI: 10.1098/rstb.2016.0155] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2016] [Indexed: 01/25/2023] Open
Abstract
Hebbian and homeostatic plasticity are two major forms of plasticity in the nervous system: Hebbian plasticity provides a synaptic basis for associative learning, whereas homeostatic plasticity serves to stabilize network activity. While achieving seemingly very different goals, these two types of plasticity interact functionally through overlapping elements in their respective mechanisms. Here, we review studies conducted in the mammalian central nervous system, summarize known circuit and molecular mechanisms of homeostatic plasticity, and compare these mechanisms with those that mediate Hebbian plasticity. We end with a discussion of 'local' homeostatic plasticity and the potential role of local homeostatic plasticity as a form of metaplasticity that modulates a neuron's future capacity for Hebbian plasticity.This article is part of the themed issue 'Integrating Hebbian and homeostatic plasticity'.
Collapse
Affiliation(s)
- Ada X Yee
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Yu-Tien Hsu
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| | - Lu Chen
- Departments of Neurosurgery, Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305-5453, USA
| |
Collapse
|
43
|
Eidsaa M, Stubbs L, Almaas E. Comparative analysis of weighted gene co-expression networks in human and mouse. PLoS One 2017; 12:e0187611. [PMID: 29161290 PMCID: PMC5697817 DOI: 10.1371/journal.pone.0187611] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/23/2017] [Indexed: 01/21/2023] Open
Abstract
The application of complex network modeling to analyze large co-expression data sets has gained traction during the last decade. In particular, the use of the weighted gene co-expression network analysis framework has allowed an unbiased and systems-level investigation of genotype-phenotype relationships in a wide range of systems. Since mouse is an important model organism for biomedical research on human disease, it is of great interest to identify similarities and differences in the functional roles of human and mouse orthologous genes. Here, we develop a novel network comparison approach which we demonstrate by comparing two gene-expression data sets from a large number of human and mouse tissues. The method uses weighted topological overlap alongside the recently developed network-decomposition method of s-core analysis, which is suitable for making gene-centrality rankings for weighted networks. The aim is to identify globally central genes separately in the human and mouse networks. By comparing the ranked gene lists, we identify genes that display conserved or diverged centrality-characteristics across the networks. This framework only assumes a single threshold value that is chosen from a statistical analysis, and it may be applied to arbitrary network structures and edge-weight distributions, also outside the context of biology. When conducting the comparative network analysis, both within and across the two species, we find a clear pattern of enrichment of transcription factors, for the homeobox domain in particular, among the globally central genes. We also perform gene-ontology term enrichment analysis and look at disease-related genes for the separate networks as well as the network comparisons. We find that gene ontology terms related to regulation and development are generally enriched across the networks. In particular, the genes FOXE3, RHO, RUNX2, ALX3 and RARA, which are disease genes in either human or mouse, are on the top-10 list of globally central genes in the human and mouse networks.
Collapse
Affiliation(s)
- Marius Eidsaa
- Department of Biotechnology, NTNU - Norwegian University of Science and Technology, N-7491 Trondheim, Norway
| | - Lisa Stubbs
- Institute for Genomic Biology, Neuroscience Program, Cell and Developmental Biology, University of Illinois at Urbana-Champaigne, Urbana, IL 61801, United States of America
| | - Eivind Almaas
- Department of Biotechnology, NTNU - Norwegian University of Science and Technology, N-7491 Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and General Practice, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- * E-mail:
| |
Collapse
|
44
|
Baertsch NA, Baker TL. Intermittent apnea elicits inactivity-induced phrenic motor facilitation via a retinoic acid- and protein synthesis-dependent pathway. J Neurophysiol 2017; 118:2702-2710. [PMID: 28814632 DOI: 10.1152/jn.00212.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 08/10/2017] [Accepted: 08/16/2017] [Indexed: 12/13/2022] Open
Abstract
Respiratory motoneuron pools must provide rhythmic inspiratory drive that is robust and reliable, yet dynamic enough to respond to respiratory challenges. One form of plasticity that is hypothesized to contribute to motor output stability by sensing and responding to inadequate respiratory neural activity is inactivity-induced phrenic motor facilitation (iPMF), an increase in inspiratory output triggered by a reduction in phrenic synaptic inputs. Evidence suggests that mechanisms giving rise to iPMF differ depending on the pattern of reduced respiratory neural activity (i.e., neural apnea). A prolonged neural apnea elicits iPMF via a spinal TNF-α-induced increase in atypical PKC activity, but little is known regarding mechanisms that elicit iPMF following intermittent neural apnea. We tested the hypothesis that iPMF triggered by intermittent neural apnea requires retinoic acid and protein synthesis. Phrenic nerve activity was recorded in urethane-anesthetized and -ventilated rats treated intrathecally with an inhibitor of retinoic acid synthesis (4-diethlyaminobenzaldehyde, DEAB), a protein synthesis inhibitor (emetine), or vehicle (artificial cerebrospinal fluid) before intermittent (5 episodes, ~1.25 min each) or prolonged (30 min) neural apnea. Both DEAB and emetine abolished iPMF elicited by intermittent neural apnea but had no effect on iPMF elicited by a prolonged neural apnea. Thus different patterns of reduced respiratory neural activity elicit phenotypically similar iPMF via distinct spinal mechanisms. Understanding mechanisms that allow respiratory motoneurons to dynamically tune their output may have important implications in the context of respiratory control disorders that involve varied patterns of reduced respiratory neural activity, such as central sleep apnea and spinal cord injury.NEW & NOTEWORTHY We identify spinal retinoic acid and protein synthesis as critical components in the cellular cascade whereby repetitive reductions in respiratory neural activity elicit rebound increases in phrenic inspiratory activity.
Collapse
Affiliation(s)
- Nathan A Baertsch
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Tracy L Baker
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
45
|
Gonzalez-Islas C, Bülow P, Wenner P. Regulation of synaptic scaling by action potential-independent miniature neurotransmission. J Neurosci Res 2017; 96:348-353. [PMID: 28782263 DOI: 10.1002/jnr.24138] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/17/2022]
Abstract
Synaptic scaling represents a homeostatic adjustment in synaptic strength that was first identified as a cell-wide mechanism to achieve firing rate homeostasis after perturbations to spiking activity levels. In this review, we consider a form of synaptic scaling that is triggered by changes in action potential-independent neurotransmitter release. This plasticity appears to be both triggered and expressed locally at the dendritic site of the synapse that experiences a perturbation. A discussion of different forms of scaling triggered by different perturbations is presented. We consider work from multiple groups supporting this form of scaling, which we call neurotransmission-based scaling. This class of homeostatic synaptic plasticity is compared in studies using hippocampal and cortical cultures, as well as in vivo work in the embryonic chick spinal cord. Despite differences in the tissues examined, there are clear similarities in neurotransmission-based scaling, which appear to be molecularly distinct from the originally described spike-based scaling.
Collapse
Affiliation(s)
- Carlos Gonzalez-Islas
- Physiology Department, Emory University, School of Medicine, Atlanta, Georgia.,Doctorado en Ciencias Biológicas, Univerisdad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Pernille Bülow
- Physiology Department, Emory University, School of Medicine, Atlanta, Georgia.,Cell Biology Department, Emory University School of Medicine, Atlanta, GA
| | - Peter Wenner
- Physiology Department, Emory University, School of Medicine, Atlanta, Georgia
| |
Collapse
|
46
|
Rothwell CM, de Hoog E, Spencer GE. The role of retinoic acid in the formation and modulation of invertebrate central synapses. J Neurophysiol 2017; 117:692-704. [PMID: 27852736 PMCID: PMC5292328 DOI: 10.1152/jn.00737.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/12/2016] [Indexed: 11/22/2022] Open
Abstract
Trophic factors can influence many aspects of nervous system function, such as neurite outgrowth, synapse formation, and synapse modulation. The vitamin A metabolite, retinoic acid, can exert trophic effects to promote neuronal survival and outgrowth in many species and is also known to modulate vertebrate hippocampal synapses. However, its role in synaptogenesis has not been well studied, and whether it can modulate existing invertebrate synapses is also not known. In this study, we first examined a potential trophic effect of retinoic acid on the formation of excitatory synapses, independently of its role in neurite outgrowth, using cultured neurons of the mollusc Lymnaea stagnalis We also investigated its role in modulating both chemical and electrical synapses between various Lymnaea neurons in cell culture. Although we found no evidence to suggest retinoic acid affected short-term synaptic plasticity in the form of post-tetanic potentiation, we did find a significant cell type-specific modulation of electrical synapses. Given the prevalence of electrical synapses in invertebrate nervous systems, these findings highlight the potential for retinoic acid to modulate network function in the central nervous system of at least some invertebrates. NEW & NOTEWORTHY This study performed the first electrophysiological analysis of the ability of the vitamin A metabolite, retinoic acid, to exert trophic influences during synaptogenesis independently of its effects in supporting neurite outgrowth. It was also the first study to examine the ability of retinoic acid to modify both chemical and electrical synapses in any invertebrate, nonchordate species. We provide evidence that all-trans retinoic acid can modify invertebrate electrical synapses of central neurons in a cell-specific manner.
Collapse
Affiliation(s)
- Cailin M Rothwell
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Eric de Hoog
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Gaynor E Spencer
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
47
|
New Insights Into the Roles of Retinoic Acid Signaling in Nervous System Development and the Establishment of Neurotransmitter Systems. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:1-84. [PMID: 28215529 DOI: 10.1016/bs.ircmb.2016.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Secreted chiefly from the underlying mesoderm, the morphogen retinoic acid (RA) is well known to contribute to the specification, patterning, and differentiation of neural progenitors in the developing vertebrate nervous system. Furthermore, RA influences the subtype identity and neurotransmitter phenotype of subsets of maturing neurons, although relatively little is known about how these functions are mediated. This review provides a comprehensive overview of the roles played by RA signaling during the formation of the central and peripheral nervous systems of vertebrates and highlights its effects on the differentiation of several neurotransmitter systems. In addition, the evolutionary history of the RA signaling system is discussed, revealing both conserved properties and alternate modes of RA action. It is proposed that comparative approaches should be employed systematically to expand our knowledge of the context-dependent cellular mechanisms controlled by the multifunctional signaling molecule RA.
Collapse
|
48
|
Fung LK, Reiss AL. Moving Toward Integrative, Multidimensional Research in Modern Psychiatry: Lessons Learned From Fragile X Syndrome. Biol Psychiatry 2016; 80:100-111. [PMID: 26868443 PMCID: PMC4912939 DOI: 10.1016/j.biopsych.2015.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
Abstract
The field of psychiatry is approaching a major inflection point. The basic science behind cognition, emotion, behavior, and social processes has been advancing rapidly in the past 20 years. However, clinical research supporting the classification system in psychiatry has not kept up with these scientific advances. To begin organizing the basic science of psychiatry in a comprehensive manner, we begin by selecting fragile X syndrome, a neurogenetic disease with cognitive-behavioral manifestations, to illustrate key concepts in an integrative, multidimensional model. Specifically, we describe key genetic and molecular mechanisms (e.g., gamma-aminobutyric acidergic dysfunction and metabotropic glutamate receptor 5-associated long-term depression) relevant to the pathophysiology of fragile X syndrome as well as neural correlates of cognitive-behavioral symptoms. We then describe what we have learned from fragile X syndrome that may be applicable to other psychiatric disorders. We conclude this review by discussing current and future opportunities in diagnosing and treating psychiatric diseases.
Collapse
Affiliation(s)
- Lawrence K. Fung
- Division of Child & Adolescent Psychiatry, Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA
| | - Allan L. Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA
| |
Collapse
|
49
|
Yee AX, Chen L. Differential regulation of spontaneous and evoked inhibitory synaptic transmission in somatosensory cortex by retinoic acid. Synapse 2016; 70:445-52. [PMID: 27348405 DOI: 10.1002/syn.21921] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/23/2016] [Indexed: 11/08/2022]
Abstract
Retinoic acid (RA), a developmental morphogen, has emerged in recent studies as a novel synaptic signaling molecule that acts in mature hippocampal neurons to modulate excitatory and inhibitory synaptic transmission in the context of homeostatic synaptic plasticity. However, it is unclear whether RA is capable of modulating neural circuits outside of the hippocampus, and if so, whether the mode of RA's action at synapses is similar to that within the hippocampal network. Here we explore for the first time RA's synaptic function outside the hippocampus and uncover a novel function of all-trans retinoic acid at inhibitory synapses. Acute RA treatment increases spontaneous inhibitory synaptic transmission in L2/3 pyramidal neurons of the somatosensory cortex, and this effect requires expression of RA's receptor RARα both pre- and post-synaptically. Intriguingly, RA does not seem to affect evoked inhibitory transmission assayed with either extracellular stimulation or direct activation of action potentials in presynaptic interneurons at connected pairs of interneurons and pyramidal neurons. Taken together, these results suggest that RA's action at synapses is not monotonous, but is diverse depending on the type of synaptic connection (excitatory versus inhibitory) and circuit (hippocampal versus cortical). Thus, synaptic signaling of RA may mediate multi-faceted regulation of synaptic plasticity. In addition to its classic roles in brain development, retinoic acid (RA) has recently been shown to regulate excitatory and inhibitory transmission in the adult brain. Here, the authors show that in layer 2/3 (L2/3) of the somatosensory cortex (S1), acute RA induces increases in spontaneous but not action-potential evoked transmission, and that this requires retinoic acid receptor (RARα) both in presynaptic PV-positive interneurons and postsynaptic pyramidal (PN) neurons.
Collapse
Affiliation(s)
- Ada X Yee
- Department of Neurosurgery, Neurosciences Program, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305-5453.,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305-5453
| | - Lu Chen
- Department of Neurosurgery, Neurosciences Program, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305-5453. .,Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305-5453.
| |
Collapse
|
50
|
Calcineurin mediates homeostatic synaptic plasticity by regulating retinoic acid synthesis. Proc Natl Acad Sci U S A 2015; 112:E5744-52. [PMID: 26443861 DOI: 10.1073/pnas.1510239112] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Homeostatic synaptic plasticity is a form of non-Hebbian plasticity that maintains stability of the network and fidelity for information processing in response to prolonged perturbation of network and synaptic activity. Prolonged blockade of synaptic activity decreases resting Ca(2+) levels in neurons, thereby inducing retinoic acid (RA) synthesis and RA-dependent homeostatic synaptic plasticity; however, the signal transduction pathway that links reduced Ca(2+)-levels to RA synthesis remains unknown. Here we identify the Ca(2+)-dependent protein phosphatase calcineurin (CaN) as a key regulator for RA synthesis and homeostatic synaptic plasticity. Prolonged inhibition of CaN activity promotes RA synthesis in neurons, and leads to increased excitatory and decreased inhibitory synaptic transmission. These effects of CaN inhibitors on synaptic transmission are blocked by pharmacological inhibitors of RA synthesis or acute genetic deletion of the RA receptor RARα. Thus, CaN, acting upstream of RA, plays a critical role in gating RA signaling pathway in response to synaptic activity. Moreover, activity blockade-induced homeostatic synaptic plasticity is absent in CaN knockout neurons, demonstrating the essential role of CaN in RA-dependent homeostatic synaptic plasticity. Interestingly, in GluA1 S831A and S845A knockin mice, CaN inhibitor- and RA-induced regulation of synaptic transmission is intact, suggesting that phosphorylation of GluA1 C-terminal serine residues S831 and S845 is not required for CaN inhibitor- or RA-induced homeostatic synaptic plasticity. Thus, our study uncovers an unforeseen role of CaN in postsynaptic signaling, and defines CaN as the Ca(2+)-sensing signaling molecule that mediates RA-dependent homeostatic synaptic plasticity.
Collapse
|