51
|
Matsumoto N, Hoshiko M, Sugo N, Fukazawa Y, Yamamoto N. Synapse-dependent and independent mechanisms of thalamocortical axon branching are regulated by neuronal activity. Dev Neurobiol 2015; 76:323-36. [PMID: 26061995 DOI: 10.1002/dneu.22317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/06/2015] [Indexed: 12/23/2022]
Abstract
Axon branching and synapse formation are critical processes for establishing precise circuit connectivity. These processes are tightly regulated by neural activity, but the relationship between them remains largely unclear. We use organotypic coculture preparations to examine the role of synapse formation in the activity-dependent axon branching of thalamocortical (TC) projections. To visualize TC axons and their presynaptic sites, two plasmids encoding DsRed and EGFP-tagged synaptophysin (SYP-EGFP) were cotransfected into a small number of thalamic neurons. Time-lapse imaging of individual TC axons showed that most branches emerged from SYP-EGFP puncta, indicating that synapse formation precedes emergences of axonal branches. We also investigated the effects of neuronal activity on axon branching and synapse formation by manipulating spontaneous firing activity of thalamic cells. An inward rectifying potassium channel, Kir2.1, and a bacterial voltage-gated sodium channel, NaChBac, were used to suppress and promote firing activity, respectively. We found suppressing neural activity reduced both axon branching and synapse formation. In contrast, increasing neural activity promoted only axonal branch formation. Time-lapse imaging of NaChBac-expressing cells further revealed that new branches frequently appeared from the locations other than SYP-EGFP puncta, indicating that enhancing activity promotes axonal branch formation due to an increase of branch emergence at nonsynaptic sites. These results suggest that presynaptic locations are hotspots for branch emergence, and that frequent firing activity can shift branch emergence to a synapse-independent process.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Maki Hoshiko
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Sugo
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical Science, University of Fukui, Eiheiji, Yoshida, 910-1193, Japan
| | - Nobuhiko Yamamoto
- Laboratory of Cellular and Molecular Neurobiology Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
52
|
Muñoz R, Edwards-Faret G, Moreno M, Zuñiga N, Cline H, Larraín J. Regeneration of Xenopus laevis spinal cord requires Sox2/3 expressing cells. Dev Biol 2015; 408:229-43. [PMID: 25797152 DOI: 10.1016/j.ydbio.2015.03.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/06/2015] [Accepted: 03/11/2015] [Indexed: 11/30/2022]
Abstract
Spinal cord regeneration is very inefficient in humans, causing paraplegia and quadriplegia. Studying model organisms that can regenerate the spinal cord in response to injury could be useful for understanding the cellular and molecular mechanisms that explain why this process fails in humans. Here, we use Xenopus laevis as a model organism to study spinal cord repair. Histological and functional analyses showed that larvae at pre-metamorphic stages restore anatomical continuity of the spinal cord and recover swimming after complete spinal cord transection. These regenerative capabilities decrease with onset of metamorphosis. The ability to study regenerative and non-regenerative stages in Xenopus laevis makes it a unique model system to study regeneration. We studied the response of Sox2(/)3 expressing cells to spinal cord injury and their function in the regenerative process. We found that cells expressing Sox2 and/or Sox3 are present in the ventricular zone of regenerative animals and decrease in non-regenerative froglets. Bromodeoxyuridine (BrdU) experiments and in vivo time-lapse imaging studies using green fluorescent protein (GFP) expression driven by the Sox3 promoter showed a rapid, transient and massive proliferation of Sox2(/)3(+) cells in response to injury in the regenerative stages. The in vivo imaging also demonstrated that Sox2(/)3(+) neural progenitor cells generate neurons in response to injury. In contrast, these cells showed a delayed and very limited response in non-regenerative froglets. Sox2 knockdown and overexpression of a dominant negative form of Sox2 disrupts locomotor and anatomical-histological recovery. We also found that neurogenesis markers increase in response to injury in regenerative but not in non-regenerative animals. We conclude that Sox2 is necessary for spinal cord regeneration and suggest a model whereby spinal cord injury activates proliferation of Sox2/3 expressing cells and their differentiation into neurons, a mechanism that is lost in non-regenerative froglets.
Collapse
Affiliation(s)
- Rosana Muñoz
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Gabriela Edwards-Faret
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Mauricio Moreno
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Nikole Zuñiga
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile
| | - Hollis Cline
- The Dorris Neuroscience Center, Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, La Jolla, CA 92037, United States
| | - Juan Larraín
- Center for Aging and Regeneration, Millennium Nucleus in Regenerative Biology, Faculty of Biological Sciences, P. Universidad Católica de Chile, Alameda 340, Santiago, Chile.
| |
Collapse
|
53
|
Sharma TP, Liu Y, Wordinger RJ, Pang IH, Clark AF. Neuritin 1 promotes retinal ganglion cell survival and axonal regeneration following optic nerve crush. Cell Death Dis 2015; 6:e1661. [PMID: 25719245 PMCID: PMC4669798 DOI: 10.1038/cddis.2015.22] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 12/11/2014] [Accepted: 01/06/2015] [Indexed: 12/16/2022]
Abstract
Neuritin 1 (Nrn1) is an extracellular glycophosphatidylinositol-linked protein that stimulates axonal plasticity, dendritic arborization and synapse maturation in the central nervous system (CNS). The purpose of this study was to evaluate the neuroprotective and axogenic properties of Nrn1 on axotomized retinal ganglion cells (RGCs) in vitro and on the in vivo optic nerve crush (ONC) mouse model. Axotomized cultured RGCs treated with recombinant hNRN1 significantly increased survival of RGCs by 21% (n=6–7, P<0.01) and neurite outgrowth in RGCs by 141% compared to controls (n=15, P<0.05). RGC transduction with AAV2-CAG–hNRN1 prior to ONC promoted RGC survival (450%, n=3–7, P<0.05) and significantly preserved RGC function by 70% until 28 days post crush (dpc) (n=6, P<0.05) compared with the control AAV2-CAG–green fluorescent protein transduction group. Significantly elevated levels of RGC marker, RNA binding protein with multiple splicing (Rbpms; 73%, n=5–8, P<0.001) and growth cone marker, growth-associated protein 43 (Gap43; 36%, n=3, P<0.01) were observed 28 dpc in the retinas of the treatment group compared with the control group. Significant increase in Gap43 (100%, n=5–6, P<0.05) expression was observed within the optic nerves of the AAV2–hNRN1 group compared to controls. In conclusion, Nrn1 exhibited neuroprotective, regenerative effects and preserved RGC function on axotomized RGCs in vitro and after axonal injury in vivo. Nrn1 is a potential therapeutic target for CNS neurodegenerative diseases.
Collapse
Affiliation(s)
- T P Sharma
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Y Liu
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - R J Wordinger
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - I-H Pang
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA
| | - A F Clark
- 1] North Texas Eye Research Institute, University of North Texas Health Science Center, Ft. Worth, TX 76107, USA [2] Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
54
|
Neuritin can normalize neural deficits of Alzheimer's disease. Cell Death Dis 2014; 5:e1523. [PMID: 25393479 PMCID: PMC4260736 DOI: 10.1038/cddis.2014.478] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 10/05/2014] [Accepted: 10/09/2014] [Indexed: 12/15/2022]
Abstract
Reductions in hippocampal neurite complexity and synaptic plasticity are believed to contribute to the progressive impairment in episodic memory and the mild cognitive decline that occur particularly in the early stages of Alzheimer's disease (AD). Despite the functional and therapeutic importance for patients with AD, intervention to rescue or normalize dendritic elaboration and synaptic plasticity is scarcely provided. Here we show that overexpression of neuritin, an activity-dependent protein, promoted neurite outgrowth and maturation of synapses in parallel with enhanced basal synaptic transmission in cultured hippocampal neurons. Importantly, exogenous application of recombinant neuritin fully restored dendritic complexity as well as spine density in hippocampal neurons prepared from Tg2576 mice, whereas it did not affect neurite branching of neurons from their wild-type littermates. We also showed that soluble recombinant neuritin, when chronically infused into the brains of Tg2576 mice, normalized synaptic plasticity in acute hippocampal slices, leading to intact long-term potentiation. By revealing the protective actions of soluble neuritin against AD-related neural defects, we provide a potential therapeutic approach for patients with AD.
Collapse
|
55
|
Menelaou E, VanDunk C, McLean DL. Differences in the morphology of spinal V2a neurons reflect their recruitment order during swimming in larval zebrafish. J Comp Neurol 2014; 522:1232-48. [PMID: 24114934 DOI: 10.1002/cne.23465] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 08/29/2013] [Accepted: 09/12/2013] [Indexed: 12/21/2022]
Abstract
Networks of neurons in spinal cord generate locomotion. However, little is known about potential differences in network architecture that underlie the production of varying speeds of movement. In larval zebrafish, as swimming speed increases, Chx10-positive V2a excitatory premotor interneurons are activated from ventral to dorsal in a topographic pattern that parallels axial motoneuron recruitment. Here, we examined whether differences in the morphology and synaptic output of V2a neurons reflect their recruitment order during swimming. To do so, we used in vivo single-cell labeling approaches to quantify the dorsoventral distribution of V2a axonal projections and synapses. Two different classes of V2a neurons are described, cells with ascending and descending axons and cells that are only descending. Among the purely descending V2a cells, more dorsal cells project longer distances than ventral ones. Proximally, all V2a neurons have axonal distributions that suggest potential connections to cells at and below their own soma positions. At more distal locations, V2a axons project dorsally, which creates a cumulative intersegmental bias to dorsally located spinal neurons. Assessments of the synapse distribution of V2a cells, reported by synaptophysin expression, support the morphological observations and also demonstrate that dorsal V2a cells have higher synapse densities proximally. Our results suggest that V2a cells with more potential output to spinal neurons are systematically engaged during increases in swimming frequency. The findings help explain patterns of axial motoneuron recruitment and set up clear predictions for future physiological studies examining the nature of spinal excitatory network connectivity as it relates to movement intensity.
Collapse
Affiliation(s)
- Evdokia Menelaou
- Department of Neurobiology, Northwestern University, Evanston, Illinois, 60208
| | | | | |
Collapse
|
56
|
Choi Y, Lee K, Ryu J, Kim HG, Jeong AY, Woo RS, Lee JH, Hyun JW, Hahn S, Kim JH, Kim HS. Neuritin attenuates cognitive function impairments in tg2576 mouse model of Alzheimer's disease. PLoS One 2014; 9:e104121. [PMID: 25101829 PMCID: PMC4125179 DOI: 10.1371/journal.pone.0104121] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 07/11/2014] [Indexed: 12/21/2022] Open
Abstract
Neuritin, also known as CPG15, is a neurotrophic factor that was initially discovered in a screen to identify genes involved in activity-dependent synaptic plasticity. Neuritin plays multiple roles in the process of neural development and synaptic plasticity, although its binding receptor(s) and downstream signaling effectors remain unclear. In this study, we found that the cortical and hippocampal expression of neuritin is reduced in the brains of Alzheimer's disease (AD) patients and demonstrated that viral-mediated expression of neuritin in the dentate gyrus of 13-month-old Tg2576 mice, an AD animal model, attenuated a deficit in learning and memory as assessed by a Morris water maze test. We also found that neuritin restored the reduction in dendritic spine density and the maturity of individual spines in primary hippocampal neuron cultures prepared from Tg2576 mice. It was also shown that viral-mediated expression of neuritin in the dentate gyrus of 7-week-old Sprague-Dawley rats increased neurogenesis in the hippocampus. Taken together, our results demonstrate that neuritin restores the reduction in dendritic spine density and the maturity of individual spines in primary hippocampal neurons from Tg2576 neurons, and also attenuates cognitive function deficits in Tg2576 mouse model of AD, suggesting that neuritin possesses a therapeutic potential for AD.
Collapse
Affiliation(s)
- Yoori Choi
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kihwan Lee
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
- National Research Laboratory for Pain, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Junghwa Ryu
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyoun Geun Kim
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - A Young Jeong
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Jun-Ho Lee
- Department of Emergency Medical Technology, Daejeon University, Daejeon, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju, Republic of Korea
| | - Seokyung Hahn
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Joung-Hun Kim
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- * E-mail: (J-HK); (H-SK)
| | - Hye-Sun Kim
- Department of Pharmacology and Biomedical Sciences, Neuroscience Research Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Seoul National University Bundang Hospital, Seoul National University College of Medicine, Sungnam, Republic of Korea
- * E-mail: (J-HK); (H-SK)
| |
Collapse
|
57
|
Gao R, Wang L, Sun J, Nie K, Jian H, Gao L, Liao X, Zhang H, Huang J, Gan S. MiR-204 promotes apoptosis in oxidative stress-induced rat Schwann cells by suppressing neuritin
expression. FEBS Lett 2014; 588:3225-32. [DOI: 10.1016/j.febslet.2014.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/29/2014] [Accepted: 07/06/2014] [Indexed: 02/04/2023]
|
58
|
|
59
|
Aberrant development and plasticity of excitatory visual cortical networks in the absence of cpg15. J Neurosci 2014; 34:3517-22. [PMID: 24599452 DOI: 10.1523/jneurosci.2955-13.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During development, experience plays a crucial role in sculpting neuronal connections. Patterned neural activity guides formation of functional neural circuits through the selective stabilization of some synapses and the pruning of others. Activity-regulated factors are fundamental to this process, but their roles in synapse stabilization and maturation is still poorly understood. CPG15, encoded by the activity-regulated gene candidate plasticity gene 15, is a small, glycosylphosphatidylinositol (GPI)-linked, extracellular protein that promotes synapse stabilization. Here we show that global knock-out of cpg15 results in abnormal postnatal development of the excitatory network in visual cortex and an associated disruption in development of visual receptive field properties. In addition, whereas repeated stimulation induced potentiation and depression in wild-type mice, the depression was slower in cpg15 knock-out mice, suggesting impairment in short-term depression-like mechanisms. These findings establish the requirement for cpg15 in activity-dependent development of the visual system and demonstrate the importance of timely excitatory network development for normal visual function.
Collapse
|
60
|
Sharma TP, McDowell CM, Liu Y, Wagner AH, Thole D, Faga BP, Wordinger RJ, Braun TA, Clark AF. Optic nerve crush induces spatial and temporal gene expression patterns in retina and optic nerve of BALB/cJ mice. Mol Neurodegener 2014; 9:14. [PMID: 24767545 PMCID: PMC4113182 DOI: 10.1186/1750-1326-9-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/18/2014] [Indexed: 12/18/2022] Open
Abstract
Background Central nervous system (CNS) trauma and neurodegenerative disorders trigger a cascade of cellular and molecular events resulting in neuronal apoptosis and regenerative failure. The pathogenic mechanisms and gene expression changes associated with these detrimental events can be effectively studied using a rodent optic nerve crush (ONC) model. The purpose of this study was to use a mouse ONC model to: (a) evaluate changes in retina and optic nerve (ON) gene expression, (b) identify neurodegenerative pathogenic pathways and (c) discover potential new therapeutic targets. Results Only 54% of total neurons survived in the ganglion cell layer (GCL) 28 days post crush. Using Bayesian Estimation of Temporal Regulation (BETR) gene expression analysis, we identified significantly altered expression of 1,723 and 2,110 genes in the retina and ON, respectively. Meta-analysis of altered gene expression (≥1.5, ≤-1.5, p < 0.05) using Partek and DAVID demonstrated 28 up and 20 down-regulated retinal gene clusters and 57 up and 41 down-regulated optic nerve clusters. Regulated gene clusters included regenerative change, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. Expression of selected genes (Vsnl1, Syt1, Synpr and Nrn1) from retinal and ON neuronal clusters were quantitatively and qualitatively examined for their relation to axonal neurodegeneration by immunohistochemistry and qRT-PCR. Conclusion A number of detrimental gene expression changes occur that contribute to trauma-induced neurodegeneration after injury to ON axons. Nrn1 (synaptic plasticity gene), Synpr and Syt1 (synaptic vesicle fusion genes), and Vsnl1 (neuron differentiation associated gene) were a few of the potentially unique genes identified that were down-regulated spatially and temporally in our rodent ONC model. Bioinformatic meta-analysis identified significant tissue-specific and time-dependent gene clusters associated with regenerative changes, synaptic plasticity, axonogenesis, neuron projection, and neuron differentiation. These ONC induced neuronal loss and regenerative failure associated clusters can be extrapolated to changes occurring in other forms of CNS trauma or in clinical neurodegenerative pathological settings. In conclusion, this study identified potential therapeutic targets to address two key mechanisms of CNS trauma and neurodegeneration: neuronal loss and regenerative failure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Abbot F Clark
- North Texas Eye Research Institute, Ft, Worth, TX USA.
| |
Collapse
|
61
|
Abstract
Antisense morpholino oligonucleotides (MOs) have become a valuable method to knock down protein levels, to block mRNA splicing, and to interfere with miRNA function. MOs are widely used to alter gene expression during development of Xenopus and zebra fish, where they are typically injected into the fertilized egg or blastomeres. Here, we present methods to use electroporation to target delivery of MOs to the central nervous system of Xenopus laevis or Xenopus tropicalis tadpoles. Briefly, MO electroporation is accomplished by injecting MO solution into the brain ventricle and driving the MOs into cells in the brain with current passing between two platinum plate electrodes, positioned on either side of the target brain area. The method is straightforward and uses standard equipment found in many neuroscience labs. A major advantage of electroporation is that it allows spatial and temporal control of MO delivery and therefore knockdown. Co-electroporation of MOs with cell-type specific fluorescent protein expression plasmids allows morphological analysis of cellular phenotypes. Furthermore, co-electroporation of MOs with rescuing plasmids allows assessment of specificity of the knockdown and phenotypic outcome. By combining MO-mediated manipulations with sophisticated assays of neuronal function, such as electrophysiological recording, behavioral assays, or in vivo time-lapse imaging of neuronal development, the functions of specific proteins and miRNAs within the developing nervous system can be elucidated. These methods can be adapted to apply antisense morpholinos to study protein and RNA function in a variety of complex tissues.
Collapse
Affiliation(s)
- Jennifer E Bestman
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
62
|
Shimada T, Sugiura H, Yamagata K. Neuritin: A therapeutic candidate for promoting axonal regeneration. World J Neurol 2013; 3:138-143. [DOI: 10.5316/wjn.v3.i4.138] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 08/09/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Following injury, the axons of the mammalian central nervous system do not regenerate. Many studies have aimed at understanding the mechanisms that prevent axonal regeneration and at designing ways to overcome the obstacles preventing axonal regrowth. These studies have identified numerous proteins as promoters of axonal regeneration. In this minireviews, we focus on neuritin as a therapeutic candidate for promoting axonal regeneration. Neuritin was first identified as a neuronal-activity-inducible gene product in the rat brain. The overexpression of neuritin in neurons or the application of neuritin to neurons induces neuritogenesis, neurite arborization, and axonal elongation both in vitro and in vivo. These morphological changes are often observed during the first step of axonal regeneration. Indeed, neuritin expression increases during axonal regeneration in the peripheral nervous system (PNS). Conversely, in a mouse model of diabetes mellitus, neuritin expression decreases in the PNS, and this reduced expression may result in deficient axonal regeneration. Neuritin is induced in the hippocampal dentate gyrus after temporal lobe epilepsy or brain ischemia; however, in these conditions, neuritin induction may exacerbate brain dysfunction through mossy fiber sprouting. Together, these findings support the hypothesis that tightly controlled regulation of neuritin may be required for the treatment of each unique axonal pathology.
Collapse
|
63
|
Lewis TL, Courchet J, Polleux F. Cell biology in neuroscience: Cellular and molecular mechanisms underlying axon formation, growth, and branching. ACTA ACUST UNITED AC 2013; 202:837-48. [PMID: 24043699 PMCID: PMC3776347 DOI: 10.1083/jcb.201305098] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proper brain wiring during development is pivotal for adult brain function. Neurons display a high degree of polarization both morphologically and functionally, and this polarization requires the segregation of mRNA, proteins, and lipids into the axonal or somatodendritic domains. Recent discoveries have provided insight into many aspects of the cell biology of axonal development including axon specification during neuronal polarization, axon growth, and terminal axon branching during synaptogenesis.
Collapse
Affiliation(s)
- Tommy L Lewis
- The Scripps Research Institute, Dorris Neuroscience Center, Department of Molecular and Cellular Neuroscience, La Jolla, CA 92037
| | | | | |
Collapse
|
64
|
Axonal localization of neuritin/CPG15 mRNA in neuronal populations through distinct 5' and 3' UTR elements. J Neurosci 2013; 33:13735-42. [PMID: 23966695 DOI: 10.1523/jneurosci.0962-13.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Many neuronal mRNAs are actively transported into distal axons. The 3' untranslated regions (UTRs) of axonal mRNAs often contain cues for their localization. The 3' UTR of neuritin mRNA was shown to be sufficient for localization into axons of hippocampal neurons. Here, we show that neuritin mRNA localizes into axons of rat sensory neurons, but this is predominantly driven by the 5' rather than 3' UTR. Neuritin mRNA shifts from cell body to axon predominantly after nerve crush injury, suggesting that it encodes a growth-associated protein. Consistent with this, overexpression of neuritin increases axon growth but only when its mRNA localizes into the axons.
Collapse
|
65
|
Serotonergic neurosecretory synapse targeting is controlled by netrin-releasing guidepost neurons in Caenorhabditis elegans. J Neurosci 2013; 33:1366-76. [PMID: 23345213 DOI: 10.1523/jneurosci.3471-12.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neurosecretory release sites lack distinct postsynaptic partners, yet target to specific circuits. This targeting specificity regulates local release of neurotransmitters and modulation of adjacent circuits. How neurosecretory release sites target to specific regions is not understood. Here we identify a molecular mechanism that governs the spatial specificity of extrasynaptic neurosecretory terminal (ENT) formation in the serotonergic neurosecretory-motor (NSM) neurons of Caenorhabditis elegans. We show that postembryonic arborization and neurosecretory terminal targeting of the C. elegans NSM neuron is dependent on the Netrin receptor UNC-40/DCC. We observe that UNC-40 localizes to specific neurosecretory terminals at the time of axon arbor formation. This localization is dependent on UNC-6/Netrin, which is expressed by nerve ring neurons that act as guideposts to instruct local arbor and release site formation. We find that both UNC-34/Enabled and MIG-10/Lamellipodin are required downstream of UNC-40 to link the sites of ENT formation to nascent axon arbor extensions. Our findings provide a molecular link between release site development and axon arborization and introduce a novel mechanism that governs the spatial specificity of serotonergic ENTs in vivo.
Collapse
|
66
|
Zito A, Cartelli D, Cappelletti G, Cariboni A, Andrews W, Parnavelas J, Poletti A, Galbiati M. Neuritin 1 promotes neuronal migration. Brain Struct Funct 2012; 219:105-18. [DOI: 10.1007/s00429-012-0487-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/16/2012] [Indexed: 11/30/2022]
|
67
|
Yao JJ, Gao XF, Chow CW, Zhan XQ, Hu CL, Mei YA. Neuritin activates insulin receptor pathway to up-regulate Kv4.2-mediated transient outward K+ current in rat cerebellar granule neurons. J Biol Chem 2012; 287:41534-45. [PMID: 23066017 PMCID: PMC3510849 DOI: 10.1074/jbc.m112.390260] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neuritin is a new neurotrophic factor discovered in a screen to identify genes involved in activity-dependent synaptic plasticity. Neuritin also plays multiple roles in the process of neural development and synaptic plasticity. The receptors for binding neuritin and its downstream signaling effectors, however, remain unclear. Here, we report that neuritin specifically increases the densities of transient outward K(+) currents (I(A)) in rat cerebellar granule neurons (CGNs) in a time- and concentration-dependent manner. Neuritin-induced amplification of I(A) is mediated by increased mRNA and protein expression of Kv4.2, the main α-subunit of I(A). Exposure of CGNs to neuritin markedly induces phosphorylation of ERK (pERK), Akt (pAkt), and mammalian target of rapamycin (pmTOR). Neuritin-induced I(A) and increased expression of Kv4.2 are attenuated by ERK, Akt, or mTOR inhibitors. Unexpectedly, pharmacological blockade of insulin receptor, but not the insulin-like growth factor 1 receptor, abrogates the effect of neuritin on I(A) amplification and Kv4.2 induction. Indeed, neuritin activates downstream signaling effectors of the insulin receptor in CGNs and HeLa. Our data reveal, for the first time, an unanticipated role of the insulin receptor in previously unrecognized neuritin-mediated signaling.
Collapse
Affiliation(s)
- Jin-Jing Yao
- Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China
| | | | | | | | | | | |
Collapse
|
68
|
Therapy development for spinal muscular atrophy in SMN independent targets. Neural Plast 2012; 2012:456478. [PMID: 22701806 PMCID: PMC3369530 DOI: 10.1155/2012/456478] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/05/2012] [Accepted: 04/06/2012] [Indexed: 12/11/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disorder, leading to progressive muscle weakness, atrophy, and sometimes premature death. SMA is caused by mutation or deletion of the survival motor neuron-1 (SMN1) gene. An effective treatment does not presently exist. Since the severity of the SMA phenotype is inversely correlated with expression levels of SMN, the SMN-encoded protein, SMN is the most important therapeutic target for development of an effective treatment for SMA. In recent years, numerous SMN independent targets and therapeutic strategies have been demonstrated to have potential roles in SMA treatment. For example, some neurotrophic, antiapoptotic, and myotrophic factors are able to promote survival of motor neurons or improve muscle strength shown in SMA mouse models or clinical trials. Plastin-3, cpg15, and a Rho-kinase inhibitor regulate axonal dynamics and might reduce the influences of SMN depletion in disarrangement of neuromuscular junction. Stem cell transplantation in SMA model mice resulted in improvement of motor behaviors and extension of survival, likely from trophic support. Although most therapies are still under investigation, these nonclassical treatments might provide an adjunctive method for future SMA therapy.
Collapse
|
69
|
Sambataro F, Pennuto M. Cell-autonomous and non-cell-autonomous toxicity in polyglutamine diseases. Prog Neurobiol 2012; 97:152-72. [DOI: 10.1016/j.pneurobio.2011.10.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 10/21/2011] [Accepted: 10/26/2011] [Indexed: 12/21/2022]
|
70
|
Antypa M, Faux C, Eichele G, Parnavelas JG, Andrews WD. Differential gene expression in migratory streams of cortical interneurons. Eur J Neurosci 2012; 34:1584-94. [PMID: 22103416 PMCID: PMC3401901 DOI: 10.1111/j.1460-9568.2011.07896.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cortical interneurons originate in the ganglionic eminences of the subpallium and migrate into the cortex in well-defined tangential streams. At the start of corticogenesis, two streams of migrating neurons are evident: a superficial one at the level of the preplate (PPL), and a deeper one at the level of the intermediate zone (IZ). Currently, little is known about the signalling mechanisms that regulate interneuron migration, and almost nothing is known about the molecules that may be involved in their choice of migratory stream. Here, we performed a microarray analysis, comparing the changes in gene expression between cells migrating in the PPL and those migrating in the IZ at embryonic day 13.5. This analysis identified genes, many of them novel, that were upregulated in one of the two streams. Moreover, polymerase chain reaction, in situ hybridization experiments and immunohistochemistry showed the expression of these genes in interneurons migrating within the PPL or IZ, suggesting that they play a role in their migration and choice of stream.
Collapse
Affiliation(s)
- Mary Antypa
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | | | | | | | | |
Collapse
|
71
|
GABA signaling promotes synapse elimination and axon pruning in developing cortical inhibitory interneurons. J Neurosci 2012; 32:331-43. [PMID: 22219294 DOI: 10.1523/jneurosci.3189-11.2012] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Accumulating evidence indicates that GABA acts beyond inhibitory synaptic transmission and regulates the development of inhibitory synapses in the vertebrate brain, but the underlying cellular mechanism is not well understood. We have combined live imaging of cortical GABAergic axons across time scales from minutes to days with single-cell genetic manipulation of GABA release to examine its role in distinct steps of inhibitory synapse formation in the mouse neocortex. We have shown previously, by genetic knockdown of GABA synthesis in developing interneurons, that GABA signaling promotes the maturation of inhibitory synapses and axons. Here we found that a complete blockade of GABA release in basket interneurons resulted in an opposite effect, a cell-autonomous increase in axon and bouton density with apparently normal synapse structures. These results not only demonstrate that GABA is unnecessary for synapse formation per se but also uncover a novel facet of GABA in regulating synapse elimination and axon pruning. Live imaging revealed that developing GABAergic axons form a large number of transient boutons, but only a subset was stabilized. Release blockade led to significantly increased bouton stability and filopodia density, increased axon branch extension, and decreased branch retraction. Our results suggest that a major component of GABA function in synapse development is transmission-mediated elimination of subsets of nascent contacts. Therefore, GABA may regulate activity-dependent inhibitory synapse formation by coordinately eliminating certain nascent contacts while promoting the maturation of other nascent synapses.
Collapse
|
72
|
Fujino T, Leslie JH, Eavri R, Chen JL, Lin WC, Flanders GH, Borok E, Horvath TL, Nedivi E. CPG15 regulates synapse stability in the developing and adult brain. Genes Dev 2012; 25:2674-85. [PMID: 22190461 DOI: 10.1101/gad.176172.111] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Use-dependent selection of optimal connections is a key feature of neural circuit development and, in the mature brain, underlies functional adaptation, such as is required for learning and memory. Activity patterns guide circuit refinement through selective stabilization or elimination of specific neuronal branches and synapses. The molecular signals that mediate activity-dependent synapse and arbor stabilization and maintenance remain elusive. We report that knockout of the activity-regulated gene cpg15 in mice delays developmental maturation of axonal and dendritic arbors visualized by anterograde tracing and diolistic labeling, respectively. Electrophysiology shows that synaptic maturation is also delayed, and electron microscopy confirms that many dendritic spines initially lack functional synaptic contacts. While circuits eventually develop, in vivo imaging reveals that spine maintenance is compromised in the adult, leading to a gradual attrition in spine numbers. Loss of cpg15 also results in poor learning. cpg15 knockout mice require more trails to learn, but once they learn, memories are retained. Our findings suggest that CPG15 acts to stabilize active synapses on dendritic spines, resulting in selective spine and arbor stabilization and synaptic maturation, and that synapse stabilization mediated by CPG15 is critical for efficient learning.
Collapse
Affiliation(s)
- Tadahiro Fujino
- The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Abstract
Branching morphology is a hallmark feature of axons and dendrites and is essential for neuronal connectivity. To understand how this develops, I analyzed the stereotyped pattern of Drosophila mushroom body (MB) neurons, which have single axons branches that extend dorsally and medially. I found that components of the Wnt/Planar Cell Polarity (PCP) pathway control MB axon branching. frizzled mutant animals showed a predominant loss of dorsal branch extension, whereas strabismus (also known as Van Gogh) mutants preferentially lost medial branches. Further results suggest that Frizzled and Strabismus act independently. Nonetheless, branching fates are determined by complex Wnt/PCP interactions, including interactions with Dishevelled and Prickle that function in a context-dependent manner. Branching decisions are MB-autonomous but non-cell-autonomous as mutant and non-mutant neurons regulate these decisions collectively. I found that Wnt/PCP components do not need to be asymmetrically localized to distinct branches to execute branching functions. However, Prickle axonal localization depends on Frizzled and Strabismus.
Collapse
Affiliation(s)
- Julian Ng
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
74
|
Fallini C, Bassell GJ, Rossoll W. Spinal muscular atrophy: the role of SMN in axonal mRNA regulation. Brain Res 2012; 1462:81-92. [PMID: 22330725 DOI: 10.1016/j.brainres.2012.01.044] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 01/19/2012] [Indexed: 01/15/2023]
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by homozygous mutations or deletions in the survival of motor neuron (SMN1) gene, encoding the ubiquitously expressed SMN protein. SMN associates with different proteins (Gemins 2-8, Unrip) to form a multimeric complex involved in the assembly of small nuclear ribonucleoprotein complexes (snRNPs). Since this activity is essential for the survival of all cell types, it still remains unclear why motor neurons are selectively vulnerable to low levels of SMN protein. Aside from its housekeeping role in the assembly of snRNPs, additional functions of SMN have been proposed. The well-documented localization of SMN in axonal transport granules and its interaction with numerous mRNA-binding proteins not involved in splicing regulation suggest a role in axonal RNA metabolism. This review will focus on the neuropathological and experimental evidence supporting a role for SMN in regulating the assembly, localization, or stability of axonal messenger ribonucleoprotein complexes (mRNPs). Furthermore, how defects in this non-canonical SMN function may contribute to the motor neuron pathology observed in SMA will be discussed. This article is part of a Special Issue entitled RNA-Binding Proteins.
Collapse
Affiliation(s)
- Claudia Fallini
- Department of Cell Biology, School of Medicine, Emory University School of Medicine, Atlanta 30322, USA
| | | | | |
Collapse
|
75
|
Characterization of neuritin as a novel angiogenic factor. Biochem Biophys Res Commun 2011; 415:608-12. [DOI: 10.1016/j.bbrc.2011.10.118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 10/26/2011] [Indexed: 12/30/2022]
|
76
|
Interaction of survival of motor neuron (SMN) and HuD proteins with mRNA cpg15 rescues motor neuron axonal deficits. Proc Natl Acad Sci U S A 2011; 108:10337-42. [PMID: 21652774 DOI: 10.1073/pnas.1104928108] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spinal muscular atrophy (SMA), caused by the deletion of the SMN1 gene, is the leading genetic cause of infant mortality. SMN protein is present at high levels in both axons and growth cones, and loss of its function disrupts axonal extension and pathfinding. SMN is known to associate with the RNA-binding protein hnRNP-R, and together they are responsible for the transport and/or local translation of β-actin mRNA in the growth cones of motor neurons. However, the full complement of SMN-interacting proteins in neurons remains unknown. Here we used mass spectrometry to identify HuD as a novel neuronal SMN-interacting partner. HuD is a neuron-specific RNA-binding protein that interacts with mRNAs, including candidate plasticity-related gene 15 (cpg15). We show that SMN and HuD form a complex in spinal motor axons, and that both interact with cpg15 mRNA in neurons. CPG15 is highly expressed in the developing ventral spinal cord and can promote motor axon branching and neuromuscular synapse formation, suggesting a crucial role in the development of motor axons and neuromuscular junctions. Cpg15 mRNA previously has been shown to localize into axonal processes. Here we show that SMN deficiency reduces cpg15 mRNA levels in neurons, and, more importantly, cpg15 overexpression partially rescues the SMN-deficiency phenotype in zebrafish. Our results provide insight into the function of SMN protein in axons and also identify potential targets for the study of mechanisms that lead to the SMA pathology and related neuromuscular diseases.
Collapse
|
77
|
Appelbaum L, Wang G, Yokogawa T, Skariah GM, Smith SJ, Mourrain P, Mignot E. Circadian and homeostatic regulation of structural synaptic plasticity in hypocretin neurons. Neuron 2010; 68:87-98. [PMID: 20920793 DOI: 10.1016/j.neuron.2010.09.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2010] [Indexed: 12/12/2022]
Abstract
Neurons exhibit rhythmic activity that ultimately affects behavior such as sleep. In living zebrafish larvae, we used time-lapse two-photon imaging of the presynaptic marker synaptophysin in hypocretin/orexin (HCRT) neurons to determine the dynamics of synaptic modifications during the day and night. We observed circadian rhythmicity in synapse number in HCRT axons. This rhythm is regulated primarily by the circadian clock but is also affected by sleep deprivation. Furthermore, NPTX2, a protein implicated in AMPA receptor clustering, modulates circadian synaptic changes. In zebrafish, nptx2b is a rhythmic gene that is mostly expressed in hypothalamic and pineal gland cells. Arrhythmic transgenic nptx2b overexpression (hcrt:NPTX2b) increases synapse number and abolishes rhythmicity in HCRT axons. Finally, hcrt:NPTX2b fish are resistant to the sleep-promoting effects of melatonin. This behavioral effect is consistent with NPTX2b-mediated increased activity of HCRT circuitry. These data provide real-time in vivo evidence of circadian and homeostatic regulation of structural synaptic plasticity.
Collapse
Affiliation(s)
- Lior Appelbaum
- Center for Narcolepsy, Department of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California 94305, USA.
| | | | | | | | | | | | | |
Collapse
|
78
|
|
79
|
Li L, Tasic B, Micheva KD, Ivanov VM, Spletter ML, Smith SJ, Luo L. Visualizing the distribution of synapses from individual neurons in the mouse brain. PLoS One 2010; 5:e11503. [PMID: 20634890 PMCID: PMC2901335 DOI: 10.1371/journal.pone.0011503] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 06/14/2010] [Indexed: 01/01/2023] Open
Abstract
Background Proper function of the mammalian brain relies on the establishment of highly specific synaptic connections among billions of neurons. To understand how complex neural circuits function, it is crucial to precisely describe neuronal connectivity and the distributions of synapses to and from individual neurons. Methods and Findings In this study, we present a new genetic synaptic labeling method that relies on expression of a presynaptic marker, synaptophysin-GFP (Syp-GFP) in individual neurons in vivo. We assess the reliability of this method and use it to analyze the spatial patterning of synapses in developing and mature cerebellar granule cells (GCs). In immature GCs, Syp-GFP is distributed in both axonal and dendritic regions. Upon maturation, it becomes strongly enriched in axons. In mature GCs, we analyzed synapses along their ascending segments and parallel fibers. We observe no differences in presynaptic distribution between GCs born at different developmental time points and thus having varied depths of projections in the molecular layer. We found that the mean densities of synapses along the parallel fiber and the ascending segment above the Purkinje cell (PC) layer are statistically indistinguishable, and higher than previous estimates. Interestingly, presynaptic terminals were also found in the ascending segments of GCs below and within the PC layer, with the mean densities two-fold lower than that above the PC layer. The difference in the density of synapses in these parts of the ascending segment likely reflects the regional differences in postsynaptic target cells of GCs. Conclusions The ability to visualize synapses of single neurons in vivo is valuable for studying synaptogenesis and synaptic plasticity within individual neurons as well as information flow in neural circuits.
Collapse
Affiliation(s)
- Ling Li
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, California, United States of America
| | - Bosiljka Tasic
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, California, United States of America
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Vsevolod M. Ivanov
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, California, United States of America
- Lynbrook High School, San Jose, California, United States of America
| | - Maria L. Spletter
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, California, United States of America
| | - Stephen J. Smith
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California, United States of America
| | - Liqun Luo
- Howard Hughes Medical Institute, Department of Biology, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
80
|
Li J, Cline HT. Visual deprivation increases accumulation of dense core vesicles in developing optic tectal synapses in Xenopus laevis. J Comp Neurol 2010; 518:2365-81. [PMID: 20437533 PMCID: PMC2980367 DOI: 10.1002/cne.22338] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Despite considerable progress in understanding the molecular components of synapses in the central nervous system, the ultrastructural rearrangements underlying synaptic development remain unclear. We used serial section transmission electron microscopy and three-dimensional reconstructions of the optic tectal neuropil of Xenopus laevis tadpoles to detect and quantify changes in synaptic ultrastructure over a 1-week period from stages 39 and 47, during which time the visual system of Xenopus tadpoles becomes functional. Synapse density, presynaptic maturation index, and number of synapses per axon bouton increase, whereas the number of DCVs per bouton decreases, between stages 39 and 47. The width of the synaptic cleft decreased and the diameter of postsynaptic profiles increased between stages 39 and 47 and then remained relatively unchanged after stage 47. We found no significant difference in synapse maturation between GABAergic and non-GABAergic synapses. To test the effect of visual experience on synaptogenesis, animals were deprived of visual experience for 3 days from stage 42 to 47. Visual deprivation decreased synapse maturation and the number of connections per bouton. Furthermore, visual deprivation increased the number of DCVs per bouton by more than twofold. The visual-deprivation-induced decrease in synaptic connections is specific to asymmetric non-GABAergic synapses; however, both symmetric GABAergic and asymmetric synapses show comparable increases in the number DCVs with visual deprivation. In both the control and the visually deprived animals, the number of DCVs per bouton is highly variable and does not correlate with either synapse maturation or the number of connected partners per bouton. These data suggest that synaptogenesis and DCV accumulation are regulated by visual experience and further suggest a complex spatial and temporal relation between DCV accumulation and synapse formation.
Collapse
Affiliation(s)
- Jianli Li
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037
| | - Hollis T. Cline
- Department of Cell Biology, The Scripps Research Institute, La Jolla, California 92037
| |
Collapse
|
81
|
Electrical stimulation and testosterone differentially enhance expression of regeneration-associated genes. Exp Neurol 2010; 223:183-91. [DOI: 10.1016/j.expneurol.2009.04.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 04/26/2009] [Accepted: 04/28/2009] [Indexed: 02/04/2023]
|
82
|
Abstract
In this issue of Neuron, Snider et al., analyze dendritic and axonal arbors of several cell types in several species. They show that general features of arbor structure are shared by the diverse cell populations, suggesting that the growth of these arbors is guided by universal principles.
Collapse
|
83
|
Netrin participates in the development of retinotectal synaptic connectivity by modulating axon arborization and synapse formation in the developing brain. J Neurosci 2009; 29:11065-77. [PMID: 19741113 DOI: 10.1523/jneurosci.0947-09.2009] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Netrin has been implicated in retinal ganglion cell (RGC) axon pathfinding in a number of species. In Xenopus laevis, RGC axons reaching their target in the optic tectum can be repelled by a netrin-1 gradient in vitro, suggesting that netrin may also function in wiring events that follow successful axon pathfinding. Here, we examined the contribution of netrin to RGC axon arborization and synapse formation at the target. Time-lapse confocal microscopy imaging of individual RGC axons coexpressing GFP-synaptobrevin and DsRed in the intact Xenopus brain demonstrated a role for deleted in colorectal cancer (DCC)-mediated netrin signaling. Microinjection of netrin-1 into the tectum induced a rapid and transient increase in presynaptic site addition that resulted in higher presynaptic site density over a 24 h observation period. Moreover, netrin induced dynamic axon branching, increasing branch addition and retraction; a behavior that ultimately increased total branch number. In contrast, microinjection of DCC function-blocking antibodies prevented the increase in presynaptic site number normally observed in control axons as well as the associated increase in branch number and axon arbor growth. Dynamic analysis of axon arbors demonstrated that the effects of anti-DCC on axon morphology and presynaptic connectivity were attributable to a specific decrease in new synapse and branch additions, without affecting the stability of existing synapses and branches. Together, these results indicate that, in the absence of DCC signaling, RGC axons fail to branch and differentiate, and support a novel role for netrin in later phases of retinotectal development.
Collapse
|
84
|
Epileptogenesis alters gene expression pattern in rats subjected to amygdala-dependent emotional learning. Neuroscience 2009; 159:468-82. [DOI: 10.1016/j.neuroscience.2008.12.060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Revised: 12/11/2008] [Accepted: 12/31/2008] [Indexed: 11/18/2022]
|
85
|
|
86
|
Park KW, Kim IH, Sun W, Kim H. Sustained Expression of Neuritin mRNA After Repeated Electroconvulsive Stimulations in the Rat Hippocampal Formation. Exp Neurobiol 2009. [DOI: 10.5607/en.2009.18.1.62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Kun Woo Park
- Department of Neurology, College of Medicine, Korea University, Seoul 136-705, Korea
| | - Il Hwan Kim
- Department of Anatomy, Brain Korea 21, College of Medicine, Korea University, Seoul 136-705, Korea
| | - Woong Sun
- Department of Anatomy, Brain Korea 21, College of Medicine, Korea University, Seoul 136-705, Korea
| | - Hyun Kim
- Department of Anatomy, Brain Korea 21, College of Medicine, Korea University, Seoul 136-705, Korea
| |
Collapse
|
87
|
Abstract
Information processing in the nervous system relies on properly localized and organized synaptic structures at the correct locations. The formation of synapses is a long and intricate process involving multiple interrelated steps. Decades of research have identified a large number of molecular components of the presynaptic compartment. In addition to neurotransmitter-containing synaptic vesicles, presynaptic terminals are defined by cytoskeletal and membrane specializations that allow highly regulated exo- and endocytosis of synaptic vesicles and that maintain precise registration with postsynaptic targets. Functional studies at multiple levels have revealed complex interactions between the transport of vesicular intermediates, the presynaptic cytoskeleton, growth cone navigation, and synaptic targets. With the advent of finer anatomical, physiological, and molecular tools, great insights have been gained toward the mechanistic dissection of functionally redundant processes controlling the specificity and dynamics of synapses. This review highlights the recent findings pertaining to the cellular and molecular regulation of presynaptic differentiation.
Collapse
Affiliation(s)
- Yishi Jin
- Division of Biological Sciences, Section of Neurobiology, Howard Hughes Medical Institute, University of California, San Diego, La Jolla, California 92093, USA.
| | | |
Collapse
|
88
|
Abstract
Imaging and molecular approaches are perfectly suited to young, transparent zebrafish (Danio rerio), where they have allowed novel functional studies of neural circuits and their links to behavior. Here, we review cutting-edge optical and genetic techniques used to dissect neural circuits in vivo and discuss their application to future studies of developing spinal circuits using living zebrafish. We anticipate that these experiments will reveal general principles governing the assembly of neural circuits that control movements.
Collapse
Affiliation(s)
- David L McLean
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA.
| | | |
Collapse
|
89
|
Fargo KN, Galbiati M, Foecking EM, Poletti A, Jones KJ. Androgen regulation of axon growth and neurite extension in motoneurons. Horm Behav 2008; 53:716-28. [PMID: 18387610 PMCID: PMC2408920 DOI: 10.1016/j.yhbeh.2008.01.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 01/11/2008] [Accepted: 01/18/2008] [Indexed: 01/10/2023]
Abstract
Androgens act on the CNS to affect motor function through interaction with a widespread distribution of intracellular androgen receptors (AR). This review highlights our work on androgens and process outgrowth in motoneurons, both in vitro and in vivo. The actions of androgens on motoneurons involve the generation of novel neuronal interactions that are mediated by the induction of androgen-dependent neurite or axonal outgrowth. Here, we summarize the experimental evidence for the androgenic regulation of the extension and regeneration of motoneuron neurites in vitro using cultured immortalized motoneurons, and axons in vivo using the hamster facial nerve crush paradigm. We place particular emphasis on the relevance of these effects to SBMA and peripheral nerve injuries.
Collapse
Affiliation(s)
- Keith N Fargo
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University Chicago, Maywood, Illinois 60153, USA.
| | | | | | | | | |
Collapse
|
90
|
Fargo KN, Alexander TD, Tanzer L, Poletti A, Jones KJ. Androgen regulates neuritin mRNA levels in an in vivo model of steroid-enhanced peripheral nerve regeneration. J Neurotrauma 2008; 25:561-6. [PMID: 18419250 PMCID: PMC9848905 DOI: 10.1089/neu.2007.0466] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Following crush injury to the facial nerve in Syrian hamsters, treatment with androgens enhances axonal regeneration rates and decreases time to recovery. It has been demonstrated in vitro that the ability of androgen to enhance neurite outgrowth in motoneurons is dependent on neuritin-a protein that is involved in the re-establisment of neuronal connectivity following traumatic damage to the central nervous system and that is under the control of several neurotrophic and neuroregenerative factors--and we have hypothesized that neuritin is a mediator of the ability of androgen to increase peripheral nerve regeneration rates in vivo. Testosterone treatment of facial nerve-axotomized hamsters resulted in an approximately 300% increase in neuritin mRNA levels 2 days post-injury. Simultaneous treatment with flutamide, an androgen receptor blocker that is known to prevent androgen enhancement of nerve regeneration, abolished the ability of testosterone to upregulate neuritin mRNA levels. In a corroborative in vitro experiment, the androgen dihydrotestosterone induced an approximately 100% increase in neuritin mRNA levels in motoneuron-neuroblastoma cells transfected with androgen receptors, but not in cells without androgen receptors. These data confirm that neuritin is under the control of androgens, and suggest that neuritin is an important effector of androgen in enhancing peripheral nerve regeneration following injury. Given that neuritin has now been shown to be involved in responses to both central and peripheral injuries, and appears to be a common effector molecule for several neurotrophic and neurotherapeutic agents, understanding the neuritin pathway is an important goal for the clinical management of traumatic nervous system injuries.
Collapse
Affiliation(s)
- Keith N Fargo
- Neuroscience Program and Department of Cell Biology, Neurobiology and Anatomy, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, USA.
| | | | | | | | | |
Collapse
|
91
|
FUJINO TADAHIRO, WU ZHEN, LIN WALTERC, PHILLIPS MARNIEA, NEDIVI ELLY. cpg15 and cpg15-2 constitute a family of activity-regulated ligands expressed differentially in the nervous system to promote neurite growth and neuronal survival. J Comp Neurol 2008; 507:1831-45. [PMID: 18265009 PMCID: PMC2828060 DOI: 10.1002/cne.21649] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Many ligands that affect nervous system development are members of gene families that function together to coordinate the assembly of complex neural circuits. cpg15/neuritin encodes an extracellular ligand that promotes neurite growth, neuronal survival, and synaptic maturation. Here we identify cpg15-2 as the only paralogue of cpg15 in the mouse and human genome. Both genes are expressed predominantly in the nervous system, where their expression is regulated by activity. cpg15-2 expression increases by more than twofold in response to kainate-induced seizures and nearly fourfold in the visual cortex in response to 24 hours of light exposure following dark adaptation. cpg15 and cpg15-2 diverge in their spatial and temporal expression profiles. cpg15-2 mRNA is most abundant in the retina and the olfactory bulb, as opposed to the cerebral cortex and the hippocampus for cpg15. In the retina, they differ in their cell-type specificity. cpg15 is expressed in retinal ganglion cells, whereas cpg15-2 is predominantly in bipolar cells. Developmentally, onset of cpg15-2 expression is delayed compared with cpg15 expression. CPG15-2 is glycosylphosphatidylinositol (GPI) anchored to the cell membrane and, like CPG15, can be released in a soluble-secreted form, but with lower efficiency. CPG15 and CPG15-2 were found to form homodimers and heterodimers with each other. In hippocampal explants and dissociated cultures, CPG15 and CPG15-2 promote neurite growth and neuronal survival with similar efficacy. Our findings suggest that CPG15 and CPG15-2 perform similar cellular functions but may play distinct roles in vivo through their cell-type- and tissue-specific transcriptional regulation.
Collapse
Affiliation(s)
- TADAHIRO FUJINO
- The Picower Institute for Learning and Memory, Departments of Brain and Cognitive Sciences and Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - ZHEN WU
- The Picower Institute for Learning and Memory, Departments of Brain and Cognitive Sciences and Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - WALTER C. LIN
- The Picower Institute for Learning and Memory, Departments of Brain and Cognitive Sciences and Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - MARNIE A. PHILLIPS
- The Picower Institute for Learning and Memory, Departments of Brain and Cognitive Sciences and Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - ELLY NEDIVI
- The Picower Institute for Learning and Memory, Departments of Brain and Cognitive Sciences and Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
92
|
Abstract
The calyx of Held is probably the largest synaptic terminal in the brain, forming a unique one-to-one connection in the auditory ventral brainstem. During early development, calyces have many collaterals, whose function is unknown. Using electrophysiological recordings and fast-calcium imaging in brain slices, we demonstrate that these collaterals are involved in synaptic transmission. We show evidence that the collaterals are pruned and that the pruning already begins 1 week before the onset of hearing. Using two-photon microscopy to image the calyx of Held in neonate rats, we report evidence that both axons and nascent calyces are structurally dynamic, showing the formation, elimination, extension, or retraction of up to 65% of their collaterals within 1 hour. The observed dynamic behavior of axons may add flexibility in the choice of postsynaptic partners and thereby contribute to ensuring that each principal cell eventually is contacted by a single calyx of Held.
Collapse
|
93
|
Abstract
The appeal of in vivo cellular imaging to any neuroscientist is not hard to understand: it is almost impossible to isolate individual neurons while keeping them and their complex interactions with surrounding tissue intact. These interactions lead to the complex network dynamics that underlie neural computation which, in turn, forms the basis of cognition, perception and consciousness. In vivo imaging allows the study of both form and function in reasonably intact preparations, often with subcellular spatial resolution, a time resolution of milliseconds and a purview of months. Recently, the limits of what can be achieved in vivo have been pushed into terrain that was previously only accessible in vitro, due to advances in both physical-imaging technology and the design of molecular contrast agents.
Collapse
|
94
|
Abstract
NMDA receptors (NMDARs) are important for neuronal development and circuit formation. The NMDAR subunits NR2A and NR2B are biophysically distinct and differentially expressed during development but their individual contribution to structural plasticity is unknown. Here we test whether NR2A and NR2B subunits have specific functions in the morphological development of tectal neurons in living Xenopus tadpoles. We use exogenous subunit expression and endogenous subunit knockdown to shift synaptic NMDAR composition toward NR2A or NR2B, as shown electrophysiologically. We analyzed the dendritic arbor structure and found evidence for both overlapping and distinct functions of NR2A and NR2B in dendritic development. Control neurons develop regions of high local branch density in their dendritic arbor, which may be important for processing topographically organized inputs. Exogenous expression of either NR2A or NR2B decreases local branch clusters, indicating a requirement for both subunits in dendritic arbor development. Knockdown of endogenous NR2A reduces local branch clusters, whereas knockdown of NR2B has no effect on branch clustering. Analysis of the underlying branch dynamics shows that exogenous NR2B-expressing neurons are more dynamic than control or exogenous NR2A-expressing neurons, demonstrating subunit-specific regulation of branch dynamics. Visual experience-dependent increases in dendritic arbor growth rate seen in control neurons are blocked in both exogenous NR2A- and NR2B-expressing neurons. These experiments indicate that NR2A and NR2B have subunit-specific properties in dendritic arbor development, but also overlapping functions, indicating a requirement for both subunits in neuronal development.
Collapse
|
95
|
Karamoysoyli E, Burnand RC, Tomlinson DR, Gardiner NJ. Neuritin mediates nerve growth factor-induced axonal regeneration and is deficient in experimental diabetic neuropathy. Diabetes 2008; 57:181-9. [PMID: 17909094 DOI: 10.2337/db07-0895] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Axonal regeneration is defective in both experimental and clinical diabetic neuropathy, contributing to loss of axonal extremities and neuronal dysfunction. The mechanisms behind this failure are not fully understood; however, a deficit in neurotrophic support and signaling has been implicated. RESEARCH DESIGN AND METHODS We investigated the expression of neuritin (also known as candidate plasticity gene 15, cpg15) in the sensory nervous system of control rats and rats with streptozotocin (STZ)-induced diabetes using microarray PCR, Western blotting, and immunocytochemical analysis. The functional role of neuritin in sensory neurons in vitro was assessed using silencing RNA. RESULTS Neuritin was expressed by a population of small-diameter neurons in the dorsal root ganglia (DRG) and was anterogradely and retrogradely transported along the sciatic nerve in vivo. Nerve growth factor (NGF) treatment induced an increase in the transcription and translation of neuritin in sensory neurons in vitro. This increase was both time and dose dependent and occurred via mitogen-activated protein kinase or phosphatidylinositol-3 kinase activation. Inhibition of neuritin using silencing RNA abolished NGF-mediated neurite outgrowth, demonstrating the crucial role played by neuritin in mediating regeneration. Neuritin levels were reduced in both the DRG and sciatic nerve of rats with 12 weeks of STZ-induced diabetes, and these deficits were reversed in vivo by treatment with NGF. CONCLUSIONS Manipulation of neuritin levels in diabetes may therefore provide a potential target for therapeutic intervention in the management of neuropathy.
Collapse
Affiliation(s)
- Eugenia Karamoysoyli
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | | | | | | |
Collapse
|
96
|
Cantallops I, Cline HT. Rapid activity-dependent delivery of the neurotrophic protein CPG15 to the axon surface of neurons in intactXenopus tadpoles. Dev Neurobiol 2008; 68:744-59. [DOI: 10.1002/dneu.20529] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
97
|
Multifunctional role of protein kinase C in regulating the formation and maturation of specific synapses. J Neurosci 2007; 27:11712-24. [PMID: 17959813 DOI: 10.1523/jneurosci.3305-07.2007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Target-dependent increases in axon growth and varicosities accompany the formation of functional synapses between Aplysia sensory neurons and specific postsynaptic neurons (L7 and not L11). The enhanced growth is regulated in part by a target-dependent increase in the secretion of sensorin, the sensory neuron neuropeptide. We report here that protein kinase C (PKC) activity is required for synapse formation by sensory neurons with L7 and for the target-dependent increases in sensorin synthesis and secretion. Blocking PKC activity reversibly blocked synapse formation and axon growth of sensory neurons contacting L7, but did not affect axon growth of sensory neurons contacting L11 or axon growth of the postsynaptic targets. Blocking PKC activity also blocked the target-induced increase in sensorin synthesis and secretion. Sensorin then activates additional signaling pathways required for synapse maturation and synapse-associated growth. Exogenous anti-sensorin antibody blocked target-induced activation and translocation into sensory neuron nuclei of p42/44 mitogen-activated protein kinase (MAPK), attenuated synapse maturation, and curtailed growth of sensory neurons contacting L7, but not the growth of sensory neurons contacting L11. Inhibitors of MAPK or phosphoinositide 3-kinase also attenuated synapse maturation and curtailed growth and varicosity formation of sensory neurons contacting L7, but not growth of sensory neurons contacting L11. These results suggest that PKC activity regulated by specific cell-cell interactions initiates the formation of specific synapses and the subsequent synthesis and release of a neuropeptide to activate additional signaling pathways required for synapse maturation.
Collapse
|
98
|
Rodríguez-González R, Hurtado O, Sobrino T, Castillo J. Neuroplasticity and cellular therapy in cerebral infarction. Cerebrovasc Dis 2007; 24 Suppl 1:167-80. [PMID: 17971653 DOI: 10.1159/000107393] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Stroke is the second to third most common cause of death in adults, and more than a third of people who survive a stroke will have severe disability. Therapeutic options currently centre on fibrinolytic treatment, but its limitations restrict use to a small proportion of patients. Although a wide range of neuroprotective substances has been effective in experimental models, they have repeatedly failed in clinical trials because of toxicity or loss of effectiveness. Recent strategies based on neuroplasticity and cellular therapy have shown significant efficacy in improving functional recovery in experimental models, although further study is still necessary to clarify how the brain responds to ischaemic damage and is able to reorganize itself in the long term. Although steps must still be taken to ensure the safety and feasibility of treatments based on neuroplasticity and cellular therapy, neurorepair strategies provide promising future therapeutic options for stroke.
Collapse
Affiliation(s)
- Raquel Rodríguez-González
- Clinical Neuroscience Research Laboratory, Division of Vascular Neurology, Department of Neurology, Hospital Clínico Universitario, University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | |
Collapse
|
99
|
Cappelletti G, Galbiati M, Ronchi C, Maggioni MG, Onesto E, Poletti A. Neuritin (cpg15) enhances the differentiating effect of NGF on neuronal PC12 cells. J Neurosci Res 2007; 85:2702-13. [PMID: 17335086 DOI: 10.1002/jnr.21235] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Neuritin is a small, highly conserved GPI-anchored protein involved in neurite outgrowth. We have analyzed the involvement of neuritin in NGF-induced differentiation of PC12 cells by investigating the time-course of neuritin expression, the effects of its overexpression or silencing, and the possible mechanisms of its regulation and action. Real-time PCR analysis has shown that neuritin gene is upregulated by NGF in PC12 cells hours before neurite outgrowth becomes appreciable. PC12 cells transfected with a plasmid expressing neuritin display a significant increase in the response to NGF: 1) in the levels of SMI312 positive phosphorylated neurofilament proteins (markers for axonal processes) and tyrosine hydroxylase; 2) in the percentage of cells bearing neurites; as well as 3) in the average length of neurites when compared to control cells. On the contrary, neuritin silencing significantly reduces neurite outgrowth. These data suggest that neuritin is a modulator of NGF-induced neurite extension in PC12 cells. We also showed that neuritin potentiated the NGF-induced differentiation of PC12 cells without affecting TrkA or EGF receptor mRNAs expression. Moreover, the S-methylisothiourea (MIU), a potent inhibitor of inducible nitric oxide synthases, partially counteracts the NGF-mediated neuritin induction. These data suggest that NGF regulates neuritin expression in PC12 cells via the signaling pathway triggered by NO. This study reports the first evidence that neuritin plays a role in modulating neurite outgrowth during the progression of NGF-induced differentiation of PC12 cells. PC12 cells could be considered a valuable model to unravel the mechanism of action of neuritin on neurite outgrowth. (c) 2007 Wiley-Liss, Inc.
Collapse
|
100
|
Campbell DS, Stringham SA, Timm A, Xiao T, Law MY, Baier H, Nonet ML, Chien CB. Slit1a inhibits retinal ganglion cell arborization and synaptogenesis via Robo2-dependent and -independent pathways. Neuron 2007; 55:231-45. [PMID: 17640525 DOI: 10.1016/j.neuron.2007.06.034] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2006] [Revised: 05/30/2007] [Accepted: 06/28/2007] [Indexed: 01/20/2023]
Abstract
Upon arriving at their targets, developing axons cease pathfinding and begin instead to arborize and form synapses. To test whether CNS arborization and synaptogenesis are controlled by Slit-Robo signaling, we followed single retinal ganglion cell (RGC) arbors over time. ast (robo2) mutant and slit1a morphant arbors had more branch tips and greater arbor area and complexity compared to wild-type and concomitantly more presumptive presynaptic sites labeled with YFP-Rab3. Increased arborization in ast was phenocopied by dominant-negative Robo2 expressed in single RGCs and rescued by full-length Robo2, indicating that Robo2 acts cell-autonomously. Time-lapse imaging revealed that ast and slit1a morphant arbors stabilized earlier than wild-type, suggesting a role for Slit-Robo signaling in preventing arbor maturation. Genetic analysis showed that Slit1a acts both through Robo2 and Robo2-independent mechanisms. Unlike previous PNS studies showing that Slits promote branching, our results show that Slits inhibit arborization and synaptogenesis in the CNS.
Collapse
Affiliation(s)
- Douglas S Campbell
- Department of Neurobiology and Anatomy, University of Utah Medical Center, Salt Lake City, UT 84132, USA.
| | | | | | | | | | | | | | | |
Collapse
|