101
|
Cheng N, Alshammari F, Hughes E, Khanbabaei M, Rho JM. Dendritic overgrowth and elevated ERK signaling during neonatal development in a mouse model of autism. PLoS One 2017; 12:e0179409. [PMID: 28609458 PMCID: PMC5469475 DOI: 10.1371/journal.pone.0179409] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (hereafter referred to as “ASD”) is a heterogeneous neurodevelopmental condition characterized by impaired social communication and interactions, and restricted, repetitive activities or interests. Alterations in network connectivity and memory function are frequently observed in autism patients, often involving the hippocampus. However, specific changes during early brain development leading to disrupted functioning remain largely unclear. Here, we investigated the development of dendritic arbor of hippocampal CA1 pyramidal neurons in the BTBR T+tf/J (BTBR) mouse model of autism. BTBR mice display the defining behavioural features of autism, and also exhibit impaired learning and memory. We found that compared to control C57BL/6J (B6) animals, the lengths of both apical and basal dendrites were significantly greater in neonatal BTBR animals. Further, basal dendrites in the BTBR mice had higher branching complexity. In contrast, cross-sectional area of the soma was unchanged. In addition, we observed a similar density of CA1 pyramidal neurons and thickness of the neuronal layer between the two strains. Thus, there was a specific, compartmentalized overgrowth of dendrites during early development in the BTBR animals. Biochemical analysis further showed that the extracellular signal-regulated kinases (ERK) pathway was up-regulated in the hippocampus of neonatal BTBR animals. Since dendritic structure is critical for information integration and relay, our data suggest that altered development of dendrites could potentially contribute to impaired hippocampal function and behavior observed in the BTBR model, and that this might be related to increased activation of the ERK pathway.
Collapse
Affiliation(s)
- Ning Cheng
- Developmental Neurosciences Research Program, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| | - Fawaz Alshammari
- O’Brien Centre for the Bachelor of Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Elizabeth Hughes
- Developmental Neurosciences Research Program, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Maryam Khanbabaei
- Developmental Neurosciences Research Program, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jong M. Rho
- Departments of Pediatrics, Clinical Neurosciences, Physiology & Pharmacology, Alberta Children’s Hospital Research Institute (ACHRI), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
102
|
Maynard KR, Hobbs JW, Sukumar M, Kardian AS, Jimenez DV, Schloesser RJ, Martinowich K. Bdnf mRNA splice variants differentially impact CA1 and CA3 dendrite complexity and spine morphology in the hippocampus. Brain Struct Funct 2017; 222:3295-3307. [PMID: 28324222 DOI: 10.1007/s00429-017-1405-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/13/2017] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is an activity-dependent neurotrophin critical for neuronal plasticity in the hippocampus. BDNF is encoded by multiple transcripts with alternative 5' untranslated regions (5'UTRS) that display activity-induced targeting to distinct subcellular compartments. While individual Bdnf 5'UTR transcripts influence dendrite morphology in cultured hippocampal neurons, it is unknown whether Bdnf splice variants impact dendrite arborization in functional classes of neurons in the intact hippocampus. Moreover, the contribution of Bdnf 5'UTR splice variants to dendritic spine density and shape has not been explored. We analyzed the structure of CA1 and CA3 dendrite arbors in transgenic mice lacking BDNF production from exon (Ex) 1, 2, 4, or 6 splice variants (Bdnf-e1, -e2, -e4, and -e6-/- mice) and found that loss of BDNF from individual Bdnf mRNA variants differentially impacts the complexity of apical and basal arbors in vivo. Consistent with the subcellular localization studies, Bdnf Ex2 and Ex6 transcripts significantly contributed to dendrite morphology in both CA1 and CA3 neurons. While Bdnf-e2-/- mice showed increased branching proximal to the soma in CA1 and CA3 apical arbors, Bdnf-e6-/- mice showed decreased apical and basal dendrite complexity. Analysis of spine morphology on Bdnf-e6-/- CA1 dendrites revealed changes in the percentage of differently sized spines on apical, but not basal, branches. These results provide further evidence that Bdnf splice variants generate a spatial code that mediates the local actions of BDNF in distinct dendritic compartments on structural and functional plasticity.
Collapse
Affiliation(s)
- Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - John W Hobbs
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Mahima Sukumar
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Alisha S Kardian
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | - Dennisse V Jimenez
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA
| | | | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, 855 North Wolfe Street, Suite 300, Baltimore, MD, 21205, USA. .,Departments of Psychiatry & Behavioral Sciences, and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
103
|
Werhane ML, Evangelista ND, Clark AL, Sorg SF, Bangen KJ, Tran M, Schiehser DM, Delano-Wood L. Pathological vascular and inflammatory biomarkers of acute- and chronic-phase traumatic brain injury. Concussion 2017; 2:CNC30. [PMID: 30202571 PMCID: PMC6094091 DOI: 10.2217/cnc-2016-0022] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022] Open
Abstract
Given the demand for developing objective methods for characterizing traumatic brain injury (TBI), research dedicated to evaluating putative biomarkers has burgeoned over the past decade. Since it is critical to elucidate the underlying pathological processes that underlie the higher diverse outcomes that follow neurotrauma, considerable efforts have been aimed at identifying biomarkers of both the acute- and chronic-phase TBI. Such information is not only critical for helping to elucidate the pathological changes that lead to poor long-term outcomes following TBI but it may also assist in the identification of possible prevention and interventions for individuals who sustain head trauma. In the current review, we discuss the potential role of vascular dysfunction and chronic inflammation in both acute- and chronic-phase TBI, and we also highlight existing studies that have investigated inflammation biomarkers associated with poorer injury outcome.
Collapse
Affiliation(s)
- Madeleine L Werhane
- San Diego State University/University of California, San Diego (SDSU/UC San Diego) Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Center of Excellence for Stress & Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Alexandra L Clark
- San Diego State University/University of California, San Diego (SDSU/UC San Diego) Joint Doctoral Program in Clinical Psychology, San Diego, CA 92120, USA
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Center of Excellence for Stress & Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Scott F Sorg
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Center of Excellence for Stress & Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - Katherine J Bangen
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Center of Excellence for Stress & Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - My Tran
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- San Diego State University (SDSU), San Diego, CA 92182, USA
| | - Dawn M Schiehser
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Center of Excellence for Stress & Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Psychiatry, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| | - Lisa Delano-Wood
- VA San Diego Healthcare System, San Diego, CA 92161, USA
- Center of Excellence for Stress & Mental Health (CESAMH), VA San Diego Healthcare System, San Diego, CA 92161, USA
- Department of Psychiatry, University of California, San Diego (UCSD), La Jolla, CA 92093, USA
| |
Collapse
|
104
|
Parent C, Wen X, Dhir SK, Ryan R, Diorio J, Zhang TY. Maternal care associates with differences in morphological complexity in the medial preoptic area. Behav Brain Res 2017; 326:22-32. [PMID: 28259675 DOI: 10.1016/j.bbr.2017.02.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/10/2017] [Accepted: 02/28/2017] [Indexed: 10/20/2022]
Abstract
The medial preoptic area (MPOA) is implicated in the expression of maternal behavior including the frequency of pup licking/grooming (LG) in the rat. Cyclic adenosine monophosphate (cAMP) responsive element-binding protein (CREB) is a transcription factor that regulates the expression of many genes. We found that lactating rats that are more maternal towards their pups showing increased licking/grooming (i.e. high-LG mothers) had increased levels of phosphorylated CREB (pCREB) in the MPOA following a nursing bout and they displayed a reduced population of greater dendritic complexity index (DCI) neurons compared to less maternal rats showing decreased licking/grooming (i.e. low-LG mothers). CREB overexpression in MPOA neuronal cultures associated with a decrease in dendritic complexity and an increase in the expression of Rem2 and brain-derived neurotrophic factor (BDNF), genes implicated in dendritic pruning. While there were no differences in Rem2 expression in virgin high and low-LG female rats, Rem2 was significantly increased in the MPOA of high-LG compared to low-LG lactating rats. CREB activity in the MPOA associates with maternal behavior and reduced dendritic complexity possibly by increasing Rem2 expression.
Collapse
Affiliation(s)
- Carine Parent
- Sackler Program for Epigenetics and Developmental Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Xianglan Wen
- Sackler Program for Epigenetics and Developmental Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Sabine K Dhir
- Sackler Program for Epigenetics and Developmental Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Richard Ryan
- Sackler Program for Epigenetics and Developmental Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Josie Diorio
- Sackler Program for Epigenetics and Developmental Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H 1R3, Canada
| | - Tie-Yuan Zhang
- Sackler Program for Epigenetics and Developmental Psychobiology at McGill University, Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada.
| |
Collapse
|
105
|
Tabbaa M, Lei K, Liu Y, Wang Z. Paternal deprivation affects social behaviors and neurochemical systems in the offspring of socially monogamous prairie voles. Neuroscience 2017; 343:284-297. [PMID: 27998780 PMCID: PMC5266501 DOI: 10.1016/j.neuroscience.2016.12.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 12/13/2022]
Abstract
Early life experiences, particularly the experience with parents, are crucial to phenotypic outcomes in both humans and animals. Although the effects of maternal deprivation on offspring well-being have been studied, paternal deprivation (PD) has received little attention despite documented associations between father absence and children health problems in humans. In the present study, we utilized the socially monogamous prairie vole (Microtus ochrogaster), which displays male-female pair bonding and bi-parental care, to examine the effects of PD on adult behaviors and neurochemical expression in the hippocampus. Male and female subjects were randomly assigned into one of two experimental groups that grew up with both the mother and father (MF) or with the mother-only (MO, to generate PD experience). Our data show that MO subjects received less parental licking/grooming and carrying and were left alone in the nest more frequently than MF subjects. At adulthood (∼75days of age), MO subjects displayed increased social affiliation (SOA) toward a conspecific compared to MF subjects, but the two groups did not differ in social recognition (SOR) and anxiety-like behavior. Interestingly, MO subjects showed consistent increases in both gene and protein expression of the brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) as well as the levels of total histone 3 and histone 3 acetylation in the hippocampus compared to MF subjects. Further, PD experience increased glucocorticoid receptor beta (GRβ) protein expression in the hippocampus of females as well as increased corticotrophin receptor 2 (CRHR2) protein expression in the hippocampus of males, but decreased CRHR2 mRNA in both sexes. Together, our data suggest that PD has a long-lasting, behavior-specific effect on SOA and alters hippocampal neurochemical systems in the vole brain. The functional role of such altered neurochemical systems in social behaviors and the potential involvement of epigenetic events should be further studied.
Collapse
Affiliation(s)
- Manal Tabbaa
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Kelly Lei
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Yan Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
106
|
Serita T, Fukushima H, Kida S. Constitutive activation of CREB in mice enhances temporal association learning and increases hippocampal CA1 neuronal spine density and complexity. Sci Rep 2017; 7:42528. [PMID: 28195219 PMCID: PMC5307365 DOI: 10.1038/srep42528] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/10/2017] [Indexed: 12/02/2022] Open
Abstract
Transcription factor CREB is believed to play essential roles in the formation of long-term memory (LTM), but not in learning and short-term memory (STM). Surprisingly, we previously showed that transgenic mice expressing a dominant active mutant of CREB (DIEDML) in the forebrain (DIEDML mice) demonstrated enhanced STM and LTM in hippocampal-dependent, rapid, one-trial learning tasks. Here we show that constitutive activation of CREB enhances hippocampal-dependent learning of temporal association in trace fear conditioning and delayed matching-to-place tasks. We then show that in DIEDML mice the apical tuft dendrites of hippocampal CA1 pyramidal neurons, required for temporal association learning, display increased spine density, especially of thin spines and of Homer1-negative spines. In contrast, the basal and apical oblique dendrites of CA1 neurons, required for rapid one-trial learning, show increased density of thin, stubby, and mushroom spines and of Homer1-positive spines. Furthermore, DIEDML mice showed increased dendritic complexity in the proximal portion of apical CA1 dendrites to the soma. In contrast, forebrain overexpression of CaMKIV, leading to enhanced LTM but not STM, show normal learning and CA1 neuron morphology. These findings suggest that dendritic region-specific morphological changes in CA1 neurons by constitutive activation of CREB may contribute to improved learning and STM.
Collapse
Affiliation(s)
- Tatsurou Serita
- Department of Bioscience, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Hotaka Fukushima
- Department of Bioscience, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan.,Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - Satoshi Kida
- Department of Bioscience, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan.,Core Research for Evolutionary Science and Technology (CREST), Japan Science and Technology Agency, Saitama 332-0012, Japan
| |
Collapse
|
107
|
Adaptor Complex 2 Controls Dendrite Morphology via mTOR-Dependent Expression of GluA2. Mol Neurobiol 2017; 55:1590-1606. [PMID: 28190237 PMCID: PMC5820378 DOI: 10.1007/s12035-017-0436-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Accepted: 02/03/2017] [Indexed: 11/26/2022]
Abstract
The formation of dendritic arbors in neurons is a highly regulated process. Among the regulators of dendritogenesis are numerous membrane proteins that are eventually internalized via clathrin-mediated endocytosis. AP2 is an adaptor complex that is responsible for recruiting endocytic machinery to internalized cargo. Its direct involvement in dendritogenesis in mammalian neurons has not yet been tested. We found that the knockdown of AP2b1 (β2-adaptin), an AP2 subunit, reduced the number of dendrites in developing rat hippocampal neurons and decreased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluA2 levels by inhibiting mechanistic/mammalian target of rapamycin (mTOR). The dendritic tree abruption that was caused by AP2b1 knockdown was rescued by the overexpression of GluA2 or restoration of the activity of the mTOR effector p70S6 kinase (S6K1). Altogether, this work provides evidence that the AP2 adaptor complex is needed for the dendritogenesis of mammalian neurons and reveals that mTOR-dependent GluA2 biosynthesis contributes to this process.
Collapse
|
108
|
Kimura E, Tohyama C. Embryonic and Postnatal Expression of Aryl Hydrocarbon Receptor mRNA in Mouse Brain. Front Neuroanat 2017; 11:4. [PMID: 28223923 PMCID: PMC5293765 DOI: 10.3389/fnana.2017.00004] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 01/17/2017] [Indexed: 12/21/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR), a member of the basic helix-loop-helix-Per-Arnt-Sim transcription factor family, plays a critical role in the developing nervous system of invertebrates and vertebrates. Dioxin, a ubiquitous environmental pollutant, avidly binds to this receptor, and maternal exposure to dioxin has been shown to impair higher brain functions and dendritic morphogenesis, possibly via an AhR-dependent mechanism. However, there is little information on AhR expression in the developing mammalian brain. To address this issue, the present study analyzed AhR mRNA expression in the brains of embryonic, juvenile, and adult mice by reverse transcription (RT)-PCR and in situ hybridization. In early brain development (embryonic day 12.5), AhR transcript was detected in the innermost cortical layer. The mRNA was also expressed in the hippocampus, cerebral cortex, cerebellum, olfactory bulb, and rostral migratory stream on embryonic day 18.5, postnatal days 3, 7, and 14, and in 12-week-old (adult) mice. Hippocampal expression was abundant in the CA1 and CA3 pyramidal and dentate gyrus granule cell layers, where expression level of AhR mRNA in 12-week old is higher than that in 7-day old. These results reveal temporal and spatial patterns of AhR mRNA expression in the mouse brain, providing the information that may contribute to the elucidation of the physiologic and toxicologic significance of AhR in the developing brain.
Collapse
Affiliation(s)
- Eiki Kimura
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of TokyoTokyo, Japan; Environmental Biology Laboratory, Faculty of Medicine, University of TsukubaTsukuba, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of TokyoTokyo, Japan; Environmental Biology Laboratory, Faculty of Medicine, University of TsukubaTsukuba, Japan
| |
Collapse
|
109
|
Ruan H, Gao J, Qi X, Tao Y, Guo X, Guo Z, Zheng L, Song Y, Liao Y, Shen W. Visual experience dependent regulation of neuronal structure and function by histone deacetylase 1 in developing Xenopus tectum in vivo. Dev Neurobiol 2017; 77:947-962. [PMID: 28033671 DOI: 10.1002/dneu.22480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/13/2016] [Accepted: 12/16/2016] [Indexed: 01/28/2023]
Abstract
Histone deacetylase 1 (HDAC1) is thought to play pivotal roles in neurogenesis and neurodegeneration. However, the role of HDAC1 in neuronal growth and structural plasticity in the developing brain in vivo remains unclear. Here, we show that in the optic tectum of Xenopus laevis, HDAC1 knockdown dramatically decreased the frequency of AMPAR-mediated synaptic currents and increased the frequency of GABAAR-mediated currents, whereas HDAC1 overexpression significantly decreased the frequency of GABAAR-mediated synaptic currents. Both HDAC1 knockdown and overexpression adversely affected dendritic arbor growth and visual experience-dependent structural plasticity. Furthermore, HDAC1 knockdown decreased BDNF expression via a mechanism that involves acetylation of specific histone H4 residues at lysine K5. In particular, the deficits in dendritic growth and visually guided avoidance behavior in HDAC1-knockdown tadpoles could be rescued by acute tectal infusion of BDNF. These results establish a relationship between HDAC1 expression, histone H4 modification and BDNF signaling in the visual-experience dependent regulation of dendritic growth, structural plasticity and function in intact animals in vivo. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 947-962, 2017.
Collapse
Affiliation(s)
- Hangze Ruan
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Juanmei Gao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Xianjie Qi
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yi Tao
- Department of Neurosurgery, Nanjing Medical University Affiliated Jiangsu Cancer Hospital, Nanjing, Jiangsu, 210029, China
| | - Xia Guo
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Zhaoyi Guo
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Lijun Zheng
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yaling Song
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yuan Liao
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Wanhua Shen
- Zhejiang Key Laboratory of Organ Development and Regeneration, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| |
Collapse
|
110
|
Nie J, Yang X. Modulation of Synaptic Plasticity by Exercise Training as a Basis for Ischemic Stroke Rehabilitation. Cell Mol Neurobiol 2017; 37:5-16. [PMID: 26910247 PMCID: PMC11482112 DOI: 10.1007/s10571-016-0348-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/11/2016] [Indexed: 12/23/2022]
Abstract
In recent years, rehabilitation of ischemic stroke draws more and more attention in the world, and has been linked to changes of synaptic plasticity. Exercise training improves motor function of ischemia as well as cognition which is associated with formation of learning and memory. The molecular basis of learning and memory might be synaptic plasticity. Research has therefore been conducted in an attempt to relate effects of exercise training to neuroprotection and neurogenesis adjacent to the ischemic injury brain. The present paper reviews the current literature addressing this question and discusses the possible mechanisms involved in modulation of synaptic plasticity by exercise training. This review shows the pathological process of synaptic dysfunction in ischemic roughly and then discusses the effects of exercise training on scaffold proteins and regulatory protein expression. The expression of scaffold proteins generally increased after training, but the effects on regulatory proteins were mixed. Moreover, the compositions of postsynaptic receptors were changed and the strength of synaptic transmission was enhanced after training. Finally, the recovery of cognition is critically associated with synaptic remodeling in an injured brain, and the remodeling occurs through a number of local regulations including mRNA translation, remodeling of cytoskeleton, and receptor trafficking into and out of the synapse. We do provide a comprehensive knowledge of synaptic plasticity enhancement obtained by exercise training in this review.
Collapse
Affiliation(s)
- Jingjing Nie
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China
| | - Xiaosu Yang
- Department of Neurology, Xiang Ya Hospital, Central South University, Xiang Ya Road 87, Changsha, 410008, Hunan, China.
| |
Collapse
|
111
|
Saligan L, Lukkahatai N, Holder G, Walitt B, Machado-Vieira R. Lower brain-derived neurotrophic factor levels associated with worsening fatigue in prostate cancer patients during repeated stress from radiation therapy. World J Biol Psychiatry 2016; 17:608-614. [PMID: 25815565 PMCID: PMC4751064 DOI: 10.3109/15622975.2015.1012227] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Fatigue during cancer treatment is associated with depression. Neurotrophic factors play a major role in depression and stress and might provide insight into mechanisms of fatigue. This study investigated the association between plasma concentrations of three neurotrophic factors (BDNF, brain-derived neurotrophic factor; GDNF, glial-derived neurotrophic factor; and SNAPIN, soluble N-ethylmaleimide sensitive fusion attachment receptor-associated protein) and initial fatigue intensification during external beam radiation therapy (EBRT) in euthymic non-metastatic prostate cancer men. METHODS Fatigue, as measured by the 13-item Functional Assessment of Cancer Therapy-Fatigue (FACT-F), and plasma neurotrophic factors were collected at baseline (prior to EBRT) and mid-EBRT. Subjects were categorized into fatigue and no fatigue groups using a > 3-point change in FACT-F scores between the two time points. Multiple linear regressions analysed the associations between fatigue and neurotrophic factors. RESULTS FACT-F scores of 47 subjects decreased from baseline (43.95 ± 1.3) to mid-EBRT (38.36 ± 1.5, P < 0.001), indicating worsening fatigue. SNAPIN levels were associated with fatigue scores (rs = 0.43, P = 0.005) at baseline. A significant decrease of BDNF concentration (P = 0.008) was found in fatigued subjects during EBRT (n = 39). CONCLUSIONS Baseline SNAPIN and decreasing BDNF levels may influence worsening fatigue during EBRT. Further investigations are warranted to confirm their role in the pathophysiology and therapeutics of fatigue.
Collapse
Affiliation(s)
- L.N. Saligan
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - N. Lukkahatai
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA,University of Nevada, Las Vegas, School of Nursing, Las Vegas, NV, USA
| | - G. Holder
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - B. Walitt
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - R. Machado-Vieira
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
112
|
Bezchlibnyk YB, Stone SSD, Hamani C, Lozano AM. High frequency stimulation of the infralimbic cortex induces morphological changes in rat hippocampal neurons. Brain Stimul 2016; 10:315-323. [PMID: 27964870 DOI: 10.1016/j.brs.2016.11.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/03/2016] [Accepted: 11/21/2016] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Although a significant subset of patients with major depressive disorder (MDD) fail to respond to medical or behavioural therapy, deep brain stimulation (DBS) applied to the subgenual cingulate cortex (SCC; sg25) has been shown to reduce depressive symptoms in a subset of patients. This area receives projections from neurons in the CA1 region and subiculum of the hippocampus (HC), a brain region implicated in the pathobiology and treatment of MDD. OBJECTIVE To assess whether high frequency stimulation (HFS) of the infralimbic cortex is associated with changes in cellular morphology in the HC. METHODS Rats were subjected to either infralimbic HFS or sham-stimulation. Measures of cellular morphology, including dendritic length and complexity, were assessed in pyramidal neurons in the CA1 region of the HC by means of the Golgi-Cox histological stain. RESULTS Dendritic length (p = 0.013) and number of branch points (p = 0.004) were significantly increased across the entire dendritic tree in animals subjected to HFS. Subsequent Scholl analysis revealed that for dendritic length these effects were localized to the region between 80 and 160 μm from the soma (p < 0.001 for either 40 μm interval) in the basal dendritic tree, while branch point number was predominantly increased between 120 and 160 μm from the soma (p < 0.001) in the apical dendritic tree. CONCLUSIONS High-frequency stimulation of the infralimbic cortex increases the complexity of apical dendrites and the length of basal dendritic trees of pyramidal neurons located in the CA1 hippocampal subfield relative to sham-stimulated animals.
Collapse
Affiliation(s)
- Yarema B Bezchlibnyk
- Department of Neurosurgery, Emory University Hospital, Atlanta, GA, United States
| | - Scellig S D Stone
- Harvard Medical School, Boston, MA, United States; Department of Neurosurgery, Boston Children's Hospital, Boston, MA, United States
| | - Clement Hamani
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Toronto Western Research Institute, Krembil Discovery Tower, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
113
|
Galati DF, Hiester BG, Jones KR. Computer Simulations Support a Morphological Contribution to BDNF Enhancement of Action Potential Generation. Front Cell Neurosci 2016; 10:209. [PMID: 27683544 PMCID: PMC5021759 DOI: 10.3389/fncel.2016.00209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/22/2016] [Indexed: 01/10/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) regulates both action potential (AP) generation and neuron morphology. However, whether BDNF-induced changes in neuron morphology directly impact AP generation is unclear. We quantified BDNF’s effect on cultured cortical neuron morphological parameters and found that BDNF stimulates dendrite growth and addition of dendrites while increasing both excitatory and inhibitory presynaptic inputs in a spatially restricted manner. To gain insight into how these combined changes in neuron structure and synaptic input impact AP generation, we used the morphological parameters we gathered to generate computational models. Simulations suggest that BDNF-induced neuron morphologies generate more APs under a wide variety of conditions. Synapse and dendrite addition have the greatest impact on AP generation. However, subtle alterations in excitatory/inhibitory synapse ratio and strength have a significant impact on AP generation when synaptic activity is low. Consistent with these simulations, BDNF rapidly enhances spontaneous activity in cortical cultures. We propose that BDNF promotes neuron morphologies that are intrinsically more efficient at translating barrages of synaptic activity into APs, which is a previously unexplored aspect of BDNF’s function.
Collapse
Affiliation(s)
- Domenico F Galati
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder Boulder, CO, USA
| | - Brian G Hiester
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder Boulder, CO, USA
| | - Kevin R Jones
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder Boulder, CO, USA
| |
Collapse
|
114
|
Nair B, Wong-Riley MTT. Transcriptional Regulation of Brain-derived Neurotrophic Factor Coding Exon IX: ROLE OF NUCLEAR RESPIRATORY FACTOR 2. J Biol Chem 2016; 291:22583-22593. [PMID: 27624937 DOI: 10.1074/jbc.m116.742304] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/09/2016] [Indexed: 01/22/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is an active neurotrophin abundantly expressed throughout the nervous system. It plays an important role in synaptic transmission, plasticity, neuronal proliferation, differentiation, survival, and death. The Bdnf gene in rodents has eight non-coding exons and only a single coding exon (IX). Despite its recognized regulation by neuronal activity, relatively little is known about its transcriptional regulation, and even less about the transcription factor candidates that may play such a role. The goal of the present study was to probe for such a candidate that may regulate exon IX in the rat Bdnf gene. Our in silico analysis revealed tandem binding sites for nuclear respiratory factor 2 (NRF-2) on the promoter of exon IX. NRF-2 is of special significance because it co-regulates the expressions of mediators of energy metabolism (cytochrome c oxidase) and mediators of neuronal activity (glutamatergic receptors). To test our hypothesis that NRF-2 also regulates the Bdnf gene, we performed electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP), promoter cloning, and site-directed mutagenesis, real-time quantitative PCR (RT-qPCR), and Western blotting analysis. Results indicate that NRF-2 functionally regulates exon IX of the rat Bdnf gene. The binding sites of NRF-2 are conserved between rats and mice. Overexpressing NRF-2 up-regulated the expression of Bdnf exon IX, whereas knocking down NRF-2 down-regulated such expression. These findings are consistent with our hypothesis that NRF-2, in addition to regulating the coupling between neuronal activity and energy metabolism, also regulates the expression of BDNF, which is intimately associated with energy-demanding neuronal activity.
Collapse
Affiliation(s)
- Bindu Nair
- From the Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Margaret T T Wong-Riley
- From the Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
115
|
The Medial Orbitofrontal Cortex Regulates Sensitivity to Outcome Value. J Neurosci 2016; 36:4600-13. [PMID: 27098701 DOI: 10.1523/jneurosci.4253-15.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/08/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED An essential component of goal-directed decision-making is the ability to maintain flexible responding based on the value of a given reward, or "reinforcer." The medial orbitofrontal cortex (mOFC), a subregion of the ventromedial prefrontal cortex, is uniquely positioned to regulate this process. We trained mice to nose poke for food reinforcers and then stimulated this region using CaMKII-driven Gs-coupled designer receptors exclusively activated by designer drugs (DREADDs). In other mice, we silenced the neuroplasticity-associated neurotrophin brain-derived neurotrophic factor (BDNF). Activation of Gs-DREADDs increased behavioral sensitivity to reinforcer devaluation, whereas Bdnf knockdown blocked sensitivity. These changes were accompanied by modifications in breakpoint ratios in a progressive ratio task, and they were recapitulated in Bdnf(+/-)mice. Replacement of BDNF selectively in the mOFC in Bdnf(+/-)mice rescued behavioral deficiencies, as well as phosphorylation of extracellular-signal regulated kinase 1/2 (ERK1/2). Thus, BDNF expression in the mOFC is both necessary and sufficient for the expression of typical effort allocation relative to an anticipated reinforcer. Additional experiments indicated that expression of the immediate-early gene c-fos was aberrantly elevated in the Bdnf(+/-)dorsal striatum, and BDNF replacement in the mOFC normalized expression. Also, systemic administration of an MAP kinase kinase inhibitor increased breakpoint ratios, whereas the addition of discrete cues bridging the response-outcome contingency rescued breakpoints in Bdnf(+/-)mice. We argue that BDNF-ERK1/2 in the mOFC is a key regulator of "online" goal-directed action selection. SIGNIFICANCE STATEMENT Goal-directed response selection often involves predicting the consequences of one's actions and the value of potential payoffs. Lesions or chemogenetic inactivation of the medial orbitofrontal cortex (mOFC) in rats induces failures in retrieving outcome identity memories (Bradfield et al., 2015), suggesting that the healthy mOFC serves to access outcome value information when it is not immediately observable and thereby guide goal-directed decision-making. Our findings suggest that the mOFC also bidirectionally regulates effort allocation for a given reward and that expression of the neurotrophin BDNF in the mOFC is both necessary and sufficient for mice to sustain stable representations of reinforcer value.
Collapse
|
116
|
Ledda F, Paratcha G. Assembly of Neuronal Connectivity by Neurotrophic Factors and Leucine-Rich Repeat Proteins. Front Cell Neurosci 2016; 10:199. [PMID: 27555809 PMCID: PMC4977320 DOI: 10.3389/fncel.2016.00199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/29/2016] [Indexed: 11/13/2022] Open
Abstract
Proper function of the nervous system critically relies on sophisticated neuronal networks interconnected in a highly specific pattern. The architecture of these connections arises from sequential developmental steps such as axonal growth and guidance, dendrite development, target determination, synapse formation and plasticity. Leucine-rich repeat (LRR) transmembrane proteins have been involved in cell-type specific signaling pathways that underlie these developmental processes. The members of this superfamily of proteins execute their functions acting as trans-synaptic cell adhesion molecules involved in target specificity and synapse formation or working in cis as cell-intrinsic modulators of neurotrophic factor receptor trafficking and signaling. In this review, we will focus on novel physiological mechanisms through which LRR proteins regulate neurotrophic factor receptor signaling, highlighting the importance of these modulatory events for proper axonal extension and guidance, tissue innervation and dendrite morphogenesis. Additionally, we discuss few examples linking this set of LRR proteins to neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine-University of Buenos Aires (UBA) Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET, School of Medicine-University of Buenos Aires (UBA) Buenos Aires, Argentina
| |
Collapse
|
117
|
Binley KE, Ng WS, Barde YA, Song B, Morgan JE. Brain-derived neurotrophic factor prevents dendritic retraction of adult mouse retinal ganglion cells. Eur J Neurosci 2016; 44:2028-39. [PMID: 27285957 PMCID: PMC4988502 DOI: 10.1111/ejn.13295] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 06/02/2016] [Accepted: 06/06/2016] [Indexed: 01/24/2023]
Abstract
We used cultured adult mouse retinae as a model system to follow and quantify the retraction of dendrites using diolistic labelling of retinal ganglion cells (RGCs) following explantation. Cell death was monitored in parallel by nuclear staining as ‘labelling’ with RGC and apoptotic markers was inconsistent and exceedingly difficult to quantify reliably. Nuclear staining allowed us to delineate a lengthy time window during which dendrite retraction can be monitored in the absence of RGC death. The addition of brain‐derived neurotrophic factor (BDNF) produced a marked reduction in dendritic degeneration, even when application was delayed for 3 days after retinal explantation. These results suggest that the delayed addition of trophic factors may be functionally beneficial before the loss of cell bodies in the course of conditions such as glaucoma.
Collapse
Affiliation(s)
- Kate E Binley
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Wai S Ng
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Yves-Alain Barde
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Cardiff, UK
| | - Bing Song
- School of Dentistry, Cardiff University, Heath Park, Cardiff, UK
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, CF24 4HQ, UK
| |
Collapse
|
118
|
The neurotrophin receptor p75 mediates gp120-induced loss of synaptic spines in aging mice. Neurobiol Aging 2016; 46:160-8. [PMID: 27498053 DOI: 10.1016/j.neurobiolaging.2016.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 03/28/2016] [Accepted: 07/01/2016] [Indexed: 01/12/2023]
Abstract
Human immunodeficiency virus 1 and its envelope protein gp120 reduce synaptodendritic complexity. However, the mechanisms contributing to this pathological feature are still not understood. The proneurotrophin brain-derived neurotrophic factor promotes synaptic simplification through the activation of the p75 neurotrophin receptor (p75NTR). Here, we have used gp120 transgenic (gp120tg) mice to investigate whether p75NTR has a role in gp120-mediated neurotoxicity. Old (∼10 months) gp120tg mice exhibited an increase in proneurotrophin brain-derived neurotrophic factor levels in the hippocampus as well as a decrease in the number of dendritic spines when compared to age-matched wild type. These effects were not observed in 3- or 6-month-old mice. To test if the reduction in spine density and morphology is caused by the activation of p75NTR, we crossed gp120tg mice with p75NTR null mice. We found that deletion of only 1 copy of the p75NTR gene in gp120tg mice is sufficient to normalize the number of hippocampal spines, strongly suggesting that the neurotoxic effect of gp120 is mediated by p75NTR. These data indicate that p75NTR antagonists could provide an adjunct therapy against synaptic simplification caused by human immunodeficiency virus 1.
Collapse
|
119
|
Peng Y, Lu Z, Li G, Piechowicz M, Anderson M, Uddin Y, Wu J, Qiu S. The autism-associated MET receptor tyrosine kinase engages early neuronal growth mechanism and controls glutamatergic circuits development in the forebrain. Mol Psychiatry 2016; 21:925-35. [PMID: 26728565 PMCID: PMC4914424 DOI: 10.1038/mp.2015.182] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/30/2015] [Accepted: 09/08/2015] [Indexed: 12/18/2022]
Abstract
The human MET gene imparts a replicated risk for autism spectrum disorder (ASD), and is implicated in the structural and functional integrity of brain. MET encodes a receptor tyrosine kinase, MET, which has a pleiotropic role in embryogenesis and modifies a large number of neurodevelopmental events. Very little is known, however, on how MET signaling engages distinct cellular events to collectively affect brain development in ASD-relevant disease domains. Here, we show that MET protein expression is dynamically regulated and compartmentalized in developing neurons. MET is heavily expressed in neuronal growth cones at early developmental stages and its activation engages small GTPase Cdc42 to promote neuronal growth, dendritic arborization and spine formation. Genetic ablation of MET signaling in mouse dorsal pallium leads to altered neuronal morphology indicative of early functional maturation. In contrast, prolonged activation of MET represses the formation and functional maturation of glutamatergic synapses. Moreover, manipulating MET signaling levels in vivo in the developing prefrontal projection neurons disrupts the local circuit connectivity made onto these neurons. Therefore, normal time-delimited MET signaling is critical in regulating the timing of neuronal growth, glutamatergic synapse maturation and cortical circuit function. Dysregulated MET signaling may lead to pathological changes in forebrain maturation and connectivity, and thus contribute to the emergence of neurological symptoms associated with ASD.
Collapse
Affiliation(s)
- Yun Peng
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004
| | - Zhongming Lu
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004,Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China, 210009
| | - Guohui Li
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004,Interdisciplinary Graduate Program in Neuroscience, School of Life Science, Arizona State University. Tempe, AZ 85287
| | - Mariel Piechowicz
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004
| | - Miranda Anderson
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004
| | - Yasin Uddin
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004
| | - Jie Wu
- Division of Neurology, Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, AZ 85013
| | - Shenfeng Qiu
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004,Interdisciplinary Graduate Program in Neuroscience, School of Life Science, Arizona State University. Tempe, AZ 85287
| |
Collapse
|
120
|
Abstract
The neurotrophin brain-derived neurotrophic factor (BDNF) has recently emerged as a possible molecular mediator of activity-dependent synaptic plasticity underlying learning and memory. Long-term potentiation (LTP) within the hippocampus and hippocampally dependent behaviors has been the primary model for examining the role of BDNF in learning and memory. However, these studies are limited by an incomplete understanding of the complex behavioral function of hippocampal circuitry, making it difficult to unravel the molecular machinery responsible for the formation and storage of these memories. In contrast, the amygdala and its role in Pavlovian fear conditioning promise to provide us with new insights into the mechanisms of BDNF-mediated synaptic plasticity during the learning and memory process. This article reviews the different levels of research on BDNF in learning and memory. The focus is primarily on the use of Pavlovian fear conditioning as a learning model that allows for the examination of the role of BDNF in the amygdala, following a single learning session and within a well-understood neural circuit.
Collapse
Affiliation(s)
- Lisa M Rattiner
- Emory University School of Medicine, Department of Psychiatry, Center for Behavioral Neuroscience, Atlanta, Georgia 30329, USA
| | | | | |
Collapse
|
121
|
Chomiak T, Hung J, Nguyen MD, Hu B. Somato-dendritic decoupling as a novel mechanism for protracted cortical maturation. BMC Biol 2016; 14:48. [PMID: 27328836 PMCID: PMC4916537 DOI: 10.1186/s12915-016-0270-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/06/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Both human and animal data indicate that disruption of the endogenously slow maturation of temporal association cortical (TeA) networks is associated with abnormal higher order cognitive development. However, the neuronal mechanisms underlying the endogenous maturation delay of the TeA are poorly understood. RESULTS Here we report a novel form of developmental plasticity that is present in the TeA. It was found that deep layer TeA neurons, but not hippocampal or primary visual neurons, exist in a protracted 'embryonic-like' state through a mechanism involving reduced somato-dendritic communication and a non-excitable somatic membrane. This mechanism of neural inactivity is present in intact tissue and shows a remarkable transition into an active somato-dendritically coupled state. The quantity of decoupled cells diminishes in a protracted and age-dependent manner, continuing into adolescence. CONCLUSIONS Based on our data, we propose a model of neural plasticity through which protracted compartmentalization and decoupling in somato-dendritic signalling plays a key role in controlling how excitable neurons are incorporated into recurrent cortical networks independent of neurogenesis.
Collapse
Affiliation(s)
- Taylor Chomiak
- Division of Translational Neuroscience, Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| | - Johanna Hung
- Division of Translational Neuroscience, Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Minh Dang Nguyen
- Division of Translational Neuroscience, Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Bin Hu
- Division of Translational Neuroscience, Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
122
|
Foltran RB, Diaz SL. BDNF isoforms: a round trip ticket between neurogenesis and serotonin? J Neurochem 2016; 138:204-21. [PMID: 27167299 DOI: 10.1111/jnc.13658] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/08/2016] [Accepted: 05/02/2016] [Indexed: 12/12/2022]
Abstract
The brain-derived neurotrophic factor, BDNF, was discovered more than 30 years ago and, like other members of the neurotrophin family, this neuropeptide is synthetized as a proneurotrophin, the pro-BDNF, which is further cleaved to yield mature BDNF. The myriad of actions of these two BDNF isoforms in the central nervous system is constantly increasing and requires the development of sophisticated tools and animal models to refine our understanding. This review is focused on BDNF isoforms, their participation in the process of neurogenesis taking place in the hippocampus of adult mammals, and the modulation of their expression by serotonergic agents. Interestingly, around this triumvirate of BDNF, serotonin, and neurogenesis, a series of recent research has emerged with apparently counterintuitive results. This calls for an exhaustive analysis of the data published so far and encourages thorough work in the quest for new hypotheses in the field. BDNF is synthetized as a pre-proneurotrophin. After removal of the pre-region, proBDNF can be cleaved by intracellular or extracellular proteases. Mature BDNF can bind TrkB receptors, promoting their homodimerization and intracellular phosphorylation. Phosphorylated-TrkB can activate three different signaling pathways. Whereas G-protein-coupled receptors can transactivate TrkB receptors, truncated forms can inhibit mBDNF signaling. Pro-BDNF binds p75(NTR) by its mature domain, whereas the pro-region binds co-receptors.
Collapse
Affiliation(s)
- Rocío Beatriz Foltran
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Fac. de Medicina - UBA, Buenos Aires, Argentina
| | - Silvina Laura Diaz
- Instituto de Biología Celular y Neurociencias Prof. E. De Robertis, CONICET-UBA, Fac. de Medicina - UBA, Buenos Aires, Argentina
| |
Collapse
|
123
|
Kumari A, Singh P, Baghel MS, Thakur M. Social isolation mediated anxiety like behavior is associated with enhanced expression and regulation of BDNF in the female mouse brain. Physiol Behav 2016; 158:34-42. [DOI: 10.1016/j.physbeh.2016.02.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 01/04/2023]
|
124
|
Impairments in dendrite morphogenesis as etiology for neurodevelopmental disorders and implications for therapeutic treatments. Neurosci Biobehav Rev 2016; 68:946-978. [PMID: 27143622 DOI: 10.1016/j.neubiorev.2016.04.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 02/08/2023]
Abstract
Dendrite morphology is pivotal for neural circuitry functioning. While the causative relationship between small-scale dendrite morphological abnormalities (shape, density of dendritic spines) and neurodevelopmental disorders is well established, such relationship remains elusive for larger-scale dendrite morphological impairments (size, shape, branching pattern of dendritic trees). Here, we summarize published data on dendrite morphological irregularities in human patients and animal models for neurodevelopmental disorders, with focus on autism and schizophrenia. We next discuss high-risk genes for these disorders and their role in dendrite morphogenesis. We finally overview recent developments in therapeutic attempts and we discuss how they relate to dendrite morphology. We find that both autism and schizophrenia are accompanied by dendritic arbor morphological irregularities, and that majority of their high-risk genes regulate dendrite morphogenesis. Thus, we present a compelling argument that, along with smaller-scale morphological impairments in dendrites (spines and synapse), irregularities in larger-scale dendrite morphology (arbor shape, size) may be an important part of neurodevelopmental disorders' etiology. We suggest that this should not be ignored when developing future therapeutic treatments.
Collapse
|
125
|
Zhao S, Yu A, Wang X, Gao X, Chen J. Post-Injury Treatment of 7,8-Dihydroxyflavone Promotes Neurogenesis in the Hippocampus of the Adult Mouse. J Neurotrauma 2016; 33:2055-2064. [PMID: 26715291 DOI: 10.1089/neu.2015.4036] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) at the moderate level of impact induces massive cell death and results in extensive dendrite degeneration in the brain, leading to persistent cognitive, sensory, and motor dysfunction. Our previous reports have shown that adult-born immature granular neurons in the dentate gyrus are the most vulnerable cell type in the hippocampus after receiving a moderate TBI with a controlled cortical impact (CCI) device. There is no effective approach to prevent immature neuron death or degeneration following TBI. Our recent study found that pretreatment of 7,8-dihydroxyflavone (DHF), a small molecule imitating brain-derived neurotrophic factor, protected immature neurons in the hippocampus from death following TBI. In the present study, we systemically treated moderate CCI-TBI mice or sham surgery mice with DHF once a day for 2 weeks via intraperitoneal injection, and then assessed the immature neurons in the hippocampus the 2nd day after the last DHF injection. We found that post-injury treatment of DHF for 2 weeks not only increased the number of adult-born immature neurons in the hippocampus, but also promoted their dendrite arborization in the injured brain following TBI. Thus, DHF may be a promising compound that can promote neurogenesis and enhance immature neuron development following TBI.
Collapse
Affiliation(s)
- Shu Zhao
- 1 Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Department of Neurosurgery, Indiana University , Indianapolis, Indiana
| | - Alex Yu
- 2 Carmel High School , Indianapolis, Indiana
| | - Xiaoting Wang
- 1 Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Department of Neurosurgery, Indiana University , Indianapolis, Indiana
| | - Xiang Gao
- 1 Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Department of Neurosurgery, Indiana University , Indianapolis, Indiana
| | - Jinhui Chen
- 1 Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Department of Neurosurgery, Indiana University , Indianapolis, Indiana
| |
Collapse
|
126
|
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
|
127
|
Kalirin is required for BDNF-TrkB stimulated neurite outgrowth and branching. Neuropharmacology 2016; 107:227-238. [PMID: 27036892 DOI: 10.1016/j.neuropharm.2016.03.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 03/03/2016] [Accepted: 03/28/2016] [Indexed: 01/19/2023]
Abstract
Exogenous brain-derived neurotrophic factor (BDNF), acting through TrkB, is known to promote neurite formation and branching. This response to BDNF was eliminated by inhibition of TrkB kinase and by specific inhibition of the GEF1 domain of Kalirin, which activates Rac1. Neurons from Kalrn knockout mice were unable to activate Rac1 in response to BDNF. BDNF-triggered neurite outgrowth was abolished when Kalrn expression was reduced using shRNA that targets all of the major Kalrn isoforms, and reduced in neurons from Kalrn knockout mice. The Kalrn isoforms expressed early in development also include a GEF2 domain that activates RhoA. However, BDNF-stimulated neurite outgrowth in Kalrn knockout neurons was rescued by expression of Kalirin-7, which includes only the GEF1 domain but lacks the GEF2 domain. Dendritic morphogenesis, which requires spatially restricted, coordinated changes in the actin cytoskeleton and in the organization of microtubules, involves essential contributions from multiple Rho GEFs. Since Tiam1, another Rho GEF, is also required for BDNF-stimulated neurite outgrowth, an inhibitory fragment of Tiam1 (PHn-CC-EX) was tested and found to interfere with both Kalirin and Tiam1 GEF activity. The prolonged TrkB activation observed in response to BDNF in Kalrn knockout neurons and the altered time course and extent of ERK, CREB and Akt activation observed in the absence of Kalrn would be expected to alter the response of these neurons to other regulatory factors.
Collapse
|
128
|
Alsina FC, Hita FJ, Fontanet PA, Irala D, Hedman H, Ledda F, Paratcha G. Lrig1 is a cell-intrinsic modulator of hippocampal dendrite complexity and BDNF signaling. EMBO Rep 2016; 17:601-16. [PMID: 26935556 DOI: 10.15252/embr.201541218] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/28/2016] [Indexed: 11/09/2022] Open
Abstract
Even though many extracellular factors have been identified as promoters of general dendritic growth and branching, little is known about the cell-intrinsic modulators that allow neurons to sculpt distinctive patterns of dendrite arborization. Here, we identify Lrig1, a nervous system-enriched LRR protein, as a key physiological regulator of dendrite complexity of hippocampal pyramidal neurons. Lrig1-deficient mice display morphological changes in proximal dendrite arborization and defects in social interaction. Specifically, knockdown of Lrig1 enhances both primary dendrite formation and proximal dendritic branching of hippocampal neurons, two phenotypes that resemble the effect of BDNF on these neurons. In addition, we show that Lrig1 physically interacts with TrkB and attenuates BDNF signaling. Gain and loss of function assays indicate that Lrig1 restricts BDNF-induced dendrite morphology. Together, our findings reveal a novel and essential role of Lrig1 in regulating morphogenic events that shape the hippocampal circuits and establish that the assembly of TrkB with Lrig1 represents a key mechanism for understanding how specific neuronal populations expand the repertoire of responses to BDNF during brain development.
Collapse
Affiliation(s)
- Fernando Cruz Alsina
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Francisco Javier Hita
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Paula Aldana Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Dolores Irala
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Håkan Hedman
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Fernanda Ledda
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Gustavo Paratcha
- Division of Molecular and Cellular Neuroscience, Institute of Cell Biology and Neuroscience (IBCN)-CONICET School of Medicine University of Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
129
|
Kim JY, Choi K, Shaker MR, Lee JH, Lee B, Lee E, Park JY, Lim MS, Park CH, Shin KS, Kim H, Geum D, Sun W. Promotion of Cortical Neurogenesis from the Neural Stem Cells in the Adult Mouse Subcallosal Zone. Stem Cells 2016; 34:888-901. [DOI: 10.1002/stem.2276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 10/19/2015] [Accepted: 11/17/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Joo Yeon Kim
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Kyuhyun Choi
- Department of Biology; Department of Life and Nanopharmaceutical Sciences; Kyung Hee University; Seoul Republic of Korea
| | - Mohammed R. Shaker
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Ju-Hyun Lee
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Boram Lee
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Eunsoo Lee
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University; Seoul Republic of Korea
| | - Mi-Sun Lim
- Graduate School of Biomedical Science and Engineering
- Hanyang Biomedical Research Institute
| | - Chang-Hwan Park
- Graduate School of Biomedical Science and Engineering
- Hanyang Biomedical Research Institute
- Department of Microbiology; College of Medicine, Hanyang University; Seoul Korea
| | - Ki Soon Shin
- Department of Biology; Department of Life and Nanopharmaceutical Sciences; Kyung Hee University; Seoul Republic of Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| | - Dongho Geum
- Department of Biomedical Sciences; Korea University College of Medicine; Seoul Korea
| | - Woong Sun
- Department of Anatomy and Division of Brain Korea 21 Plus Biomedical Science; Korea University College of Medicine; Seoul Korea
| |
Collapse
|
130
|
Chattarji S, Tomar A, Suvrathan A, Ghosh S, Rahman MM. Neighborhood matters: divergent patterns of stress-induced plasticity across the brain. Nat Neurosci 2015; 18:1364-75. [PMID: 26404711 DOI: 10.1038/nn.4115] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023]
Abstract
The fact that exposure to severe stress leads to the development of psychiatric disorders serves as the basic rationale for animal models of stress disorders. Clinical and neuroimaging studies have shown that three brain areas involved in learning and memory--the hippocampus, amygdala and prefrontal cortex--undergo distinct structural and functional changes in individuals with stress disorders. These findings from patient studies pose several challenges for animal models of stress disorders. For instance, why does stress impair cognitive function, yet enhance fear and anxiety? Can the same stressful experience elicit contrasting patterns of plasticity in the hippocampus, amygdala and prefrontal cortex? How does even a brief exposure to traumatic stress lead to long-lasting behavioral abnormalities? Thus, animal models of stress disorders must not only capture the unique spatio-temporal features of structural and functional alterations in these brain areas, but must also provide insights into the underlying neuronal plasticity mechanisms. This Review will address some of these key questions by describing findings from animal models on how stress-induced plasticity varies across different brain regions and thereby gives rise to the debilitating emotional and cognitive symptoms of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Sumantra Chattarji
- Centre for Brain Development and Repair, Institute of Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| | - Anupratap Tomar
- Laboratory for Circuit and Behavioral Physiology, RIKEN Brain Science Institute, Wakoshi, Saitama, Japan
| | - Aparna Suvrathan
- Department of Neurobiology, Stanford University, Stanford, California, USA
| | - Supriya Ghosh
- Department of Neurobiology, University of Chicago, Chicago, Illinois, USA
| | - Mohammed Mostafizur Rahman
- Centre for Brain Development and Repair, Institute of Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| |
Collapse
|
131
|
Bathina S, Das UN. Brain-derived neurotrophic factor and its clinical implications. Arch Med Sci 2015; 11:1164-78. [PMID: 26788077 PMCID: PMC4697050 DOI: 10.5114/aoms.2015.56342] [Citation(s) in RCA: 752] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/05/2014] [Indexed: 01/09/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal survival and growth, serves as a neurotransmitter modulator, and participates in neuronal plasticity, which is essential for learning and memory. It is widely expressed in the CNS, gut and other tissues. BDNF binds to its high affinity receptor TrkB (tyrosine kinase B) and activates signal transduction cascades (IRS1/2, PI3K, Akt), crucial for CREB and CBP production, that encode proteins involved in β cell survival. BDNF and insulin-like growth factor-1 have similar downstream signaling mechanisms incorporating both p-CAMK and MAPK that increase the expression of pro-survival genes. Brain-derived neurotrophic factor regulates glucose and energy metabolism and prevents exhaustion of β cells. Decreased levels of BDNF are associated with neurodegenerative diseases with neuronal loss, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis and Huntington's disease. Thus, BDNF may be useful in the prevention and management of several diseases including diabetes mellitus.
Collapse
Affiliation(s)
- Siresha Bathina
- Bio-Science Research Center, Gayatri Vidya Parishad College of Engineering, Visakhapatnam, India
| | - Undurti N. Das
- Bio-Science Research Center, Gayatri Vidya Parishad College of Engineering, Visakhapatnam, India
- UND Life Sciences, USA
| |
Collapse
|
132
|
Purine nucleosides in neuroregeneration and neuroprotection. Neuropharmacology 2015; 104:226-42. [PMID: 26577017 DOI: 10.1016/j.neuropharm.2015.11.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/20/2022]
Abstract
In the present review, we stress the importance of the purine nucleosides, adenosine and guanosine, in protecting the nervous system, both centrally and peripherally, via activation of their receptors and intracellular signalling mechanisms. A most novel part of the review focus on the mechanisms of neuronal regeneration that are targeted by nucleosides, including a recently identified action of adenosine on axonal growth and microtubule dynamics. Discussion on the role of the purine nucleosides transversally with the most established neurotrophic factors, e.g. brain derived neurotrophic factor (BDNF), glial derived neurotrophic factor (GDNF), is also focused considering the intimate relationship between some adenosine receptors, as is the case of the A2A receptors, and receptors for neurotrophins. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
|
133
|
Ehrlich DE, Josselyn SA. Plasticity-related genes in brain development and amygdala-dependent learning. GENES BRAIN AND BEHAVIOR 2015; 15:125-43. [PMID: 26419764 DOI: 10.1111/gbb.12255] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022]
Abstract
Learning about motivationally important stimuli involves plasticity in the amygdala, a temporal lobe structure. Amygdala-dependent learning involves a growing number of plasticity-related signaling pathways also implicated in brain development, suggesting that learning-related signaling in juveniles may simultaneously influence development. Here, we review the pleiotropic functions in nervous system development and amygdala-dependent learning of a signaling pathway that includes brain-derived neurotrophic factor (BDNF), extracellular signaling-related kinases (ERKs) and cyclic AMP-response element binding protein (CREB). Using these canonical, plasticity-related genes as an example, we discuss the intersection of learning-related and developmental plasticity in the immature amygdala, when aversive and appetitive learning may influence the developmental trajectory of amygdala function. We propose that learning-dependent activation of BDNF, ERK and CREB signaling in the immature amygdala exaggerates and accelerates neural development, promoting amygdala excitability and environmental sensitivity later in life.
Collapse
Affiliation(s)
- D E Ehrlich
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA.,Department of Otolaryngology, NYU Langone School of Medicine, New York, NY, USA
| | - S A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychology, University of Toronto, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
134
|
PLD1 participates in BDNF-induced signalling in cortical neurons. Sci Rep 2015; 5:14778. [PMID: 26437780 PMCID: PMC4594037 DOI: 10.1038/srep14778] [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] [Received: 06/26/2015] [Accepted: 09/09/2015] [Indexed: 01/07/2023] Open
Abstract
The brain-derived neurotrophic factor BDNF plays a critical role in neuronal development and the induction of L-LTP at glutamatergic synapses in several brain regions. However, the cellular and molecular mechanisms underlying these BDNF effects have not been firmly established. Using in vitro cultures of cortical neurons from knockout mice for Pld1 and Rsk2, BDNF was observed to induce a rapid RSK2-dependent activation of PLD and to stimulate BDNF ERK1/2-CREB and mTor-S6K signalling pathways, but these effects were greatly reduced in Pld1(-/-) neurons. Furthermore, phospho-CREB did not accumulate in the nucleus, whereas overexpression of PLD1 amplified the BDNF-dependent nuclear recruitment of phospho-ERK1/2 and phospho-CREB. This BDNF retrograde signalling was prevented in cells silenced for the scaffolding protein PEA15, a protein which complexes with PLD1, ERK1/2, and RSK2 after BDNF treatment. Finally PLD1, ERK1/2, and RSK2 partially colocalized on endosomal structures, suggesting that these proteins are part of the molecular module responsible for BDNF signalling in cortical neurons.
Collapse
|
135
|
Abstract
The nervous system is populated by numerous types of neurons, each bearing a dendritic arbor with a characteristic morphology. These type-specific features influence many aspects of a neuron's function, including the number and identity of presynaptic inputs and how inputs are integrated to determine firing properties. Here, we review the mechanisms that regulate the construction of cell type-specific dendrite patterns during development. We focus on four aspects of dendrite patterning that are particularly important in determining the function of the mature neuron: (a) dendrite shape, including branching pattern and geometry of the arbor; (b) dendritic arbor size;
Collapse
Affiliation(s)
| | - Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138;
| | - Jeremy N Kay
- Departments of Neurobiology and Ophthalmology, Duke University School of Medicine, Durham, North Carolina 27710;
| |
Collapse
|
136
|
PKA Inhibitor H89 (N-[2-p-bromocinnamylamino-ethyl]-5-isoquinolinesulfonamide) Attenuates Synaptic Dysfunction and Neuronal Cell Death following Ischemic Injury. Neural Plast 2015; 2015:374520. [PMID: 26448879 PMCID: PMC4584069 DOI: 10.1155/2015/374520] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/27/2015] [Accepted: 03/17/2015] [Indexed: 11/26/2022] Open
Abstract
The cyclic AMP-dependent protein kinase (PKA), which activates prosurvival signaling proteins, has been implicated in the expression of long-term potentiation and hippocampal long-term memory. It has come to light that H89 commonly known as the PKA inhibitor have diverse roles in the nervous system that are unrelated to its role as a PKA inhibitor. We have investigated the role of H89 in ischemic and reperfusion injury. First, we examined the expression of postsynaptic density protein 95 (PSD95), microtubule-associated protein 2 (MAP2), and synaptophysin in mouse brain after middle cerebral artery occlusion injury. Next, we examined the role of H89 pretreatment on the expression of brain-derived neurotrophic factor (BDNF), PSD95, MAP2, and the apoptosis regulators Bcl2 and cleaved caspase-3 in cultured neuroblastoma cells exposed to hypoxia and reperfusion injury. In addition, we investigated the alteration of AKT activation in H89 pretreated neuroblastoma cells under hypoxia and reperfusion injury. The data suggest that H89 may contribute to brain recovery after ischemic stroke by regulating neuronal death and proteins related to synaptic plasticity.
Collapse
|
137
|
Sammons RP, Keck T. Adult plasticity and cortical reorganization after peripheral lesions. Curr Opin Neurobiol 2015; 35:136-41. [PMID: 26313527 DOI: 10.1016/j.conb.2015.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 06/29/2015] [Accepted: 08/04/2015] [Indexed: 11/26/2022]
Abstract
Following loss of input due to peripheral lesions, functional reorganization occurs in the deprived cortical region in adults. Over a period of hours to months, cells in the lesion projection zone (LPZ) begin to respond to novel stimuli. This reorganization is mediated by two processes: a reduction of inhibition in a gradient throughout the cortex and input remapping via sprouting of axonal arbors from cortical regions spatially adjacent to the LPZ, and strengthening of pre-existing subthreshold inputs. Together these inputs facilitate receptive field remapping of cells in the LPZ. Recent experiments have revealed time courses and potential interactions of the mechanisms associated with functional reorganization, suggesting that large scale reorganization in the adult may utilize plasticity mechanisms prominent during development.
Collapse
Affiliation(s)
- Rosanna P Sammons
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK; MRC Centre for Developmental Neurobiology, King's College London, London, UK
| | - Tara Keck
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| |
Collapse
|
138
|
Valnegri P, Puram SV, Bonni A. Regulation of dendrite morphogenesis by extrinsic cues. Trends Neurosci 2015; 38:439-47. [PMID: 26100142 DOI: 10.1016/j.tins.2015.05.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 01/19/2023]
Abstract
Dendrites play a central role in the integration and flow of information in the nervous system. The morphogenesis and maturation of dendrites is hence an essential step in the establishment of neuronal connectivity. Recent studies have uncovered crucial functions for extrinsic cues in the development of dendrites. We review the contribution of secreted polypeptide growth factors, contact-mediated proteins, and neuronal activity in distinct phases of dendrite development. We also highlight how extrinsic cues influence local and global intracellular mechanisms of dendrite morphogenesis. Finally, we discuss how these studies have advanced our understanding of neuronal connectivity and have shed light on the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Pamela Valnegri
- Department of Anatomy and Neurobiology, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Sidharth V Puram
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Azad Bonni
- Department of Anatomy and Neurobiology, Washington University in St Louis School of Medicine, St Louis, MO 63110, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
139
|
Mastication as a Stress-Coping Behavior. BIOMED RESEARCH INTERNATIONAL 2015; 2015:876409. [PMID: 26090453 PMCID: PMC4450283 DOI: 10.1155/2015/876409] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/21/2014] [Accepted: 01/05/2015] [Indexed: 11/29/2022]
Abstract
Exposure to chronic stress induces various physical and mental effects that may ultimately lead to disease. Stress-related disease has become a global health problem. Mastication (chewing) is an effective behavior for coping with stress, likely due to the alterations chewing causes in the activity of the hypothalamic-pituitary-adrenal axis and autonomic nervous system. Mastication under stressful conditions attenuates stress-induced increases in plasma corticosterone and catecholamines, as well as the expression of stress-related substances, such as neurotrophic factors and nitric oxide. Further, chewing reduces stress-induced changes in central nervous system morphology, especially in the hippocampus and hypothalamus. In rodents, chewing or biting on wooden sticks during exposure to various stressors reduces stress-induced gastric ulcer formation and attenuates spatial cognitive dysfunction, anxiety-like behavior, and bone loss. In humans, some studies demonstrate that chewing gum during exposure to stress decreases plasma and salivary cortisol levels and reduces mental stress, although other studies report no such effect. Here, we discuss the neuronal mechanisms that underline the interactions between masticatory function and stress-coping behaviors in animals and humans.
Collapse
|
140
|
Grados M, Sung HM, Kim S, Srivastava S. Genetic findings in obsessive-compulsive disorder connect to brain-derived neutrophic factor and mammalian target of rapamycin pathways: implications for drug development. Drug Dev Res 2015; 75:372-83. [PMID: 25195581 DOI: 10.1002/ddr.21223] [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] [Indexed: 01/18/2023]
Abstract
Traditional pharmacological approaches to the treatment of obsessive-compulsive disorder (OCD) are based on affecting serotonergic and dopaminergic transmission in the central nervous system. However, genetic epidemiology findings are pointing to glutamate pathways and developmental genes as etiological in OCD. A review of recent genetic findings in OCD is conducted, and bioinformatics approaches are used to locate pathways relevant to neuroprotection. The OCD susceptibility genes DLGAP1, RYR3, PBX1-MEIS2, LMX1A and candidate genes BDNF and GRIN2B are components of the neuronal growth, differentiation and neurogenesis pathways BDNF-mTOR. These pathways are emerging as a promising area of research for the development of neuroprotective pharmaceuticals. Emergent genetic epidemiologic data on OCD and repetitive behaviors may support new approaches for pharmacological discovery. Neuroprotective approaches that take into consideration glutamate-mediated BDNF-mTOR pathways are suggested by OCD susceptibility genes.
Collapse
Affiliation(s)
- Marco Grados
- Division of Child & Adolescent Psychiatry, Johns Hopkins University School of Medicine, 1800 Orleans St.-12th floor, Baltimore, MD, 21287, USA
| | | | | | | |
Collapse
|
141
|
Sconce M, Churchill M, Moore C, Meshul C. Intervention with 7,8-dihydroxyflavone blocks further striatal terminal loss and restores motor deficits in a progressive mouse model of Parkinson’s disease. Neuroscience 2015; 290:454-71. [DOI: 10.1016/j.neuroscience.2014.12.080] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/16/2014] [Accepted: 12/18/2014] [Indexed: 01/22/2023]
|
142
|
Ca(2+) signaling initiated by canonical transient receptor potential channels in dendritic development. Neurosci Bull 2015; 31:351-6. [PMID: 25732528 DOI: 10.1007/s12264-014-1511-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/22/2014] [Indexed: 12/25/2022] Open
Abstract
The spatial patterns of dendritic structures diverge in different types of neurons as adaptations to their unique functions. Although different intracellular mechanisms underlying dendritic morphogenesis have been suggested, it is evident that the elevation in intracellular Ca(2+) levels plays a major role in the process. Canonical transient receptor potential (TRPC) channels, known to be non-selective Ca(2+)-permeable cation channels, act as environmental detectors to sense and transduce extracellular signals into different intracellular responses, including the regulation of dendritic growth, via Ca(2+) influx. Here, we review recent advances in the understanding of Ca(2+) signaling, especially signals mediated by Ca(2+) influx via TRPC channels, and the underlying molecular events in dendritic development.
Collapse
|
143
|
Wu YK, Fujishima K, Kengaku M. Differentiation of apical and basal dendrites in pyramidal cells and granule cells in dissociated hippocampal cultures. PLoS One 2015; 10:e0118482. [PMID: 25705877 PMCID: PMC4338060 DOI: 10.1371/journal.pone.0118482] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/18/2015] [Indexed: 11/21/2022] Open
Abstract
Hippocampal pyramidal cells and dentate granule cells develop morphologically distinct dendritic arbors, yet also share some common features. Both cell types form a long apical dendrite which extends from the apex of the cell soma, while short basal dendrites are developed only in pyramidal cells. Using quantitative morphometric analyses of mouse hippocampal cultures, we evaluated the differences in dendritic arborization patterns between pyramidal and granule cells. Furthermore, we observed and described the final apical dendrite determination during dendritic polarization by time-lapse imaging. Pyramidal and granule cells in culture exhibited similar dendritic patterns with a single principal dendrite and several minor dendrites so that the cell types were not readily distinguished by appearance. While basal dendrites in granule cells are normally degraded by adulthood in vivo, cultured granule cells retained their minor dendrites. Asymmetric growth of a single principal dendrite harboring the Golgi was observed in both cell types soon after the onset of dendritic growth. Time-lapse imaging revealed that up until the second week in culture, final principal dendrite designation was not stabilized, but was frequently replaced by other minor dendrites. Before dendritic polarity was stabilized, the Golgi moved dynamically within the soma and was repeatedly repositioned at newly emerging principal dendrites. Our results suggest that polarized growth of the apical dendrite is regulated by cell intrinsic programs, while regression of basal dendrites requires cue(s) from the extracellular environment in the dentate gyrus. The apical dendrite designation is determined from among multiple growing dendrites of young developing neurons.
Collapse
Affiliation(s)
- You Kure Wu
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kazuto Fujishima
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
| | - Mineko Kengaku
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
144
|
De Giorgio A, Granato A. Reduced density of dendritic spines in pyramidal neurons of rats exposed to alcohol during early postnatal life. Int J Dev Neurosci 2015; 41:74-9. [PMID: 25644892 DOI: 10.1016/j.ijdevneu.2015.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/07/2015] [Accepted: 01/29/2015] [Indexed: 11/24/2022] Open
Abstract
Dendritic spines are the main postsynaptic sites of excitatory connections of neocortical pyramidal neurons. Alterations of spine shape, number, and density can be observed in different mental diseases, including those caused by developmental alcohol exposure. Pyramidal neurons of layer 2/3 are the most abundant cells of the neocortex and represent the main source of associative cortico-cortical connections. These neurons are essential for higher functions mediated by the cortex such as feature selection and perceptual grouping. Furthermore, their connections have been shown to be altered in experimental models of fetal alcohol spectrum disorders. Here, we used a Golgi-like tracing method to study the spine density of layer 2/3 associative pyramidal neurons in the somatosensory cortex of adult rats exposed to alcohol during the first postnatal week. The main result of the present study is represented by the decreased spine density in the apical dendrite of alcohol-treated rats, as compared to controls. As to the basal dendritic tree, there were no significant differences between the experimental and the control group. A decreased density of dendritic spines in the apical dendrite may impair the excitatory input onto pyramidal neurons, thus resulting in a widespread alteration of the cortical information flow.
Collapse
Affiliation(s)
- Andrea De Giorgio
- Department of Psychology, Catholic University, Largo A. Gemelli 1, 20123 Milan, Italy.
| | - Alberto Granato
- Department of Psychology, Catholic University, Largo A. Gemelli 1, 20123 Milan, Italy.
| |
Collapse
|
145
|
Ryoo K, Hwang SG, Kim KJ, Choi EJ. RC3/neurogranin negatively regulates extracellular signal-regulated kinase pathway through its interaction with Ras. Mol Cell Biochem 2014; 402:33-40. [DOI: 10.1007/s11010-014-2311-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/20/2014] [Indexed: 12/19/2022]
|
146
|
Molecular regulation of synaptogenesis during associative learning and memory. Brain Res 2014; 1621:239-51. [PMID: 25485772 DOI: 10.1016/j.brainres.2014.11.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 01/06/2023]
Abstract
Synaptogenesis plays a central role in associative learning and memory. The biochemical pathways that underlie synaptogenesis are complex and incompletely understood. Nevertheless, research has so far identified three conceptually distinct routes to synaptogenesis: cell-cell contact mediated by adhesion proteins, cell-cell biochemical signaling from astrocytes and other cells, and neuronal signaling through classical ion channels and cell surface receptors. The cell adhesion pathways provide the physical substrate to the new synaptic connection, while cell-cell signaling may provide a global or regional signal, and the activity-dependent pathways provide the neuronal specificity that is required for the new synapses to produce functional neuronal networks capable of storing associative memories. These three aspects of synaptogenesis require activation of a variety of interacting biochemical pathways that converge on the actin cytoskeleton and strengthen the synapse in an information-dependent manner. This article is part of a Special Issue titled SI: Brain and Memory.
Collapse
|
147
|
Joo W, Hippenmeyer S, Luo L. Neurodevelopment. Dendrite morphogenesis depends on relative levels of NT-3/TrkC signaling. Science 2014; 346:626-9. [PMID: 25359972 DOI: 10.1126/science.1258996] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neurotrophins regulate diverse aspects of neuronal development and plasticity, but their precise in vivo functions during neural circuit assembly in the central brain remain unclear. We show that the neurotrophin receptor tropomyosin-related kinase C (TrkC) is required for dendritic growth and branching of mouse cerebellar Purkinje cells. Sparse TrkC knockout reduced dendrite complexity, but global Purkinje cell knockout had no effect. Removal of the TrkC ligand neurotrophin-3 (NT-3) from cerebellar granule cells, which provide major afferent input to developing Purkinje cell dendrites, rescued the dendrite defects caused by sparse TrkC disruption in Purkinje cells. Our data demonstrate that NT-3 from presynaptic neurons (granule cells) is required for TrkC-dependent competitive dendrite morphogenesis in postsynaptic neurons (Purkinje cells)--a previously unknown mechanism of neural circuit development.
Collapse
Affiliation(s)
- William Joo
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA. Neurosciences Program, Stanford University, Stanford, CA 94305, USA
| | - Simon Hippenmeyer
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Liqun Luo
- Howard Hughes Medical Institute and Department of Biology, Stanford University, Stanford, CA 94305, USA. Neurosciences Program, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
148
|
Martin JL, Finsterwald C. Cooperation between BDNF and glutamate in the regulation of synaptic transmission and neuronal development. Commun Integr Biol 2014. [DOI: 10.4161/cib.13761] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
149
|
Abstract
The complex, branched morphology of dendrites is a cardinal feature of neurons and has been used as a criterion for cell type identification since the beginning of neurobiology. Regulated dendritic outgrowth and branching during development form the basis of receptive fields for neurons and are essential for the wiring of the nervous system. The cellular and molecular mechanisms of dendritic morphogenesis have been an intensely studied area. In this review, we summarize the major experimental systems that have contributed to our understandings of dendritic development as well as the intrinsic and extrinsic mechanisms that instruct the neurons to form cell type-specific dendritic arbors.
Collapse
|
150
|
The role of the retromer complex in aging-related neurodegeneration: a molecular and genomic review. Mol Genet Genomics 2014; 290:413-27. [PMID: 25332075 DOI: 10.1007/s00438-014-0939-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/10/2014] [Indexed: 10/24/2022]
Abstract
The retromer coat complex is a vital component of the intracellular trafficking mechanism sorting cargo from the endosomes to the trans-Golgi network or to the cell surface. In recent years, genes encoding components of the retromer coat complex and members of the vacuolar protein sorting 10 (Vps10) family of receptors, which play pleiotropic functions in protein trafficking and intracellular/intercellular signaling in neuronal and non-neuronal cells and are primary cargos of the retromer complex, have been implicated as genetic risk factors for sporadic and autosomal dominant forms of several neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and frontotemporal lobar degeneration. In addition to their functions in protein trafficking, the members of the Vps10 receptor family (sortilin, SorL1, SorCS1, SorCS2, and SorCS3) modulate neurotrophic signaling pathways. Both sortilin and SorCS2 act as cell surface receptors to mediate acute responses to proneurotrophins. In addition, sortilin can modulate the intracellular response to brain-derived neurotrophic factor (BDNF) by direct control of BDNF levels and regulating anterograde trafficking of Trk receptors to the synapse. This review article summarizes the emerging data from this rapidly growing field of intracellular trafficking signaling in the pathogenesis of neurodegeneration.
Collapse
|