1
|
Leahy SN, Vita DJ, Broadie K. PTPN11/Corkscrew Activates Local Presynaptic Mapk Signaling to Regulate Synapsin, Synaptic Vesicle Pools, and Neurotransmission Strength, with a Dual Requirement in Neurons and Glia. J Neurosci 2024; 44:e1077232024. [PMID: 38471782 PMCID: PMC11044113 DOI: 10.1523/jneurosci.1077-23.2024] [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: 06/04/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024] Open
Abstract
Cytoplasmic protein tyrosine phosphatase nonreceptor type 11 (PTPN11) and Drosophila homolog Corkscrew (Csw) regulate the mitogen-activated protein kinase (MAPK) pathway via a conserved autoinhibitory mechanism. Disease-causing loss-of-function (LoF) and gain-of-function (GoF) mutations both disrupt this autoinhibition to potentiate MAPK signaling. At the Drosophila neuromuscular junction glutamatergic synapse, LoF/GoF mutations elevate transmission strength and reduce activity-dependent synaptic depression. In both sexes of LoF/GoF mutations, the synaptic vesicles (SV)-colocalized synapsin phosphoprotein tether is highly elevated at rest, but quickly reduced with stimulation, suggesting a larger SV reserve pool with greatly heightened activity-dependent recruitment. Transmission electron microscopy of mutants reveals an elevated number of SVs clustered at the presynaptic active zones, suggesting that the increased vesicle availability is causative for the elevated neurotransmission. Direct neuron-targeted extracellular signal-regulated kinase (ERK) GoF phenocopies both increased local presynaptic MAPK/ERK signaling and synaptic transmission strength in mutants, confirming the presynaptic regulatory mechanism. Synapsin loss blocks this elevation in both presynaptic PTPN11 and ERK mutants. However, csw null mutants cannot be rescued by wild-type Csw in neurons: neurotransmission is only rescued by expressing Csw in both neurons and glia simultaneously. Nevertheless, targeted LoF/GoF mutations in either neurons or glia alone recapitulate the elevated neurotransmission. Thus, PTPN11/Csw mutations in either cell type are sufficient to upregulate presynaptic function, but a dual requirement in neurons and glia is necessary for neurotransmission. Taken together, we conclude that PTPN11/Csw acts in both neurons and glia, with LoF and GoF similarly upregulating MAPK/ERK signaling to enhance presynaptic Synapsin-mediated SV trafficking.
Collapse
Affiliation(s)
- Shannon N Leahy
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| | - Dominic J Vita
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| | - Kendal Broadie
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| |
Collapse
|
2
|
Neumann S, Kuteykin-Teplyakov K, Heumann R. Neuronal Protection by Ha-RAS-GTPase Signaling through Selective Downregulation of Plasmalemmal Voltage-Dependent Anion Channel-1. Int J Mol Sci 2024; 25:3030. [PMID: 38474278 DOI: 10.3390/ijms25053030] [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: 01/10/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
The small GTPase RAS acts as a plasma membrane-anchored intracellular neurotrophin counteracting neuronal degeneration in the brain, but the underlying molecular mechanisms are largely unknown. In transgenic mice expressing constitutively activated V12-Ha-RAS selectively in neurons, proteome analysis uncovered a 70% decrease in voltage-dependent anion channel-1 (VDAC-1) in the cortex and hippocampus. We observed a corresponding reduction in the levels of mRNA splicing variant coding for plasma membrane-targeted VDAC-1 (pl-VDAC-1) while mRNA levels for mitochondrial membrane VDAC-1 (mt-VDAC-1) remained constant. In primary cortical neurons derived from V12-Ha-RAS animals, a decrease in pl-VDAC-1 mRNA levels was observed, accompanied by a concomitant reduction in the ferricyanide reductase activity associated with VDAC-1 protein. Application of MEK inhibitor U0126 to transgenic cortical neurons reconstituted pl-VDAC-1 mRNA to reach wild-type levels. Excitotoxic glutamate-induced cell death was strongly attenuated in transgenic V12-Ha-RAS overexpressing cortical cultures. Consistently, a neuroprotective effect could also be achieved in wild-type cortical cultures by the extracellular application of channel-blocking antibody targeting the N-terminus of VDAC-1. These results may encourage novel therapeutic approaches toward blocking pl-VDAC-1 by monoclonal antibody targeting for complementary treatments in transplantation and neurodegenerative disease.
Collapse
Affiliation(s)
- Sebastian Neumann
- Department of Biochemistry II-Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Konstantin Kuteykin-Teplyakov
- Department of Biochemistry II-Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Rolf Heumann
- Department of Biochemistry II-Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr-Universität Bochum, 44801 Bochum, Germany
| |
Collapse
|
3
|
Guhathakurta D, Petrušková A, Akdaş EY, Perelló-Amorós B, Frischknecht R, Anni D, Weiss EM, Walter M, Fejtová A. Hydroxynorketamine, but not ketamine, acts via α7 nicotinic acetylcholine receptor to control presynaptic function and gene expression. Transl Psychiatry 2024; 14:47. [PMID: 38253622 PMCID: PMC10803733 DOI: 10.1038/s41398-024-02744-y] [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: 04/20/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Ketamine is clinically used fast-acting antidepressant. Its metabolite hydroxynorketamine (HNK) shows a robust antidepressant effect in animal studies. It is unclear, how these chemically distinct compounds converge on similar neuronal effects. While KET acts mostly as N-methyl-d-aspartate receptor (NMDAR) antagonist, the molecular target of HNK remains enigmatic. Here, we show that KET and HNK converge on rapid inhibition of glutamate release by reducing the release competence of synaptic vesicles and induce nuclear translocation of pCREB that controls expression of neuroplasticity genes connected to KET- and HNK-mediated antidepressant action. Ro25-6981, a selective antagonist of GluN2B, mimics effect of KET indicating that GluN2B-containing NMDAR might mediate the presynaptic effect of KET. Selective antagonist of α7 nicotinic acetylcholine receptors (α7nAChRs) or genetic deletion of Chrna7, its pore-forming subunit, fully abolishes HNK-induced synaptic and nuclear regulations, but leaves KET-dependent cellular effects unaffected. Thus, KET or HNK-induced modulation of synaptic transmission and nuclear translocation of pCREB can be mediated by selective signaling via NMDAR or α7nAChRs, respectively. Due to the rapid metabolism of KET to HNK, it is conceivable that subsequent modulation of glutamatergic and cholinergic neurotransmission affects circuits in a cell-type-specific manner and contributes to the therapeutic potency of KET. This finding promotes further exploration of new combined medications for mood disorders.
Collapse
Affiliation(s)
- Debarpan Guhathakurta
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Aneta Petrušková
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- National Institute of Mental Health, Klecany, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Enes Yağız Akdaş
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bartomeu Perelló-Amorós
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Renato Frischknecht
- Department of Biology, Animal Physiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Anni
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva-Maria Weiss
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Jena, Jena, Germany
| | - Anna Fejtová
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
4
|
Qu W, Jeong A, Zhong R, Thieschafer JS, Gram A, Li L. Deletion of Small GTPase H-Ras Rescues Memory Deficits and Reduces Amyloid Plaque-Associated Dendritic Spine Loss in Transgenic Alzheimer's Mice. Mol Neurobiol 2023; 60:495-511. [PMID: 36287323 PMCID: PMC10771223 DOI: 10.1007/s12035-022-03082-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/27/2022] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder, affecting millions of lives without a cure. While the molecular mechanism of AD remains obscure, emerging evidence suggests that small GTPases, a group of GTP-binding proteins that regulate a plethora of essential cellular events, modulate the pathogenic process of AD. Among those, the small GTPase H-Ras, extensively studied in cancer, regulates synaptic function, and both upstream and downstream signaling pathways of H-Ras have been implicated in AD. However, the role of H-Ras per se in AD pathogenesis had not been explored previously. In the present study, the impact of Hras deletion on cognitive function and amyloid pathology was investigated in transgenic APP/PS1 mice of AD. Behavioral assessments showed that the absence of Hras rescued spatial memory deficit in APP/PS1 mice at 9 months of age. The pathological evaluation demonstrated that Hras deletion reduced cortical amyloid deposition and astrogliosis. Furthermore, Hras deficiency protected against amyloid plaque-associated loss of dendritic spines in APP/PS1 mice. Intriguingly, canonical signaling pathways downstream of H-Ras were not affected by the absence of Hras in the brain. Unbiased transcriptomic analysis revealed that lack of H-Ras affected the expression of select genes in the brain of AD mice and identified a novel connection between H-Ras and Annexin A4, a calcium-dependent phospholipid-binding protein that has been shown to regulate membrane repair, neuroinflammation, and calcium homeostasis. Taken together, these data indicate that H-Ras modifies the pathogenic process of AD and may serve as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Wenhui Qu
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA
- Department of Pathology & Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Angela Jeong
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Rui Zhong
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Josslen S Thieschafer
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Andrea Gram
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ling Li
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
5
|
Leahy SN, Song C, Vita DJ, Broadie K. FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 2023; 21:e3001969. [PMID: 36701299 PMCID: PMC9879533 DOI: 10.1371/journal.pbio.3001969] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Noonan syndrome (NS) and NS with multiple lentigines (NSML) cognitive dysfunction are linked to SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) gain-of-function (GoF) and loss-of-function (LoF), respectively. In Drosophila disease models, we find both SHP2 mutations from human patients and corkscrew (csw) homolog LoF/GoF elevate glutamatergic transmission. Cell-targeted RNAi and neurotransmitter release analyses reveal a presynaptic requirement. Consistently, all mutants exhibit reduced synaptic depression during high-frequency stimulation. Both LoF and GoF mutants also show impaired synaptic plasticity, including reduced facilitation, augmentation, and post-tetanic potentiation. NS/NSML diseases are characterized by elevated MAPK/ERK signaling, and drugs suppressing this signaling restore normal neurotransmission in mutants. Fragile X syndrome (FXS) is likewise characterized by elevated MAPK/ERK signaling. Fragile X Mental Retardation Protein (FMRP) binds csw mRNA and neuronal Csw protein is elevated in Drosophila fragile X mental retardation 1 (dfmr1) nulls. Moreover, phosphorylated ERK (pERK) is increased in dfmr1 and csw null presynaptic boutons. We find presynaptic pERK activation in response to stimulation is reduced in dfmr1 and csw nulls. Trans-heterozygous csw/+; dfmr1/+ recapitulate elevated presynaptic pERK activation and function, showing FMRP and Csw/SHP2 act within the same signaling pathway. Thus, a FMRP and SHP2 MAPK/ERK regulative mechanism controls basal and activity-dependent neurotransmission strength.
Collapse
Affiliation(s)
- Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Dominic J. Vita
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
6
|
Blok LER, Boon M, van Reijmersdal B, Höffler KD, Fenckova M, Schenck A. Genetics, molecular control and clinical relevance of habituation learning. Neurosci Biobehav Rev 2022; 143:104883. [PMID: 36152842 DOI: 10.1016/j.neubiorev.2022.104883] [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: 04/22/2022] [Revised: 08/08/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
Habituation is the most fundamental form of learning. As a firewall that protects our brain from sensory overload, it is indispensable for cognitive processes. Studies in humans and animal models provide increasing evidence that habituation is affected in autism and related monogenic neurodevelopmental disorders (NDDs). An integrated application of habituation assessment in NDDs and their animal models has unexploited potential for neuroscience and medical care. With the aim to gain mechanistic insights, we systematically retrieved genes that have been demonstrated in the literature to underlie habituation. We identified 258 evolutionarily conserved genes across species, describe the biological processes they converge on, and highlight regulatory pathways and drugs that may alleviate habituation deficits. We also summarize current habituation paradigms and extract the most decisive arguments that support the crucial role of habituation for cognition in health and disease. We conclude that habituation is a conserved, quantitative, cognition- and disease-relevant process that can connect preclinical and clinical work, and hence is a powerful tool to advance research, diagnostics, and treatment of NDDs.
Collapse
Affiliation(s)
- Laura Elisabeth Rosalie Blok
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Marina Boon
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Boyd van Reijmersdal
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Kira Daniela Höffler
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| | - Michaela Fenckova
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands; Department of Molecular Biology and Genetics, Faculty of Science, University of South Bohemia in Ceske Budejovice, Branisovska 31, 37005, Ceske Budejovice, Czech Republic.
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, the Netherlands.
| |
Collapse
|
7
|
Costa RO, Martins LF, Tahiri E, Duarte CB. Brain-derived neurotrophic factor-induced regulation of RNA metabolism in neuronal development and synaptic plasticity. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1713. [PMID: 35075821 DOI: 10.1002/wrna.1713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) plays multiple roles in the nervous system, including in neuronal development, in long-term synaptic potentiation in different brain regions, and in neuronal survival. Alterations in these regulatory mechanisms account for several diseases of the nervous system. The synaptic effects of BDNF mediated by activation of tropomyosin receptor kinase B (TrkB) receptors are partly mediated by stimulation of local protein synthesis which is now considered a ubiquitous feature in both presynaptic and postsynaptic compartments of the neuron. The capacity to locally synthesize proteins is of great relevance at several neuronal developmental stages, including during neurite development, synapse formation, and stabilization. The available evidence shows that the effects of BDNF-TrkB signaling on local protein synthesis regulate the structure and function of the developing and mature synapses. While a large number of studies have illustrated a wide range of effects of BDNF on the postsynaptic proteome, a growing number of studies also point to presynaptic effects of the neurotrophin in the local regulation of the protein composition at the presynaptic level. Here, we will review the latest evidence on the role of BDNF in local protein synthesis, comparing the effects on the presynaptic and postsynaptic compartments. Additionally, we overview the relevance of BDNF-associated local protein synthesis in neuronal development and synaptic plasticity, at the presynaptic and postsynaptic compartments, and their relevance in terms of disease. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Export and Localization > RNA Localization.
Collapse
Affiliation(s)
- Rui O Costa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Luís F Martins
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
- Molecular Neurobiology Laboratory, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - Emanuel Tahiri
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Carlos B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
8
|
Plasticity of visual evoked potentials in patients with neurofibromatosis type 1. Clin Neurophysiol 2022; 142:220-227. [DOI: 10.1016/j.clinph.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022]
|
9
|
Vasic V, Jones MSO, Haslinger D, Knaus LS, Schmeisser MJ, Novarino G, Chiocchetti AG. Translating the Role of mTOR- and RAS-Associated Signalopathies in Autism Spectrum Disorder: Models, Mechanisms and Treatment. Genes (Basel) 2021; 12:genes12111746. [PMID: 34828352 PMCID: PMC8624393 DOI: 10.3390/genes12111746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/23/2022] Open
Abstract
Mutations affecting mTOR or RAS signaling underlie defined syndromes (the so-called mTORopathies and RASopathies) with high risk for Autism Spectrum Disorder (ASD). These syndromes show a broad variety of somatic phenotypes including cancers, skin abnormalities, heart disease and facial dysmorphisms. Less well studied are the neuropsychiatric symptoms such as ASD. Here, we assess the relevance of these signalopathies in ASD reviewing genetic, human cell model, rodent studies and clinical trials. We conclude that signalopathies have an increased liability for ASD and that, in particular, ASD individuals with dysmorphic features and intellectual disability (ID) have a higher chance for disruptive mutations in RAS- and mTOR-related genes. Studies on rodent and human cell models confirm aberrant neuronal development as the underlying pathology. Human studies further suggest that multiple hits are necessary to induce the respective phenotypes. Recent clinical trials do only report improvements for comorbid conditions such as epilepsy or cancer but not for behavioral aspects. Animal models show that treatment during early development can rescue behavioral phenotypes. Taken together, we suggest investigating the differential roles of mTOR and RAS signaling in both human and rodent models, and to test drug treatment both during and after neuronal development in the available model systems.
Collapse
Affiliation(s)
- Verica Vasic
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (V.V.); (M.J.S.)
| | - Mattson S. O. Jones
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
| | - Denise Haslinger
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Lisa S. Knaus
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Michael J. Schmeisser
- Institute for Microscopic Anatomy and Neurobiology, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (V.V.); (M.J.S.)
- Focus Program Translational Neurosciences (FTN), University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria; (L.S.K.); (G.N.)
| | - Andreas G. Chiocchetti
- Autism Therapy and Research Center of Excellence, Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, 60528 Frankfurt am Main, Germany; (M.S.O.J.); (D.H.)
- Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, 60528 Frankfurt am Main, Germany
- Correspondence: ; Tel.: +49-69-6301-80658
| |
Collapse
|
10
|
Noyes NC, Phan A, Davis RL. Memory suppressor genes: Modulating acquisition, consolidation, and forgetting. Neuron 2021; 109:3211-3227. [PMID: 34450024 PMCID: PMC8542634 DOI: 10.1016/j.neuron.2021.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/15/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023]
Abstract
The brain has a remarkable but underappreciated capacity to limit memory formation and expression. The term "memory suppressor gene" was coined in 1998 as an attempt to explain emerging reports that some genes appeared to limit memory. At that time, only a handful of memory suppressor genes were known, and they were understood to work by limiting cAMP-dependent consolidation. In the intervening decades, almost 100 memory suppressor genes with diverse functions have been discovered that affect not only consolidation but also acquisition and forgetting. Here we highlight the surprising extent to which biological limits are placed on memory formation through reviewing the literature on memory suppressor genes. In this review, we present memory suppressors within the framework of their actions on different memory operations: acquisition, consolidation, and forgetting. This is followed by a discussion of the reasons why there may be a biological need to limit memory formation.
Collapse
Affiliation(s)
- Nathaniel C Noyes
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Anna Phan
- Department of Biological Sciences, University of Alberta, 11355 Saskatchewan Drive, Edmonton, AB T6G 2E9, Canada
| | - Ronald L Davis
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
11
|
Mir S, Ashraf S, Saeed M, Rahman AU, Ul-Haq Z. Protonation states at different pH, conformational changes and impact of glycosylation in synapsin Ia. Phys Chem Chem Phys 2021; 23:16718-16729. [PMID: 34318818 DOI: 10.1039/d1cp00531f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synapsin I (SynI) is the most abundant brain phosphoprotein present at presynaptic terminals that regulates neurotransmitter release, clustering of synaptic vesicles (SVs) at active zones, and stimulates synaptogenesis and neurite outgrowth. Earlier studies have established that SynI displays pH-dependent tethering of SVs to actin filaments and exhibits a maximum binding around neutral pH, however, the effect of pH shift from acidic to basic on the conformational stability of SynI has not been explored yet. Another important aspect of SynIa is its O-GlcNAcylation (O-GlcNac) at the Thr87 position, which is responsible for the positive regulation of synaptic plasticity linked to learning and memory in mice. Furthermore, reduced levels of O-GlcNAc have been observed in Alzheimer's disease, suggesting a possible link to deficits in synaptic plasticity. In this study, the effect of pH and glycosylation on the structure and functional stability of SynIa is determined through molecular dynamics (MD) simulation approach. The 3D structure of SynIa was established via threading-based homology modeling methods. It was observed that the structure of SynIa adopts extended conformational changes as the pH shifts from acidic to basic, resulting in a compact conformation at pH 8.0. Moreover, the results obtained by comparing the glycosylated and unglycosylated protein indicated that the glycan moiety imparts stability to the protein by forming intramolecular hydrogen bond interactions with the protein residues. The results indicate that although O-GlcNAc moieties do not induce a significant change in SynIa structure they minimize protein dynamics, likely leading to enhanced protein stability.
Collapse
Affiliation(s)
- Sonia Mir
- H.E.J., Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | | | | | | | | |
Collapse
|
12
|
Komorowska-Müller JA, Ravichandran KA, Zimmer A, Schürmann B. Cannabinoid receptor 2 deletion influences social memory and synaptic architecture in the hippocampus. Sci Rep 2021; 11:16828. [PMID: 34413398 PMCID: PMC8376893 DOI: 10.1038/s41598-021-96285-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Although the cannabinoid receptor 2 (CB2R) is often thought to play a role mainly outside the brain several publications unequivocally showed the presence of CB2R on hippocampal principal neurons. Activation of CB2R produced a long-lasting membrane potential hyperpolarization, altered the input/output function of CA2/3 principal neurons and produced alterations in gamma oscillations. However, other cellular, molecular and behavioral consequences of hippocampal CB2R signaling have not been studied in detail. Here we demonstrate that the deletion of CB2 leads to a highly significant increase in hippocampal synapsin-I expression levels and particle density, as well as increased vesicular GABA transporter (vGAT) levels. This phenotype was restricted to females and not observed in males. Furthermore, we demonstrate an impairment of social memory in CB2 deficient mice. Our results thus demonstrate that the lack of CB2R leads to changes in the hippocampal synaptic landscape and reveals an important sex-specific difference in endocannabinoid signaling. This study supports a significant role of the CB2R in modulation of different types of memory despite its low expression levels in the brain and provides more insight into a sex-specific role of CB2R in synaptic architecture.
Collapse
Affiliation(s)
- Joanna Agnieszka Komorowska-Müller
- Institute for Molecular Psychiatry, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bldg. 76, 53127, Bonn, Germany
| | - Kishore Aravind Ravichandran
- Institute for Molecular Psychiatry, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bldg. 76, 53127, Bonn, Germany
| | - Andreas Zimmer
- Institute for Molecular Psychiatry, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bldg. 76, 53127, Bonn, Germany.
| | - Britta Schürmann
- Institute for Molecular Psychiatry, Medical Faculty, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, Bldg. 76, 53127, Bonn, Germany
| |
Collapse
|
13
|
Calleja-Felipe M, Wojtas MN, Diaz-González M, Ciceri D, Escribano R, Ouro A, Morales M, Knafo S. FORTIS: a live-cell assay to monitor AMPA receptors using pH-sensitive fluorescence tags. Transl Psychiatry 2021; 11:324. [PMID: 34045447 PMCID: PMC8160262 DOI: 10.1038/s41398-021-01457-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/09/2021] [Accepted: 05/18/2021] [Indexed: 11/26/2022] Open
Abstract
The real-time live fluorescent monitoring of surface AMPA receptors (AMPARs) could open new opportunities for drug discovery and phenotypic screening concerning neuropsychiatric disorders. We have developed FORTIS, a tool based on pH sensitivity capable of detecting subtle changes in surface AMPARs at a neuronal population level. The expression of SEP-GluA1 or pHuji-GluA1 recombinant AMPAR subunits in mammalian neurons cultured in 96-well plates enables surface AMPARs to be monitored with a microplate reader. Thus, FORTIS can register rapid changes in surface AMPARs induced by drugs or genetic modifications without having to rely on conventional electrophysiology or imaging. By combining FORTIS with pharmacological manipulations, basal surface AMPARs, and plasticity-like changes can be monitored. We expect that employing FORTIS to screen for changes in surface AMPARs will accelerate both neuroscience research and drug discovery.
Collapse
Affiliation(s)
- María Calleja-Felipe
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain
| | - Magdalena Natalia Wojtas
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Marta Diaz-González
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dalila Ciceri
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain
| | - Raúl Escribano
- Fundación Biofísica Bizkaia/Biofisika Bizkaia Fundazioa (FBB), Barrio Sarriena s/n, Leioa, E-48940, Spain
| | - Alberto Ouro
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Miguel Morales
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain
| | - Shira Knafo
- Department of Physiology and Cell Biology, Faculty of Health Sciences, The National Institute for Biotechnology in the Negev, and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
- Instituto Biofisika (UPV/EHU, CSIC), University of the Basque Country, Leioa, E-48940, Spain.
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain.
| |
Collapse
|
14
|
Shil SK, Kagawa Y, Umaru BA, Nanto-Hara F, Miyazaki H, Yamamoto Y, Kobayashi S, Suzuki C, Abe T, Owada Y. Ndufs4 ablation decreases synaptophysin expression in hippocampus. Sci Rep 2021; 11:10969. [PMID: 34040028 PMCID: PMC8155116 DOI: 10.1038/s41598-021-90127-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 05/04/2021] [Indexed: 02/04/2023] Open
Abstract
Altered function of mitochondrial respiratory chain in brain cells is related to many neurodegenerative diseases. NADH Dehydrogenase (Ubiquinone) Fe-S protein 4 (Ndufs4) is one of the subunits of mitochondrial complex I and its mutation in human is associated with Leigh syndrome. However, the molecular biological role of Ndufs4 in neuronal function is poorly understood. In this study, upon Ndufs4 expression confirmation in NeuN-positive neurons, and GFAP-positive astrocytes in WT mouse hippocampus, we found significant decrease of mitochondrial respiration in Ndufs4-KO mouse hippocampus. Although there was no change in the number of NeuN positive neurons in Ndufs4-KO hippocampus, the expression of synaptophysin, a presynaptic protein, was significantly decreased. To investigate the detailed mechanism, we silenced Ndufs4 in Neuro-2a cells and we observed shorter neurite lengths with decreased expression of synaptophysin. Furthermore, western blot analysis for phosphorylated extracellular regulated kinase (pERK) revealed that Ndufs4 silencing decreases the activity of ERK signalling. These results suggest that Ndufs4-modulated mitochondrial activity may be involved in neuroplasticity via regulating synaptophysin expression.
Collapse
Affiliation(s)
- Subrata Kumar Shil
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yoshiteru Kagawa
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Banlanjo Abdulaziz Umaru
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Fumika Nanto-Hara
- Division of Animal Metabolism and Nutrition, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan
| | - Hirofumi Miyazaki
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yui Yamamoto
- Department of Anatomy, Tohoku Medical and Pharmaceutical University, Sendai, 981-0905, Japan
| | - Shuhei Kobayashi
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Chitose Suzuki
- Department of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Takaaki Abe
- Department of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
15
|
Olevska A, Spanagel R, Bernardi RE. Impaired contextual fear conditioning in RasGRF2 mutant mice is likely Ras-ERK-dependent. Neurobiol Learn Mem 2021; 181:107435. [PMID: 33831510 DOI: 10.1016/j.nlm.2021.107435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 03/08/2021] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Ras/Raf/MEK/ERK (Ras-ERK) signaling has been shown to play an important role in fear acquisition. However, little information is known regarding the mechanisms that contribute to the regulation of this pathway in terms of the learning of conditioned fears. Ras Guanine Nucleotide Releasing Factor 2 (RasGRF2) is one of two guanine nucleotide exchange factors (GEF) that regulates the Ras-ERK signaling pathway in a Ca2+-dependent manner via control of the cycling of Ras isoforms between an inactive and active state. Here we sought to determine the role of RasGRF2 on contextual fear conditioning in RasGRF2 knockout (KO) and their wild type (WT) counterparts. Male KO and WT mice underwent a single session of contextual fear conditioning (12 min, 4 unsignaled shocks), followed by either daily 12-min retention trials or the molecular analysis of Ras activation and pERK1/2 activity. KO mice showed an impaired acquisition of contextual fear, as demonstrated by reduced freezing during fear conditioning and 24-hr retention tests relative to WT mice. Ras analysis following fear conditioning demonstrated a reduction in Ras activation in the hippocampus as well as a reduction in pERK1/2 in the CA1 region of the hippocampus in KO mice, suggesting that the decrease in fear conditioning in KO mice is at least in part due to the impairment of Ras-ERK signaling in the hippocampus during learning. These data indicate a role for RasGRF2 in contextual fear conditioning in mice that may be Ras-ERK-dependent.
Collapse
Affiliation(s)
- Anastasia Olevska
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany.
| |
Collapse
|
16
|
Bentea E, Villers A, Moore C, Funk AJ, O’Donovan SM, Verbruggen L, Lara O, Janssen P, De Pauw L, Declerck NB, DePasquale EAK, Churchill MJ, Sato H, Hermans E, Arckens L, Meshul CK, Ris L, McCullumsmith RE, Massie A. Corticostriatal dysfunction and social interaction deficits in mice lacking the cystine/glutamate antiporter. Mol Psychiatry 2021; 26:4754-4769. [PMID: 32366950 PMCID: PMC7609546 DOI: 10.1038/s41380-020-0751-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/07/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
Abstract
The astrocytic cystine/glutamate antiporter system xc- represents an important source of extracellular glutamate in the central nervous system, with potential impact on excitatory neurotransmission. Yet, its function and importance in brain physiology remain incompletely understood. Employing slice electrophysiology and mice with a genetic deletion of the specific subunit of system xc-, xCT (xCT-/- mice), we uncovered decreased neurotransmission at corticostriatal synapses. This effect was partly mitigated by replenishing extracellular glutamate levels, indicating a defect linked with decreased extracellular glutamate availability. We observed no changes in the morphology of striatal medium spiny neurons, the density of dendritic spines, or the density or ultrastructure of corticostriatal synapses, indicating that the observed functional defects are not due to morphological or structural abnormalities. By combining electron microscopy with glutamate immunogold labeling, we identified decreased intracellular glutamate density in presynaptic terminals, presynaptic mitochondria, and in dendritic spines of xCT-/- mice. A proteomic and kinomic screen of the striatum of xCT-/- mice revealed decreased expression of presynaptic proteins and abnormal kinase network signaling, that may contribute to the observed changes in postsynaptic responses. Finally, these corticostriatal deregulations resulted in a behavioral phenotype suggestive of autism spectrum disorder in the xCT-/- mice; in tests sensitive to corticostriatal functioning we recorded increased repetitive digging behavior and decreased sociability. To conclude, our findings show that system xc- plays a previously unrecognized role in regulating corticostriatal neurotransmission and influences social preference and repetitive behavior.
Collapse
Affiliation(s)
- Eduard Bentea
- grid.8767.e0000 0001 2290 8069Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Agnès Villers
- grid.8364.90000 0001 2184 581XDepartment of Neurosciences, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Cynthia Moore
- grid.410404.50000 0001 0165 2383Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR USA
| | - Adam J. Funk
- grid.267337.40000 0001 2184 944XDepartment of Neurosciences, University of Toledo College of Medicine, Toledo, OH USA
| | - Sinead M. O’Donovan
- grid.267337.40000 0001 2184 944XDepartment of Neurosciences, University of Toledo College of Medicine, Toledo, OH USA
| | - Lise Verbruggen
- grid.8767.e0000 0001 2290 8069Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Olaya Lara
- grid.8767.e0000 0001 2290 8069Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Pauline Janssen
- grid.8767.e0000 0001 2290 8069Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Laura De Pauw
- grid.8767.e0000 0001 2290 8069Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Noemi B. Declerck
- grid.8767.e0000 0001 2290 8069Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Erica A. K. DePasquale
- grid.239573.90000 0000 9025 8099Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA ,grid.24827.3b0000 0001 2179 9593Department of Biomedical Informatics, University of Cincinnati, Cincinnati, OH USA
| | - Madeline J. Churchill
- grid.410404.50000 0001 0165 2383Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR USA
| | - Hideyo Sato
- grid.260975.f0000 0001 0671 5144Department of Medical Technology, Faculty of Medicine, Laboratory of Biochemistry and Molecular Biology, Niigata University, Niigata, Japan
| | - Emmanuel Hermans
- grid.7942.80000 0001 2294 713XInstitute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Lutgarde Arckens
- grid.5596.f0000 0001 0668 7884Laboratory of Neuroplasticity and Neuroproteomics, and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, Leuven, Belgium
| | - Charles K. Meshul
- grid.410404.50000 0001 0165 2383Research Services, Neurocytology Laboratory, Veterans Affairs Medical Center, Portland, OR USA ,grid.5288.70000 0000 9758 5690Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR USA
| | - Laurence Ris
- grid.8364.90000 0001 2184 581XDepartment of Neurosciences, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Robert E. McCullumsmith
- grid.267337.40000 0001 2184 944XDepartment of Neurosciences, University of Toledo College of Medicine, Toledo, OH USA
| | - Ann Massie
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
17
|
Gruol DL, Hernandez RV, Roberts A. Alcohol Enhances Responses to High Frequency Stimulation in Hippocampus from Transgenic Mice with Increased Astrocyte Expression of IL-6. Cell Mol Neurobiol 2020; 41:1299-1310. [PMID: 32562098 DOI: 10.1007/s10571-020-00902-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Recent studies show that alcohol exposure can induce glial production of neuroimmune factors in the CNS. Of these, IL-6 has gained attention because it is involved in a number of important physiological and pathophysiological processes that could be affected by alcohol-induced CNS production of IL-6, particularly under conditions of excessive alcohol use. For example, IL-6 has been shown to play a role in hippocampal behaviors and synaptic plasticity (long-term potentiation; LTP) associated with memory and learning. Surprisingly, in our in vitro studies of LTP at the Schaffer collateral to CA1 pyramidal neuron synapse in hippocampus from transgenic mice that express elevated levels of astrocyte produced IL-6 (TG), LTP was not altered by the increased levels of IL-6. However, exposure to acute alcohol revealed neuroadaptive changes that served to protect LTP against the alcohol-induced reduction of LTP observed in hippocampus from non-transgenic control mice (WT). Here we examined the induction phase of LTP to assess if presynaptic neuroadaptive changes occurred in the hippocampus of TG mice that contributed to the resistance of LTP to alcohol. Results are consistent with a role for IL-6-induced neuroadaptive effects on presynaptic mechanisms involved in transmitter release in the resistance of LTP to alcohol in hippocampus from the TG mice. These actions are important with respect to a role for IL-6 in physiological and pathophysiological processes in the CNS and in CNS actions of alcohol, especially when excessive alcohol used is comorbid with conditions associated with elevated levels of IL-6 in the CNS.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| | - Ruben V Hernandez
- Neuroscience Department, SR301, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
18
|
Poston RG, Murphy L, Rejepova A, Ghaninejad-Esfahani M, Segales J, Mulligan K, Saha RN. Certain ortho-hydroxylated brominated ethers are promiscuous kinase inhibitors that impair neuronal signaling and neurodevelopmental processes. J Biol Chem 2020; 295:6120-6137. [PMID: 32229587 PMCID: PMC7196656 DOI: 10.1074/jbc.ra119.011138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/23/2020] [Indexed: 12/20/2022] Open
Abstract
The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body β-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.
Collapse
Affiliation(s)
- Robert G Poston
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Lillian Murphy
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ayna Rejepova
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Mina Ghaninejad-Esfahani
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Joshua Segales
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343
| | - Kimberly Mulligan
- Department of Biological Sciences, Center for Interdisciplinary Molecular Biology: Education, Research and Advancement (CIMERA), California State University, Sacramento, California 95819
| | - Ramendra N Saha
- Molecular and Cell Biology Department, School of Natural Sciences, University of California, Merced, Merced, California 95343.
| |
Collapse
|
19
|
Paw-Min-Thein-Oo, Sakimoto Y, Kida H, Mitsushima D. Proximodistal Heterogeneity in Learning-promoted Pathway-specific Plasticity at Dorsal CA1 Synapses. Neuroscience 2020; 437:184-195. [PMID: 32360699 DOI: 10.1016/j.neuroscience.2020.04.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 01/28/2023]
Abstract
Contextual learning requires the delivery of AMPA receptors to CA1 synapses in the dorsal hippocampus. However, proximodistal heterogeneity of pathway-specific plasticity remains unclear. Here, we examined the proximodistal heterogeneity in learning-induced plasticity at the CA1 synapses with inputs from the entorhinal cortex layer III (ECIII) or from CA3. We subjected male rats to an inhibitory avoidance task and prepared acute hippocampal slices for whole-cell patch clamp experiments, where we stimulated ECIII-CA1 or CA3-CA1 input fibers to analyze evoked excitatory postsynaptic currents (EPSCs). Compared to untrained controls, trained rats exhibited higher AMPA/NMDA current ratios at CA3-CA1 synapses of proximal and intermediate, but not distal CA1 neurons, which suggested that region-specific plasticity occurred after learning. Moreover, trained rats exhibited higher AMPA/NMDA current ratios at ECIII-CA1 synapses of intermediate and distal, but not proximal CA1 neurons. These findings suggested the presence of proximodistal heterogeneity in pathway-specific postsynaptic plasticity. Regarding presynaptic plasticity, training slightly, but significantly increased the paired-pulse ratios of CA3-CA1 synapses of proximal and intermediate, but not distal CA1 neurons. Moreover, trained rats exhibited higher paired-pulse ratios at ECIII-CA1 synapses of intermediate and distal, but not proximal CA1 neurons, which suggested region-specific presynaptic plasticity. Finally, learning was clearly prevented by the bilateral microinjection of a plasticity blocker in the proximal or intermediate, but not distal CA1 subfields, which suggested functional heterogeneity along the proximodistal axis. Understanding region- and pathway-specific plasticity at dorsal CA1 synapses could aid in controlling encoded memory.
Collapse
Affiliation(s)
- Paw-Min-Thein-Oo
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Yuya Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Hiroyuki Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan
| | - Dai Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi 755-8505, Japan; The Research Institute for Time Studies, Yamaguchi University, Yamaguchi 753-8511, Japan.
| |
Collapse
|
20
|
Chen QY, Zhang ZL, Liu Q, Chen CJ, Zhang XK, Xu PY, Zhuo M. Presynaptic long-term potentiation requires extracellular signal-regulated kinases in the anterior cingulate cortex. Mol Pain 2020; 16:1744806920917245. [PMID: 32264746 PMCID: PMC7144679 DOI: 10.1177/1744806920917245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Extracellular signal-regulated kinases are widely expressed protein kinases in neurons, which serve as important intracellular signaling molecules for central plasticity such as long-term potentiation. Recent studies demonstrate that there are two major forms of long-term potentiation in cortical areas related to pain: postsynaptic long-term potentiation and presynaptic long-term potentiation. In particular, presynaptic long-term potentiation in the anterior cingulate cortex has been shown to contribute to chronic pain-related anxiety. In this review, we briefly summarized the components and roles of extracellular signal-regulated kinases in neuronal signaling, especially in the presynaptic long-term potentiation of anterior cingulate cortex, and discuss the possible molecular mechanisms and functional implications in pain-related emotional disorders.
Collapse
Affiliation(s)
- Qi-Yu Chen
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhi-Ling Zhang
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qin Liu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao-Jun Chen
- Department of Neurology, Guangzhou Chinese Medical Integrated Hospital (Huadu), Guangdong, China
| | - Xiao-Kang Zhang
- The First Affiliated Hospital of Gan-Nan Medical University, Ganzhopu, China
| | - Ping-Yi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
21
|
Pytka K, Dawson N, Tossell K, Ungless MA, Plevin R, Brett RR, Bushell TJ. Mitogen-activated protein kinase phosphatase-2 deletion modifies ventral tegmental area function and connectivity and alters reward processing. Eur J Neurosci 2020; 52:2838-2852. [PMID: 31989721 DOI: 10.1111/ejn.14688] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 01/06/2020] [Accepted: 01/17/2020] [Indexed: 11/30/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate normal brain functioning, and their dysfunction is implicated in a number of brain disorders. Thus, there is great interest in understanding the signalling systems that control MAPK functioning. One family of proteins that contribute to this process, the mitogen-activated protein kinase phosphatases (MKPs), directly inactivate MAPKs through dephosphorylation. Recent studies have identified novel functions of MKPs in foetal development, the immune system, cancer and synaptic plasticity and memory. In the present study, we performed an unbiased investigation using MKP-2-/- mice to assess whether MKP-2 plays a global role in modulating brain function. Local cerebral glucose utilization is significantly increased in the ventral tegmental area (VTA) of MKP-2-/- mice, with connectivity analysis revealing alterations in VTA functional connectivity, including a significant reduction in connectivity to the nucleus accumbens and hippocampus. In addition, spontaneous excitatory postsynaptic current frequency, but not amplitude, onto putative dopamine neurons in the VTA is increased in MKP-2-/- mice, which indicates that increased excitatory drive may account for the increased VTA glucose utilization. Consistent with modified VTA function and connectivity, in behavioural tests MKP-2-/- mice exhibited increased sucrose preference and impaired amphetamine-induced hyperlocomotion. Overall, these data reveal that MKP-2 plays a role in modulating VTA function and that its dysfunction may contribute to brain disorders in which altered reward processing is present.
Collapse
Affiliation(s)
- Karolina Pytka
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK.,Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Neil Dawson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK.,Division of Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | - Kyoko Tossell
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Mark A Ungless
- MRC London Institute of Medical Sciences (LMS), London, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, UK
| | - Robin Plevin
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Ros R Brett
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Trevor J Bushell
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
22
|
Kang M, Lee YS. The impact of RASopathy-associated mutations on CNS development in mice and humans. Mol Brain 2019; 12:96. [PMID: 31752929 PMCID: PMC6873535 DOI: 10.1186/s13041-019-0517-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 01/04/2023] Open
Abstract
The RAS signaling pathway is involved in the regulation of developmental processes, including cell growth, proliferation, and differentiation, in the central nervous system (CNS). Germline mutations in the RAS signaling pathway genes are associated with a group of neurodevelopmental disorders, collectively called RASopathy, which includes neurofibromatosis type 1, Noonan syndrome, cardio-facio-cutaneous syndrome, and Costello syndrome. Most mutations associated with RASopathies increase the activity of the RAS-ERK signaling pathway, and therefore, most individuals with RASopathies share common phenotypes, such as a short stature, heart defects, facial abnormalities, and cognitive impairments, which are often accompanied by abnormal CNS development. Recent studies using mouse models of RASopathies demonstrated that particular mutations associated with each disorder disrupt CNS development in a mutation-specific manner. Here, we reviewed the recent literatures that investigated the developmental role of RASopathy-associated mutations using mutant mice, which provided insights into the specific contribution of RAS-ERK signaling molecules to CNS development and the subsequent impact on cognitive function in adult mice.
Collapse
Affiliation(s)
- Minkyung Kang
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea. .,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 03080, South Korea.
| |
Collapse
|
23
|
Fu T, Wang J, Ding Y, Zhang Y, Han S, Li J. Modulation of cPKCγ on Synapsin-Ia/b-Specific Phosphorylation Sites in the Developing Visual Cortex of Mice. Invest Ophthalmol Vis Sci 2019; 60:2676-2684. [PMID: 31242289 DOI: 10.1167/iovs.19-26675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To explore the role of synapsin-Ia/b in visual cortical plasticity, the dynamic changes in total protein expression (T-) and conventional protein kinase C (cPKC)γ-modulated phosphorylation (P-) levels of synapsin-Ia/b were observed in the developing visual cortex of mice. Methods The Western blot analysis was used to determine the levels of T- and P-synapsin-Ia/b at site of Ser9, 549, and 603; the cPKCγ gene wild-type (cPKCγ+/+) and knockout (cPKCγ-/-) mice were applied to explore the modulation of cPKCγ on synapsin-Ia/b phosphorylation status in visual cortex of mice at postnatal 7 to 60 days (P7-P60, n = 6 per group). Results The results showed that T-synapsin-Ia/b protein levels significantly increased at P14 to P35 and peaked at P42 to 60 (P < 0.001) in visual cortex when compared with that of P7 cPKCγ+/+ mice, and cPKCγ-/- did not affect this pattern of T-synapsin-Ia/b protein expressions. For synapsin-Ia/b phosphorylation status, the levels of P-Ser9 and 603 synapsin-Ia/b significantly elevated at P21 to P28 (P < 0.05 or 0.001), and then went down and maintained at lower levels at P35 to P60 (P < 0.05 or 0.001) compared with P7 cPKCγ+/+ mice. In addition, the cPKCγ gene knockout could significantly (P < 0.001) inhibit both the increase and decrease of P-Ser9 and 603 synapsin-Ia/b levels when compared with cPKCγ+/+ mice at P7 to P60. However, there were no significant changes of P-Ser549 synapsin-Ia/b in the developing visual cortex of both cPKCγ+/+ and cPKCγ-/- mice at P7 to P60. Conclusions These results suggested that both protein expression levels and cPKCγ-modulated phosphorylation status at Ser9 and 603 of synapsin-Ia/b may play important role in developing visual cortex of mice.
Collapse
Affiliation(s)
- Tao Fu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Jing Wang
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Yichao Ding
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Science Key Laboratory, Beijing, China
| | - Yunxia Zhang
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Song Han
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Junfa Li
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Holter MC, Hewitt LT, Koebele SV, Judd JM, Xing L, Bimonte-Nelson HA, Conrad CD, Araki T, Neel BG, Snider WD, Newbern JM. The Noonan Syndrome-linked Raf1L613V mutation drives increased glial number in the mouse cortex and enhanced learning. PLoS Genet 2019; 15:e1008108. [PMID: 31017896 PMCID: PMC6502435 DOI: 10.1371/journal.pgen.1008108] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 05/06/2019] [Accepted: 03/22/2019] [Indexed: 12/19/2022] Open
Abstract
RASopathies are a family of related syndromes caused by mutations in regulators of the RAS/Extracellular Regulated Kinase 1/2 (ERK1/2) signaling cascade that often result in neurological deficits. RASopathy mutations in upstream regulatory components, such as NF1, PTPN11/SHP2, and RAS have been well-characterized, but mutation-specific differences in the pathogenesis of nervous system abnormalities remain poorly understood, especially those involving mutations downstream of RAS. Here, we assessed cellular and behavioral phenotypes in mice expressing a Raf1L613V gain-of-function mutation associated with the RASopathy, Noonan Syndrome. We report that Raf1L613V/wt mutants do not exhibit a significantly altered number of excitatory or inhibitory neurons in the cortex. However, we observed a significant increase in the number of specific glial subtypes in the forebrain. The density of GFAP+ astrocytes was significantly increased in the adult Raf1L613V/wt cortex and hippocampus relative to controls. OLIG2+ oligodendrocyte progenitor cells were also increased in number in mutant cortices, but we detected no significant change in myelination. Behavioral analyses revealed no significant changes in voluntary locomotor activity, anxiety-like behavior, or sociability. Surprisingly, Raf1L613V/wt mice performed better than controls in select aspects of the water radial-arm maze, Morris water maze, and cued fear conditioning tasks. Overall, these data show that increased astrocyte and oligodendrocyte progenitor cell (OPC) density in the cortex coincides with enhanced cognition in Raf1L613V/wt mutants and further highlight the distinct effects of RASopathy mutations on nervous system development and function. The RASopathies are a large and complex family of syndromes caused by mutations in the RAS/MAPK signaling cascade with no known cure. Individuals with these syndromes often present with heart defects, craniofacial differences, and neurological abnormalities, such as developmental delay, cognitive changes, epilepsy, and an increased risk of autism. However, there is wide variation in the extent of intellectual ability between individuals. It is currently unclear how different RASopathy mutations affect brain development. Here, we describe the cellular and behavioral consequences of a mutation in a gene called Raf1 that is associated with a common RASopathy, Noonan Syndrome. We find that mice harboring a mutation in Raf1 show moderate increases in the number of two subsets of glial cells, which is also observed in a number of other RASopathy brain samples. Surprisingly, we found that Raf1 mutant mice show improved performance in several learning and memory tasks. Our work highlights potential mutation-specific changes in RASopathy brain function and helps set the framework for future personalized therapeutic approaches.
Collapse
Affiliation(s)
- Michael C. Holter
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Lauren. T. Hewitt
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
- Arizona Alzheimer’s Consortium, Phoenix, Arizona, United States of America
| | - Jessica M. Judd
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
| | - Lei Xing
- Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
- Arizona Alzheimer’s Consortium, Phoenix, Arizona, United States of America
| | - Cheryl D. Conrad
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
| | - Toshiyuki Araki
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, United States of America
| | - Benjamin G. Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, United States of America
| | - William D. Snider
- Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Jason M. Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
25
|
Sun C, Zhu L, Ma R, Ren J, Wang J, Gao S, Yang D, Ning K, Ling B, Lu B, Chen X, Xu J. Astrocytic miR-324-5p is essential for synaptic formation by suppressing the secretion of CCL5 from astrocytes. Cell Death Dis 2019; 10:141. [PMID: 30760705 PMCID: PMC6374376 DOI: 10.1038/s41419-019-1329-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/01/2018] [Accepted: 01/04/2019] [Indexed: 12/18/2022]
Abstract
There is accumulating evidence that astrocytes play an important role in synaptic formation, plasticity, and pruning. Dicer and the fine-tuning of microRNA (miRNA) network are important for maintaining the normal functions of central nervous system and dysregulation of miRNAs is implicated in neurological disorders. However, little is known about the role of Dicer and miRNAs of astrocytes in the homeostasis of synapse as well as its plasticity. By selectively deleting Dicer in postnatal astrocytes, Dicer-deficient mice exhibited reactive astrogliosis and deficits in dendritic spine formation. Astrocyte-conditioned medium (ACM) collected from Dicer-null astrocytes caused synapse degeneration in cultured primary neurons. The expression of chemokine ligand 5 (CCL5) elevated in Dicer-deleted astrocytes which led to the significant augmentation of secreted CCL5 in ACM. In neurons treated with Dicer KO-ACM, CCL5 supplementation inhibited MAPK/CREB signaling pathway and exacerbated the synaptic formation deficiency, while CCL5 knockdown partially rescued the synapse degeneration. Moreover, we validated CCL5 as miR-324-5p targeted gene. ACM collected from miR-324-5p antagomir-transfected astrocytes mimicked the effect of CCL5 treatment on inhibiting synapse formation and MAPK/CREB signaling in Dicer KO-ACM-cocultured neurons. Furthermore, decreased miR-324-5p expression and elevated CCL5 expression were observed in the brain of aging mice. Our work reveals the non-cell-autonomous roles of astroglial miRNAs in regulation of astrocytic secretory milieu and neuronal synaptogenesis, implicating the loss or misregulation of astroglial miRNA network may contribute to neuroinflammation, neurodegeneration, and aging.
Collapse
Affiliation(s)
- Chenxi Sun
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Zhu
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rongjie Ma
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Ren
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Wang
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shane Gao
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Danjing Yang
- East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ke Ning
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Bin Ling
- The Second People's Hospital of Yunnan Province, Kunming, China.
| | - Bing Lu
- East Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Xu Chen
- Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, China.
| | - Jun Xu
- East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
26
|
Lesuis SL, Lucassen PJ, Krugers HJ. Early life stress impairs fear memory and synaptic plasticity; a potential role for GluN2B. Neuropharmacology 2019; 149:195-203. [PMID: 30641077 DOI: 10.1016/j.neuropharm.2019.01.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 01/01/2023]
Abstract
Programming of the brain by early life stress has been associated with alterations in structure and function of the dorsal hippocampus. Yet, the underlying molecular mechanisms remain largely elusive. In this study, we examined the effects of early life stress (ELS) - by housing mouse dams with limited nesting and bedding material from postnatal days 2-9 and examined in 6 month old offspring; 1) auditory fear conditioning, 2) expression of the hippocampal N-methyl-d-aspartate receptor (NMDA-R) subunits 2A and 2B (GluN2A, GluN2B), and expression of PSD-95 and synaptophysin, and 3) short- and long-term (LTP) synaptic plasticity. Given its critical role in NMDA receptor function and synaptic plasticity, we further examined the role of GluN2B in effects of ELS on synaptic plasticity and fear memory formation. We demonstrate that ELS impaired fear memory in 6 month old mice and decreased hippocampal LTP as well as the paired-pulse ratio (PPR). ELS also reduced hippocampal GluN2B expression. Interestingly, pharmacological blockade of GluN2B with the selective antagonist Ro25 6981 was less effective to reduce synaptic plasticity in ELS mice, and was also ineffective to impair memory retrieval in ELS mice. These studies suggest that ELS reduces hippocampal synaptic plasticity and fear memory formation and hampers GluN2B receptor function. As such, GluN2B may provide an important target for future strategies to prevent lasting ELS effects on cognition.
Collapse
Affiliation(s)
- Sylvie L Lesuis
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands.
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| | - Harm J Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, the Netherlands
| |
Collapse
|
27
|
Lin X, Liu T, Li P, He Z, Zhong Y, Cui H, Luo J, Wang Y, Tang T. iTRAQ-Based Proteomics Analysis Reveals the Effect of Rhubarb in Rats with Ischemic Stroke. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6920213. [PMID: 30112417 PMCID: PMC6077657 DOI: 10.1155/2018/6920213] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/05/2018] [Accepted: 06/14/2018] [Indexed: 12/23/2022]
Abstract
Background. Rhubarb, a traditional Chinese medicine, promotes viscera and remove blood stasis. Rhubarb is skilled in smoothening meridians, improving blood circulation which exhibits better efficacy on cerebral ischemic stroke. In this study, we aimed to analyze the underlying mechanisms of rhubarb which treated rats of middle cerebral artery occlusion (MCAO) model according to an iTRAQ-based proteomics and bioinformatics analysis. 30 rats were randomly allocated into three groups including sham group (SG), model group (MG), and rhubarb group (RG). Rhubarb group was given a gavage of rhubarb decoction at dose of 3 g/kg and the remaining groups were prepared with normal saline by gavage. Rats from MG and RG were induced into MCAO model. The effects of rhubarb were estimated by Modified Neurological Severity Score (mNSS) and cerebral infarct volume. The brain tissues were measured via the quantitative proteomic approach of iTRAQ coupled to liquid chromatography-tandem mass spectrometry (LC-MS/MS). Furthermore, the bioinformatics analysis of overlapping differentially expression proteins (DEPs) was conducted by DAVID, KEGG, and Cytoscape. Specific selective DEPs were validated by Western blotting. Rats treated with rhubarb after MCAO showed a significant reduction on mNSS and cerebral infarct volume compared with MG. In MG versus SG and RG versus MG, we identified a total of 4578 proteins, of which 287 were DEPs. There were 76 overlapping DEPs between MG versus SG and RG versus MG. Through bioinformatics analysis, 14 associated pathways were searched including cGMP-PKG signaling pathway, tuberculosis, synaptic vesicle cycle, amyotrophic lateral sclerosis, long-term potentiation, and so on. 76 overlapping DEPs mainly involved synaptic vesicle cycling biological processes based on GO annotation. Further, the selective overlapping DEPs were verified at the protein level by using Western blotting. Our present study reveals that rhubarb highlights promising neuroprotective effect. Rhubarb exerts novel therapeutic action via modulating multiple proteins, targets, and pathways.
Collapse
Affiliation(s)
- Xiangping Lin
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Tao Liu
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
- Department of Gerontology, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, 830000 Urumqi, China
| | - Pengfei Li
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Zehui He
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yuanyuan Zhong
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Hanjin Cui
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Jiekun Luo
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Yang Wang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Tao Tang
- Laboratory of Ethnopharmacology, Institute of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 410008 Changsha, China
| |
Collapse
|
28
|
Coleman B, Topalidou I, Ailion M. Modulation of Gq-Rho Signaling by the ERK MAPK Pathway Controls Locomotion in Caenorhabditis elegans. Genetics 2018; 209:523-535. [PMID: 29615470 PMCID: PMC5972424 DOI: 10.1534/genetics.118.300977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/29/2018] [Indexed: 12/17/2022] Open
Abstract
The heterotrimeric G protein Gq regulates neuronal activity through distinct downstream effector pathways. In addition to the canonical Gq effector phospholipase Cβ, the small GTPase Rho was recently identified as a conserved effector of Gq. To identify additional molecules important for Gq signaling in neurons, we performed a forward genetic screen in the nematode Caenorhabditis elegans for suppressors of the hyperactivity and exaggerated waveform of an activated Gq mutant. We isolated two mutations affecting the MAP kinase scaffold protein KSR-1 and found that KSR-1 modulates locomotion downstream of, or in parallel to, the Gq-Rho pathway. Through epistasis experiments, we found that the core ERK MAPK cascade is required for Gq-Rho regulation of locomotion, but that the canonical ERK activator LET-60/Ras may not be required. Through neuron-specific rescue experiments, we found that the ERK pathway functions in head acetylcholine neurons to control Gq-dependent locomotion. Additionally, expression of activated LIN-45/Raf in head acetylcholine neurons is sufficient to cause an exaggerated waveform phenotype and hypersensitivity to the acetylcholinesterase inhibitor aldicarb, similar to an activated Gq mutant. Taken together, our results suggest that the ERK MAPK pathway modulates the output of Gq-Rho signaling to control locomotion behavior in C. elegans.
Collapse
Affiliation(s)
- Brantley Coleman
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Irini Topalidou
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| | - Michael Ailion
- Department of Biochemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
29
|
Shah S, Brock EJ, Ji K, Mattingly RR. Ras and Rap1: A tale of two GTPases. Semin Cancer Biol 2018; 54:29-39. [PMID: 29621614 DOI: 10.1016/j.semcancer.2018.03.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
Ras oncoproteins play pivotal roles in both the development and maintenance of many tumor types. Unfortunately, these proteins are difficult to directly target using traditional pharmacological strategies, in part due to their lack of obvious binding pockets or allosteric sites. This obstacle has driven a considerable amount of research into pursuing alternative ways to effectively inhibit Ras, examples of which include inducing mislocalization to prevent Ras maturation and inactivating downstream proteins in Ras-driven signaling pathways. Ras proteins are archetypes of a superfamily of small GTPases that play specific roles in the regulation of many cellular processes, including vesicle trafficking, nuclear transport, cytoskeletal rearrangement, and cell cycle progression. Several other superfamily members have also been linked to the control of normal and cancer cell growth and survival. For example, Rap1 has high sequence similarity to Ras, has overlapping binding partners, and has been demonstrated to both oppose and mimic Ras-driven cancer phenotypes. Rap1 plays an important role in cell adhesion and integrin function in a variety of cell types. Mechanistically, Ras and Rap1 cooperate to initiate and sustain ERK signaling, which is activated in many malignancies and is the target of successful therapeutics. Here we review the role activated Rap1 in ERK signaling and other downstream pathways to promote invasion and cell migration and metastasis in various cancer types.
Collapse
Affiliation(s)
- Seema Shah
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ethan J Brock
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kyungmin Ji
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Raymond R Mattingly
- Program in Cancer Biology, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
30
|
Zhang X, Li Q, Wang L, Liu ZJ, Zhong Y. Active Protection: Learning-Activated Raf/MAPK Activity Protects Labile Memory from Rac1-Independent Forgetting. Neuron 2018; 98:142-155.e4. [DOI: 10.1016/j.neuron.2018.02.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/20/2017] [Accepted: 02/23/2018] [Indexed: 12/20/2022]
|
31
|
Hamada S, Ohtsuka T. CAST: Its molecular structure and phosphorylation-dependent regulation of presynaptic plasticity. Neurosci Res 2018; 127:25-32. [DOI: 10.1016/j.neures.2017.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 11/16/2022]
|
32
|
Kwon SH, Han JK, Choi M, Kwon YJ, Kim SJ, Yi EH, Shin JC, Cho IH, Kim BH, Jeong Kim S, Ye SK. Dysfunction of Microglial STAT3 Alleviates Depressive Behavior via Neuron-Microglia Interactions. Neuropsychopharmacology 2017; 42:2072-2086. [PMID: 28480882 PMCID: PMC5561349 DOI: 10.1038/npp.2017.93] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 12/22/2022]
Abstract
Neuron-microglia interactions have a crucial role in maintaining the neuroimmune system. The balance of neuroimmune system has emerged as an important process in the pathophysiology of depression. However, how neuron-microglia interactions contribute to major depressive disorders has been poorly understood. Herein, we demonstrated that microglia-derived synaptic changes induced antidepressive-like behavior by using microglia-specific signal transducer and activator of transcription 3 (STAT3) knockout (KO) (STAT3fl/fl;LysM-Cre+/-) mice. We found that microglia-specific STAT3 KO mice showed antidepressive-like behavior in the forced swim, tail suspension, sucrose preference, and open-field tests. Surprisingly, the secretion of macrophage colony-stimulating factor (M-CSF) was increased from neuronal cells in the brains of STAT3fl/fl;LysM-Cre+/- mice. Moreover, the phosphorylation of antidepressant-targeting mediators and brain-derived neurotrophic factor expression were increased in the brains of STAT3fl/fl;LysM-Cre+/- mice as well as in neuronal cells in response to M-CSF stimulation. Importantly, the miniature excitatory postsynaptic current frequency in the medial prefrontal cortex was increased in STAT3fl/fl;LysM-Cre+/- mice and in the M-CSF treatment group. Collectively, microglial STAT3 regulates depression-related behaviors via neuronal M-CSF-mediated synaptic activity, suggesting that inhibition of microglial STAT3 might be a new therapeutic strategy for depression.
Collapse
Affiliation(s)
- Sun-Ho Kwon
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea,Neuro-Immune Information Storage Network Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea,Biomedical Science Project (BK21[PLUS]), Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeong-Kyu Han
- Neuro-Immune Information Storage Network Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Brain and Cognitive Sciences, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Moonseok Choi
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong-Jin Kwon
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Yi
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae-Cheon Shin
- Pohang Center for Evaluation of Biomaterials, Pohang, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Byung-Hak Kim
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea,Biomedical Science Project (BK21[PLUS]), Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang Jeong Kim
- Neuro-Immune Information Storage Network Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea,Biomedical Science Project (BK21[PLUS]), Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Brain and Cognitive Sciences, Seoul National University Graduate School, Seoul, Republic of Korea,Department of Physiology, Seoul National University College of Medicine, Seoul, South Korea. Tel: +82 2 740 8229, Fax: +82 2 763 9667, E-mail:
| | - Sang-Kyu Ye
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea,Neuro-Immune Information Storage Network Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea,Biomedical Science Project (BK21[PLUS]), Seoul National University College of Medicine, Seoul, Republic of Korea,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, Republic of Korea,Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea. Tel: +82 2 740 8281, Fax: +82 2 745 7996, E-mail:
| |
Collapse
|
33
|
Borrie SC, Brems H, Legius E, Bagni C. Cognitive Dysfunctions in Intellectual Disabilities: The Contributions of the Ras-MAPK and PI3K-AKT-mTOR Pathways. Annu Rev Genomics Hum Genet 2017; 18:115-142. [DOI: 10.1146/annurev-genom-091416-035332] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Sarah C. Borrie
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Eric Legius
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Claudia Bagni
- Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00173 Rome, Italy
| |
Collapse
|
34
|
Schreiber J, Grimbergen LA, Overwater I, Vaart TVD, Stedehouder J, Schuhmacher AJ, Guerra C, Kushner SA, Jaarsma D, Elgersma Y. Mechanisms underlying cognitive deficits in a mouse model for Costello Syndrome are distinct from other RASopathy mouse models. Sci Rep 2017; 7:1256. [PMID: 28455524 PMCID: PMC5430680 DOI: 10.1038/s41598-017-01218-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 03/24/2017] [Indexed: 01/07/2023] Open
Abstract
RASopathies, characterized by germline mutations in genes encoding proteins of the RAS-ERK signaling pathway, show overlapping phenotypes, which manifest themselves with a varying severity of intellectual disability. However, it is unclear to what extent they share the same downstream pathophysiology that underlies the cognitive deficits. Costello syndrome (CS) is a rare RASopathy caused by activating mutations in the HRAS gene. Here we investigated the mechanisms underlying the cognitive deficits of HRas G12V/G12V mice. HRas G12V/G12V mice showed robust upregulation of ERK signaling, neuronal hypertrophy, increased brain volume, spatial learning deficits, and impaired mGluR-dependent long-term depression (LTD). In contrast, long-term potentiation (LTP), which is affected in other RASopathy mouse models was unaffected. Treatment with lovastatin, a HMG-CoA-Reductase inhibitor which has been shown to rescue the behavioral phenotypes of mouse models of NF1 and Noonan syndrome, was unable to restore ERK signaling and the cognitive deficits of HRas G12V/G12V mice. Administration of a potent mitogen-activated protein kinase (MEK) inhibitor rescued the ERK upregulation and the mGluR-LTD deficit of HRas G12V/G12V mice, but failed to rescue the cognitive deficits. Taken together, this study indicates that the fundamental molecular and cellular mechanisms underlying the cognitive aspects of different RASopathies are remarkably distinct, and may require disease specific treatments.
Collapse
Affiliation(s)
- Jadwiga Schreiber
- Department of Neuroscience, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Laura-Anne Grimbergen
- Department of Neuroscience, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Iris Overwater
- Department of Neuroscience, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Thijs van der Vaart
- Department of Neuroscience, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Jeffrey Stedehouder
- Department of Psychiatry Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Alberto J Schuhmacher
- Molecular Oncology, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Carmen Guerra
- Molecular Oncology, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Steven A Kushner
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
- Department of Psychiatry Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Dick Jaarsma
- Department of Neuroscience, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands
| | - Ype Elgersma
- Department of Neuroscience, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands.
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Centre Rotterdam, 3015, CN Rotterdam, The Netherlands.
| |
Collapse
|
35
|
Cheng Q, Song SH, Augustine GJ. Calcium-Dependent and Synapsin-Dependent Pathways for the Presynaptic Actions of BDNF. Front Cell Neurosci 2017; 11:75. [PMID: 28392759 PMCID: PMC5364187 DOI: 10.3389/fncel.2017.00075] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/02/2017] [Indexed: 11/19/2022] Open
Abstract
We used cultured hippocampal neurons to determine the signaling pathways mediating brain-derived neurotrophic factor (BDNF) regulation of spontaneous glutamate and GABA release. BDNF treatment elevated calcium concentration in presynaptic terminals; this calcium signal reached a peak within 1 min and declined in the sustained presence of BDNF. This BDNF-induced transient rise in presynaptic calcium was reduced by SKF96365, indicating that BDNF causes presynaptic calcium influx via TRPC channels. BDNF treatment increased the frequency of miniature excitatory postsynaptic currents (mEPSCs). This response consisted of two components: a transient component that peaked within 1 min of initiating BDNF application and a second component that was sustained, at a lower mEPSC frequency, for the duration of BDNF application. The initial transient component was greatly reduced by removing external calcium or by treatment with SKF96365, as well as by Pyr3, a selective blocker of TRPC3 channels. In contrast, the sustained component was unaffected in these conditions but was eliminated by U0126, an inhibitor of the MAP kinase (MAPK) pathway, as well as by genetic deletion of synapsins in neurons from a synapsin triple knock-out (TKO) mouse. Thus, two pathways mediate the ability of BDNF to enhance spontaneous glutamate release: the transient component arises from calcium influx through TRPC3 channels, while the sustained component is mediated by MAPK phosphorylation of synapsins. We also examined the ability of these two BDNF-dependent pathways to regulate spontaneous release of the inhibitory neurotransmitter, GABA. BDNF had no effect on the frequency of spontaneous miniature inhibitory postsynaptic currents (mIPSCs) in neurons from wild-type (WT) mice, but surprisingly did increase mIPSC frequency in synapsin TKO mice. This covert BDNF response was blocked by removal of external calcium or by treatment with SKF96365 or Pyr3, indicating that it results from calcium influx mediated by TRPC3 channels. Thus, the BDNF-activated calcium signaling pathway can also enhance spontaneous GABA release, though this effect is suppressed by synapsins under normal physiological conditions.
Collapse
Affiliation(s)
- Qing Cheng
- Department of Neurobiology, Duke University Medical Center Durham, NC, USA
| | - Sang-Ho Song
- Center for Functional Connectomics, Korea Institute of Science and TechnologySeoul, South Korea; Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingapore, Singapore; Institute of Molecular and Cell BiologySingapore, Singapore
| | - George J Augustine
- Center for Functional Connectomics, Korea Institute of Science and TechnologySeoul, South Korea; Lee Kong Chian School of Medicine, Nanyang Technological UniversitySingapore, Singapore; Institute of Molecular and Cell BiologySingapore, Singapore
| |
Collapse
|
36
|
Physiological and Pathological Roles of 15-Deoxy-Δ12,14-Prostaglandin J2 in the Central Nervous System and Neurological Diseases. Mol Neurobiol 2017; 55:2227-2248. [DOI: 10.1007/s12035-017-0435-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/03/2017] [Indexed: 12/29/2022]
|
37
|
Zhou M, Greenhill S, Huang S, Silva TK, Sano Y, Wu S, Cai Y, Nagaoka Y, Sehgal M, Cai DJ, Lee YS, Fox K, Silva AJ. CCR5 is a suppressor for cortical plasticity and hippocampal learning and memory. eLife 2016; 5. [PMID: 27996938 PMCID: PMC5213777 DOI: 10.7554/elife.20985] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 12/19/2016] [Indexed: 11/13/2022] Open
Abstract
Although the role of CCR5 in immunity and in HIV infection has been studied widely, its role in neuronal plasticity, learning and memory is not understood. Here, we report that decreasing the function of CCR5 increases MAPK/CREB signaling, long-term potentiation (LTP), and hippocampus-dependent memory in mice, while neuronal CCR5 overexpression caused memory deficits. Decreasing CCR5 function in mouse barrel cortex also resulted in enhanced spike timing dependent plasticity and consequently, dramatically accelerated experience-dependent plasticity. These results suggest that CCR5 is a powerful suppressor for plasticity and memory, and CCR5 over-activation by viral proteins may contribute to HIV-associated cognitive deficits. Consistent with this hypothesis, the HIV V3 peptide caused LTP, signaling and memory deficits that were prevented by Ccr5 knockout or knockdown. Overall, our results demonstrate that CCR5 plays an important role in neuroplasticity, learning and memory, and indicate that CCR5 has a role in the cognitive deficits caused by HIV.
Collapse
Affiliation(s)
- Miou Zhou
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Stuart Greenhill
- Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom.,School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Shan Huang
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Tawnie K Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yoshitake Sano
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Shumin Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, United States
| | - Ying Cai
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yoshiko Nagaoka
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, United States
| | - Megha Sehgal
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Denise J Cai
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| | - Yong-Seok Lee
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States.,Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kevin Fox
- Cardiff School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Alcino J Silva
- Departments of Neurobiology, Psychology, Psychiatry, Integrative Center for Learning and Memory and Brain Research Institute, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
38
|
Louros SR, Osterweil EK. Perturbed proteostasis in autism spectrum disorders. J Neurochem 2016; 139:1081-1092. [PMID: 27365114 PMCID: PMC5215415 DOI: 10.1111/jnc.13723] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/10/2016] [Accepted: 06/24/2016] [Indexed: 12/30/2022]
Abstract
Dynamic changes in synaptic strength rely on de novo protein synthesis and protein degradation by the ubiquitin proteasome system (UPS). Disruption of either of these cellular processes will result in significant impairments in synaptic plasticity and memory formation. Mutations in several genes encoding regulators of mRNA translation and members of the UPS have been associated with an increased risk for the development of autism spectrum disorders. It is possible that these mutations result in a similar imbalance in protein homeostasis (proteostasis) at the synapse. This review will summarize recent work investigating the role of the UPS in synaptic plasticity at glutamatergic synapses, and propose that dysfunctional proteostasis is a common consequence of several genetic mutations linked to autism spectrum disorders.
Dynamic changes in synaptic strength rely on de novo protein synthesis and protein degradation by the ubiquitin proteasome system (UPS). Disruption of either of these cellular processes will result in significant impairments in synaptic plasticity and memory formation. Mutations in several genes encoding regulators of mRNA translation (i.e. FMR1) and protein degradation (i.e. UBE3A) have been associated with an increased risk for autism spectrum disorders and intellectual disability (ASD/ID). These mutations similarly disrupt protein homeostasis (proteostasis). Compensatory changes that reset the rate of proteostasis may contribute to the neurological symptoms of ASD/ID. This review summarizes recent work investigating the role of the UPS in synaptic plasticity at glutamatergic synapses, and proposes that dysfunctional proteostasis is a common consequence of several genetic mutations linked to ASD.
This article is part of a mini review series: “Synaptic Function and Dysfunction in Brain Diseases”.
Collapse
Affiliation(s)
- Susana R Louros
- Centre for Integrative Physiology/Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Emily K Osterweil
- Centre for Integrative Physiology/Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| |
Collapse
|
39
|
Tahmasebi L, Komaki A, Karamian R, Shahidi S, Sarihi A, Komaki H. Interaction between paired-pulse facilitation and long-term potentiation during the stimulation of the cannabinoid and vanilloid systems in the dentate gyrus. Brain Res 2016; 1643:27-34. [DOI: 10.1016/j.brainres.2016.04.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 02/09/2016] [Accepted: 04/25/2016] [Indexed: 10/21/2022]
|
40
|
Vail G, Cheng A, Han YR, Zhao T, Du S, Loy MMT, Herrup K, Plummer MR. ATM protein is located on presynaptic vesicles and its deficit leads to failures in synaptic plasticity. J Neurophysiol 2016; 116:201-9. [PMID: 27075534 PMCID: PMC4961758 DOI: 10.1152/jn.00006.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/12/2016] [Indexed: 01/07/2023] Open
Abstract
Ataxia telangiectasia is a multisystemic disorder that includes a devastating neurodegeneration phenotype. The ATM (ataxia-telangiectasia mutated) protein is well-known for its role in the DNA damage response, yet ATM is also found in association with cytoplasmic vesicular structures: endosomes and lysosomes, as well as neuronal synaptic vesicles. In keeping with this latter association, electrical stimulation of the Schaffer collateral pathway in hippocampal slices from ATM-deficient mice does not elicit normal long-term potentiation (LTP). The current study was undertaken to assess the nature of this deficit. Theta burst-induced LTP was reduced in Atm(-/-) animals, with the reduction most pronounced at burst stimuli that included 6 or greater trains. To assess whether the deficit was associated with a pre- or postsynaptic failure, we analyzed paired-pulse facilitation and found that it too was significantly reduced in Atm(-/-) mice. This indicates a deficit in presynaptic function. As further evidence that these synaptic effects of ATM deficiency were presynaptic, we used stochastic optical reconstruction microscopy. Three-dimensional reconstruction revealed that ATM is significantly more closely associated with Piccolo (a presynaptic marker) than with Homer1 (a postsynaptic marker). These results underline how, in addition to its nuclear functions, ATM plays an important functional role in the neuronal synapse where it participates in the regulation of presynaptic vesicle physiology.
Collapse
Affiliation(s)
- Graham Vail
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Aifang Cheng
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; and
| | - Yu Ray Han
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Teng Zhao
- Department of Physics, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Shengwang Du
- Department of Physics, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Michael M T Loy
- Department of Physics, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Karl Herrup
- Division of Life Science and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong; and
| | - Mark R Plummer
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey;
| |
Collapse
|
41
|
Mitogen-Activated Protein Kinase Phosphatase-2 Deletion Impairs Synaptic Plasticity and Hippocampal-Dependent Memory. J Neurosci 2016; 36:2348-54. [PMID: 26911683 DOI: 10.1523/jneurosci.3825-15.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) regulate brain function and their dysfunction is implicated in a number of brain disorders, including Alzheimer's disease. Thus, there is great interest in understanding the signaling systems that control MAPK function. One family of proteins that contribute to this process, the mitogen-activated protein kinase phosphatases (MKPs), directly inactivate MAPKs through dephosphorylation. Recent studies have identified novel functions of MKPs in development, the immune system, and cancer. However, a significant gap in our knowledge remains in relation to their role in brain functioning. Here, using transgenic mice where the Dusp4 gene encoding MKP-2 has been knocked out (MKP-2(-/-) mice), we show that long-term potentiation is impaired in MKP-2(-/-) mice compared with MKP-2(+/+) controls whereas neuronal excitability, evoked synaptic transmission, and paired-pulse facilitation remain unaltered. Furthermore, spontaneous EPSC (sEPSC) frequency was increased in acute slices and primary hippocampal cultures prepared from MKP-2(-/-) mice with no effect on EPSC amplitude observed. An increase in synapse number was evident in primary hippocampal cultures, which may account for the increase in sEPSC frequency. In addition, no change in ERK activity was detected in both brain tissue and primary hippocampal cultures, suggesting that the effects of MKP-2 deletion were MAPK independent. Consistent with these alterations in hippocampal function, MKP-2(-/-) mice show deficits in spatial reference and working memory when investigated using the Morris water maze. These data show that MKP-2 plays a role in regulating hippocampal function and that this effect may be independent of MAPK signaling.
Collapse
|
42
|
Krueger JN, Moore SJ, Parent R, McKinney BC, Lee A, Murphy GG. A novel mouse model of the aged brain: Over-expression of the L-type voltage-gated calcium channel Ca V1.3. Behav Brain Res 2016; 322:241-249. [PMID: 27368417 DOI: 10.1016/j.bbr.2016.06.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 06/09/2016] [Accepted: 06/27/2016] [Indexed: 11/16/2022]
Abstract
The aged population is growing rapidly, which has sparked tremendous interest in elucidating mechanisms of aging in both the body and the brain. Animal models have become an indispensable tool in biomedical science, but because of the cost and extended timeframe associated with aging animals to appropriate time points, studies that rely on using aged animals are often not feasible. Somewhat surprisingly, there are relatively few animal models that have been specifically engineered to mimic physiological changes known to occur during "normal" aging. Developing transgenic animal models that faithfully mimic key aspects of aging would likely be of great utility in studying both age-related deficits in the absence of overt pathology as well as an adjunct for transgenic models of diseases where aging is a primary risk factor. In particular, there are several alterations in the aged brain that are amenable to being modeled genetically. We have focused on one key aspect that has been repeatedly demonstrated in aged animals - an increase in the L-type voltage-gated calcium channel CaV1.3. Here we present a novel transgenic mouse line in which expression of CaV1.3 is increased by approximately 50% in the forebrain of young mice. These mice do not display any overt physical or non-cognitive deficits, exhibiting normal exploratory behavior, motor function, and affective-like responses, suggesting that these mice can be successfully deployed to assess the impact of an "aged brain" in a variety of conditions.
Collapse
Affiliation(s)
- Jamie N Krueger
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Shannon J Moore
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Rachel Parent
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brandon C McKinney
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Amy Lee
- Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, United States
| | - Geoffrey G Murphy
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States; Department of Physiology, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
43
|
Ryu HH, Lee YS. Cell type-specific roles of RAS-MAPK signaling in learning and memory: Implications in neurodevelopmental disorders. Neurobiol Learn Mem 2016; 135:13-21. [PMID: 27296701 DOI: 10.1016/j.nlm.2016.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/28/2016] [Accepted: 06/09/2016] [Indexed: 01/17/2023]
Abstract
The RAS-mitogen-activated protein kinase (MAPK) signaling pathway plays critical roles in brain function, including learning and memory. Mutations of molecules in the RAS-MAPK pathway are associated with a group of disorders called RASopathies, which include Noonan syndrome, neurofibromatosis type 1, Costello syndrome, Noonan syndrome with multiple lentigines, Legius syndrome, and cardio-facio-cutaneous syndrome. RASopathies share certain clinical symptoms, including craniofacial abnormalities, heart defects, delayed growth, and cognitive deficits such as learning disabilities, while each individual syndrome also displays unique phenotypes. Recent studies using mouse models of RASopathies showed that each disorder may have a distinct molecular and cellular etiology depending on the cellular specificity of the mutated molecules. Here, we review the cell-type specific roles of the regulators of the RAS-MAPK pathway in cognitive function (learning and memory) and their contribution to the development of RASopathies. We also discussed recent technical advances in analyzing cell type-specific transcriptomes and proteomes in the nervous system. Understanding specific mechanisms for these similar but distinct disorders would facilitate the development of mechanism-based individualized treatment for RASopathies.
Collapse
Affiliation(s)
- Hyun-Hee Ryu
- Department of Life Science, College of Natural Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, South Korea; Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Yong-Seok Lee
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.
| |
Collapse
|
44
|
Park JC, Jeong WJ, Kim MY, Min D, Choi KY. Retinoic-acid-mediated HRas stabilization induces neuronal differentiation of neural stem cells during brain development. J Cell Sci 2016; 129:2997-3007. [PMID: 27185863 DOI: 10.1242/jcs.184366] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/06/2016] [Indexed: 01/15/2023] Open
Abstract
Ras signaling is tightly regulated during neural stem cell (NSC) differentiation, and defects in this pathway result in aberrant brain development. However, the mechanism regulating Ras signaling during NSC differentiation was unknown. Here, we show that stabilized HRas specifically induces neuronal differentiation of NSCs. Lentivirus-mediated HRas overexpression and knockdown resulted in stimulation and inhibition, respectively, of NSC differentiation into neuron in the ex vivo embryo. Retinoic acid, an active metabolite of vitamin A, promoted neuronal differentiation of NSCs by stabilizing HRas, and HRas knockdown blocked the retinoic acid effect. Vitamin-A-deficient mice displayed abnormal brain development with reduced HRas levels and a reduced thickness of the postmitotic region containing differentiated neurons. All of these abnormal phenotypes were rescued with the restoration of HRas protein levels achieved upon feeding with a retinoic-acid-supplemented diet. In summary, this study shows that retinoic acid stabilizes HRas protein during neurogenesis, and that this is required for NSC differentiation into neurons and murine brain development.
Collapse
Affiliation(s)
- Jong-Chan Park
- Translational Research Center for Protein Function Control, Yonsei University, Seoul 120-749, Korea Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Woo-Jeong Jeong
- Translational Research Center for Protein Function Control, Yonsei University, Seoul 120-749, Korea Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - Mi-Yeon Kim
- Translational Research Center for Protein Function Control, Yonsei University, Seoul 120-749, Korea Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| | - DoSik Min
- Translational Research Center for Protein Function Control, Yonsei University, Seoul 120-749, Korea Department of Molecular Biology, College of Natural Science, Pusan National University, Pusan 609-735, Korea
| | - Kang-Yell Choi
- Translational Research Center for Protein Function Control, Yonsei University, Seoul 120-749, Korea Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Korea
| |
Collapse
|
45
|
Yamanaka M, Tian Z, Darvish-Ghane S, Zhuo M. Pre-LTP requires extracellular signal-regulated kinase in the ACC. Mol Pain 2016; 12:12/0/1744806916647373. [PMID: 27178245 PMCID: PMC4956388 DOI: 10.1177/1744806916647373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023] Open
Abstract
The extracellular signal-regulated kinase is an important protein kinase for cortical plasticity. Long-term potentiation in the anterior cingulate cortex is believed to play important roles in chronic pain, fear, and anxiety. Previous studies of extracellular signal-regulated kinase are mainly focused on postsynaptic form of long-term potentiation (post-long-term potentiation). Little is known about the relationship between extracellular signal-regulated kinase and presynaptic long-term potentiation (pre-long-term potentiation) in cortical synapses. In this study, we examined whether pre-long-term potentiation in the anterior cingulate cortex requires the activation of presynaptic extracellular signal-regulated kinase. We found that p42/p44 mitogen-activated protein kinase inhibitors, PD98059 and U0126, suppressed the induction of pre-long-term potentiation. By contrast, these inhibitors did not affect the maintenance of pre-long-term potentiation. Using pharmacological inhibitors, we found that pre-long-term potentiation recorded for 1 h did not require transcriptional or translational processes. Our results strongly indicate that the activation of presynaptic extracellular signal-regulated kinase is required for the induction of pre-long-term potentiation, and this involvement may explain the contribution of extracellular signal-regulated kinase to mood disorders.
Collapse
Affiliation(s)
- Manabu Yamanaka
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Shanxi, China Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Zhen Tian
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Shanxi, China Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Soroush Darvish-Ghane
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Shanxi, China Department of Physiology, Faculty of Medicine, University of Toronto, Ontario, Canada
| |
Collapse
|
46
|
Schmitz SK, King C, Kortleven C, Huson V, Kroon T, Kevenaar JT, Schut D, Saarloos I, Hoetjes JP, de Wit H, Stiedl O, Spijker S, Li KW, Mansvelder HD, Smit AB, Cornelisse LN, Verhage M, Toonen RF. Presynaptic inhibition upon CB1 or mGlu2/3 receptor activation requires ERK/MAPK phosphorylation of Munc18-1. EMBO J 2016; 35:1236-50. [PMID: 27056679 DOI: 10.15252/embj.201592244] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 03/02/2016] [Indexed: 01/22/2023] Open
Abstract
Presynaptic cannabinoid (CB1R) and metabotropic glutamate receptors (mGluR2/3) regulate synaptic strength by inhibiting secretion. Here, we reveal a presynaptic inhibitory pathway activated by extracellular signal-regulated kinase (ERK) that mediates CB1R- and mGluR2/3-induced secretion inhibition. This pathway is triggered by a variety of events, from foot shock-induced stress to intense neuronal activity, and induces phosphorylation of the presynaptic protein Munc18-1. Mimicking constitutive phosphorylation of Munc18-1 results in a drastic decrease in synaptic transmission. ERK-mediated phosphorylation of Munc18-1 ultimately leads to degradation by the ubiquitin-proteasome system. Conversely, preventing ERK-dependent Munc18-1 phosphorylation increases synaptic strength. CB1R- and mGluR2/3-induced synaptic inhibition and depolarization-induced suppression of excitation (DSE) are reduced upon ERK/MEK pathway inhibition and further reduced when ERK-dependent Munc18-1 phosphorylation is blocked. Thus, ERK-dependent Munc18-1 phosphorylation provides a major negative feedback loop to control synaptic strength upon activation of presynaptic receptors and during intense neuronal activity.
Collapse
Affiliation(s)
- Sabine K Schmitz
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Cillian King
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Christian Kortleven
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Vincent Huson
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Tim Kroon
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Josta T Kevenaar
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Desiree Schut
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Ingrid Saarloos
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Joost P Hoetjes
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Heidi de Wit
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Oliver Stiedl
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular & Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Lennart Niels Cornelisse
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| | - Ruud F Toonen
- Department of Functional Genomics and Clinical Genetics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit (VU) and VU Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Wu J, Chen H, Li H, Tang Y, Yang L, Cao S, Qin D. Antidepressant Potential of Chlorogenic Acid-Enriched Extract from Eucommia ulmoides Oliver Bark with Neuron Protection and Promotion of Serotonin Release through Enhancing Synapsin I Expression. Molecules 2016; 21:260. [PMID: 26927040 PMCID: PMC6274286 DOI: 10.3390/molecules21030260] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022] Open
Abstract
Eucommia ulmoides Oliver (E. ulmoides) is a traditional Chinese medicine with many beneficial effects, used as a tonic medicine in China and other countries. Chlorogenic acid (CGA) is an important compound in E. ulmoides with neuroprotective, cognition improvement and other pharmacological effects. However, it is unknown whether chlorogenic acid-enriched Eucommia ulmoides Oliver bark has antidepressant potential through neuron protection, serotonin release promotion and penetration of blood-cerebrospinal fluid barrier. In the present study, we demonstrated that CGA could stimulate axon and dendrite growth and promote serotonin release through enhancing synapsin I expression in the cells of fetal rat raphe neurons in vitro. More importantly, CGA-enriched extract of E. ulmoides (EUWE) at 200 and 400 mg/kg/day orally administered for 7 days showed antidepressant-like effects in the tail suspension test of KM mice. Furthermore, we also found CGA could be detected in the the cerebrospinal fluid of the rats orally treated with EUWE and reach the level of pharmacological effect for neuroprotection by UHPLC-ESI-MS/MS. The findings indicate CGA is able to cross the blood-cerebrospinal fluid barrier to exhibit its neuron protection and promotion of serotonin release through enhancing synapsin I expression. This is the first report of the effect of CGA on promoting 5-HT release through enhancing synapsin I expression and CGA-enriched EUWE has antidepressant-like effect in vivo. EUWE may be developed as the natural drugs for the treatment of depression.
Collapse
Affiliation(s)
- Jianming Wu
- Department of Pharmacology, School of Pharmacy, Sichuan Medical University, Luzhou 86646-000, Sichuan, China.
| | - Haixia Chen
- Department of Pharmacology, School of Pharmacy, Sichuan Medical University, Luzhou 86646-000, Sichuan, China.
| | - Hua Li
- Department of Pharmacology, School of Pharmacy, Sichuan Medical University, Luzhou 86646-000, Sichuan, China.
| | - Yong Tang
- Department of Pharmacology, School of Pharmacy, Sichuan Medical University, Luzhou 86646-000, Sichuan, China.
| | - Le Yang
- Chengdu Analytical Applications Center, Shimadzu (China) Co. Ltd., Chengdu 86610-063, Sichuan, China.
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Sichuan Medical University, Luzhou 86646-000, Sichuan, China.
| | - Dalian Qin
- Department of Pharmacology, School of Pharmacy, Sichuan Medical University, Luzhou 86646-000, Sichuan, China.
| |
Collapse
|
48
|
Xing L, Larsen RS, Bjorklund GR, Li X, Wu Y, Philpot BD, Snider WD, Newbern JM. Layer specific and general requirements for ERK/MAPK signaling in the developing neocortex. eLife 2016; 5. [PMID: 26848828 PMCID: PMC4758957 DOI: 10.7554/elife.11123] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 01/04/2016] [Indexed: 12/11/2022] Open
Abstract
Aberrant signaling through the Raf/MEK/ERK (ERK/MAPK) pathway causes pathology in a family of neurodevelopmental disorders known as 'RASopathies' and is implicated in autism pathogenesis. Here, we have determined the functions of ERK/MAPK signaling in developing neocortical excitatory neurons. Our data reveal a critical requirement for ERK/MAPK signaling in the morphological development and survival of large Ctip2+ neurons in layer 5. Loss of Map2k1/2 (Mek1/2) led to deficits in corticospinal tract formation and subsequent corticospinal neuron apoptosis. ERK/MAPK hyperactivation also led to reduced corticospinal axon elongation, but was associated with enhanced arborization. ERK/MAPK signaling was dispensable for axonal outgrowth of layer 2/3 callosal neurons. However, Map2k1/2 deletion led to reduced expression of Arc and enhanced intrinsic excitability in both layers 2/3 and 5, in addition to imbalanced synaptic excitation and inhibition. These data demonstrate selective requirements for ERK/MAPK signaling in layer 5 circuit development and general effects on cortical pyramidal neuron excitability. DOI:http://dx.doi.org/10.7554/eLife.11123.001 In the nervous system, cells called neurons form networks that relay information in the form of electrical signals around the brain and the rest of the body. Typically, an electrical signal travels from branch-like structures at one end of the cell, through the cell body and then along a long fiber called an axon to reach junctions with another neurons. The connections between neurons start to form as the nervous system develops in the embryo, and any errors or delays in this process can cause severe neurological disorders and intellectual disabilities. For example, genetic mutations affecting a communication system within cells known as the ERK/MAPK pathway can lead to a family of syndromes called the “RASopathies”. Abnormalities in this pathway may also contribute to certain types of autism. However, it is not clear how alterations to the ERK/MAPK pathway cause these conditions. Xing et al. investigated whether ERK/MAPK signaling regulates the formation of connections between neurons and the activity of neurons in mouse brains. The experiments showed that the growth of axons that extend from an area of the brain called the cerebral cortex towards the spinal cord are particularly sensitive to changes in the level of signaling through the ERK/MAPK pathway. On the other hand, inhibiting the pathway has relatively little effect on the growth of axons within the cerebral cortex. Further experiments showed that many neurons in the cerebral cortex require the ERK/MAPK pathway to activate genes that alter neuronal activity and the strength of the connections between neurons. Xing et al.’s findings suggest that defects in the connections between the cerebral cortex and different regions of the nervous system may contribute to the symptoms observed in patients with conditions linked to alterations in ERK/MAPK activity. Future studies will focus on understanding the molecular mechanisms by which ERK/MAPK pathway influences the organization and activity of neuron circuits during the development of the nervous system. DOI:http://dx.doi.org/10.7554/eLife.11123.002
Collapse
Affiliation(s)
- Lei Xing
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Rylan S Larsen
- Allen Institute for Brain Science, Seattle, United States
| | | | - Xiaoyan Li
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Yaohong Wu
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Benjamin D Philpot
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Carolina Institute for Developmental Disabilities, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - William D Snider
- University of North Carolina Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, United States.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Carolina Institute for Developmental Disabilities, The University of North Carolina School of Medicine, Chapel Hill, United States
| | - Jason M Newbern
- School of Life Sciences, Arizona State University, Tempe, United States
| |
Collapse
|
49
|
Greenwood MP, Greenwood M, Mecawi AS, Antunes-Rodrigues J, Paton JFR, Murphy D. Rasd1, a small G protein with a big role in the hypothalamic response to neuronal activation. Mol Brain 2016; 9:1. [PMID: 26739966 PMCID: PMC4704412 DOI: 10.1186/s13041-015-0182-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/23/2015] [Indexed: 12/28/2022] Open
Abstract
Background Rasd1 is a member of the Ras family of monomeric G proteins that was first identified as a dexamethasone inducible gene in the pituitary corticotroph cell line AtT20. Using microarrays we previously identified increased Rasd1 mRNA expression in the rat supraoptic nucleus (SON) and paraventricular nucleus (PVN) of the hypothalamus in response to increased plasma osmolality provoked by fluid deprivation and salt loading. RASD1 has been shown to inhibit adenylyl cyclase activity in vitro resulting in the inhibition of the cAMP-PKA-CREB signaling pathway. Therefore, we tested the hypothesis that RASD1 may inhibit cAMP stimulated gene expression in the brain. Results We show that Rasd1 is expressed in vasopressin neurons of the PVN and SON, within which mRNA levels are induced by hyperosmotic cues. Dexamethasone treatment of AtT20 cells decreased forskolin stimulation of c-Fos, Nr4a1 and phosphorylated CREB expression, effects that were mimicked by overexpression of Rasd1, and inhibited by knockdown of Rasd1. These effects were dependent upon isoprenylation, as both farnesyltransferase inhibitor FTI-277 and CAAX box deletion prevented Rasd1 inhibition of cAMP-induced gene expression. Injection of lentiviral vector into rat SON expressing Rasd1 diminished, whereas CAAX mutant increased, cAMP inducible genes in response to osmotic stress. Conclusions We have identified two mechanisms of Rasd1 induction in the hypothalamus, one by elevated glucocorticoids in response to stress, and one in response to increased plasma osmolality resulting from osmotic stress. We propose that the abundance of RASD1 in vasopressin expressing neurons, based on its inhibitory actions on CREB phosphorylation, is an important mechanism for controlling the transcriptional responses to stressors in both the PVN and SON. These effects likely occur through modulation of cAMP-PKA-CREB signaling pathway in the brain.
Collapse
Affiliation(s)
| | - Mingkwan Greenwood
- School of Clinical Sciences, University of Bristol, Bristol, BS1 3NY, UK.
| | - Andre S Mecawi
- School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil. .,Department of Physiology, University of Malaya, Kuala Lumpur, 50603, Malaysia. .,Department of Physiological Sciences, Biology Institute, Federal Rural University of Rio de Janeiro, Seropedica, Rio de Janeiro, Brazil.
| | | | - Julian F R Paton
- School of Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK.
| | - David Murphy
- School of Clinical Sciences, University of Bristol, Bristol, BS1 3NY, UK. .,Department of Physiology, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
50
|
Dileone M, Ranieri F, Florio L, Capone F, Musumeci G, Leoni C, Mordillo-Mateos L, Tartaglia M, Zampino G, Di Lazzaro V. Differential Effects of HRAS Mutation on LTP-Like Activity Induced by Different Protocols of Repetitive Transcranial Magnetic Stimulation. Brain Stimul 2016; 9:33-8. [DOI: 10.1016/j.brs.2015.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 11/30/2022] Open
|