1
|
Benjamin Kacerovsky J, Murai KK. Stargazing: Monitoring subcellular dynamics of brain astrocytes. Neuroscience 2015; 323:84-95. [PMID: 26162237 DOI: 10.1016/j.neuroscience.2015.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 06/28/2015] [Accepted: 07/01/2015] [Indexed: 01/21/2023]
Abstract
Astrocytes are major non-neuronal cell types in the central nervous system that regulate a variety of processes in the brain including synaptic transmission, neurometabolism, and cerebrovasculature tone. Recent discoveries have revealed that astrocytes perform very specialized and heterogeneous roles in brain homeostasis and function. Exactly how astrocytes fulfill such diverse roles in the brain remains to be fully understood and is an active area of research. In this review, we focus on the complex subcellular anatomical features of protoplasmic gray matter astrocytes in the mature, healthy brain that likely empower these cells with the ability to detect and respond to changes in neuronal and synaptic activity. In particular, we discuss how intricate processes on astrocytes allow these cells to communicate with neurons and their synapses and strategically deliver specific cellular organelles such as mitochondria and ribosomes to active compartments within the neuropil. Understanding the properties of these structural elements will lead to a better understanding of how astrocytes function in the healthy and diseased brain.
Collapse
Affiliation(s)
- J Benjamin Kacerovsky
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada
| | - K K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, The Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, Quebec H3G 1A4, Canada.
| |
Collapse
|
2
|
Chinchalongporn V, Koppensteiner P, Prè D, Thangnipon W, Bilo L, Arancio O. Connectivity and circuitry in a dish versus in a brain. ALZHEIMERS RESEARCH & THERAPY 2015; 7:44. [PMID: 26045718 PMCID: PMC4456047 DOI: 10.1186/s13195-015-0129-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In order to understand and find therapeutic strategies for neurological disorders, disease models that recapitulate the connectivity and circuitry of patients’ brain are needed. Owing to many limitations of animal disease models, in vitro neuronal models using patient-derived stem cells are currently being developed. However, prior to employing neurons as a model in a dish, they need to be evaluated for their electrophysiological properties, including both passive and active membrane properties, dynamics of neurotransmitter release, and capacity to undergo synaptic plasticity. In this review, we survey recent attempts to study these issues in human induced pluripotent stem cell-derived neurons. Although progress has been made, there are still many hurdles to overcome before human induced pluripotent stem cell-derived neurons can fully recapitulate all of the above physiological properties of adult mature neurons. Moreover, proper integration of neurons into pre-existing circuitry still needs to be achieved. Nevertheless, in vitro neuronal stem cell-derived models hold great promise for clinical application in neurological diseases in the future.
Collapse
Affiliation(s)
- Vorapin Chinchalongporn
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA ; Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom 73170 Thailand
| | - Peter Koppensteiner
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA ; Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Deborah Prè
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA
| | - Wipawan Thangnipon
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom 73170 Thailand
| | - Leonilda Bilo
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA ; Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University of Naples, 80131 Naples, Italy
| | - Ottavio Arancio
- Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA ; Taub Institute for Research on Alzheimer's Disease and the Aging Brain P&S Bldg, Room 12-420D, Columbia University, New York, NY 10032 USA ; Columbia Stem Cell Initiative, CUMC, New York, NY 10032 USA
| |
Collapse
|
3
|
Kazim SF, Cardenas-Aguayo MDC, Arif M, Blanchard J, Fayyaz F, Grundke-Iqbal I, Iqbal K. Sera from children with autism induce autistic features which can be rescued with a CNTF small peptide mimetic in rats. PLoS One 2015; 10:e0118627. [PMID: 25769033 PMCID: PMC4359103 DOI: 10.1371/journal.pone.0118627] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/21/2015] [Indexed: 12/29/2022] Open
Abstract
Autism is a neurodevelopmental disorder characterized clinically by impairments in social interaction and verbal and non-verbal communication skills as well as restricted interests and repetitive behavior. It has been hypothesized that altered brain environment including an imbalance in neurotrophic support during early development contributes to the pathophysiology of autism. Here we report that sera from children with autism which exhibited abnormal levels of various neurotrophic factors induced cell death and oxidative stress in mouse primary cultured cortical neurons. The effects of sera from autistic children were rescued by pre-treatment with a ciliary neurotrophic factor (CNTF) small peptide mimetic, Peptide 6 (P6), which was previously shown to exert its neuroprotective effect by modulating CNTF/JAK/STAT pathway and LIF signaling and by enhancing brain derived neurotrophic factor (BDNF) expression. Similar neurotoxic effects and neuroinflammation were observed in young Wistar rats injected intracerebroventricularly with autism sera within hours after birth. The autism sera injected rats demonstrated developmental delay and deficits in social communication, interaction, and novelty. Both the neurobiological changes and the behavioral autistic phenotype were ameliorated by P6 treatment. These findings implicate the involvement of neurotrophic imbalance during early brain development in the pathophysiology of autism and a proof of principle of P6 as a potential therapeutic strategy for autism.
Collapse
Affiliation(s)
- Syed Faraz Kazim
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- Neural and Behavioral Science Graduate Program, State University of New York (SUNY) Downstate Medical Center, Brooklyn, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
| | - Maria del Carmen Cardenas-Aguayo
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Mohammad Arif
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Julie Blanchard
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Fatima Fayyaz
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Inge Grundke-Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
| | - Khalid Iqbal
- Inge Grundke-Iqbal Research Floor, Department of Neurochemistry, New York State Institute for Basic Research in Developmental Disabilities (NYSIBR), Staten Island, New York, United States of America
- SUNY Downstate/NYSIBR Center for Developmental Neuroscience (CDN), Staten Island, New York, United States of America
- * E-mail:
| |
Collapse
|
4
|
Saliu A, Adise S, Xian S, Kudelska K, Rodríguez-Contreras A. Natural and lesion-induced decrease in cell proliferation in the medial nucleus of the trapezoid body during hearing development. J Comp Neurol 2014; 522:971-85. [PMID: 24115041 PMCID: PMC4082979 DOI: 10.1002/cne.23473] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 09/06/2013] [Accepted: 09/17/2013] [Indexed: 02/03/2023]
Abstract
The functional interactions between neurons and glial cells that are important for nervous system function are presumably established during development from the activity of progenitor cells. In this study we examined proliferation of progenitor cells in the medial nucleus of the trapezoid body (MNTB) located in the rat auditory brainstem. We performed DNA synthesis labeling experiments to demonstrate changes in cell proliferation activity during postnatal stages of development. An increase in cell proliferation correlated with MNTB growth and the presence of S100β-positive astrocytes among MNTB neurons. In additional experiments we analyzed the fate of newly born cells. At perinatal ages, newly born cells colabeled with the astrocyte marker S100β in higher numbers than when cells were generated at postnatal day 6. Furthermore, we identified newly born cells that were colabeled with caspase-3 immunohistochemistry and performed comparative experiments to demonstrate that there is a natural decrease in cell proliferation activity during postnatal development in rats, mice, gerbils, and ferrets. Lastly, we found that there is a stronger decrease in MNTB cell proliferation after performing bilateral lesions of the auditory periphery in rats. Altogether, these results identify important stages in the development of astrocytes in the MNTB and provide evidence that the proliferative activity of the progenitor cells is developmentally regulated. We propose that the developmental reduction in cell proliferation may reflect coordinated signaling between the auditory brainstem and the auditory periphery. J. Comp. Neurol. 522:971–985, 2014.
Collapse
Affiliation(s)
- Aminat Saliu
- Biology Department, City College of New York, New York, New York, 10031
| | | | | | | | | |
Collapse
|
5
|
A time course analysis of the electrophysiological properties of neurons differentiated from human induced pluripotent stem cells (iPSCs). PLoS One 2014; 9:e103418. [PMID: 25072157 PMCID: PMC4114788 DOI: 10.1371/journal.pone.0103418] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 07/02/2014] [Indexed: 11/19/2022] Open
Abstract
Many protocols have been designed to differentiate human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs) into neurons. Despite the relevance of electrophysiological properties for proper neuronal function, little is known about the evolution over time of important neuronal electrophysiological parameters in iPSC-derived neurons. Yet, understanding the development of basic electrophysiological characteristics of iPSC-derived neurons is critical for evaluating their usefulness in basic and translational research. Therefore, we analyzed the basic electrophysiological parameters of forebrain neurons differentiated from human iPSCs, from day 31 to day 55 after the initiation of neuronal differentiation. We assayed the developmental progression of various properties, including resting membrane potential, action potential, sodium and potassium channel currents, somatic calcium transients and synaptic activity. During the maturation of iPSC-derived neurons, the resting membrane potential became more negative, the expression of voltage-gated sodium channels increased, the membrane became capable of generating action potentials following adequate depolarization and, at day 48–55, 50% of the cells were capable of firing action potentials in response to a prolonged depolarizing current step, of which 30% produced multiple action potentials. The percentage of cells exhibiting miniature excitatory post-synaptic currents increased over time with a significant increase in their frequency and amplitude. These changes were associated with an increase of Ca2+ transient frequency. Co-culturing iPSC-derived neurons with mouse glial cells enhanced the development of electrophysiological parameters as compared to pure iPSC-derived neuronal cultures. This study demonstrates the importance of properly evaluating the electrophysiological status of the newly generated neurons when using stem cell technology, as electrophysiological properties of iPSC-derived neurons mature over time.
Collapse
|
6
|
Szepietowska B, Zhu W, Czyzyk J, Eid T, Sherwin RS. EphA5-EphrinA5 interactions within the ventromedial hypothalamus influence counterregulatory hormone release and local glutamine/glutamate balance during hypoglycemia. Diabetes 2013; 62:1282-8. [PMID: 23274893 PMCID: PMC3609598 DOI: 10.2337/db12-0982] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Activation of β-cell EphA5 receptors by its ligand ephrinA5 from adjacent β-cells has been reported to decrease insulin secretion during hypoglycemia. Given the similarities between islet and ventromedial hypothalamus (VMH) glucose sensing, we tested the hypothesis that the EphA5/ephrinA5 system might function within the VMH during hypoglycemia to stimulate counterregulatory hormone release as well. Counterregulatory responses and glutamine/glutamate concentrations in the VMH were assessed during a hyperinsulinemic-hypoglycemic glucose clamp study in chronically catheterized awake male Sprague-Dawley rats that received an acute VMH microinjection of ephrinA5-Fc, chronic VMH knockdown, or overexpression of ephrinA5 using an adenoassociated viral construct. Local stimulation of VMH EphA5 receptors by ephrinA5-Fc or ephrinA5 overexpression increased, whereas knockdown of VMH ephrinA5 reduced counterregulatory responses during hypoglycemia. Overexpression of VMH ephrinA5 transiently increased local glutamate concentrations, whereas ephrinA5 knockdown produced profound suppression of VMH interstitial fluid glutamine concentrations in the basal state and during hypoglycemia. Changes in ephrinA5/EphA5 interactions within the VMH, a key brain glucose-sensing region, act in concert with islets to restore glucose homeostasis during acute hypoglycemia, and its effect on counterregulation may be mediated by changes in glutamate/glutamine cycling.
Collapse
Affiliation(s)
- Barbara Szepietowska
- Department of Internal Medicine and Endocrinology, Yale University School of Medicine, New Haven, Connecticut
| | - Wanling Zhu
- Department of Internal Medicine and Endocrinology, Yale University School of Medicine, New Haven, Connecticut
| | - Jan Czyzyk
- Department of Pathology and Laboratory Medicine, University of Rochester, Rochester, New York
| | - Tore Eid
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Robert S. Sherwin
- Department of Internal Medicine and Endocrinology, Yale University School of Medicine, New Haven, Connecticut
- Corresponding author: Robert S. Sherwin,
| |
Collapse
|
7
|
Chang PKY, Verbich D, McKinney RA. AMPA receptors as drug targets in neurological disease - advantages, caveats, and future outlook. Eur J Neurosci 2012; 35:1908-16. [DOI: 10.1111/j.1460-9568.2012.08165.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Jones EV, Cook D, Murai KK. A neuron-astrocyte co-culture system to investigate astrocyte-secreted factors in mouse neuronal development. Methods Mol Biol 2012; 814:341-352. [PMID: 22144317 DOI: 10.1007/978-1-61779-452-0_22] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Astrocytes secrete factors that promote neuron survival, synapse formation, and plasticity. Understanding how these factors perform these roles requires a robust in vitro system that can effectively assess the impact of individual glial factors on neuronal properties. A classical approach to studying neuron-glial interactions in vitro uses a system where dissociated embryonic rat neurons are suspended over a feeder layer of rat astrocytes. Here, we describe a useful "sandwich" co-culture system where postnatal mouse hippocampal neurons are grown suspended above a feeder layer of mouse hippocampal astrocytes. We demonstrate that neurons in these cultures remain healthy beyond 3 weeks in vitro and develop more synapses compared to neurons grown without astrocytes. An advantage of this method is that astrocytes and neurons can be prepared separately from postnatal transgenic or knock-out mouse lines allowing one to study, for example, how wild-type neurons develop in the presence of astrocytes from a knock-out mouse line that lacks the expression of a specific astrocyte-secreted factor. We find this culture system to be a convenient and powerful approach to study the contribution of astrocyte-secreted molecules to neuron development.
Collapse
Affiliation(s)
- Emma V Jones
- Centre for Research in Neuroscience, Montreal General Hospital, Montreal, QC, Canada
| | | | | |
Collapse
|
9
|
Eph receptors and ephrins in neuron-astrocyte communication at synapses. Glia 2011; 59:1567-78. [DOI: 10.1002/glia.21226] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 07/05/2011] [Indexed: 12/24/2022]
|
10
|
Pokrzywa M, Dacklin I, Vestling M, Hultmark D, Lundgren E, Cantera R. Uptake of aggregating transthyretin by fat body in a Drosophila model for TTR-associated amyloidosis. PLoS One 2010; 5:e14343. [PMID: 21179560 PMCID: PMC3002944 DOI: 10.1371/journal.pone.0014343] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 11/11/2010] [Indexed: 01/26/2023] Open
Abstract
Background A functional link has been established between the severe neurodegenerative disorder Familial amyloidotic polyneuropathy and the enhanced propensity of the plasma protein transthyretin (TTR) to form aggregates in patients with single point mutations in the TTR gene. Previous work has led to the establishment of an experimental model based on transgenic expression of normal or mutant forms of human TTR in Drosophila flies. Remarkably, the severity of the phenotype was greater in flies that expressed a single copy than with two copies of the mutated gene. Methodology/Principal Findings In this study, we analyze the distribution of normal and mutant TTR in transgenic flies, and the ultrastructure of TTR-positive tissues to clarify if aggregates and/or amyloid filaments are formed. We report the formation of intracellular aggregates of 20 nm spherules and amyloid filaments in thoracic adipose tissue and in brain glia, two tissues that do not express the transgene. The formation of aggregates of nanospherules increased with age and was more considerable in flies with two copies of mutated TTR. Treatment of human neuronal cells with protein extracts prepared from TTR flies of different age showed that the extracts from older flies were less toxic than those from younger flies. Conclusions/Significance These findings suggest that the uptake of TTR from the circulation and its subsequent segregation into cytoplasmic quasi-crystalline arrays of nanospherules is part of a mechanism that neutralizes the toxic effect of TTR.
Collapse
|
11
|
Drosophila glial glutamate transporter Eaat1 is regulated by fringe-mediated notch signaling and is essential for larval locomotion. J Neurosci 2010; 30:14446-57. [PMID: 20980602 DOI: 10.1523/jneurosci.1021-10.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In the mammalian CNS, glial cells expressing excitatory amino acid transporters (EAATs) tightly regulate extracellular glutamate levels to control neurotransmission and protect neurons from excitotoxic damage. Dysregulated EAAT expression is associated with several CNS pathologies in humans, yet mechanisms of EAAT regulation and the importance of glutamate transport for CNS development and function in vivo remain incompletely understood. Drosophila is an advanced genetic model with only a single high-affinity glutamate transporter termed Eaat1. We found that Eaat1 expression in CNS glia is regulated by the glycosyltransferase Fringe, which promotes neuron-to-glia signaling through the Delta-Notch ligand-receptor pair during embryogenesis. We made Eaat1 loss-of-function mutations and found that homozygous larvae could not perform the rhythmic peristaltic contractions required for crawling. We found no evidence for excitotoxic cell death or overt defects in the development of neurons and glia, and the crawling defect could be induced by postembryonic inactivation of Eaat1. Eaat1 fully rescued locomotor activity when expressed in only a limited subpopulation of glial cells situated near potential glutamatergic synapses within the CNS neuropil. Eaat1 mutants had deficits in the frequency, amplitude, and kinetics of synaptic currents in motor neurons whose rhythmic patterns of activity may be regulated by glutamatergic neurotransmission among premotor interneurons; similar results were seen with pharmacological manipulations of glutamate transport. Our findings indicate that Eaat1 expression is promoted by Fringe-mediated neuron-glial communication during development and suggest that Eaat1 plays an essential role in regulating CNS neural circuits that control locomotion in Drosophila.
Collapse
|
12
|
Araque A, Navarrete M. Glial cells in neuronal network function. Philos Trans R Soc Lond B Biol Sci 2010; 365:2375-81. [PMID: 20603358 DOI: 10.1098/rstb.2009.0313] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Numerous evidence demonstrates that astrocytes, a type of glial cell, are integral functional elements of the synapses, responding to neuronal activity and regulating synaptic transmission and plasticity. Consequently, they are actively involved in the processing, transfer and storage of information by the nervous system, which challenges the accepted paradigm that brain function results exclusively from neuronal network activity, and suggests that nervous system function actually arises from the activity of neuron-glia networks. Most of our knowledge of the properties and physiological consequences of the bidirectional communication between astrocytes and neurons resides at cellular and molecular levels. In contrast, much less is known at higher level of complexity, i.e. networks of cells, and the actual impact of astrocytes in the neuronal network function remains largely unexplored. In the present article, we summarize the current evidence that supports the notion that astrocytes are integral components of nervous system networks and we discuss some functional properties of intercellular signalling in neuron-glia networks.
Collapse
Affiliation(s)
- Alfonso Araque
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain.
| | | |
Collapse
|
13
|
Zhang X, Poo MM. Progress in neural plasticity. SCIENCE CHINA-LIFE SCIENCES 2010; 53:322-329. [DOI: 10.1007/s11427-010-0062-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 01/19/2010] [Indexed: 11/25/2022]
|
14
|
Crispino M, Cefaliello C, Kaplan B, Giuditta A. Protein synthesis in nerve terminals and the glia-neuron unit. Results Probl Cell Differ 2010; 48:243-67. [PMID: 19554280 DOI: 10.1007/400_2009_9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The progressive philogenetic lengthening of axonal processes and the increase in complexity of terminal axonal arborizations markedly augmented the demands of the neuronal cytoplasmic mass on somatic gene expression. It is proposed that in an adaptive response to this challenge, novel gene expression functions developed in the axon compartment, consisting of axonal and presynaptic translation systems that rely on the delivery of transcripts synthesized in adjacent glial cells. Such intercellular mode of gene expression would allow more rapid plastic changes to occur in spatially restricted neuronal domains, down to the size of individual synapses. The cell body contribution to local gene expression in well-differentiated neurons remains to be defined. The history of this concept and the experimental evidence supporting its validity are critically discussed in this article. The merit of this perspective lies with the recognition that plasticity events represent a major occurrence in the brain, and that they largely occur at synaptic sites, including presynaptic endings.
Collapse
Affiliation(s)
- Marianna Crispino
- Department of Biological Sciences, University of Naples Federico II, Naples, Italy
| | | | | | | |
Collapse
|
15
|
Bolton MM, Eroglu C. Look who is weaving the neural web: glial control of synapse formation. Curr Opin Neurobiol 2009; 19:491-7. [PMID: 19879129 DOI: 10.1016/j.conb.2009.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 09/18/2009] [Indexed: 10/20/2022]
Abstract
Historically, our understanding of synapse formation has been shaped by studies focusing on neurons. However, with advancements in live imaging techniques and molecular and genetic tools we are rapidly uncovering new roles for glia in synapse formation and function. Contact-mediated signals from glia instruct dendrites to become receptive to synaptic partners. Glia-secreted factors coordinate the assembly and apposition of pre-synaptic and post-synaptic specializations. Glial cells also provide cues that are required for synaptic maturation and remodeling of spines both during development and in the adult. As we continue to learn about glial contributions to synapse formation and maintenance, it is likely that glia-derived signals will emerge as potential therapeutic targets for diseases that involve aberrant circuit function such as autism, epilepsy and Alzheimer's Disease.
Collapse
Affiliation(s)
- M McLean Bolton
- Department of Cell Biology, Duke University Medical Center, 333A Nanaline Duke Bldg., Box 3709, Durham, NC 27710, United States
| | | |
Collapse
|
16
|
Pardo-Villamizar C, Zimmerman A. Inflammation and Neuroimmunity in the Pathogenesis of Autism. AUTISM : THE INTERNATIONAL JOURNAL OF RESEARCH AND PRACTICE 2009. [DOI: 10.1201/9781420068870-c12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
17
|
Pfrieger FW. Roles of glial cells in synapse development. Cell Mol Life Sci 2009; 66:2037-47. [PMID: 19308323 PMCID: PMC2705714 DOI: 10.1007/s00018-009-0005-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2008] [Revised: 01/20/2009] [Accepted: 02/10/2009] [Indexed: 11/29/2022]
Abstract
Brain function relies on communication among neurons via highly specialized contacts, the synapses, and synaptic dysfunction lies at the heart of age-, disease-, and injury-induced defects of the nervous system. For these reasons, the formation-and repair-of synaptic connections is a major focus of neuroscience research. In this review, I summarize recent evidence that synapse development is not a cell-autonomous process and that its distinct phases depend on assistance from the so-called glial cells. The results supporting this view concern synapses in the central nervous system as well as neuromuscular junctions and originate from experimental models ranging from cell cultures to living flies, worms, and mice. Peeking at the future, I will highlight recent technical advances that are likely to revolutionize our views on synapse-glia interactions in the developing, adult and diseased brain.
Collapse
Affiliation(s)
- Frank W Pfrieger
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, University of Strasbourg, 5, rue Louis Pasteur, 67084, Strasbourg, France.
| |
Collapse
|