151
|
Smith MR, Burman P, Sadahiro M, Kidd BA, Dudley JT, Morishita H. Integrative Analysis of Disease Signatures Shows Inflammation Disrupts Juvenile Experience-Dependent Cortical Plasticity. eNeuro 2016; 3:ENEURO.0240-16.2016. [PMID: 28101530 PMCID: PMC5241709 DOI: 10.1523/eneuro.0240-16.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/01/2016] [Accepted: 11/12/2016] [Indexed: 01/04/2023] Open
Abstract
Throughout childhood and adolescence, periods of heightened neuroplasticity are critical for the development of healthy brain function and behavior. Given the high prevalence of neurodevelopmental disorders, such as autism, identifying disruptors of developmental plasticity represents an essential step for developing strategies for prevention and intervention. Applying a novel computational approach that systematically assessed connections between 436 transcriptional signatures of disease and multiple signatures of neuroplasticity, we identified inflammation as a common pathological process central to a diverse set of diseases predicted to dysregulate plasticity signatures. We tested the hypothesis that inflammation disrupts developmental cortical plasticity in vivo using the mouse ocular dominance model of experience-dependent plasticity in primary visual cortex. We found that the administration of systemic lipopolysaccharide suppressed plasticity during juvenile critical period with accompanying transcriptional changes in a particular set of molecular regulators within primary visual cortex. These findings suggest that inflammation may have unrecognized adverse consequences on the postnatal developmental trajectory and indicate that treating inflammation may reduce the burden of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Milo R. Smith
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Poromendro Burman
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Masato Sadahiro
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Brian A. Kidd
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Joel T. Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Hirofumi Morishita
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
152
|
Abstract
Amblyopia is a developmental brain disorder in which vision is lost due to asymmetric or inadequate visual stimulation early in life. Although amblyopia is responsive to treatment if therapy is initiated early, treatment of older children and adults is usually unsuccessful due to closure of a window of cortical brain plasticity. Extensive basic research has been devoted to understanding modulators in shaping the visual cortex during the critical period of plasticity, and to providing potential clinical applications of neurotransmitters in the treatment of amblyopia. Current pharmacological treatments are reviewed from basic science research extending into clinical use, focusing on the acetylcholinesterase inhibitor donezepil, serotonin receptor inhibitor fluoxetine, dopamine precursors carbidopa-levodopa, and catecholamine modulator citicoline.
Collapse
Affiliation(s)
- Charlotte Gore
- a Boston Children's Hospital , Boston , Massachusetts , USA
| | - Carolyn Wu
- a Boston Children's Hospital , Boston , Massachusetts , USA
| |
Collapse
|
153
|
Cell-Autonomous Regulation of Dendritic Spine Density by PirB. eNeuro 2016; 3:eN-NWR-0089-16. [PMID: 27752542 PMCID: PMC5054304 DOI: 10.1523/eneuro.0089-16.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/17/2016] [Accepted: 09/23/2016] [Indexed: 01/27/2023] Open
Abstract
Synapse density on cortical pyramidal neurons is modulated by experience. This process is highest during developmental critical periods, when mechanisms of synaptic plasticity are fully engaged. In mouse visual cortex, the critical period for ocular dominance (OD) plasticity coincides with the developmental pruning of synapses. At this time, mice lacking paired Ig-like receptor B (PirB) have excess numbers of dendritic spines on L5 neurons; these spines persist and are thought to underlie the juvenile-like OD plasticity observed in adulthood. Here we examine whether PirB is required specifically in excitatory neurons to exert its effect on dendritic spine and synapse density during the critical period. In mice with a conditional allele of PirB (PirBfl/fl), PirB was deleted only from L2/3 cortical pyramidal neurons in vivo by timed in utero electroporation of Cre recombinase. Sparse mosaic expression of Cre produced neurons lacking PirB in a sea of wild-type neurons and glia. These neurons had significantly elevated dendritic spine density, as well as increased frequency of miniature EPSCs, suggesting that they receive a greater number of synaptic inputs relative to Cre– neighbors. The effect of cell-specific PirB deletion on dendritic spine density was not accompanied by changes in dendritic branching complexity or axonal bouton density. Together, results imply a neuron-specific, cell-autonomous action of PirB on synaptic density in L2/3 pyramidal cells of visual cortex. Moreover, they are consistent with the idea that PirB functions normally to corepress spine density and synaptic plasticity, thereby maintaining headroom for cells to encode ongoing experience-dependent structural change throughout life.
Collapse
|
154
|
Layer-specific cholinergic control of human and mouse cortical synaptic plasticity. Nat Commun 2016; 7:12826. [PMID: 27604129 PMCID: PMC5025530 DOI: 10.1038/ncomms12826] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 08/04/2016] [Indexed: 02/02/2023] Open
Abstract
Individual cortical layers have distinct roles in information processing. All layers receive cholinergic inputs from the basal forebrain (BF), which is crucial for cognition. Acetylcholinergic receptors are differentially distributed across cortical layers, and recent evidence suggests that different populations of BF cholinergic neurons may target specific prefrontal cortical (PFC) layers, raising the question of whether cholinergic control of the PFC is layer dependent. Here we address this issue and reveal dendritic mechanisms by which endogenous cholinergic modulation of synaptic plasticity is opposite in superficial and deep layers of both mouse and human neocortex. Our results show that in different cortical layers, spike timing-dependent plasticity is oppositely regulated by the activation of nicotinic acetylcholine receptors (nAChRs) either located on dendrites of principal neurons or on GABAergic interneurons. Thus, layer-specific nAChR expression allows functional layer-specific control of cortical processing and plasticity by the BF cholinergic system, which is evolutionarily conserved from mice to humans.
Collapse
|
155
|
Sugihara H, Chen N, Sur M. Cell-specific modulation of plasticity and cortical state by cholinergic inputs to the visual cortex. JOURNAL OF PHYSIOLOGY, PARIS 2016; 110:37-43. [PMID: 27840211 PMCID: PMC5769868 DOI: 10.1016/j.jphysparis.2016.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 12/18/2022]
Abstract
Acetylcholine (ACh) modulates diverse vital brain functions. Cholinergic neurons from the basal forebrain innervate a wide range of cortical areas, including the primary visual cortex (V1), and multiple cortical cell types have been found to be responsive to ACh. Here we review how different cell types contribute to different cortical functions modulated by ACh. We specifically focus on two major cortical functions: plasticity and cortical state. In layer II/III of V1, ACh acting on astrocytes and somatostatin-expressing inhibitory neurons plays critical roles in these functions. Cell type specificity of cholinergic modulation points towards the growing understanding that even diffuse neurotransmitter systems can mediate specific functions through specific cell classes and receptors.
Collapse
Affiliation(s)
- Hiroki Sugihara
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Naiyan Chen
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A(∗)STAR, Republic of Singapore
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
156
|
Lunghi C, Morrone MC, Secci J, Caputo R. Binocular Rivalry Measured 2 Hours After Occlusion Therapy Predicts the Recovery Rate of the Amblyopic Eye in Anisometropic Children. Invest Ophthalmol Vis Sci 2016; 57:1537-46. [PMID: 27046118 DOI: 10.1167/iovs.15-18419] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Recent studies on adults have shown that short-term monocular deprivation boosts the deprived eye signal in binocular rivalry, reflecting homeostatic plasticity. Here we investigate whether homeostatic plasticity is present also during occlusion therapy for moderate amblyopia. METHODS Binocular rivalry and visual acuity (using Snellen charts for children) were measured in 10 children (mean age 6.2 ± 1 years) with moderate anisometropic amblyopia before the beginning of treatment and at four intervals during occlusion therapy (2 hours, 1, 2, and 5 months). Visual stimuli were orthogonal gratings presented dichoptically through ferromagnetic goggles and children reported verbally visual rivalrous perception. Bangerter filters were applied on the spectacle lens over the best eye for occlusion therapy. RESULTS Two hours of occlusion therapy increased the nonamblyopic eye predominance over the amblyopic eye compared with pretreatment measurements, consistent with the results in adults. The boost of the nonamblyopic eye was still present after 1 month of treatment, steadily decreasing afterward to reach pretreatment levels after 2 months of continuous occlusion. Across subjects, the increase in nonamblyopic eye predominance observed after 2 hours of occlusion correlated (rho = -0.65, P = 0.04) with the visual acuity improvement of the amblyopic eye measured after 2 months of treatment. CONCLUSIONS Homeostatic plasticity operates during occlusion therapy for moderate amblyopia and the increase in nonamblyopic eye dominance observed at the beginning of treatment correlates with the amblyopic eye recovery rate. These results suggest that binocular rivalry might be used to monitor visual cortical plasticity during occlusion therapy, although further investigations on larger clinical populations are needed to validate the predictive power of the technique.
Collapse
Affiliation(s)
- Claudia Lunghi
- Department of Translational Research on New Technologies in Medicine and Surgery University of Pisa, Pisa, Italy 2Institute of Neuroscience, CNR, Pisa, Italy
| | - Maria Concetta Morrone
- Department of Translational Research on New Technologies in Medicine and Surgery University of Pisa, Pisa, Italy 3Scientific Institute Stella Maris (IRCSS), Calambrone (Pisa), Italy
| | - Jacopo Secci
- Clinical Ophthalmology Unit, Anna Meyer Children's University Hospital, Florence, Italy
| | - Roberto Caputo
- Clinical Ophthalmology Unit, Anna Meyer Children's University Hospital, Florence, Italy
| |
Collapse
|
157
|
Arvaniti M, Jensen MM, Soni N, Wang H, Klein AB, Thiriet N, Pinborg LH, Muldoon PP, Wienecke J, Imad Damaj M, Kohlmeier KA, Gondré-Lewis MC, Mikkelsen JD, Thomsen MS. Functional interaction between Lypd6 and nicotinic acetylcholine receptors. J Neurochem 2016; 138:806-20. [PMID: 27344019 PMCID: PMC5017906 DOI: 10.1111/jnc.13718] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 06/22/2016] [Accepted: 06/24/2016] [Indexed: 01/15/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) affect multiple physiological functions in the brain and their functions are modulated by regulatory proteins of the Lynx family. Here, we report for the first time a direct interaction of the Lynx protein LY6/PLAUR domain-containing 6 (Lypd6) with nAChRs in human brain extracts, identifying Lypd6 as a novel regulator of nAChR function. Using protein cross-linking and affinity purification from human temporal cortical extracts, we demonstrate that Lypd6 is a synaptically enriched membrane-bound protein that binds to multiple nAChR subtypes in the human brain. Additionally, soluble recombinant Lypd6 protein attenuates nicotine-induced hippocampal inward currents in rat brain slices and decreases nicotine-induced extracellular signal-regulated kinase phosphorylation in PC12 cells, suggesting that binding of Lypd6 is sufficient to inhibit nAChR-mediated intracellular signaling. We further show that perinatal nicotine exposure in rats (4 mg/kg/day through minipumps to dams from embryonic day 7 to post-natal day 21) significantly increases Lypd6 protein levels in the hippocampus in adulthood, which did not occur after exposure to nicotine in adulthood only. Our findings suggest that Lypd6 is a versatile inhibitor of cholinergic signaling in the brain, and that Lypd6 is dysregulated by nicotine exposure during early development. Regulatory proteins of the Lynx family modulate the function of nicotinic receptors (nAChRs). We report for the first time that the Lynx protein Lypd6 binds to nAChRs in human brain extracts, and that recombinant Lypd6 decreases nicotine-induced ERK phosphorylation and attenuates nicotine-induced hippocampal inward currents. Our findings suggest that Lypd6 is a versatile inhibitor of cholinergic signaling in the brain.
Collapse
Affiliation(s)
- Maria Arvaniti
- Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Majbrit M Jensen
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Neeraj Soni
- Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Hong Wang
- Laboratory for Neurodevelopment, Department of Anatomy, Howard University College of Medicine, Washington, District of Columbia, USA
| | - Anders B Klein
- Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Nathalie Thiriet
- Laboratory of Experimental and Clinical Neurosciences, University of Poitiers, Poitiers, France
| | - Lars H Pinborg
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark.,Epilepsy Clinic, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Pretal P Muldoon
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jacob Wienecke
- Department of Nutrition, Exercise and Sport & Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kristi A Kohlmeier
- Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Marjorie C Gondré-Lewis
- Laboratory for Neurodevelopment, Department of Anatomy, Howard University College of Medicine, Washington, District of Columbia, USA
| | - Jens D Mikkelsen
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Morten S Thomsen
- Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark. .,Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
158
|
Lyukmanova EN, Shulepko MA, Shenkarev ZO, Bychkov ML, Paramonov AS, Chugunov AO, Kulbatskii DS, Arvaniti M, Dolejsi E, Schaer T, Arseniev AS, Efremov RG, Thomsen MS, Dolezal V, Bertrand D, Dolgikh DA, Kirpichnikov MP. Secreted Isoform of Human Lynx1 (SLURP-2): Spatial Structure and Pharmacology of Interactions with Different Types of Acetylcholine Receptors. Sci Rep 2016; 6:30698. [PMID: 27485575 PMCID: PMC4971505 DOI: 10.1038/srep30698] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/06/2016] [Indexed: 11/12/2022] Open
Abstract
Human-secreted Ly-6/uPAR-related protein-2 (SLURP-2) regulates the growth and differentiation of epithelial cells. Previously, the auto/paracrine activity of SLURP-2 was considered to be mediated via its interaction with the α3β2 subtype of the nicotinic acetylcholine receptors (nAChRs). Here, we describe the structure and pharmacology of a recombinant analogue of SLURP-2. Nuclear magnetic resonance spectroscopy revealed a 'three-finger' fold of SLURP-2 with a conserved β-structural core and three protruding loops. Affinity purification using cortical extracts revealed that SLURP-2 could interact with the α3, α4, α5, α7, β2, and β4 nAChR subunits, revealing its broader pharmacological profile. SLURP-2 inhibits acetylcholine-evoked currents at α4β2 and α3β2-nAChRs (IC50 ~0.17 and >3 μM, respectively) expressed in Xenopus oocytes. In contrast, at α7-nAChRs, SLURP-2 significantly enhances acetylcholine-evoked currents at concentrations <1 μM but induces inhibition at higher concentrations. SLURP-2 allosterically interacts with human M1 and M3 muscarinic acetylcholine receptors (mAChRs) that are overexpressed in CHO cells. SLURP-2 was found to promote the proliferation of human oral keratinocytes via interactions with α3β2-nAChRs, while it inhibited cell growth via α7-nAChRs. SLURP-2/mAChRs interactions are also probably involved in the control of keratinocyte growth. Computer modeling revealed possible SLURP-2 binding to the 'classical' orthosteric agonist/antagonist binding sites at α7 and α3β2-nAChRs.
Collapse
Affiliation(s)
- E. N. Lyukmanova
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - M. A. Shulepko
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - Z. O. Shenkarev
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
- Moscow Institute of Physics and Technology, Institutskiy Pereulok 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| | - M. L. Bychkov
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - A. S. Paramonov
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - A. O. Chugunov
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - D. S. Kulbatskii
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - M. Arvaniti
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark
| | - Eva Dolejsi
- Institute of Physiology, Academy of Sciences of the Czech Republic (public research institution), Prague, 14220, Czech Republic
| | - T. Schaer
- HiQScreen Sàrl, 6 rte de Compois, 1222, Vésenaz, Geneva, Switzerland
| | - A. S. Arseniev
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Moscow Institute of Physics and Technology, Institutskiy Pereulok 9, Dolgoprudny, Moscow Region 141700, Russian Federation
| | - R. G. Efremov
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- National Research University Higher School of Economics, Myasnitskaya ul. 20, 101000 Moscow, Russia
| | - M. S. Thomsen
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 160, DK-2100 Copenhagen, Denmark
| | - V. Dolezal
- Institute of Physiology, Academy of Sciences of the Czech Republic (public research institution), Prague, 14220, Czech Republic
| | - D. Bertrand
- HiQScreen Sàrl, 6 rte de Compois, 1222, Vésenaz, Geneva, Switzerland
| | - D. A. Dolgikh
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| | - M. P. Kirpichnikov
- Lomonosov Moscow State University, Leninskie Gori 1, Moscow 119234, Russian Federation
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya Street 16/10, Moscow 117997, Russian Federation
| |
Collapse
|
159
|
Thomsen MS, Arvaniti M, Jensen MM, Shulepko MA, Dolgikh DA, Pinborg LH, Härtig W, Lyukmanova EN, Mikkelsen JD. Lynx1 and Aβ1-42 bind competitively to multiple nicotinic acetylcholine receptor subtypes. Neurobiol Aging 2016; 46:13-21. [PMID: 27460145 DOI: 10.1016/j.neurobiolaging.2016.06.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 01/13/2023]
Abstract
Lynx1 regulates synaptic plasticity in the brain by regulating nicotinic acetylcholine receptors (nAChRs). It is not known to which extent Lynx1 can bind to endogenous nAChR subunits in the brain or how this interaction is affected by Alzheimer's disease pathology. We apply affinity purification to demonstrate that a water-soluble variant of human Lynx1 (Ws-Lynx1) isolates α3, α4, α5, α6, α7, β2, and β4 nAChR subunits from human and rat cortical extracts, and rat midbrain and olfactory bulb extracts, suggesting that Lynx1 forms complexes with multiple nAChR subtypes in the human and rodent brain. Incubation with Ws-Lynx1 decreases nicotine-mediated extracellular signal-regulated kinase phosphorylation in PC12 cells and striatal neurons, indicating that binding of Ws-Lynx1 is sufficient to inhibit signaling downstream of nAChRs. The effect of nicotine in PC12 cells is independent of α7 or α4β2 nAChRs, suggesting that Lynx1 can affect the function of native non-α7, non-α4β2 nAChR subtypes. We further show that Lynx1 and oligomeric β-amyloid1-42 compete for binding to several nAChR subunits, that Ws-Lynx1 prevents β-amyloid1-42-induced cytotoxicity in cortical neurons, and that cortical Lynx1 levels are decreased in a transgenic mouse model with concomitant β-amyloid and tau pathology. Our data suggest that Lynx1 binds to multiple nAChR subtypes in the brain and that this interaction might have functional and pathophysiological implications in relation to Alzheimer's disease.
Collapse
Affiliation(s)
- Morten S Thomsen
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark; Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Maria Arvaniti
- Department of Drug Design & Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Majbrit M Jensen
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Mikhail A Shulepko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry A Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| | - Lars H Pinborg
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark; Epilepsy Clinic, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Ekaterina N Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia; Lomonosov Moscow State University, Moscow, Russia
| | - Jens D Mikkelsen
- Neurobiology Research Unit, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
160
|
Scheyltjens I, Arckens L. The Current Status of Somatostatin-Interneurons in Inhibitory Control of Brain Function and Plasticity. Neural Plast 2016; 2016:8723623. [PMID: 27403348 PMCID: PMC4923604 DOI: 10.1155/2016/8723623] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/12/2016] [Indexed: 12/05/2022] Open
Abstract
The mammalian neocortex contains many distinct inhibitory neuronal populations to balance excitatory neurotransmission. A correct excitation/inhibition equilibrium is crucial for normal brain development, functioning, and controlling lifelong cortical plasticity. Knowledge about how the inhibitory network contributes to brain plasticity however remains incomplete. Somatostatin- (SST-) interneurons constitute a large neocortical subpopulation of interneurons, next to parvalbumin- (PV-) and vasoactive intestinal peptide- (VIP-) interneurons. Unlike the extensively studied PV-interneurons, acknowledged as key components in guiding ocular dominance plasticity, the contribution of SST-interneurons is less understood. Nevertheless, SST-interneurons are ideally situated within cortical networks to integrate unimodal or cross-modal sensory information processing and therefore likely to be important mediators of experience-dependent plasticity. The lack of knowledge on SST-interneurons partially relates to the wide variety of distinct subpopulations present in the sensory neocortex. This review informs on those SST-subpopulations hitherto described based on anatomical, molecular, or electrophysiological characteristics and whose functional roles can be attributed based on specific cortical wiring patterns. A possible role for these subpopulations in experience-dependent plasticity will be discussed, emphasizing on learning-induced plasticity and on unimodal and cross-modal plasticity upon sensory loss. This knowledge will ultimately contribute to guide brain plasticity into well-defined directions to restore sensory function and promote lifelong learning.
Collapse
Affiliation(s)
- Isabelle Scheyltjens
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, 3000 Leuven, Belgium
| | - Lutgarde Arckens
- Laboratory of Neuroplasticity and Neuroproteomics, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
161
|
Embryonic interneurons from the medial, but not the caudal ganglionic eminence trigger ocular dominance plasticity in adult mice. Brain Struct Funct 2016; 222:539-547. [PMID: 27165433 DOI: 10.1007/s00429-016-1232-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
Abstract
The maturation of cortical inhibition provided by parvalbumin-containing basket cells derived from the medial ganglionic eminence (MGE) is a key event in starting the enhanced visual cortical plasticity during the critical period. Although it is generally assumed that a further increase in inhibition closes the critical period again, it was recently shown that embryonic interneurons derived from the MGE can induce an additional, artificial critical period when injected into the visual cortex of young mice. It has, however, remained open whether this effect was indeed specific for MGE-derived cells, and whether critical period-like plasticity could also be induced in fully adult animals. To clarify these issues, we injected explants from either the MGE or the caudal ganglionic eminence (CGE) into the visual cortices of fully adult mice, and performed monocular deprivation 33 days later for 4 days. Animals implanted with MGE cells, but not with CGE cells, showed marked ocular dominance plasticity. Immunohistochemistry confirmed that the injected cells from both sources migrated far in the host cortex, that most developed into neurons producing GABA, and that only cells from the MGE expressed parvalbumin. Thus, our results confirm that the plasticity-inducing effect of embryonic interneurons is specific for cells from the MGE, and is independent of the host animal's age.
Collapse
|
162
|
Faure G, Shelukhina IV, Porowinska D, Shulepko MA, Lyukmanova EN, Dolgikh DA, Spirova EN, Kasheverov IE, Utkin YN, Corringer JP, Tsetlin VI. Interaction of three-finger proteins from snake venoms and from mammalian brain with the cys-loop receptors and their models. DOKL BIOCHEM BIOPHYS 2016; 468:193-196. [PMID: 27417718 DOI: 10.1134/s1607672916030091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Indexed: 05/14/2025]
Abstract
With the use of surface plasmon resonance (SPR) it was shown that ws-Lynx1, a water-soluble analog of the three-finger membrane-bound protein Lynx1, that modulates the activity of brain nicotinic acetylcholine receptors (nAChRs), interacts with the acetylcholine-binding protein (AChBP) with high affinity, K D = 62 nM. This result agrees with the earlier demonstrated competition of ws-Lynx1 with radioiodinated α-bungarotoxin for binding to AChBP. For the first time it was shown that ws-Lynx1 binds to GLIC, prokaryotic Cys-loop receptor (K D = 1.3 μM). On the contrary, SPR revealed that α-cobratoxin, a three-finger protein from cobra venom, does not bind to GLIC. Obtained results indicate that SPR is a promising method for analysis of topography of ws-Lynx1 binding sites using its mutants and those of AChBP and GLIC.
Collapse
Affiliation(s)
- G Faure
- Pasteur Institute, Paris, France.
| | - I V Shelukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - D Porowinska
- Pasteur Institute, Paris, France
- Biochemistry Department, Nicolaus Copernicus University, Torun, Poland
| | - M A Shulepko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - E N Lyukmanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - D A Dolgikh
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - E N Spirova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - I E Kasheverov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - Yu N Utkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | | | - V I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| |
Collapse
|
163
|
Organization, evolution and functions of the human and mouse Ly6/uPAR family genes. Hum Genomics 2016; 10:10. [PMID: 27098205 PMCID: PMC4839075 DOI: 10.1186/s40246-016-0074-2] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/14/2016] [Indexed: 01/08/2023] Open
Abstract
Members of the lymphocyte antigen-6 (Ly6)/urokinase-type plasminogen activator receptor (uPAR) superfamily of proteins are cysteine-rich proteins characterized by a distinct disulfide bridge pattern that creates the three-finger Ly6/uPAR (LU) domain. Although the Ly6/uPAR family proteins share a common structure, their expression patterns and functions vary. To date, 35 human and 61 mouse Ly6/uPAR family members have been identified. Based on their subcellular localization, these proteins are further classified as GPI-anchored on the cell membrane, or secreted. The genes encoding Ly6/uPAR family proteins are conserved across different species and are clustered in syntenic regions on human chromosomes 8, 19, 6 and 11, and mouse Chromosomes 15, 7, 17, and 9, respectively. Here, we review the human and mouse Ly6/uPAR family gene and protein structure and genomic organization, expression, functions, and evolution, and introduce new names for novel family members.
Collapse
|
164
|
Adelson JD, Sapp RW, Brott BK, Lee H, Miyamichi K, Luo L, Cheng S, Djurisic M, Shatz CJ. Developmental Sculpting of Intracortical Circuits by MHC Class I H2-Db and H2-Kb. Cereb Cortex 2016; 26:1453-1463. [PMID: 25316337 PMCID: PMC4785944 DOI: 10.1093/cercor/bhu243] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Synapse pruning is an activity-regulated process needed for proper circuit sculpting in the developing brain. Major histocompatibility class I (MHCI) molecules are regulated by activity, but little is known about their role in the development of connectivity in cortex. Here we show that protein for 2 MHCI molecules H2-Kb and H2-Db is associated with synapses in the visual cortex. Pyramidal neurons in mice lacking H2-Kb and H2-Db (KbDb KO) have more extensive cortical connectivity than normal. Modified rabies virus tracing was used to monitor the extent of pyramidal cell connectivity: Horizontal connectivity is greater in the visual cortex of KbDb KO mice. Basal dendrites of L2/3 pyramids, where many horizontal connections terminate, are more highly branched and have elevated spine density in the KO. Furthermore, the density of axonal boutons is elevated within L2/3 of mutant mice. These increases are accompanied by elevated miniature excitatory postsynaptic current frequency, consistent with an increase in functional synapses. This functional and anatomical increase in intracortical connectivity is also associated with enhanced ocular dominance plasticity that persists into adulthood. Thus, these MHCI proteins regulate sculpting of local cortical circuits and in their absence, the excess connectivity can function as a substrate for cortical plasticity throughout life.
Collapse
Affiliation(s)
| | | | | | - Hanmi Lee
- Departments of Biology and Neurobiology and Bio-X
| | | | - Liqun Luo
- Department of Biology, Stanford University, Stanford, CA94305, USA
| | - Sarah Cheng
- Departments of Biology and Neurobiology and Bio-X
| | | | | |
Collapse
|
165
|
Maconachie GDE, Gottlob I. The challenges of amblyopia treatment. Biomed J 2016; 38:510-6. [PMID: 27013450 PMCID: PMC6138377 DOI: 10.1016/j.bj.2015.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/08/2015] [Indexed: 11/22/2022] Open
Abstract
The treatment of amblyopia, particularly anisometropic (difference in refractive correction) and/or strabismic (turn of one eye) amblyopia has long been a challenge for many clinicians. Achieving optimum outcomes, where the amblyopic eye reaches a visual acuity similar to the fellow eye, is often impossible in many patients. Part of this challenge has resulted from a previous lack of scientific evidence for amblyopia treatment that was highlight by a systematic review by Snowdon et al. in 1998. Since this review, a number of publications have revealed new findings in the treatment of amblyopia. This includes the finding that less intensive occlusion treatments can be successful in treating amblyopia. A relationship between adherence to treatment and visual acuity has also been established and has been shown to be influenced by the use of intervention material. In addition, there is growing evidence of that a period of glasses wearing only can significantly improve visual acuity alone without any other modes of treatment. This review article reports findings since the Snowdon's report.
Collapse
Affiliation(s)
- Gail D E Maconachie
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, RKSCB, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Irene Gottlob
- Ulverscroft Eye Unit, Department of Neuroscience, Psychology and Behaviour, University of Leicester, RKSCB, Leicester Royal Infirmary, Leicester LE2 7LX, UK.
| |
Collapse
|
166
|
Wu M, Liu CZ, Joiner WJ. Structural Analysis and Deletion Mutagenesis Define Regions of QUIVER/SLEEPLESS that Are Responsible for Interactions with Shaker-Type Potassium Channels and Nicotinic Acetylcholine Receptors. PLoS One 2016; 11:e0148215. [PMID: 26828958 PMCID: PMC4735452 DOI: 10.1371/journal.pone.0148215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 01/13/2016] [Indexed: 12/16/2022] Open
Abstract
Ly6 proteins are endogenous prototoxins found in most animals. They show striking structural and functional parallels to snake α-neurotoxins, including regulation of ion channels and cholinergic signaling. However, the structural contributions of Ly6 proteins to regulation of effector molecules is poorly understood. This question is particularly relevant to the Ly6 protein QUIVER/SLEEPLESS (QVR/SSS), which has previously been shown to suppress excitability and synaptic transmission by upregulating potassium (K) channels and downregulating nicotinic acetylcholine receptors (nAChRs) in wake-promoting neurons to facilitate sleep in Drosophila. Using deletion mutagenesis, co-immunoprecipitations, ion flux assays, surface labeling and confocal microscopy, we demonstrate that only loop 2 is required for many of the previously described properties of SSS in transfected cells, including interactions with K channels and nAChRs. Collectively our data suggest that QVR/SSS, and by extension perhaps other Ly6 proteins, target effector molecules using limited protein motifs. Mapping these motifs may be useful in rational design of drugs that mimic or suppress Ly6-effector interactions to modulate nervous system function.
Collapse
Affiliation(s)
- Meilin Wu
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America
| | - Clifford Z Liu
- UCSD undergraduate program, Marshall College, University of California San Diego, La Jolla, California, United States of America
| | - William J Joiner
- Department of Pharmacology, University of California San Diego, La Jolla, California, United States of America.,Center for Circadian Biology, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
167
|
Abstract
UNLABELLED Experience-dependent cortical plasticity declines with age. At the molecular level, experience-dependent proteolytic activity of tissue plasminogen activator (tPA) becomes restricted in the adult brain if mice are raised in standard cages. Understanding the mechanism for the loss of permissive proteolytic activity is therefore a key link for improving function in adult brains. Using the mouse primary visual cortex (V1) as a model, we demonstrate that tPA activity in V1 can be unmasked following 4 d of monocular deprivation when the mice older than 2 months are raised in standard cages by the genetic removal of Lynx1, a negative regulator of adult plasticity. This was also associated with the reduction of stubby and thin spine density and enhancement of ocular dominance shift in adult V1 of Lynx1 knock-out (KO) mice. These structural and functional changes were tPA-dependent because genetic removal of tPA in Lynx1 KO mice can block the monocular deprivation-dependent reduction of dendritic spine density, whereas both genetic and adult specific inhibition of tPA activity can ablate the ocular dominance shift in Lynx1 KO mice. Our work demonstrates that the adult brain has an intrinsic potential for experience-dependent elevation of proteolytic activity to express juvenile-like structural and functional changes but is effectively limited by Lynx1 if mice are raised in standard cages. Insights into the Lynx1-tPA plasticity mechanism may provide novel therapeutic targets for adult brain disorders. SIGNIFICANCE STATEMENT Experience-dependent proteolytic activity of tissue plasminogen activator (tPA) becomes restricted in the adult brain in correlation with the decline in cortical plasticity when mice are raised in standard cages. We demonstrated that removal of Lynx1, one of negative regulators of plasticity, unmasks experience-dependent tPA elevation in visual cortex of adult mice reared in standard cages. This proteolytic elevation facilitated dendritic spine reduction and ocular dominance plasticity in adult visual cortex. This is the first demonstration of adult brain to retain the intrinsic capacity to elevate tPA in an experience-dependent manner but is effectively limited by Lynx1. tPA-Lynx1 may potentially be a new candidate mechanism for interventions that were shown to activate plasticity in adult brain.
Collapse
|
168
|
Lennartsson A, Arner E, Fagiolini M, Saxena A, Andersson R, Takahashi H, Noro Y, Sng J, Sandelin A, Hensch TK, Carninci P. Remodeling of retrotransposon elements during epigenetic induction of adult visual cortical plasticity by HDAC inhibitors. Epigenetics Chromatin 2015; 8:55. [PMID: 26673794 PMCID: PMC4678690 DOI: 10.1186/s13072-015-0043-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/09/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The capacity for plasticity in the adult brain is limited by the anatomical traces laid down during early postnatal life. Removing certain molecular brakes, such as histone deacetylases (HDACs), has proven to be effective in recapitulating juvenile plasticity in the mature visual cortex (V1). We investigated the chromatin structure and transcriptional control by genome-wide sequencing of DNase I hypersensitive sites (DHSS) and cap analysis of gene expression (CAGE) libraries after HDAC inhibition by valproic acid (VPA) in adult V1. RESULTS We found that VPA reliably reactivates the critical period plasticity and induces a dramatic change of chromatin organization in V1 yielding significantly greater accessibility distant from promoters, including at enhancer regions. VPA also induces nucleosome eviction specifically from retrotransposon (in particular SINE) elements. The transiently accessible SINE elements overlap with transcription factor-binding sites of the Fox family. Mapping of transcription start site activity using CAGE revealed transcription of epigenetic and neural plasticity-regulating genes following VPA treatment, which may help to re-program the genomic landscape and reactivate plasticity in the adult cortex. CONCLUSIONS Treatment with HDAC inhibitors increases accessibility to enhancers and repetitive elements underlying brain-specific gene expression and reactivation of visual cortical plasticity.
Collapse
Affiliation(s)
- Andreas Lennartsson
- />Department of Biosciences and Nutrition, NOVUM, Karolinska Institutet, Stockholm, Sweden
- />Genome Science Lab, RIKEN, Hirosawa, Wako-shi, Saitama 351-0198 Japan
| | - Erik Arner
- />Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Institute, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Michela Fagiolini
- />Lab for Neuronal Circuit Development, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198 Japan
- />F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Alka Saxena
- />Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Institute, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Robin Andersson
- />Department of Biology and BRIC, The Bioinformatics Centre, University of Copenhagen, Copenhagen, Denmark
| | - Hazuki Takahashi
- />Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Institute, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Yukihiko Noro
- />Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Institute, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Judy Sng
- />Lab for Neuronal Circuit Development, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198 Japan
- />F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115 USA
- />Department of Pharmacology, National University of Singapore, 10 Medical Drive 05-34, Singapore, Singapore
| | - Albin Sandelin
- />Department of Biology and BRIC, The Bioinformatics Centre, University of Copenhagen, Copenhagen, Denmark
| | - Takao K. Hensch
- />Lab for Neuronal Circuit Development, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-shi, Saitama, 351-0198 Japan
- />F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115 USA
- />Department of Molecular and Cellular Biology, Center for Brain Science, Harvard University, 52 Oxford Street, Cambridge, MA 02138 USA
| | - Piero Carninci
- />Division of Genomic Technologies, RIKEN Center for Life Science Technologies, RIKEN Yokohama Institute, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| |
Collapse
|
169
|
Beshara S, Beston BR, Pinto JGA, Murphy KM. Effects of Fluoxetine and Visual Experience on Glutamatergic and GABAergic Synaptic Proteins in Adult Rat Visual Cortex. eNeuro 2015; 2:ENEURO.0126-15.2015. [PMID: 26730408 PMCID: PMC4698542 DOI: 10.1523/eneuro.0126-15.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 01/12/2023] Open
Abstract
Fluoxetine has emerged as a novel treatment for persistent amblyopia because in adult animals it reinstates critical period-like ocular dominance plasticity and promotes recovery of visual acuity. Translation of these results from animal models to the clinic, however, has been challenging because of the lack of understanding of how this selective serotonin reuptake inhibitor affects glutamatergic and GABAergic synaptic mechanisms that are essential for experience-dependent plasticity. An appealing hypothesis is that fluoxetine recreates a critical period (CP)-like state by shifting synaptic mechanisms to be more juvenile. To test this we studied the effect of fluoxetine treatment in adult rats, alone or in combination with visual deprivation [monocular deprivation (MD)], on a set of highly conserved presynaptic and postsynaptic proteins (synapsin, synaptophysin, VGLUT1, VGAT, PSD-95, gephyrin, GluN1, GluA2, GluN2B, GluN2A, GABAAα1, GABAAα3). We did not find evidence that fluoxetine shifted the protein amounts or balances to a CP-like state. Instead, it drove the balances in favor of the more mature subunits (GluN2A, GABAAα1). In addition, when fluoxetine was paired with MD it created a neuroprotective-like environment by normalizing the glutamatergic gain found in adult MDs. Together, our results suggest that fluoxetine treatment creates a novel synaptic environment dominated by GluN2A- and GABAAα1-dependent plasticity.
Collapse
Affiliation(s)
- Simon Beshara
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Brett R. Beston
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, Ontario L8S 4K1, Canada
- Department of Psychology, Neuroscience & Behavior, McMaster University, Hamilton, Ontario L8S 4K1, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, L5L 1C6, ON
| | - Joshua G. A. Pinto
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, Ontario L8S 4K1, Canada
- Health Care Investment Banking, Credit Suisse AG, San Francisco, CA 94108
| | - Kathryn M. Murphy
- McMaster Integrative Neuroscience Discovery and Study (MiNDS) Program, McMaster University, Hamilton, Ontario L8S 4K1, Canada
- Department of Psychology, Neuroscience & Behavior, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
170
|
Kang JI, Huppé-Gourgues F, Vaucher E. Pharmacological Mechanisms of Cortical Enhancement Induced by the Repetitive Pairing of Visual/Cholinergic Stimulation. PLoS One 2015; 10:e0141663. [PMID: 26513575 PMCID: PMC4626033 DOI: 10.1371/journal.pone.0141663] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/12/2015] [Indexed: 11/18/2022] Open
Abstract
Repetitive visual training paired with electrical activation of cholinergic projections to the primary visual cortex (V1) induces long-term enhancement of cortical processing in response to the visual training stimulus. To better determine the receptor subtypes mediating this effect the selective pharmacological blockade of V1 nicotinic (nAChR), M1 and M2 muscarinic (mAChR) or GABAergic A (GABAAR) receptors was performed during the training session and visual evoked potentials (VEPs) were recorded before and after training. The training session consisted of the exposure of awake, adult rats to an orientation-specific 0.12 CPD grating paired with an electrical stimulation of the basal forebrain for a duration of 1 week for 10 minutes per day. Pharmacological agents were infused intracortically during this period. The post-training VEP amplitude was significantly increased compared to the pre-training values for the trained spatial frequency and to adjacent spatial frequencies up to 0.3 CPD, suggesting a long-term increase of V1 sensitivity. This increase was totally blocked by the nAChR antagonist as well as by an M2 mAChR subtype and GABAAR antagonist. Moreover, administration of the M2 mAChR antagonist also significantly decreased the amplitude of the control VEPs, suggesting a suppressive effect on cortical responsiveness. However, the M1 mAChR antagonist blocked the increase of the VEP amplitude only for the high spatial frequency (0.3 CPD), suggesting that M1 role was limited to the spread of the enhancement effect to a higher spatial frequency. More generally, all the drugs used did block the VEP increase at 0.3 CPD. Further, use of each of the aforementioned receptor antagonists blocked training-induced changes in gamma and beta band oscillations. These findings demonstrate that visual training coupled with cholinergic stimulation improved perceptual sensitivity by enhancing cortical responsiveness in V1. This enhancement is mainly mediated by nAChRs, M2 mAChRs and GABAARs. The M1 mAChR subtype appears to be involved in spreading the enhancement of V1 cortical responsiveness to adjacent neurons.
Collapse
Affiliation(s)
- Jun-Il Kang
- École d’optométrie, Université de Montréal, CP 6128 succursale centre-ville, Montréal, Qc, H3C 3J7, Canada
- Département de Neuroscience, Université de Montréal, CP 6128 succursale centre-ville, Montréal, Qc, H3C 3J7, Canada
| | - Frédéric Huppé-Gourgues
- École d’optométrie, Université de Montréal, CP 6128 succursale centre-ville, Montréal, Qc, H3C 3J7, Canada
| | - Elvire Vaucher
- École d’optométrie, Université de Montréal, CP 6128 succursale centre-ville, Montréal, Qc, H3C 3J7, Canada
- * E-mail:
| |
Collapse
|
171
|
Sale A, Berardi N. Active training for amblyopia in adult rodents. Front Behav Neurosci 2015; 9:281. [PMID: 26578911 PMCID: PMC4621305 DOI: 10.3389/fnbeh.2015.00281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/06/2015] [Indexed: 11/13/2022] Open
Abstract
Amblyopia is the most diffused form of visual function impairment affecting one eye, with a prevalence of 1–5% in the total world population. Amblyopia is usually caused by an early functional imbalance between the two eyes, deriving from anisometropia, strabismus, or congenital cataract, leading to severe deficits in visual acuity, contrast sensitivity and stereopsis. While amblyopia can be efficiently treated in children, it becomes irreversible in adults, as a result of a dramatic decline in visual cortex plasticity which occurs at the end of the critical period (CP) in the primary visual cortex. Notwithstanding this widely accepted dogma, recent evidence in animal models and in human patients have started to challenge this view, revealing a previously unsuspected possibility to enhance plasticity in the adult visual system and to achieve substantial visual function recovery. Among the new proposed intervention strategies, non invasive procedures based on environmental enrichment, physical exercise or visual perceptual learning (vPL) appear particularly promising in terms of future applicability in the clinical setting. In this survey, we will review recent literature concerning the application of these behavioral intervention strategies to the treatment of amblyopia, with a focus on possible underlying molecular and cellular mechanisms.
Collapse
Affiliation(s)
- Alessandro Sale
- Neuroscience Institute, National Research Council Pisa, Italy
| | - Nicoletta Berardi
- Neuroscience Institute, National Research Council Pisa, Italy ; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence Florence, Italy
| |
Collapse
|
172
|
Jitsuki S, Nakajima W, Takemoto K, Sano A, Tada H, Takahashi-Jitsuki A, Takahashi T. Nogo Receptor Signaling Restricts Adult Neural Plasticity by Limiting Synaptic AMPA Receptor Delivery. Cereb Cortex 2015; 26:427-439. [PMID: 26472557 PMCID: PMC4677985 DOI: 10.1093/cercor/bhv232] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Experience-dependent plasticity is limited in the adult brain, and its molecular and cellular mechanisms are poorly understood. Removal of the myelin-inhibiting signaling protein, Nogo receptor (NgR1), restores adult neural plasticity. Here we found that, in NgR1-deficient mice, whisker experience-driven synaptic α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) insertion in the barrel cortex, which is normally complete by 2 weeks after birth, lasts into adulthood. In vivo live imaging by two-photon microscopy revealed more AMPAR on the surface of spines in the adult barrel cortex of NgR1-deficient than on those of wild-type (WT) mice. Furthermore, we observed that whisker stimulation produced new spines in the adult barrel cortex of mutant but not WT mice, and that the newly synthesized spines contained surface AMPAR. These results suggest that Nogo signaling limits plasticity by restricting synaptic AMPAR delivery in coordination with anatomical plasticity.
Collapse
Affiliation(s)
- Susumu Jitsuki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Waki Nakajima
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Kiwamu Takemoto
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.,JST, PRESTO, Saitama 332-0012, Japan
| | - Akane Sano
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Hirobumi Tada
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Aoi Takahashi-Jitsuki
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Takuya Takahashi
- Department of Physiology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
173
|
Letzkus J, Wolff S, Lüthi A. Disinhibition, a Circuit Mechanism for Associative Learning and Memory. Neuron 2015; 88:264-76. [PMID: 26494276 DOI: 10.1016/j.neuron.2015.09.024] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
174
|
Morishita H, Cabungcal JH, Chen Y, Do KQ, Hensch TK. Prolonged Period of Cortical Plasticity upon Redox Dysregulation in Fast-Spiking Interneurons. Biol Psychiatry 2015; 78:396-402. [PMID: 25758057 PMCID: PMC4514575 DOI: 10.1016/j.biopsych.2014.12.026] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/29/2014] [Accepted: 12/31/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND Oxidative stress and the specific impairment of perisomatic gamma-aminobutyric acid circuits are hallmarks of the schizophrenic brain and its animal models. Proper maturation of these fast-spiking inhibitory interneurons normally defines critical periods of experience-dependent cortical plasticity. METHODS Here, we linked these processes by genetically inducing a redox dysregulation restricted to such parvalbumin-positive cells and examined the impact on critical period plasticity using the visual system as a model (3-6 mice/group). RESULTS Oxidative stress was accompanied by a significant loss of perineuronal nets, which normally enwrap mature fast-spiking cells to limit adult plasticity. Accordingly, the neocortex remained plastic even beyond the peak of its natural critical period. These effects were not seen when redox dysregulation was targeted in excitatory principal cells. CONCLUSIONS A cell-specific regulation of redox state thus balances plasticity and stability of cortical networks. Mistimed developmental trajectories of brain plasticity may underlie, in part, the pathophysiology of mental illness. Such prolonged developmental plasticity may, in turn, offer a therapeutic opportunity for cognitive interventions targeting brain plasticity in schizophrenia.
Collapse
Affiliation(s)
- Hirofumi Morishita
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Jan-Harry Cabungcal
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1008 Prilly-Lausanne, Switzerland
| | - Ying Chen
- School of Pharmacy, University of Colorado at Denver, Boulder, CO USA
| | - Kim Q. Do
- Department of Psychiatry, Center for Psychiatric Neuroscience, Centre Hospitalier Universitaire Vaudois and University of Lausanne, 1008 Prilly-Lausanne, Switzerland
| | - Takao K. Hensch
- FM Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA,Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA,Correspondence to:
| |
Collapse
|
175
|
Lee HK, Whitt JL. Cross-modal synaptic plasticity in adult primary sensory cortices. Curr Opin Neurobiol 2015; 35:119-26. [PMID: 26310109 DOI: 10.1016/j.conb.2015.08.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 12/25/2022]
Abstract
Sensory loss leads to widespread adaptation of brain circuits to allow an organism to navigate its environment with its remaining senses, which is broadly referred to as cross-modal plasticity. Such adaptation can be observed even in the primary sensory cortices, and falls into two distinct categories: recruitment of the deprived sensory cortex for processing the remaining senses, which we term 'cross-modal recruitment', and experience-dependent refinement of the spared sensory cortices referred to as 'compensatory plasticity.' Here we will review recent studies demonstrating that cortical adaptation to sensory loss involves LTP/LTD and homeostatic synaptic plasticity. Cross-modal synaptic plasticity is observed in adults, hence cross-modal sensory deprivation may be an effective way to promote plasticity in adult primary sensory cortices.
Collapse
Affiliation(s)
- Hey-Kyoung Lee
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, United States.
| | - Jessica L Whitt
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD 21218, United States
| |
Collapse
|
176
|
Boyce WT, Kobor MS. Development and the epigenome: the 'synapse' of gene-environment interplay. Dev Sci 2015; 18:1-23. [PMID: 25546559 DOI: 10.1111/desc.12282] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/14/2014] [Indexed: 12/21/2022]
Abstract
This paper argues that there is a revolution afoot in the developmental science of gene-environment interplay. We summarize, for an audience of developmental researchers and clinicians, how epigenetic processes - chromatin structural modifications that regulate gene expression without changing DNA sequences - may offer a strong, parsimonious account for the convergence of genetic and contextual variation in the genesis of adaptive and maladaptive development. Epigenetic processes may play a plausible explanatory role in understanding: divergent trajectories and sexual dimorphisms in brain development; statistical interactions between genes and environments; the biological embedding of early psychosocial adversities; the linkages of such adversities to disorders of mental health; the striking individual variation in the strength of those linkages; the molecular origins of critical and sensitive periods; and the transgenerational inheritance of risk and protection. Taken together, these arguments converge in a claim that epigenetic processes constitute a promising and illuminating point of connection - a 'synapse' - between genes and environments.
Collapse
Affiliation(s)
- W Thomas Boyce
- Departments of Pediatrics and Psychiatry, University of California, San Francisco, USA; Child and Brain Development Program, Canadian Institute for Advanced Research, Canada
| | | |
Collapse
|
177
|
Liu H, Li Y, Wang Y, Wang X, An X, Wang S, Chen L, Liu G, Yang Y. The distinct role of NR2B subunit in the enhancement of visual plasticity in adulthood. Mol Brain 2015; 8:49. [PMID: 26282667 PMCID: PMC4539718 DOI: 10.1186/s13041-015-0141-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 08/10/2015] [Indexed: 01/23/2023] Open
Abstract
Background Experience-dependent plasticity is confined to the critical period of early postnatal life, and declines dramatically thereafter. This attenuation promotes the stabilization of cortical circuits, but also limits functional recovery of several brain diseases. The cognitive functions and synaptic plasticity in the hippocampus and prefrontal cortex are elevated following chronic magnesium treatment. Here, we explored the effect of magnesium treatment on visual plasticity and the potential clinical significance. Results Visual plasticity in adult mice was dramatically enhanced following magnesium treatment, which was concurrent with an increase in the expression of NR2 subunits of N-methyl-D-aspartate receptors. Blockade of NR2B activity in both the induction and expression periods of plasticity prevented this reinstatement. However, the plasticity restored via a decrease in cortical inhibition was independent on the activation of NR2B, indicating a different underlying mechanism. The functional excitatory synapses on layer 2/3 pyramidal neurons were increased following magnesium supplementation. Moreover, the synaptic and neuronal responses were reminiscent of that within the critical period, and this rejuvenation of adult visual cortex facilitated the recovery of visual functions in amblyopia. Conclusions Collectively, our data reveal two distinct mechanisms underlying the restoration of visual plasticity in adulthood, and the rejuvenation of adult visual cortex following magnesium treatment provides a new avenue to develop clinical therapies for adult amblyopia, as well as to explore plasticity-based treatment of other brain diseases, such as stroke and aphasia. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0141-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hanxiao Liu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, China
| | - Yue Li
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Yan Wang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Xinxing Wang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Xu An
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Siying Wang
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Lin Chen
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, China
| | - Guosong Liu
- Tsinghua-Peking Centre for Life Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yupeng Yang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Diseases, and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
178
|
Wu M, Puddifoot CA, Taylor P, Joiner WJ. Mechanisms of inhibition and potentiation of α4β2 nicotinic acetylcholine receptors by members of the Ly6 protein family. J Biol Chem 2015; 290:24509-18. [PMID: 26276394 DOI: 10.1074/jbc.m115.647248] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Indexed: 11/06/2022] Open
Abstract
α4β2 nicotinic acetylcholine receptors (nAChRs) are abundantly expressed throughout the central nervous system and are thought to be the primary target of nicotine, the main addictive substance in cigarette smoking. Understanding the mechanisms by which these receptors are regulated may assist in developing compounds to selectively interfere with nicotine addiction. Here we report previously unrecognized modulatory properties of members of the Ly6 protein family on α4β2 nAChRs. Using a FRET-based Ca(2+) flux assay, we found that the maximum response of α4β2 receptors to agonist was strongly inhibited by Ly6h and Lynx2 but potentiated by Ly6g6e. The mechanisms underlying these opposing effects appear to be fundamentally distinct. Receptor inhibition by Lynx2 was accompanied by suppression of α4β2 expression at the cell surface, even when assays were preceded by chronic exposure of cells to an established chaperone, nicotine. Receptor inhibition by Lynx2 also was resistant to pretreatment with extracellular phospholipase C, which cleaves lipid moieties like those that attach Ly6 proteins to the plasma membrane. In contrast, potentiation of α4β2 activity by Ly6g6e was readily reversible by pretreatment with phospholipase C. Potentiation was also accompanied by slowing of receptor desensitization and an increase in peak currents. Collectively our data support roles for Lynx2 and Ly6g6e in intracellular trafficking and allosteric potentiation of α4β2 nAChRs, respectively.
Collapse
Affiliation(s)
| | | | - Palmer Taylor
- From the Department of Pharmacology, Biomedical Sciences Graduate Program, Neuroscience Graduate Program, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093-0636
| | - William J Joiner
- From the Department of Pharmacology, Biomedical Sciences Graduate Program, Neuroscience Graduate Program, Center for Circadian Biology, and
| |
Collapse
|
179
|
Lunghi C, Berchicci M, Morrone MC, Di Russo F. Short-term monocular deprivation alters early components of visual evoked potentials. J Physiol 2015; 593:4361-72. [PMID: 26119530 DOI: 10.1113/jp270950] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 06/22/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Short-term monocular deprivation in adult humans produces a perceptual boost of the deprived eye reflecting homeostatic plasticity. Visual evoked potentials (VEPs) to transient stimuli change after 150 min of monocular deprivation in adult humans. The amplitude of the C1 component of the VEP at a latency of about 100 ms increases for the deprived eye and decreases for the non-deprived eye after deprivation, the two effects being highly negatively correlated. Similarly, the evoked alpha rhythm increases after deprivation for the deprived eye and decreases for the non-deprived eye. The data demonstrate that primary visual cortex excitability is altered by a short period of monocular deprivation, reflecting homeostatic plasticity. ABSTRACT Very little is known about plasticity in the adult visual cortex. In recent years psychophysical studies have shown that short-term monocular deprivation alters visual perception in adult humans. Specifically, after 150 min of monocular deprivation the deprived eye strongly dominates the dynamics of binocular rivalry, reflecting homeostatic plasticity. Here we investigate the neural mechanisms underlying this form of short-term visual cortical plasticity by measuring visual evoked potentials (VEPs) on the scalp of adult humans during monocular stimulation before and after 150 min of monocular deprivation. We found that monocular deprivation had opposite effects on the amplitude of the earliest component of the VEP (C1) for the deprived and non-deprived eye stimulation. C1 amplitude increased (+66%) for the deprived eye, while it decreased (-29%) for the non-deprived eye. Source localization analysis confirmed that the C1 originates in the primary visual cortex. We further report that following monocular deprivation, the amplitude of the peak of the evoked alpha spectrum increased on average by 23% for the deprived eye and decreased on average by 10% for the non-deprived eye, indicating a change in cortical excitability. These results indicate that a brief period of monocular deprivation alters interocular balance in the primary visual cortex of adult humans by both boosting the activity of the deprived eye and reducing the activity of the non-deprived eye. This indicates a high level of residual homeostatic plasticity in the adult human primary visual cortex, probably mediated by a change in cortical excitability.
Collapse
Affiliation(s)
- Claudia Lunghi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, via Savi 10, 56126, Pisa, Italy.,Institute of Neuroscience, CNR, via Moruzzi 1, 56127, Pisa, Italy
| | - Marika Berchicci
- Department of Movement, Human and Health Sciences, University of Rome 'Foro Italico', Piazza Lauro de Bosis 15, 00135, Rome (RM), Italy
| | - M Concetta Morrone
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, via Savi 10, 56126, Pisa, Italy.,Scientific Institute Stella Maris (IRCSS), Calambrone (Pisa), Italy
| | - Francesco Di Russo
- Department of Movement, Human and Health Sciences, University of Rome 'Foro Italico', Piazza Lauro de Bosis 15, 00135, Rome (RM), Italy.,Santa Lucia Foundation IRCCS, via Ardeatina 280, 00144, Rome (RM), Italy
| |
Collapse
|
180
|
Zhang Y. Why do we study animal toxins? DONG WU XUE YAN JIU = ZOOLOGICAL RESEARCH 2015; 36:183-222. [PMID: 26228472 PMCID: PMC4790257 DOI: 10.13918/j.issn.2095-8137.2015.4.183] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/25/2015] [Indexed: 12/31/2022]
Abstract
Venom (toxins) is an important trait evolved along the evolutionary tree of animals. Our knowledges on venoms, such as their origins and loss, the biological relevance and the coevolutionary patterns with other organisms are greatly helpful in understanding many fundamental biological questions, i.e., the environmental adaptation and survival competition, the evolution shaped development and balance of venoms, and the sophisticated correlations among venom, immunity, body power, intelligence, their genetic basis, inherent association, as well as the cost-benefit and trade-offs of biological economy. Lethal animal envenomation can be found worldwide. However, from foe to friend, toxin studies have led lots of important discoveries and exciting avenues in deciphering and fighting human diseases, including the works awarded the Nobel Prize and lots of key clinic therapeutics. According to our survey, so far, only less than 0.1% of the toxins of the venomous animals in China have been explored. We emphasize on the similarities shared by venom and immune systems, as well as the studies of toxin knowledge-based physiological toxin-like proteins/peptides (TLPs). We propose the natural pairing hypothesis. Evolution links toxins with humans. Our mission is to find out the right natural pairings and interactions of our body elements with toxins, and with endogenous toxin-like molecules. Although, in nature, toxins may endanger human lives, but from a philosophical point of view, knowing them well is an effective way to better understand ourselves. So, this is why we study toxins.
Collapse
Affiliation(s)
- Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of The Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming Yunnan 650223,
| |
Collapse
|
181
|
Do KQ, Cuenod M, Hensch TK. Targeting Oxidative Stress and Aberrant Critical Period Plasticity in the Developmental Trajectory to Schizophrenia. Schizophr Bull 2015; 41:835-46. [PMID: 26032508 PMCID: PMC4466197 DOI: 10.1093/schbul/sbv065] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a neurodevelopmental disorder reflecting a convergence of genetic risk and early life stress. The slow progression to first psychotic episode represents both a window of vulnerability as well as opportunity for therapeutic intervention. Here, we consider recent neurobiological insight into the cellular and molecular components of developmental critical periods and their vulnerability to redox dysregulation. In particular, the consistent loss of parvalbumin-positive interneuron (PVI) function and their surrounding perineuronal nets (PNNs) as well as myelination in patient brains is consistent with a delayed or extended period of circuit instability. This linkage to critical period triggers (PVI) and brakes (PNN, myelin) implicates mistimed trajectories of brain development in mental illness. Strategically introduced antioxidant treatment or later reinforcement of molecular brakes may then offer a novel prophylactic psychiatry.
Collapse
Affiliation(s)
- Kim Q. Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Takao K. Hensch
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, Cambridge, MA,*To whom correspondence should be addressed; Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, US; tel: +1-617-384-5882; fax: +1-617-495-4038; e-mail:
| |
Collapse
|
182
|
Checks and balances on cholinergic signaling in brain and body function. Trends Neurosci 2015; 38:448-58. [DOI: 10.1016/j.tins.2015.05.007] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 05/19/2015] [Accepted: 05/25/2015] [Indexed: 02/07/2023]
|
183
|
Ly6h regulates trafficking of alpha7 nicotinic acetylcholine receptors and nicotine-induced potentiation of glutamatergic signaling. J Neurosci 2015; 35:3420-30. [PMID: 25716842 DOI: 10.1523/jneurosci.3630-14.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
α7 nAChRs are expressed widely throughout the brain, where they are important for synaptic signaling, gene transcription, and plastic changes that regulate sensory processing, cognition, and neural responses to chronic nicotine exposure. However, the mechanisms by which α7 nAChRs are regulated are poorly understood. Here we show that trafficking of α7-subunits is controlled by endogenous membrane-associated prototoxins in the Ly6 family. In particular, we find that Ly6h reduces cell-surface expression and calcium signaling by α7 nAChRs. We detect Ly6h in several rat brain regions, including the hippocampus, where we find it is both necessary and sufficient to limit the magnitude of α7-mediated currents. Consistent with such a regulatory function, knockdown of Ly6h in rat hippocampal pyramidal neurons enhances nicotine-induced potentiation of glutamatergic mEPSC amplitude, which is known to be mediated by α7 signaling. Collectively our data suggest a novel cellular role for Ly6 proteins in regulating nAChRs, which may be relevant to plastic changes in the nervous system including rewiring of glutamatergic circuitry during nicotine addiction.
Collapse
|
184
|
Trachtenberg JT. Competition, inhibition, and critical periods of cortical plasticity. Curr Opin Neurobiol 2015; 35:44-8. [PMID: 26126153 DOI: 10.1016/j.conb.2015.06.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/15/2015] [Indexed: 11/17/2022]
Abstract
Maturation of cortical inhibition just after eye opening is a necessary precedent for the emergence of competitive, experience-dependent ocular dominance plasticity in the visual cortex. What inhibition is doing in this context, though, is not clear. Here I outline new hypotheses on the roles of somatic and dendritic inhibition in the opening and closure of critical periods, and their roles in the competitive processes therein.
Collapse
|
185
|
Bochner DN, Sapp RW, Adelson JD, Zhang S, Lee H, Djurisic M, Syken J, Dan Y, Shatz CJ. Blocking PirB up-regulates spines and functional synapses to unlock visual cortical plasticity and facilitate recovery from amblyopia. Sci Transl Med 2015; 6:258ra140. [PMID: 25320232 DOI: 10.1126/scitranslmed.3010157] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
During critical periods of development, the brain easily changes in response to environmental stimuli, but this neural plasticity declines by adulthood. By acutely disrupting paired immunoglobulin-like receptor B (PirB) function at specific ages, we show that PirB actively represses neural plasticity throughout life. We disrupted PirB function either by genetically introducing a conditional PirB allele into mice or by minipump infusion of a soluble PirB ectodomain (sPirB) into mouse visual cortex. We found that neural plasticity, as measured by depriving mice of vision in one eye and testing ocular dominance, was enhanced by this treatment both during the critical period and when PirB function was disrupted in adulthood. Acute blockade of PirB triggered the formation of new functional synapses, as indicated by increases in miniature excitatory postsynaptic current (mEPSC) frequency and spine density on dendrites of layer 5 pyramidal neurons. In addition, recovery from amblyopia--the decline in visual acuity and spine density resulting from long-term monocular deprivation--was possible after a 1-week infusion of sPirB after the deprivation period. Thus, neural plasticity in adult visual cortex is actively repressed and can be enhanced by blocking PirB function.
Collapse
Affiliation(s)
- David N Bochner
- Department of Biology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Richard W Sapp
- Department of Biology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Jaimie D Adelson
- Department of Biology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Siyu Zhang
- Division of Neurobiology, Department of Molecular and Cell Biology and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hanmi Lee
- Department of Biology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Maja Djurisic
- Department of Biology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA
| | - Josh Syken
- Vertex Pharmaceuticals Inc., Cambridge, MA 02139, USA
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology and Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Carla J Shatz
- Department of Biology and Bio-X, James H. Clark Center, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
186
|
Kaut O, Schmitt I, Hofmann A, Hoffmann P, Schlaepfer TE, Wüllner U, Hurlemann R. Aberrant NMDA receptor DNA methylation detected by epigenome-wide analysis of hippocampus and prefrontal cortex in major depression. Eur Arch Psychiatry Clin Neurosci 2015; 265:331-41. [PMID: 25571874 DOI: 10.1007/s00406-014-0572-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/19/2014] [Indexed: 12/21/2022]
Abstract
Current perspectives on the molecular underpinnings of major depressive disorder (MDD) posit a mechanistic role of epigenetic DNA modifications in mediating the interaction between environmental risk factors and a genetic predisposition. However, conclusive evidence for differential methylation signatures in the brain's epigenome of MDD patients as compared to controls is still lacking. To address this issue, we conducted a pilot study including an epigenome-wide methylation analysis in six individuals diagnosed with recurrent MDD and six control subjects matched for age and gender, with a priori focus on the hippocampus and prefrontal cortex as pathophysiologically relevant candidate regions. Our analysis revealed differential methylation profiles of 11 genes in hippocampus and 20 genes in prefrontal cortex, five of which were selected for replication of the methylation status using pyrosequencing. Among these replicated targets, GRIN2A was found to be hypermethylated in both prefrontal cortex and hippocampus. This finding may be of particular functional relevance as GRIN2A encodes the glutamatergic N-methyl-D-aspartate receptor subunit epsilon-1 (NR2A) and is known to be involved in a plethora of synaptic plasticity-related regulatory processes probably disturbed in MDD.
Collapse
Affiliation(s)
- Oliver Kaut
- Department of Neurology, University of Bonn, Sigmund-Freud-Str. 25, 53105, Bonn, Germany,
| | | | | | | | | | | | | |
Collapse
|
187
|
Progressive maturation of silent synapses governs the duration of a critical period. Proc Natl Acad Sci U S A 2015; 112:E3131-40. [PMID: 26015564 DOI: 10.1073/pnas.1506488112] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
During critical periods, all cortical neural circuits are refined to optimize their functional properties. The prevailing notion is that the balance between excitation and inhibition determines the onset and closure of critical periods. In contrast, we show that maturation of silent glutamatergic synapses onto principal neurons was sufficient to govern the duration of the critical period for ocular dominance plasticity in the visual cortex of mice. Specifically, postsynaptic density protein-95 (PSD-95) was absolutely required for experience-dependent maturation of silent synapses, and its absence before the onset of critical periods resulted in lifelong juvenile ocular dominance plasticity. Loss of PSD-95 in the visual cortex after the closure of the critical period reinstated silent synapses, resulting in reopening of juvenile-like ocular dominance plasticity. Additionally, silent synapse-based ocular dominance plasticity was largely independent of the inhibitory tone, whose developmental maturation was independent of PSD-95. Moreover, glutamatergic synaptic transmission onto parvalbumin-positive interneurons was unaltered in PSD-95 KO mice. These findings reveal not only that PSD-95-dependent silent synapse maturation in visual cortical principal neurons terminates the critical period for ocular dominance plasticity but also indicate that, in general, once silent synapses are consolidated in any neural circuit, initial experience-dependent functional optimization and critical periods end.
Collapse
|
188
|
Lunghi C, Emir UE, Morrone MC, Bridge H. Short-term monocular deprivation alters GABA in the adult human visual cortex. Curr Biol 2015; 25:1496-501. [PMID: 26004760 DOI: 10.1016/j.cub.2015.04.021] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 02/24/2015] [Accepted: 04/13/2015] [Indexed: 11/19/2022]
Abstract
Neuroplasticity is a fundamental property of the nervous system that is maximal early in life, within the critical period [1-3]. Resting GABAergic inhibition is necessary to trigger ocular dominance plasticity and to modulate the onset and offset of the critical period [4, 5]. GABAergic inhibition also plays a crucial role in neuroplasticity of adult animals: the balance between excitation and inhibition in the primary visual cortex (V1), measured at rest, modulates the susceptibility of ocular dominance to deprivation [6-10]. In adult humans, short-term monocular deprivation strongly modifies ocular balance, unexpectedly boosting the deprived eye, reflecting homeostatic plasticity [11, 12]. There is no direct evidence, however, to support resting GABAergic inhibition in homeostatic plasticity induced by visual deprivation. Here, we tested the hypothesis that GABAergic inhibition, measured at rest, is reduced by deprivation, as demonstrated by animal studies. GABA concentration in V1 of adult humans was measured using ultra-high-field 7T magnetic resonance spectroscopy before and after short-term monocular deprivation. After monocular deprivation, resting GABA concentration decreased in V1 but was unaltered in a control parietal area. Importantly, across participants, the decrease in GABA strongly correlated with the deprived eye perceptual boost measured by binocular rivalry. Furthermore, after deprivation, GABA concentration measured during monocular stimulation correlated with the deprived eye dominance. We suggest that reduction in resting GABAergic inhibition triggers homeostatic plasticity in adult human V1 after a brief period of abnormal visual experience. These results are potentially useful for developing new therapeutic strategies that could exploit the intrinsic residual plasticity of the adult human visual cortex.
Collapse
Affiliation(s)
- Claudia Lunghi
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; Institute of Neuroscience, CNR, 56124 Pisa, Italy
| | - Uzay E Emir
- FMRIB, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK
| | - Maria Concetta Morrone
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; Scientific Institute Stella Maris (IRCSS), Calambrone, 56018 Pisa, Italy.
| | - Holly Bridge
- FMRIB, University of Oxford, Oxford, Oxfordshire OX3 9DU, UK
| |
Collapse
|
189
|
Song S, Mitchell DE, Crowder NA, Duffy KR. Postnatal accumulation of intermediate filaments in the cat and human primary visual cortex. J Comp Neurol 2015; 523:2111-26. [PMID: 25823892 DOI: 10.1002/cne.23781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 03/16/2015] [Accepted: 03/17/2015] [Indexed: 12/25/2022]
Abstract
A principal characteristic of the mammalian visual system is its high capacity for plasticity in early postnatal development during a time commonly referred to as the critical period. The progressive diminution of plasticity with age is linked to the emergence of a collection of molecules called molecular brakes that reduce plasticity and stabilize neural circuits modified by earlier visual experiences. Manipulation of braking molecules either pharmacologically or though experiential alteration enhances plasticity and promotes recovery from visual impairment. The stability of neural circuitry is increased by intermediate filamentous proteins of the cytoskeleton such as neurofilaments and α-internexin. We examined levels of these intermediate filaments within cat and human primary visual cortex (V1) across development to determine whether they accumulate following a time course consistent with a molecular brake. In both species, levels of intermediate filaments increased considerably throughout early postnatal life beginning shortly after the peak of the critical period, with the highest levels measured in adults. Neurofilament phosphorylation was also observed to increase throughout development, raising the possibility that posttranslational modification by phosphorylation reduces plasticity due to increased protein stability. Finally, an approach to scale developmental time points between species is presented that compares the developmental profiles of intermediate filaments between cats and humans. Although causality between intermediate filaments and plasticity was not directly tested in this study, their accumulation relative to the critical period indicates that they may contribute to the decline in plasticity with age, and may also constrain the success of treatments for visual disorders applied in adulthood.
Collapse
Affiliation(s)
- Seoho Song
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, Nova Scotia, Canada, B3H 4R2
| | - Donald E Mitchell
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, Nova Scotia, Canada, B3H 4R2
| | - Nathan A Crowder
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, Nova Scotia, Canada, B3H 4R2
| | - Kevin R Duffy
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, Nova Scotia, Canada, B3H 4R2
| |
Collapse
|
190
|
Hirano T, Yanai S, Omotehara T, Hashimoto R, Umemura Y, Kubota N, Minami K, Nagahara D, Matsuo E, Aihara Y, Shinohara R, Furuyashiki T, Mantani Y, Yokoyama T, Kitagawa H, Hoshi N. The combined effect of clothianidin and environmental stress on the behavioral and reproductive function in male mice. J Vet Med Sci 2015; 77:1207-15. [PMID: 25960033 PMCID: PMC4638285 DOI: 10.1292/jvms.15-0188] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neonicotinoids, some of the most widely used pesticides in the world, act as agonists to
the nicotinic acetylcholine receptors (nAChRs) of insects, resulting in death from
abnormal excitability. Neonicotinoids unexpectedly became a major topic as a compelling
cause of honeybee colony collapse disorder, which is damaging crop production that
requires pollination worldwide. Mammal nAChRs appear to have a certain affinity for
neonicotinoids with lower levels than those of insects; there is thus rising concern about
unpredictable adverse effects of neonicotinoids on vertebrates. We hypothesized that the
effects of neonicotinoids would be enhanced under a chronic stressed condition, which is
known to alter the expression of targets of neonicotinoids, i.e.,
neuronal nAChRs. We performed immunohistochemical and behavioral analyses in male mice
actively administered a neonicotinoid, clothianidin (CTD; 0, 10, 50 and 250 mg/kg/day),
for 4 weeks under an unpredictable chronic stress procedure. Vacuolated seminiferous
epithelia and a decrease in the immunoreactivity of the antioxidant enzyme glutathione
peroxidase 4 were observed in the testes of the CTD+stress mice. In an open field test,
although the locomotor activities were not affected, the anxiety-like behaviors of the
mice were elevated by both CTD and stress. The present study demonstrates that the
behavioral and reproductive effects of CTD become more serious in combination with
environmental stress, which may reflect our actual situation of multiple exposure.
Collapse
Affiliation(s)
- Tetsushi Hirano
- Laboratory of Molecular Morphology, Department of Animal Science, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
191
|
Kobayashi Y, Ye Z, Hensch TK. Clock genes control cortical critical period timing. Neuron 2015; 86:264-75. [PMID: 25801703 PMCID: PMC4392344 DOI: 10.1016/j.neuron.2015.02.036] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 01/25/2015] [Accepted: 02/18/2015] [Indexed: 01/05/2023]
Abstract
Circadian rhythms control a variety of physiological processes, but whether they may also time brain development remains largely unknown. Here, we show that circadian clock genes control the onset of critical period plasticity in the neocortex. Within visual cortex of Clock-deficient mice, the emergence of circadian gene expression was dampened, and the maturation of inhibitory parvalbumin (PV) cell networks slowed. Loss of visual acuity in response to brief monocular deprivation was concomitantly delayed and rescued by direct enhancement of GABAergic transmission. Conditional deletion of Clock or Bmal1 only within PV cells recapitulated the results of total Clock-deficient mice. Unique downstream gene sets controlling synaptic events and cellular homeostasis for proper maturation and maintenance were found to be mis-regulated by Clock deletion specifically within PV cells. These data demonstrate a developmental role for circadian clock genes outside the suprachiasmatic nucleus, which may contribute mis-timed brain plasticity in associated mental disorders.
Collapse
Affiliation(s)
- Yohei Kobayashi
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Zhanlei Ye
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Takao K Hensch
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA; F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
192
|
Quantitative profiling of brain lipid raft proteome in a mouse model of fragile X syndrome. PLoS One 2015; 10:e0121464. [PMID: 25849048 PMCID: PMC4388542 DOI: 10.1371/journal.pone.0121464] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 02/12/2015] [Indexed: 11/19/2022] Open
Abstract
Fragile X Syndrome, a leading cause of inherited intellectual disability and autism, arises from transcriptional silencing of the FMR1 gene encoding an RNA-binding protein, Fragile X Mental Retardation Protein (FMRP). FMRP can regulate the expression of approximately 4% of brain transcripts through its role in regulation of mRNA transport, stability and translation, thus providing a molecular rationale for its potential pleiotropic effects on neuronal and brain circuitry function. Several intracellular signaling pathways are dysregulated in the absence of FMRP suggesting that cellular deficits may be broad and could result in homeostatic changes. Lipid rafts are specialized regions of the plasma membrane, enriched in cholesterol and glycosphingolipids, involved in regulation of intracellular signaling. Among transcripts targeted by FMRP, a subset encodes proteins involved in lipid biosynthesis and homeostasis, dysregulation of which could affect the integrity and function of lipid rafts. Using a quantitative mass spectrometry-based approach we analyzed the lipid raft proteome of Fmr1 knockout mice, an animal model of Fragile X syndrome, and identified candidate proteins that are differentially represented in Fmr1 knockout mice lipid rafts. Furthermore, network analysis of these candidate proteins reveals connectivity between them and predicts functional connectivity with genes encoding components of myelin sheath, axonal processes and growth cones. Our findings provide insight to aid identification of molecular and cellular dysfunctions arising from Fmr1 silencing and for uncovering shared pathologies between Fragile X syndrome and other autism spectrum disorders.
Collapse
|
193
|
Morishita H, Kundakovic M, Bicks L, Mitchell A, Akbarian S. Interneuron epigenomes during the critical period of cortical plasticity: Implications for schizophrenia. Neurobiol Learn Mem 2015; 124:104-10. [PMID: 25849095 DOI: 10.1016/j.nlm.2015.03.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
Abstract
Schizophrenia, a major psychiatric disorder defined by delusions and hallucinations, among other symptoms, often with onset in early adulthood, is potentially associated with molecular and cellular alterations in parvalbumin-expressing fast spiking interneurons and other constituents of the cortical inhibitory GABAergic circuitry. The underlying mechanisms, including the role of disease-associated risk factors operating in adolescence such as drug abuse and social stressors, remain incompletely understood. Here, we summarize emerging findings from animal models, highlighting the ability of parvalbuminergic interneurons (PVI) to induce, during the juvenile period, long-term plastic changes in prefrontal and visual cortex, thereby altering perception, cognition and behavior in the adult. Of note, molecular alterations in PVI from subjects with schizophrenia, including downregulated expression of a subset of GABAergic genes, have also been found in juvenile stress models of the disorder. Some of the transcriptional alterations observed in schizophrenia postmortem brain could be linked to changes in the epigenetic architecture of GABAergic gene promoters, including dysregulated DNA methylation, histone modification patterns and disruption of promoter-enhancer interactions at site of chromosomal loop formations. Therefore, we predict that, in the not-to-distant future, PVI- and other cell-type specific epigenomic mappings in the animal model and human brain will provide novel insights into the pathophysiology of schizophrenia and related psychotic diseases, including the role of cortical GABAergic circuitry in shaping long-term plasticity and cognitive function of the cerebral cortex.
Collapse
Affiliation(s)
- Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, United States; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States.
| | - Marija Kundakovic
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Lucy Bicks
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Amanda Mitchell
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, United States; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, United States.
| |
Collapse
|
194
|
Impaired synaptic plasticity in the visual cortex of mice lacking α7-nicotinic receptor subunit. Neuroscience 2015; 294:166-71. [PMID: 25797465 DOI: 10.1016/j.neuroscience.2015.03.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/11/2015] [Accepted: 03/11/2015] [Indexed: 01/12/2023]
Abstract
The primary visual cortex (V1) is the first step in visual information processing and its function may be modulated by acetylcholine through nicotinic receptors (nAChRs). Since our previous work demonstrated that visual acuity and cortical spatial resolution limit were significantly reduced in α7 knock-out (KO) mice in the absence of retinal alterations, we decided to characterize the contribution of homomeric α7 nicotinic receptors (α7nAChRs) to visual information processing at the cortical level. We evaluated long-term forms of synaptic plasticity in occipital slices containing V1 from α7 KO mice and in wild-type (WT) slices perfused with nAChRs selective blocking agents. In α7 KO mice slices, electrophysiological recordings demonstrated the absence of long-term potentiation (LTP) and long-term depression (LTD) in layer II/III after the stimulation of different intracortical pathways (layer IV or II/III). Furthermore, the acute and selective blockade of α7nAChRs in slices from WT mice with either α-bungarotoxin or methyllycaconitine did not alter the expression of LTP and LTD. Conversely, the perfusion with the unspecific nAChRs antagonist mecamylamine impaired LTP and LTD. Our results suggest the presence of impaired synaptic plasticity in the V1 of α7 KO mice and indicate a different contribution of nAChRs to visual cortex function.
Collapse
|
195
|
Jensen MM, Arvaniti M, Mikkelsen JD, Michalski D, Pinborg LH, Härtig W, Thomsen MS. Prostate stem cell antigen interacts with nicotinic acetylcholine receptors and is affected in Alzheimer's disease. Neurobiol Aging 2015; 36:1629-1638. [PMID: 25680266 DOI: 10.1016/j.neurobiolaging.2015.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 12/08/2014] [Accepted: 01/03/2015] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder involving impaired cholinergic neurotransmission and dysregulation of nicotinic acetylcholine receptors (nAChRs). Ly-6/neurotoxin (Lynx) proteins have been shown to modulate cognition and neural plasticity by binding to nAChR subtypes and modulating their function. Hence, changes in nAChR regulatory proteins such as Lynx proteins could underlie the dysregulation of nAChRs in AD. Using Western blotting, we detected bands corresponding to the Lynx proteins prostate stem cell antigen (PSCA) and Lypd6 in human cortex indicating that both proteins are present in the human brain. We further showed that PSCA forms stable complexes with the α4 nAChR subunit and decreases nicotine-induced extracellular-signal regulated kinase phosphorylation in PC12 cells. In addition, we analyzed protein levels of PSCA and Lypd6 in postmortem tissue of medial frontal gyrus from AD patients and found significantly increased PSCA levels (approximately 70%). In contrast, no changes in Lypd6 levels were detected. In concordance with our findings in AD patients, PSCA levels were increased in the frontal cortex of triple transgenic mice with an AD-like pathology harboring human transgenes that cause both age-dependent β-amyloidosis and tauopathy, whereas Tg2576 mice, which display β-amyloidosis only, had unchanged PSCA levels compared to wild-type animals. These findings identify PSCA as a nAChR-binding protein in the human brain that is affected in AD, suggesting that PSCA-nAChR interactions may be involved in the cognitive dysfunction observed in AD.
Collapse
Affiliation(s)
- Majbrit M Jensen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Maria Arvaniti
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jens D Mikkelsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | - Lars H Pinborg
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Epilepsy Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Morten S Thomsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
196
|
Abstract
A continuing debate in language acquisition research is whether there are critical periods (CPs) in development during which the system is most responsive to environmental input. Recent advances in neurobiology provide a mechanistic explanation of CPs, with the balance between excitatory and inhibitory processes establishing the onset and molecular brakes establishing the offset of windows of plasticity. In this article, we review the literature on human speech perception development within the context of this CP model, highlighting research that reveals the interplay of maturational and experiential influences at key junctures in development and presenting paradigmatic examples testing CP models in human subjects. We conclude with a discussion of how a mechanistic understanding of CP processes changes the nature of the debate: The question no longer is, "Are there CPs?" but rather what processes open them, keep them open, close them, and allow them to be reopened.
Collapse
Affiliation(s)
- Janet F Werker
- Department of Psychology, University of British Columbia, Vancouver British Columbia V6T 1Z4, Canada;
| | | |
Collapse
|
197
|
Ji D, Li L, Zhang S, Li H. Identification of a Ly-6 superfamily gene expressed in lateral line neuromasts in zebrafish. Dev Genes Evol 2015; 225:47-53. [PMID: 25586305 DOI: 10.1007/s00427-015-0487-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 01/05/2015] [Indexed: 02/02/2023]
Abstract
Lymphocyte antigen-6 (Ly-6) superfamily members have been identified in zebrafish, but the expression and function of these Ly-6 genes remain largely unknown. Posterior lateral line (pLL) system is produced by migrating pLL primordium (pLLp). Chemokine signaling, Notch, Wnt, and fibroblast growth factor (FGF) signaling regulate migration of pLLp cells and formation of neuromasts. However, the mechanism of neuromast deposition remains to be explored. Identification of novel genes expressed in pLLp will certainly help the study of such a process. Here we identified a Ly-6 gene called neuromast-expressed gpi-anchored lymphocyte antigen-6 (negaly6), which was specifically expressed in neuromast. Quantitative real-time PCR (qRT-PCR) analysis showed that negaly6 started to be expressed at 24 hpf, and whole-mount in situ hybridization analysis indicated that negaly6 was highly expressed in the trailing zone of pLLp and mature neuromast. Furthermore, negaly6 expression was inhibited by FGF signaling antagonist but not by Wnt signaling agonist or antagonist. Collectively, these data indicate that negaly6 may be associated with the regulation of neuromast deposition via FGF signaling pathway.
Collapse
Affiliation(s)
- Dongrui Ji
- Institute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | | | | | | |
Collapse
|
198
|
Hartley CA, Lee FS. Sensitive periods in affective development: nonlinear maturation of fear learning. Neuropsychopharmacology 2015; 40:50-60. [PMID: 25035083 PMCID: PMC4262897 DOI: 10.1038/npp.2014.179] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 07/08/2014] [Accepted: 07/11/2014] [Indexed: 12/11/2022]
Abstract
At specific maturational stages, neural circuits enter sensitive periods of heightened plasticity, during which the development of both brain and behavior are highly receptive to particular experiential information. A relatively advanced understanding of the regulatory mechanisms governing the initiation, closure, and reinstatement of sensitive period plasticity has emerged from extensive research examining the development of the visual system. In this article, we discuss a large body of work characterizing the pronounced nonlinear changes in fear learning and extinction that occur from childhood through adulthood, and their underlying neural substrates. We draw upon the model of sensitive period regulation within the visual system, and present burgeoning evidence suggesting that parallel mechanisms may regulate the qualitative changes in fear learning across development.
Collapse
Affiliation(s)
- Catherine A Hartley
- Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY, USA,Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA,Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, 1300 York Ave, New York, NY 10065, USA, Tel: +212 746 3781, Fax: +212 746 5755, E-mail:
| | - Francis S Lee
- Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY, USA,Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
199
|
Duffy KR, Bukhamseen DH, Smithen MJ, Mitchell DE. Binocular eyelid closure promotes anatomical but not behavioral recovery from monocular deprivation. Vision Res 2014; 114:151-60. [PMID: 25536470 DOI: 10.1016/j.visres.2014.12.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 12/12/2014] [Accepted: 12/14/2014] [Indexed: 10/24/2022]
Abstract
Deprivation of patterned vision of frontal eyed mammals early in postnatal life alters structural and functional attributes of neurones in the central visual pathways, and can produce severe impairments of the vision of the deprived eye that resemble the visual loss observed in human amblyopia. A traditional approach to treatment of amblyopia has been the occlusion of the stronger fellow eye in order to force use of the weaker eye and thereby strengthen its connections in the visual cortex. Although this monocular treatment strategy can be effective at promoting recovery of visual acuity of the amblyopic eye, such binocular visual functions as stereoscopic vision often remain impaired due in part to the lack of concordant vision during the period of unilateral occlusion. The recent development of binocular approaches for treatment of amblyopia that improve the possibility for binocular interaction have achieved success in promoting visual recovery. The full and rapid recovery of visual acuity observed in amblyopic kittens placed in complete darkness is an example of a binocular treatment whose success may in part derive from a restored balance of visually-driven neural activity. In the current study we examined as an alternative to dark rearing the efficacy of binocular lid suture (BLS) to stimulate anatomical and visual recovery from a preceding amblyogenic period of monocular deprivation. In the dorsal lateral geniculate nucleus (dLGN) of monocularly deprived kittens, darkness or BLS for 10days produced a complete recovery of neurone soma size within initially deprived layers. The growth of neurone somata within initially deprived dLGN layers after darkness or BLS was accompanied by an increase in neurotrophin-4/5 labeling within these layers. Although anatomical recovery was observed in both recovery conditions, BLS failed to promote any improvement of the visual acuity of the deprived eye no matter whether it followed immediately or was delayed with respect to the prior period of monocular deprivation. Notwithstanding the lack of visual recovery with BLS, all animals in the BLS condition that were subsequently placed in darkness exhibited a substantial recovery of visual acuity in the amblyopic eye. We conclude that the balanced binocular visual input provided by BLS does not stimulate the collection of neural events necessary to support recovery from amblyopia. The complete absence of visually-driven activity that occurs with dark rearing evidently plays an important role in the recovery process.
Collapse
Affiliation(s)
- Kevin R Duffy
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, NS B3H 4R2, Canada.
| | - Dalia H Bukhamseen
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, NS B3H 4R2, Canada
| | - Matthew J Smithen
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, NS B3H 4R2, Canada
| | - Donald E Mitchell
- Department of Psychology and Neuroscience, Dalhousie University, Life Sciences Centre, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
200
|
Three-finger snake neurotoxins and Ly6 proteins targeting nicotinic acetylcholine receptors: pharmacological tools and endogenous modulators. Trends Pharmacol Sci 2014; 36:109-23. [PMID: 25528970 DOI: 10.1016/j.tips.2014.11.003] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 11/04/2014] [Accepted: 11/10/2014] [Indexed: 02/07/2023]
Abstract
Snake venom neurotoxins and lymphocyte antigen 6 (Ly6) proteins, most of the latter being membrane tethered by a glycosylphosphatidylinositol (GPI) anchor, have a variety of biological activities, but their three-finger (3F) folding combines them in one Ly6/neurotoxin family. Subsets of two groups, represented by α-neurotoxins and Lynx1, respectively, interact with nicotinic acetylcholine receptors (nAChR) and, hence, are of therapeutic interest for the treatment of neurodegenerative diseases, pain, and cancer. Information on the mechanisms of action and 3D structure of the binding sites, which is required for drug design, is available from the 3D structure of α-neurotoxin complexes with nAChR models. Here, I compare the structural and functional features of α-neurotoxins versus Lynx1 and its homologs to get a clearer picture of Lynx1-nAChR interactions that is necessary for fundamental science and practical applications.
Collapse
|