1
|
Nakaya Y, Kosukegawa S, Kobayashi S, Hirose K, Kitano K, Mayahara K, Takei H, Motoyoshi M, Kobayashi M. Insulin potentiates inhibitory synaptic currents between fast-spiking and pyramidal neurons in the rat insular cortex. Neuropharmacology 2023:109649. [PMID: 37393988 DOI: 10.1016/j.neuropharm.2023.109649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/04/2023]
Abstract
Insulin plays roles in brain functions such as neural development and plasticity and is reported to be involved in dementia and depression. However, little information is available on the insulin-mediated modulation of electrophysiological activities, especially in the cerebral cortex. This study examined how insulin modulates the neural activities of inhibitory neurons and inhibitory postsynaptic currents (IPSCs) in rat insular cortex (IC; either sex) by multiple whole-cell patch-clamp recordings. We demonstrated that insulin increased the repetitive spike firing rate with a decrease in the threshold potential without changing the resting membrane potentials and input resistance of fast-spiking GABAergic neurons (FSNs). Next, we found a dose-dependent enhancement of unitary IPSCs (uIPSCs) by insulin in the connections from FSNs to pyramidal neurons (PNs). The insulin-induced enhancement of uIPSCs accompanied decreases in the paired-pulse ratio, suggesting that insulin increases GABA release from presynaptic terminals. The finding of miniature IPSC recordings of the increased frequency without changing the amplitude supports this hypothesis. Insulin had little effect on uIPSCs under the coapplication of S961, an insulin receptor antagonist, or lavendustin A, an inhibitor of tyrosine kinase. The PI3-K inhibitor wortmannin or the PKB/Akt inhibitors, deguelin and Akt inhibitor VIII, blocked the insulin-induced enhancement of uIPSCs. Intracellular application of Akt inhibitor VIII to presynaptic FSNs also blocked insulin-induced enhancement of uIPSCs. In contrast, uIPSCs were enhanced by insulin in combination with the MAPK inhibitor PD98059. These results suggest that insulin facilitates the inhibition of PNs by increases in FSN firing frequency and IPSCs from FSNs to PNs. (250 words).
Collapse
Affiliation(s)
- Yuka Nakaya
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Satoshi Kosukegawa
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Satomi Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Biology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kensuke Hirose
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Pedodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kouhei Kitano
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Kotoe Mayahara
- Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hiroki Takei
- Department of Dentistry, Saitama Prefectural Children's Medical Center, 1-2, Shintoshin, Chuo-ku, Saitama-shi, 3330-8777, Japan
| | - Mitsuru Motoyoshi
- Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Department of Orthodontics, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Masayuki Kobayashi
- Department of Pharmacology, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan; Division of Oral and Craniomaxillofacial Research, Dental Research Center, Nihon University School of Dentistry, 1-8-13 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| |
Collapse
|
2
|
Xue CY, Gao T, Mao E, Kou ZZ, Dong L, Gao F. Hippocampus Insulin Receptors Regulate Episodic and Spatial Memory Through Excitatory/Inhibitory Balance. ASN Neuro 2023; 15:17590914231206657. [PMID: 37908089 PMCID: PMC10621302 DOI: 10.1177/17590914231206657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/25/2023] [Accepted: 09/23/2023] [Indexed: 11/02/2023] Open
Abstract
It is well known that the hippocampus is a vital brain region playing a key role in both episodic and spatial memory. Insulin receptors (InsRs) are densely distributed in the hippocampus and are important for its function. However, the effects of InsRs on the function of the specific hippocampal cell types remain elusive. In this study, hippocampal InsRs knockout mice had impaired episodic and spatial memory. GABAergic neurons and glutamatergic neurons in the hippocampus are involved in the balance between excitatory and inhibitory (E/I) states and participate in the processes of episodic and spatial memory. InsRs are located mainly at excitatory neurons in the hippocampus, whereas 8.5% of InsRs are glutamic acid decarboxylase 2 (GAD2)::Ai9-positive (GABAergic) neurons. Next, we constructed a transgenic mouse system in which InsR expression was deleted from GABAergic (glutamate decarboxylase 2::InsRfl/fl, GAD2Cre::InsRfl/fl) or glutamatergic neurons (vesicular glutamate transporter 2::InsRfl/fl,Vglut2Cre::InsRfl/fl). Our results showed that in comparison to the InsRfl/fl mice, both episodic and spatial memory were lower in GAD2Cre::InsRfl/fl and Vglut2Cre::InsRfl/fl. In addition, both GAD2Cre::InsRfl/fl and Vglut2Cre::InsRfl/fl were associated with more anxiety and lower glucose tolerance. These findings reveal that hippocampal InsRs might be crucial for episodic and spatial memory through E/I balance hippocampal regulation.
Collapse
Affiliation(s)
- Cai-Yan Xue
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Tian Gao
- Division of Health Management, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
| | - E Mao
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an, China
| | - Zhen-Zhen Kou
- Department of Anatomy, Histology and Embryology & K. K. Leung Brain Research Centre, Fourth Military Medical University, Xi’an, China
| | - Ling Dong
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
3
|
Falling Short: The Contribution of Central Insulin Receptors to Gait Dysregulation in Brain Aging. Biomedicines 2022; 10:biomedicines10081923. [PMID: 36009470 PMCID: PMC9405648 DOI: 10.3390/biomedicines10081923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/23/2022] Open
Abstract
Insulin resistance, which manifests as a reduction of insulin receptor signaling, is known to correlate with pathological changes in peripheral tissues as well as in the brain. Central insulin resistance has been associated with impaired cognitive performance, decreased neuronal health, and reduced brain metabolism; however, the mechanisms underlying central insulin resistance and its impact on brain regions outside of those associated with cognition remain unclear. Falls are a leading cause of both fatal and non-fatal injuries in the older population. Despite this, there is a paucity of work focused on age-dependent alterations in brain regions associated with ambulatory control or potential therapeutic approaches to target these processes. Here, we discuss age-dependent alterations in central modalities that may contribute to gait dysregulation, summarize current data supporting the role of insulin signaling in the brain, and highlight key findings that suggest insulin receptor sensitivity may be preserved in the aged brain. Finally, we present novel results showing that administration of insulin to the somatosensory cortex of aged animals can alter neuronal communication, cerebral blood flow, and the motivation to ambulate, emphasizing the need for further investigations of intranasal insulin as a clinical management strategy in the older population.
Collapse
|
4
|
Wang X, Gao C, Zhang Y, Hu S, Qiao Y, Zhao Z, Gou L, Song J, Wang Q. Overexpression of mGluR7 in the Prefrontal Cortex Attenuates Autistic Behaviors in Mice. Front Cell Neurosci 2021; 15:689611. [PMID: 34335187 PMCID: PMC8319395 DOI: 10.3389/fncel.2021.689611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is associated with a range of abnormalities pertaining to socialization, communication, repetitive behaviors, and restricted interests. Owing to its complexity, the etiology of ASD remains incompletely understood. The presynaptic G protein-coupled glutamate receptor metabotropic glutamate receptor 7 (mGluR7) is known to be essential for synaptic transmission and is also tightly linked with ASD incidence. Herein, we report that prefrontal cortex (PFC) mGluR7 protein levels were decreased in C57BL/6J mice exposed to valproic acid (VPA) and BTBR T+ Itpr3tf/J mice. The overexpression of mGluR7 in the PFC of these mice using a lentiviral vector was sufficient to reduce the severity of ASD-like behavioral patterns such that animals exhibited decreases in abnormal social interactions and communication, anxiety-like, and stereotyped/repetitive behaviors. Intriguingly, patch-clamp recordings revealed that the overexpression of mGluR7 suppressed neuronal excitability by inhibiting action potential discharge frequencies, together with enhanced action potential threshold and increased rheobase. These data offer a scientific basis for the additional study of mGluR7 as a promising therapeutic target in ASD and related neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiaona Wang
- Department of Nuclear Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Chao Gao
- Department of Rehabilitation, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yaodong Zhang
- Department of Nuclear Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Shunan Hu
- Department of Nuclear Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Yidan Qiao
- Department of Pathology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Zhengqin Zhao
- Department of Nuclear Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Lingshan Gou
- Center for Genetic Medicine, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, China
| | - Jijun Song
- Henan Infectious Disease Hospital, The Sixth People's Hospital of Zhengzhou, Zhengzhou, China
| | - Qi Wang
- Department of Histology and Embryology, Guizhou Medical University, Guizhou, China
| |
Collapse
|
5
|
Khalid M, Raza H, M. Driessen T, J. Lee P, Tejwani L, Sami A, Nawaz M, Mehmood Baig S, Lim J, Kaukab Raja G. Genetic Risk of Autism Spectrum Disorder in a Pakistani Population. Genes (Basel) 2020; 11:genes11101206. [PMID: 33076578 PMCID: PMC7602870 DOI: 10.3390/genes11101206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of complex multifactorial neurodevelopmental and neuropsychiatric disorders in children characterized by impairment of communication and social interaction. Several genes with associated single nucleotide polymorphisms (SNPs) have been identified for ASD in different genetic association studies, meta-analyses, and genome-wide association studies (GWAS). However, associations between different SNPs and ASD vary from population to population. Four SNPs in genes CNTNAP2, EIF4E, ATP2B2, CACNA1C, and SNP rs4307059 (which is found between CDH9 and CDH10 genes) have been identified and reported as candidate risk factors for ASD. The aim of the present study was, for the first time, to assess the association of SNPs in these genes with ASD in the Pakistani population. PCR-based genotyping was performed using allele-specific primers in 93 ASD and 93 control Pakistani individuals. All genetic associations, genotype frequencies, and allele frequencies were computed as odds’ ratios (ORs) using logistic regression with a threshold of p ≤ 0.01 to determine statistical significance. We found that the homozygous genotypes of mutant T alleles of CNTNAP2 and ATP2B2 were significantly associated with Pakistani ASD patients in unadjusted ORs (p < 0.01), but their significance score was lost in the adjusted model. Other SNPs such as rs4307059, rs17850950 of EIF4E, and rs1006737 of CACNA1C were not statistically significant. Based on this, we conclude that SNPs are not associated with, or are not the main cause of, autism in the Pakistani population, indicating the involvement of additional players, which need to be investigated in future studies in a large population size. One of the limitations of present study is its small sample size. However, this study, being the first on Pakistani ASD patients, may lay the foundations for future studies in larger samples.
Collapse
Affiliation(s)
- Madiha Khalid
- Department of Biochemistry, University Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi 46000, Pakistan; (M.K.); (A.S.)
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Hashim Raza
- Pakistan Institute of Medical Sciences, Islamabad 44000, Pakistan;
| | - Terri M. Driessen
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA;
| | - Paul J. Lee
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; (P.J.L.); (L.T.)
| | - Leon Tejwani
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; (P.J.L.); (L.T.)
| | - Abdul Sami
- Department of Biochemistry, University Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi 46000, Pakistan; (M.K.); (A.S.)
| | - Muhammad Nawaz
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden;
| | - Shahid Mehmood Baig
- Human Molecular Genetics Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan;
| | - Janghoo Lim
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA;
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA; (P.J.L.); (L.T.)
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06510, USA
- Correspondence: (J.L.); (G.K.R.); Tel.: +1-203-737-6268 (J.L.); +92-(051)-9062-742 (G.K.R.)
| | - Ghazala Kaukab Raja
- Department of Biochemistry, University Institute of Biochemistry and Biotechnology, PMAS Arid Agriculture University, Rawalpindi 46000, Pakistan; (M.K.); (A.S.)
- Correspondence: (J.L.); (G.K.R.); Tel.: +1-203-737-6268 (J.L.); +92-(051)-9062-742 (G.K.R.)
| |
Collapse
|
6
|
Erzurumlu RS, Gaspar P. How the Barrel Cortex Became a Working Model for Developmental Plasticity: A Historical Perspective. J Neurosci 2020; 40:6460-6473. [PMID: 32817388 PMCID: PMC7486654 DOI: 10.1523/jneurosci.0582-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 01/08/2023] Open
Abstract
For half a century now, the barrel cortex of common laboratory rodents has been an exceptionally useful model for studying the formation of topographically organized maps, neural patterning, and plasticity, both in development and in maturity. We present a historical perspective on how barrels were discovered, and how thereafter, they became a workhorse for developmental neuroscientists and for studies on brain plasticity and activity-dependent modeling of brain circuits. What is particularly remarkable about this sensory system is a cellular patterning that is induced by signals derived from the sensory receptors surrounding the snout whiskers and transmitted centrally to the brainstem (barrelettes), the thalamus (barreloids), and the neocortex (barrels). Injury to the sensory receptors shortly after birth leads to predictable pattern alterations at all levels of the system. Mouse genetics have increased our understanding of how barrels are constructed and revealed the interplay of the molecular programs that direct axon growth and cell specification, with activity-dependent mechanisms. There is an ever-rising interest in this sensory system as a neurobiological model to study development of somatotopy, patterning, and plasticity at both the morphologic and physiological levels. This article is part of a group of articles commemorating the 50th anniversary of the Society for Neuroscience.
Collapse
Affiliation(s)
- Reha S Erzurumlu
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale, Paris Brain Institute, Sorbonne Universités, Paris, France 75013
| |
Collapse
|
7
|
Yan Y, Yang H, Xie Y, Ding Y, Kong D, Yu H. Research Progress on Alzheimer's Disease and Resveratrol. Neurochem Res 2020; 45:989-1006. [PMID: 32162143 DOI: 10.1007/s11064-020-03007-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), a common irreversible neurodegenerative disease characterized by amyloid-β plaques, neurofibrillary tangles, and changes in tau phosphorylation, is accompanied by memory loss and symptoms of cognitive dysfunction. Increases in disease incidence due to the ageing of the population have placed a great burden on society. To date, the mechanism of AD and the identities of adequate drugs for AD prevention and treatment have eluded the medical community. It has been confirmed that phytochemicals have certain neuroprotective effects against AD. For example, some progress has been made in research on the use of resveratrol, a natural polyphenolic phytochemical, for the prevention and treatment of AD in recent years. Elucidation of the pathogenesis of AD will create a solid foundation for drug treatment. In addition, research on resveratrol, including its mechanism of action, the roles of signalling pathways and its therapeutic targets, will provide new ideas for AD treatment, which is of great significance. In this review, we discuss the possible relationships between AD and the following factors: synapses, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPARs), silent information regulator 1 (SIRT1), and estrogens. We also discuss the findings of previous studies regarding these relationships in the context of AD treatment and further summarize research progress related to resveratrol treatment.
Collapse
Affiliation(s)
- Yan Yan
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Huihuang Yang
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuxun Xie
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Yuanlin Ding
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China
| | - Danli Kong
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| | - Haibing Yu
- The Department of Epidemiology and Health Statistics, Public Health School of Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
8
|
Domanski APF, Booker SA, Wyllie DJA, Isaac JTR, Kind PC. Cellular and synaptic phenotypes lead to disrupted information processing in Fmr1-KO mouse layer 4 barrel cortex. Nat Commun 2019; 10:4814. [PMID: 31645553 PMCID: PMC6811545 DOI: 10.1038/s41467-019-12736-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
Sensory hypersensitivity is a common and debilitating feature of neurodevelopmental disorders such as Fragile X Syndrome (FXS). How developmental changes in neuronal function culminate in network dysfunction that underlies sensory hypersensitivities is unknown. By systematically studying cellular and synaptic properties of layer 4 neurons combined with cellular and network simulations, we explored how the array of phenotypes in Fmr1-knockout (KO) mice produce circuit pathology during development. We show that many of the cellular and synaptic pathologies in Fmr1-KO mice are antagonistic, mitigating circuit dysfunction, and hence may be compensatory to the primary pathology. Overall, the layer 4 network in the Fmr1-KO exhibits significant alterations in spike output in response to thalamocortical input and distorted sensory encoding. This developmental loss of layer 4 sensory encoding precision would contribute to subsequent developmental alterations in layer 4-to-layer 2/3 connectivity and plasticity observed in Fmr1-KO mice, and circuit dysfunction underlying sensory hypersensitivity.
Collapse
Affiliation(s)
- Aleksander P F Domanski
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK.
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
- Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
- Developmental Synaptic Plasticity Section, NINDS, NIH, Bethesda, MD, 20892, USA.
| | - Sam A Booker
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
- Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - David J A Wyllie
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
- Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
- Centre for Brain Development and Repair, NCBS, GKVK Campus, Bangalore, 560065, India
| | - John T R Isaac
- Developmental Synaptic Plasticity Section, NINDS, NIH, Bethesda, MD, 20892, USA.
- Janssen Neuroscience, J&J London Innovation Centre, J&J London Innovation Centre, One Chapel Place, London, W1G 0B, UK.
| | - Peter C Kind
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
- Patrick Wild Centre, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
- Centre for Brain Development and Repair, NCBS, GKVK Campus, Bangalore, 560065, India.
| |
Collapse
|