251
|
Synaptic capture-mediated long-lasting long-term potentiation is strongly dependent on mRNA translation. Neuroreport 2010; 20:1572-6. [PMID: 19841599 DOI: 10.1097/wnr.0b013e328332e021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the CA1 region of mice hippocampal slices, a strong tetanic stimulation of an input pathway triggers a long-lasting long-term potentiation (L-LTP), which requires protein synthesis for the development of its late phase. A weak tetanic stimulation of one pathway, which is incapable of triggering protein synthesis on its own, can nonetheless induce L-LTP if it is preceded by a strong stimulation of another pathway (synaptic capture-mediated L-LTP). We found that anisomycin (25 microM), a translational inhibitor, impaired the strong stimulation-induced L-LTP more severely when the drug was applied during the whole experiment than when delivered only around the induction period. Taking advantage of this phenomenon, we showed that the synaptic capture-mediated L-LTP was strongly dependent on mRNA translation.
Collapse
|
252
|
Tsc2-Rheb signaling regulates EphA-mediated axon guidance. Nat Neurosci 2010; 13:163-72. [PMID: 20062052 PMCID: PMC2812631 DOI: 10.1038/nn.2477] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 11/24/2009] [Indexed: 12/11/2022]
Abstract
Tuberous sclerosis complex is a disease caused by mutations in the TSC1 or TSC2 genes, which encode a protein complex that inhibits mTOR kinase signaling by inactivating the Rheb GTPase. Activation of mTOR promotes the formation of benign tumors in various organs while the mechanisms underlying the neurological symptoms of the disease remain largely unknown. Here, we report that in mice Tsc2 haploinsufficiency causes aberrant retinogeniculate projections that suggest defects in EphA receptor-dependent axon guidance. We also show that EphA receptor activation by ephrin-A ligands in neurons leads to inhibition of ERK1/2 kinase activity and decreased inhibition of Tsc2 by ERK1/2. Thus, ephrin stimulation inactivates the mTOR pathway by enhancing Tsc2 activity. Furthermore, Tsc2 deficiency and hyperactive Rheb constitutively activate mTOR and inhibit ephrin-induced growth cone collapse. Our results demonstrate that TSC2-Rheb-mTOR signaling cooperates with the ephrin-Eph receptor system to control axon guidance in the visual system.
Collapse
|
253
|
Ortega JA, Alcántara S. BDNF/MAPK/ERK-induced BMP7 expression in the developing cerebral cortex induces premature radial glia differentiation and impairs neuronal migration. ACTA ACUST UNITED AC 2009; 20:2132-44. [PMID: 20038543 DOI: 10.1093/cercor/bhp275] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During development of the mammalian nervous system, a combination of genetic and environmental factors governs the sequential generation of neurons and glia and the initial establishment of the neural circuitry. Here, we demonstrate that brain-derived neurotrophic factor (BDNF), one of those local acting factors, induces Bone Morphogenetic Protein 7 (BMP7) expression in embryonic neurons by activating Mitogen-Activated Protein Kinase/Extracellular signal-Regulated Kinase signaling and by the negative regulation of p53/p73 function. We also show that intraventricular injection of BMP7 at midgestation induces the early differentiation of radial glia into glial precursors and astrocytes and the expression of mature glial markers such as the antiadhesive protein SC1. As a result of this precocious radial glia maturation, the laminar distribution of late-born pyramidal neurons is altered, most likely by the termination of radial glia ability to support neuronal migration and the early neuronal detachment from the glial rail. We propose a mechanism for BDNF regulation of BMP7 in which local activity-driven BDNF-induced BMP7 expression at the end of neurogenesis instructs competent precursors to generate astrocytes. Such a mechanism might ensure synchronic neuronal and glial maturation at the beginning of cortical activity.
Collapse
Affiliation(s)
- Juan Alberto Ortega
- Unit of Cell Biology, Department of Experimental Pathology and Therapeutics, School of Medicine, University of Barcelona, 08907 L'Hospitalet de Llobregat, Spain
| | | |
Collapse
|
254
|
Ito HT, Schuman EM. Distance-dependent homeostatic synaptic scaling mediated by a-type potassium channels. Front Cell Neurosci 2009; 3:15. [PMID: 20076774 PMCID: PMC2806179 DOI: 10.3389/neuro.03.015.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 11/09/2009] [Indexed: 11/13/2022] Open
Abstract
Many lines of evidence suggest that the efficacy of synapses on CA1 pyramidal neuron dendrites increases as a function of distance from the cell body. The strength of an individual synapse is also dynamically modulated by activity-dependent synaptic plasticity, which raises the question as to how a neuron can reconcile individual synaptic changes with the maintenance of the proximal-to-distal gradient of synaptic strength along the dendrites. As the density of A-type potassium channels exhibits a similar gradient from proximal (low)-to-distal (high) dendrites, the A-current may play a role in coordinating local synaptic changes with the global synaptic strength gradient. Here we describe a form of homeostatic plasticity elicited by conventional activity blockade (with tetrodotoxin) coupled with a block of the A-type potassium channel. Following A-type potassium channel inhibition for 12 h, recordings from CA1 somata revealed a significantly higher miniature excitatory postsynaptic current (mEPSC) frequency, whereas in dendritic recordings, there was no change in mEPSC frequency. Consistent with mEPSC recordings, we observed a significant increase in AMPA receptor density in stratum pyramidale but not stratum radiatum. Based on these data, we propose that the differential distribution of A-type potassium channels along the apical dendrites may create a proximal-to-distal membrane potential gradient. This gradient may regulate AMPA receptor distribution along the same axis. Taken together, our results indicate that A-type potassium channels play an important role in controlling synaptic strength along the dendrites, which may help to maintain the computational capacity of the neuron.
Collapse
Affiliation(s)
- Hiroshi T Ito
- Division of Biology, California Institute of Technology Pasadena, CA, USA
| | | |
Collapse
|
255
|
A reporter of local dendritic translocation shows plaque- related loss of neural system function in APP-transgenic mice. J Neurosci 2009; 29:12636-40. [PMID: 19812338 DOI: 10.1523/jneurosci.1948-09.2009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although neuronal communication is thought to be summated within local dendritic segments, no technique is currently available to monitor activity in vivo at this level of resolution. To overcome this challenge, we developed an optical reporter of neuronal activity using the coding sequence of Venus, flanked by short stretches of the 5'- and 3'-untranslated regions from calcium/calmodulin-dependent kinase IIalpha (CAMKIIalpha). This reporter takes advantage of the fact that CAMKIIalpha mRNA is transported to the dendrite and locally translated in an activity-dependent manner. Using adeno-associated virus, we used this reporter to study neuronal activity in adult mice. Exposure of the mice to an enriched environment led to enhancement of Venus expression in dendritic segments of somatosensory cortex, demonstrating in vivo that dendritic mRNA translocation and local translation occur in response to physiologically relevant stimuli. We then used this system to examine the impact of Alzheimer-related local amyloid-beta deposits on neural system function to test the hypothesis that plaques are toxic. In APPswe/PS1dE9 (APP/PS1) mice, neurons close to plaques, and dendritic segments close to plaques, both showed diminished fluorescent intensity and therefore neuronal activity. In contrast to wild-type mice, fluorescent intensity in neurons near plaques in transgenic mice did not increase after environmental enrichment. These data indicate that neuronal activity in dendritic segments and neurons in the vicinity of a plaque is decreased compared with wild-type mice, supporting the idea that plaques are a focal lesion leading to impaired neural system function.
Collapse
|
256
|
Dreixler JC, Barone FC, Shaikh AR, Du E, Roth S. Mitogen-activated protein kinase p38alpha and retinal ischemic preconditioning. Exp Eye Res 2009; 89:782-90. [PMID: 19631642 PMCID: PMC2782459 DOI: 10.1016/j.exer.2009.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Revised: 07/08/2009] [Accepted: 07/09/2009] [Indexed: 12/11/2022]
Abstract
In previous studies, inhibition of mitogen-activated protein kinase (MAP) p38 significantly improved recovery and attenuated apoptosis after retinal ischemia in rats. Yet, ischemic preconditioning (IPC) attenuated the ischemia-induced increase in p38 expression. We hypothesized that p38 was required for induction of ischemic tolerance by IPC. We examined the mechanisms of involvement of p38 in IPC neuroprotection. IPC or ischemia was induced in rat retina in vivo. Recovery after ischemia performed 24h after IPC was assessed functionally (electroretinography) and histologically at 7d after ischemia in the presence or absence of inhibition of p38. We examined the role of p38alpha in the mimicking of IPC produced by opening mitochondrial KATP channels using diazoxide, or stimulation of p38 activation by anisomycin. The importance of adenosine receptors in p38 activation after IPC was assessed using specific blockers of adenosine A1 and A2a receptors. Interfering RNA (siRNA) or SB203580 was used to block p38alpha. Phosphorylated p38 levels were measured. Phosphorylated p38 protein increased with IPC. Interfering RNA (siRNA) to p38alpha prior to IPC, or inhibiting p38 activation with SB203580, with ischemia following 24h later, significantly attenuated the neuroprotective effect of IPC. Anisomycin administered to increase p38 mimicked IPC, an effect blocked by SB203580. IPC-mimicking with diazoxide, an opener of mitochondrial KATP channels, was diminished with p38alpha siRNA. Adenosine receptor blockade did not decrease the elevated levels of phosphorylated p38 after IPC. Specific inhibition of p38alpha suggests that this MAPK is involved in the protective effects of IPC, and that p38 is downstream of mitochondrial KATP channels, but not adenosine receptors, in this neuroprotection.
Collapse
Affiliation(s)
- John C. Dreixler
- Department of Anesthesia and Critical Care, the University of Chicago, Chicago, Illinois
| | - Frank C. Barone
- Department of Neurology, Downstate Medical Center, State Univ. of New York, Brooklyn, New York
| | - Afzhal R. Shaikh
- Department of Anesthesia and Critical Care, the University of Chicago, Chicago, Illinois
| | - Eugenie Du
- Department of Anesthesia and Critical Care, the University of Chicago, Chicago, Illinois
| | - Steven Roth
- Department of Anesthesia and Critical Care, the University of Chicago, Chicago, Illinois
| |
Collapse
|
257
|
Zhong L, Cherry T, Bies CE, Florence MA, Gerges NZ. Neurogranin enhances synaptic strength through its interaction with calmodulin. EMBO J 2009; 28:3027-39. [PMID: 19713936 PMCID: PMC2736013 DOI: 10.1038/emboj.2009.236] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 07/23/2009] [Indexed: 11/09/2022] Open
Abstract
Learning-correlated plasticity at CA1 hippocampal excitatory synapses is dependent on neuronal activity and NMDA receptor (NMDAR) activation. However, the molecular mechanisms that transduce plasticity stimuli to postsynaptic potentiation are poorly understood. Here, we report that neurogranin (Ng), a neuron-specific and postsynaptic protein, enhances postsynaptic sensitivity and increases synaptic strength in an activity- and NMDAR-dependent manner. In addition, Ng-mediated potentiation of synaptic transmission mimics and occludes long-term potentiation (LTP). Expression of Ng mutants that lack the ability to bind to, or dissociate from, calmodulin (CaM) fails to potentiate synaptic transmission, strongly suggesting that regulated Ng-CaM binding is necessary for Ng-mediated potentiation. Moreover, knocking-down Ng blocked LTP induction. Thus, Ng-CaM interaction can provide a mechanistic link between induction and expression of postsynaptic potentiation.
Collapse
Affiliation(s)
- Ling Zhong
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Tiffani Cherry
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Christine E Bies
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Matthew A Florence
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nashaat Z Gerges
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
258
|
Waterhouse EG, Xu B. New insights into the role of brain-derived neurotrophic factor in synaptic plasticity. Mol Cell Neurosci 2009; 42:81-9. [PMID: 19577647 PMCID: PMC2748315 DOI: 10.1016/j.mcn.2009.06.009] [Citation(s) in RCA: 261] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2009] [Accepted: 06/25/2009] [Indexed: 12/14/2022] Open
Abstract
Substantial evidence indicates that brain-derived neurotrophic factor (BDNF) plays a crucial role in synaptic plasticity. Long-lasting synaptic plasticity is restricted to active synapses and requires new protein synthesis. Recent work has identified local protein synthesis as an important source for new protein during the expression of enduring synaptic plasticity. This review discusses recent progress in understanding the mechanisms that restrict the action of BDNF to active synapses and by which BDNF mediates chemical and structural modifications of individual synapses, placing an emphasis on the role of local protein synthesis in these processes.
Collapse
Affiliation(s)
- Emily G. Waterhouse
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Baoji Xu
- Department of Pharmacology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
259
|
Dendritic synthesis and release of the neuropeptide galanin: Morphological evidence from studies on rat locus coeruleus neurons. J Comp Neurol 2009; 516:199-212. [DOI: 10.1002/cne.22105] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
260
|
Manadas B, Santos AR, Szabadfi K, Gomes JR, Garbis SD, Fountoulakis M, Duarte CB. BDNF-Induced Changes in the Expression of the Translation Machinery in Hippocampal Neurons: Protein Levels and Dendritic mRNA. J Proteome Res 2009; 8:4536-52. [DOI: 10.1021/pr900366x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Bruno Manadas
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| | - Ana Rita Santos
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| | - Krisztina Szabadfi
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| | - João R. Gomes
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| | - Spiros D. Garbis
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| | - Michael Fountoulakis
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| | - Carlos B. Duarte
- Center for Neuroscience and Cell Biology, Department of Zoology, University of Coimbra, Coimbra, Portugal, and Foundation for Biomedical Research of the Academy of Athens, Athens, Greece
| |
Collapse
|
261
|
Li C, Bassell GJ, Sasaki Y. Fragile X Mental Retardation Protein is Involved in Protein Synthesis-Dependent Collapse of Growth Cones Induced by Semaphorin-3A. Front Neural Circuits 2009; 3:11. [PMID: 19826618 PMCID: PMC2759364 DOI: 10.3389/neuro.04.011.2009] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 08/20/2009] [Indexed: 11/13/2022] Open
Abstract
Fragile X syndrome, the most frequent form of familial mental retardation, is caused by mutation of the Fmr1 gene. Fmr1 encodes the fragile X mental retardation protein (FMRP), an mRNA binding protein regulating local, postsynaptic mRNA translation along dendrites necessary for long-term synaptic plasticity. However, recent studies on FMRP localization in axons and growth cones suggest a possible function in the regulation of local protein synthesis needed for axon guidance. Here, we have demonstrated that FMRP is involved in axonal and growth cone responses induced by the axon guidance factor, Semaphorin-3A (Sema3A). In cultured hippocampal neurons from wild type mice, Sema3A-induced growth cone collapse was protein synthesis-dependent. In contrast, Sema3A-induced growth cone collapse was attenuated in Fmr1 knock-out (KO) neurons and insensitive to protein synthesis inhibitors, suggesting that FMRP is involved in protein synthesis-dependent growth cone collapse. Sema3A increased phosphorylation of eukaryotic initiation factor 4E (eIF4E), an indicator of local translation, in distal axons and growth cones of wild type, but not Fmr1 KO neurons. Furthermore, Sema3A rapidly induced a protein synthesis-dependent increase in levels of microtubule associated protein 1B (MAP1B) in distal axons of wild type neurons, but this response was attenuated in Fmr1 KO neurons. These results suggest a possible role of FMRP to regulate local translation and axonal protein localization in response to Sema3A. This study reveals a new link between FMRP and semaphorin signaling in vitro, and raises the possibility that FMRP may have a critical role in semaphorin signaling in axon guidance during brain development.
Collapse
Affiliation(s)
- Chanxia Li
- Department of Cell Biology, Emory University Atlanta, GA, USA
| | | | | |
Collapse
|
262
|
Andreassi C, Riccio A. To localize or not to localize: mRNA fate is in 3'UTR ends. Trends Cell Biol 2009; 19:465-74. [PMID: 19716303 DOI: 10.1016/j.tcb.2009.06.001] [Citation(s) in RCA: 267] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Revised: 06/17/2009] [Accepted: 06/18/2009] [Indexed: 01/30/2023]
Abstract
Translation of localized mRNA is a fast and efficient way of reacting to extracellular stimuli with the added benefit of providing spatial resolution to the cellular response. The efficacy of this adaptive response ultimately relies on the ability to express a particular protein at the right time and in the right place. Although mRNA localization is a mechanism shared by most organisms, it is especially relevant in highly polarized cells, such as differentiated neurons. 3'-Untranslated regions (3'UTRs) of mRNAs are critical both for the targeting of transcripts to specific subcellular compartments and for translational control. Here we review recent studies that indicate how, in response to extracellular cues, nuclear and cytoplasmic remodeling of the 3'UTR contributes to mRNA localization and local protein synthesis.
Collapse
Affiliation(s)
- Catia Andreassi
- MRC Laboratory for Molecular and Cell Biology, University College London, London WC1E 6BT, UK
| | | |
Collapse
|
263
|
Bestman JE, Cline HT. The Relationship between Dendritic Branch Dynamics and CPEB-Labeled RNP Granules Captured in Vivo. Front Neural Circuits 2009; 3:10. [PMID: 19753328 PMCID: PMC2742666 DOI: 10.3389/neuro.04.010.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Accepted: 08/15/2009] [Indexed: 11/13/2022] Open
Abstract
Cytoplasmic Polyadenylation Element Binding protein (CPEB) is an RNA binding protein involved in dendritic delivery of mRNA and activity-dependent, polyadenylation-induced translation of mRNAs in the dendritic arbor. CPEB affects learning and memory and impacts neuronal morphological and synaptic plasticity. In neurons, CPEB is concentrated in ribonucleoprotein (RNP) granules that distribute throughout the dendritic arbor and localize near synapses, suggesting that the trafficking of RNP granules is important for CPEB function. We tagged full-length CPEB and an inactive mutant CPEB with fluorescent proteins, then imaged rapid dendritic branch dynamics and RNP distribution using two-photon time-lapse microscopy of neurons in the optic tectum of living Xenopus laevis tadpoles. Though the inactive CPEB mutant transports mRNA in the dendritic arbor, its expression interferes with CPEB-dependent translation because it is incapable of activity-triggered mRNA polyadenylation. In dendrites, the distributions of the active and inactive CPEB-containing RNP granules do not differ; the RNP granules are dense and their positions do not correlate with sites of rapid dendritic branch dynamics or the eventual fate of the dendritic branches. Because CPEB's sensitivity to activity-dependent signaling does not alter its dendritic distribution, it indicates that active sites in the dendritic arbor are not targeted for RNP granule localization. Nevertheless, inactive CPEB accumulates in granules in terminal dendritic branches, supporting the hypothesis that upon activation CPEB and its mRNA cargo are released from granules and are then available for dendritic translation.
Collapse
Affiliation(s)
- Jennifer E Bestman
- Department of Cell Biology, The Scripps Research Institute La Jolla, CA, USA
| | | |
Collapse
|
264
|
Sebeo J, Hsiao K, Bozdagi O, Dumitriu D, Ge Y, Zhou Q, Benson DL. Requirement for protein synthesis at developing synapses. J Neurosci 2009; 29:9778-93. [PMID: 19657031 PMCID: PMC2771567 DOI: 10.1523/jneurosci.2613-09.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 06/15/2009] [Indexed: 01/08/2023] Open
Abstract
Activity and protein synthesis act cooperatively to generate persistent changes in synaptic responses. This forms the basis for enduring memory in adults. Activity also shapes neural circuits developmentally, but whether protein synthesis plays a congruent function in this process is poorly understood. Here, we show that brief periods of global or local protein synthesis inhibition decrease the synaptic vesicles available for fusion and increase synapse elimination. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is a critical target; its levels are controlled by rapid turnover, and blocking its activity or knocking it down recapitulates the effects of protein synthesis inhibition. Mature presynaptic terminals show decreased sensitivity to protein synthesis inhibition, and resistance coincides with a developmental switch in regulation from CaMKII to PKA (protein kinase A). These findings demonstrate a novel mechanism regulating presynaptic activity and synapse elimination during development, and suggest that protein translation acts coordinately with activity to selectively stabilize appropriate synaptic interactions.
Collapse
Affiliation(s)
| | | | | | | | - Yongchao Ge
- Department of Neurology, Mount Sinai School of Medicine, New York, New York 10029
| | - Qiang Zhou
- Department of Neurology, Mount Sinai School of Medicine, New York, New York 10029
| | | |
Collapse
|
265
|
Secreted amyloid precursor protein-α upregulates synaptic protein synthesis by a protein kinase G-dependent mechanism. Neurosci Lett 2009; 460:92-6. [DOI: 10.1016/j.neulet.2009.05.040] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Revised: 05/13/2009] [Accepted: 05/14/2009] [Indexed: 01/24/2023]
|
266
|
Hengst U, Deglincerti A, Kim HJ, Jeon NL, Jaffrey SR. Axonal elongation triggered by stimulus-induced local translation of a polarity complex protein. Nat Cell Biol 2009; 11:1024-30. [PMID: 19620967 PMCID: PMC2724225 DOI: 10.1038/ncb1916] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 04/23/2009] [Indexed: 12/11/2022]
Abstract
During development, axon growth rates are precisely regulated to provide temporal control over pathfinding. The precise temporal regulation of axonal growth is a key step in the formation of functional synapses and the proper patterning of the nervous system. The rate of axonal elongation is increased by factors such as netrin-1 and nerve growth factor (NGF), which stimulate axon outgrowth using incompletely defined pathways. To clarify the mechanism of netrin-1- and NGF-stimulated axon growth, we explored the role of local protein translation. We found that intra-axonal protein translation is required for stimulated, but not basal, axon outgrowth. To identify the mechanism of translation-dependent outgrowth, we examined the PAR complex, a cytoskeleton regulator. We found that the PAR complex, like local translation, is required for stimulated, but not basal, outgrowth. Par3 mRNA is localized to developing axons, and NGF and netrin-1 trigger its local translation. Selective ablation of Par3 mRNA from axons abolishes the outgrowth-promoting effect of NGF. These results identify a new role for local translation and the PAR complex in axonal outgrowth.
Collapse
Affiliation(s)
- Ulrich Hengst
- Department of Pharmacology, Weill Medical College, Cornell University, NY 10065, USA
| | | | | | | | | |
Collapse
|
267
|
Jourdi H, Hsu YT, Zhou M, Qin Q, Bi X, Baudry M. Positive AMPA receptor modulation rapidly stimulates BDNF release and increases dendritic mRNA translation. J Neurosci 2009; 29:8688-97. [PMID: 19587275 PMCID: PMC2761758 DOI: 10.1523/jneurosci.6078-08.2009] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 05/18/2009] [Accepted: 05/19/2009] [Indexed: 11/21/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) stimulates local dendritic mRNA translation and is involved in formation and consolidation of memory. 2H,3H,6aH-pyrrolidino[2'',1''-3',2']1,3-oxazino[6',5'-5,4]-benzo[e]1,4-dioxan-10-one (CX614), one of the best-studied positive AMPA receptor modulators (also known as ampakines), increases BDNF mRNA and protein and facilitates long-term potentiation (LTP) induction. Several other ampakines also improve performance in various behavioral and learning tasks. Since local dendritic protein synthesis has been implicated in LTP stabilization and in memory consolidation, this study investigated whether CX614 could influence synaptic plasticity by upregulating dendritic protein translation. CX614 treatment of primary neuronal cultures and acute hippocampal slices rapidly activated the translation machinery and increased local dendritic protein synthesis. CX614-induced activation of translation was blocked by K252a [(9S,10R,12R)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid methyl ester], CNQX, APV, and TTX, and was inhibited in the presence of an extracellular BDNF scavenger, TrkB-Fc. The acute effect of CX614 on translation was mediated by increased BDNF release as demonstrated with a BDNF scavenging assay using TrkB-Fc during CX614 treatment of cultured primary neurons and was blocked by nifedipine, ryanodine, and lack of extracellular Ca(2+) in acute hippocampal slices. Finally, CX614, like BDNF, rapidly increased dendritic translation of an exogenous translation reporter. Together, our results demonstrate that positive modulation of AMPA receptors rapidly stimulates dendritic translation, an effect mediated by BDNF secretion and TrkB receptor activation. They also suggest that increased BDNF secretion and stimulation of local protein synthesis contribute to the effects of ampakines on synaptic plasticity.
Collapse
Affiliation(s)
- Hussam Jourdi
- Neurobiology, University of Southern California, Los Angeles, California 90089, and
| | - Yu-Tien Hsu
- Neurobiology, University of Southern California, Los Angeles, California 90089, and
| | - Miou Zhou
- Neurobiology, University of Southern California, Los Angeles, California 90089, and
| | - Qingyu Qin
- Western University of Health Sciences, Pomona, California 91766
| | - Xiaoning Bi
- Western University of Health Sciences, Pomona, California 91766
| | - Michel Baudry
- Neurobiology, University of Southern California, Los Angeles, California 90089, and
| |
Collapse
|
268
|
Slipczuk L, Bekinschtein P, Katche C, Cammarota M, Izquierdo I, Medina JH. BDNF activates mTOR to regulate GluR1 expression required for memory formation. PLoS One 2009; 4:e6007. [PMID: 19547753 PMCID: PMC2695538 DOI: 10.1371/journal.pone.0006007] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 05/27/2009] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The mammalian target of Rapamycin (mTOR) kinase plays a key role in translational control of a subset of mRNAs through regulation of its initiation step. In neurons, mTOR is present at the synaptic region, where it modulates the activity-dependent expression of locally-translated proteins independently of mRNA synthesis. Indeed, mTOR is necessary for different forms of synaptic plasticity and long-term memory (LTM) formation. However, little is known about the time course of mTOR activation and the extracellular signals governing this process or the identity of the proteins whose translation is regulated by this kinase, during mnemonic processing. METHODOLOGY/PRINCIPAL FINDINGS Here we show that consolidation of inhibitory avoidance (IA) LTM entails mTOR activation in the dorsal hippocampus at the moment of and 3 h after training and is associated with a rapid and rapamycin-sensitive increase in AMPA receptor GluR1 subunit expression, which was also blocked by intra-hippocampal delivery of GluR1 antisense oligonucleotides (ASO). In addition, we found that pre- or post-training administration of function-blocking anti-BDNF antibodies into dorsal CA1 hampered IA LTM retention, abolished the learning-induced biphasic activation of mTOR and its readout, p70S6K and blocked GluR1 expression, indicating that BDNF is an upstream factor controlling mTOR signaling during fear-memory consolidation. Interestingly, BDNF ASO hindered LTM retention only when given into dorsal CA1 1 h after but not 2 h before training, suggesting that BDNF controls the biphasic requirement of mTOR during LTM consolidation through different mechanisms: an early one involving BDNF already available at the moment of training, and a late one, happening around 3 h post-training that needs de novo synthesis of this neurotrophin. CONCLUSIONS/SIGNIFICANCE IN CONCLUSION, OUR FINDINGS DEMONSTRATE THAT: 1) mTOR-mediated mRNA translation is required for memory consolidation during at least two restricted time windows; 2) this kinase acts downstream BDNF in the hippocampus and; 3) it controls the increase of synaptic GluR1 necessary for memory consolidation.
Collapse
Affiliation(s)
- Leandro Slipczuk
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Pedro Bekinschtein
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Cynthia Katche
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Martín Cammarota
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Centro de Memoria, Instituto de Pesquisas Biomedicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brasil
| | - Iván Izquierdo
- Centro de Memoria, Instituto de Pesquisas Biomedicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brasil
| | - Jorge H. Medina
- Instituto de Biología Celular y Neurociencias, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Centro de Memoria, Instituto de Pesquisas Biomedicas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brasil
| |
Collapse
|
269
|
Wang DO, Kim SM, Zhao Y, Hwang H, Miura SK, Sossin WS, Martin KC. Synapse- and stimulus-specific local translation during long-term neuronal plasticity. Science 2009; 324:1536-40. [PMID: 19443737 PMCID: PMC2821090 DOI: 10.1126/science.1173205] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Long-term memory and synaptic plasticity require changes in gene expression and yet can occur in a synapse-specific manner. Messenger RNA localization and regulated translation at synapses are thus critical for establishing synapse specificity. Using live-cell microscopy of photoconvertible fluorescent protein translational reporters, we directly visualized local translation at synapses during long-term facilitation of Aplysia sensory-motor synapses. Translation of the reporter required multiple applications of serotonin, was spatially restricted to stimulated synapses, was transcript- and stimulus-specific, and occurred during long-term facilitation but not during long-term depression of sensory-motor synapses. Translational regulation only occurred in the presence of a chemical synapse and required calcium signaling in the postsynaptic motor neuron. Thus, highly regulated local translation occurs at synapses during long-term plasticity and requires trans-synaptic signals.
Collapse
Affiliation(s)
- Dan Ohtan Wang
- Department of Psychiatry and Biobehavioral Sciences, University of California-Los Angeles (UCLA), BSRB 390B, 615 Charles E. Young Drive South, Los Angeles, CA 90095-1737, USA
| | | | | | | | | | | | | |
Collapse
|
270
|
Abstract
Mechanisms of neuronal mRNA localization and translation are of considerable biological interest. Spatially regulated mRNA translation contributes to cell-fate decisions and axon guidance during development, as well as to long-term synaptic plasticity in adulthood. The Fragile-X Mental Retardation protein (FMRP/dFMR1) is one of the best-studied neuronal translational control molecules and here we describe the identification and early characterization of proteins likely to function in the dFMR1 pathway. Induction of the dFMR1 in sevenless-expressing cells of the Drosophila eye causes a disorganized (rough) eye through a mechanism that requires residues necessary for dFMR1/FMRP's translational repressor function. Several mutations in dco, orb2, pAbp, rm62, and smD3 genes dominantly suppress the sev-dfmr1 rough-eye phenotype, suggesting that they are required for dFMR1-mediated processes. The encoded proteins localize to dFMR1-containing neuronal mRNPs in neurites of cultured neurons, and/or have an effect on dendritic branching predicted for bona fide neuronal translational repressors. Genetic mosaic analyses indicate that dco, orb2, rm62, smD3, and dfmr1 are dispensable for translational repression of hid, a microRNA target gene, known to be repressed in wing discs by the bantam miRNA. Thus, the encoded proteins may function as miRNA- and/or mRNA-specific translational regulators in vivo.
Collapse
|
271
|
Falley K, Schütt J, Iglauer P, Menke K, Maas C, Kneussel M, Kindler S, Wouters FS, Richter D, Kreienkamp HJ. Shank1 mRNA: dendritic transport by kinesin and translational control by the 5'untranslated region. Traffic 2009; 10:844-57. [PMID: 19416473 DOI: 10.1111/j.1600-0854.2009.00912.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Dendritic mRNA transport coupled with local regulation of translation enables neurons to selectively alter the protein composition of individual postsynaptic sites. We have analyzed dendritic localization of shank1 mRNAs; shank proteins (shank1-3) are scaffolding molecules of the postsynaptic density (PSD) of excitatory synapses, which are crucial for PSD assembly and the formation of dendritic spines. Live cell imaging demonstrates saltatory movements of shank1 mRNA containing granules along microtubules in both anterograde and retrograde directions. A population of brain messenger ribonucleoprotein particles (mRNPs) containing shank1 mRNAs associates with the cargo-binding domain of the motor protein KIF5C. Through expression of dominant negative proteins, we show that dendritic targeting of shank1 mRNA granules involves KIF5C and the KIF5-associated RNA-binding protein staufen1. While transport of shank1 mRNAs follows principles previously outlined for other dendritic transcripts, shank1 mRNAs are distinguished by their translational regulation. Translation is strongly inhibited by a GC-rich 5(')untranslated region; in addition, internal ribosomal entry sites previously detected in other dendritic transcripts are absent in the shank1 mRNA. A concept emerges from our data in which dendritic transport of different mRNAs occurs collectively via a staufen1- and KIF5-dependent pathway, whereas their local translation is controlled individually by unique cis-acting elements.
Collapse
Affiliation(s)
- Katrin Falley
- Institut für Humangenetik, Universitätsklinikum Hamburg-Eppendorf; Martinistrasse 52; 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
272
|
Trembleau A. [Axonal mRNAs: from histochemical visualization to functional analyses]. ACTA ACUST UNITED AC 2009; 203:65-73. [PMID: 19358812 DOI: 10.1051/jbio/2009012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The vertebrate neuron axon has long been considered as devoid of the protein synthesis machinery. During the early nineties however, the cytochemical visualization of identified mRNAs within certain rodent neuron axons challenged this dogma of cellular neurobiology. The aim of this paper is to illustrate, taking mainly the mouse olfactory system as an example, conceptual and methodological approaches developed in particular in my group, that aim at identifying the function of these axonal mRNAs.
Collapse
Affiliation(s)
- Alain Trembleau
- Equipe Avenir/INSERM Développement et Plasticité des Réseaux Neuronaux, CNRS UMR 7102, Université Pierre et Marie Curie - Paris 6, Boîte 12, 9 quai St Bernard, 75005 Paris, France.
| |
Collapse
|
273
|
Subcellular neuropharmacology: the importance of intracellular targeting. Trends Pharmacol Sci 2009; 30:203-11. [DOI: 10.1016/j.tips.2009.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 01/09/2009] [Accepted: 01/21/2009] [Indexed: 01/03/2023]
|
274
|
Cambray S, Pedraza N, Rafel M, Garí E, Aldea M, Gallego C. Protein kinase KIS localizes to RNA granules and enhances local translation. Mol Cell Biol 2009; 29:726-35. [PMID: 19015237 PMCID: PMC2630681 DOI: 10.1128/mcb.01180-08] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2008] [Revised: 08/22/2008] [Accepted: 11/10/2008] [Indexed: 01/11/2023] Open
Abstract
The regulation of mRNA transport is a fundamental process for cytoplasmic sorting of transcripts and spatially controlled translational derepression once properly localized. There is growing evidence that translation is locally modulated as a result of specific synaptic inputs. However, the underlying molecular mechanisms that regulate this translational process are just emerging. We show that KIS, a serine/threonine kinase functionally related to microtubule dynamics and axon development, interacts with three proteins found in RNA granules: KIF3A, NonO, and eEF1A. KIS localizes to RNA granules and colocalizes with the KIF3A kinesin and the beta-actin mRNA in cultured cortical neurons. In addition, KIS is found associated with KIF3A and 10 RNP-transported mRNAs in brain extracts. The results of knockdown experiments indicate that KIS is required for normal neurite outgrowth. More important, the kinase activity of KIS stimulates 3' untranslated region-dependent local translation in neuritic projections. We propose that KIS is a component of the molecular device that modulates translation in RNA-transporting granules as a result of local signals.
Collapse
Affiliation(s)
- Serafí Cambray
- Departament de Ciències Mèdiques Bàsiques, IRBLLEIDA, Universitat de Lleida, Lleida, Catalonia, Spain
| | | | | | | | | | | |
Collapse
|
275
|
Object-place recognition learning triggers rapid induction of plasticity-related immediate early genes and synaptic proteins in the rat dentate gyrus. Neural Plast 2009; 2008:269097. [PMID: 19190776 PMCID: PMC2631155 DOI: 10.1155/2008/269097] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2008] [Accepted: 10/22/2008] [Indexed: 12/14/2022] Open
Abstract
Long-term recognition memory requires protein synthesis, but little is known about the coordinate regulation of specific genes. Here, we examined expression of the plasticity-associated immediate early genes (Arc, Zif268, and Narp) in the dentate gyrus following long-term object-place recognition learning in rats. RT-PCR analysis from dentate gyrus tissue collected shortly after training did not reveal learning-specific changes in Arc mRNA expression. In situ hybridization and immunohistochemistry were therefore used to assess possible sparse effects on gene expression. Learning about objects increased the density of granule cells expressing Arc, and to a lesser extent Narp, specifically in the dorsal blade of the dentate gyrus, while Zif268 expression was elevated across both blades. Thus, object-place recognition triggers rapid, blade-specific upregulation of plasticity-associated immediate early genes. Furthermore, Western blot analysis of dentate gyrus homogenates demonstrated concomitant upregulation of three postsynaptic density proteins (Arc, PSD-95, and α-CaMKII) with key roles in long-term synaptic plasticity and long-term memory.
Collapse
|
276
|
Vuppalanchi D, Willis DE, Twiss JL. Regulation of mRNA transport and translation in axons. Results Probl Cell Differ 2009; 48:193-224. [PMID: 19582411 DOI: 10.1007/400_2009_16] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Movement of mRNAs into axons occurs by active transport by microtubules through the activity of molecular motor proteins. mRNAs are sequestered into granular-like particles, referred to as transport ribonucleoprotein particles (RNPs) that mediate transport into the axonal compartment. The interaction of mRNA binding proteins with targeted mRNA is a key event in regulating axonal mRNA localization and subsequent localized translation of mRNAs. Several growth-modulating stimuli have been shown to regulate axonal mRNA localization. These do so by activating specific intracellular signaling pathways that converge upon RNA binding proteins and other components of the transport RNP to regulate their activity specifically. Transport can be both positively and negatively regulated by individual stimuli with regard to individual mRNAs. Consequently, there is exquisite specificity for regulating the axon's composition of mRNAs and proteins that control expression in the axon. Finally, recent studies indicate that axotomy can also trigger changes in axonal mRNA composition by specifically shifting the populations of mRNAs that are transported into distal axons.
Collapse
|
277
|
Santos S, Carvalho A, Caldeira M, Duarte C. Regulation of AMPA receptors and synaptic plasticity. Neuroscience 2009; 158:105-25. [DOI: 10.1016/j.neuroscience.2008.02.037] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 01/02/2008] [Accepted: 02/13/2008] [Indexed: 10/22/2022]
|
278
|
Tiruchinapalli DM, Caron MG, Keene JD. Activity-dependent expression of ELAV/Hu RBPs and neuronal mRNAs in seizure and cocaine brain. J Neurochem 2008; 107:1529-43. [DOI: 10.1111/j.1471-4159.2008.05718.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
279
|
Besse F, Ephrussi A. Translational control of localized mRNAs: restricting protein synthesis in space and time. Nat Rev Mol Cell Biol 2008; 9:971-80. [PMID: 19023284 DOI: 10.1038/nrm2548] [Citation(s) in RCA: 271] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
280
|
Abbas AK, Dozmorov M, Li R, Huang FS, Hellberg F, Danielson J, Tian Y, Ekström J, Sandberg M, Wigström H. Persistent LTP without triggered protein synthesis. Neurosci Res 2008; 63:59-65. [PMID: 19013486 DOI: 10.1016/j.neures.2008.10.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 10/10/2008] [Accepted: 10/16/2008] [Indexed: 11/26/2022]
Abstract
Protein synthesis is believed to be involved in stabilizing synaptic plasticity. Effects lasting longer than about 2-3h are considered to require synthesis of new proteins, implying a functional separation between early (E) and late (L) components. However, the issue of constitutive vs. new protein synthesis is still unclear, especially in young animals. Here, we examined the effects of two protein synthesis inhibitors, anisomycin and emetine, on long-term-potentiation (LTP) in CA1 area of hippocampal slices from 12- to 20-day-old rats. Either drug was applied from -30 min to +30 min with respect to LTP induction, a time window previously reported to be critical. However, the LTP remained stable under the entire recording period of 4h (anisomycin), or 8h (emetine). Proper preparation of emetine solution was evidenced by the fact that, in separate experiments, prolonged treatment with emetine gradually blocked baseline responses. Although no corresponding effect was observed with anisomycin, the drug was judged to be potent by its ability to inhibit yeast growth. The ability of anisomycin to inhibit protein synthesis was further confirmed by radiolabeling experiments assessing the degree of leucine incorporation. Our data suggest that LTP up to at least 8h is not dependent on triggered protein synthesis but can be attained by utilizing proteins already available at induction time.
Collapse
Affiliation(s)
- Abdul-Karim Abbas
- Department of Medical Biophysics, Institute of Neuroscience and Physiology, Gothenburg University, Box 433, 40530 Gothenburg, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
281
|
Gao X, Smith GM, Chen J. Impaired dendritic development and synaptic formation of postnatal-born dentate gyrus granular neurons in the absence of brain-derived neurotrophic factor signaling. Exp Neurol 2008; 215:178-90. [PMID: 19014937 DOI: 10.1016/j.expneurol.2008.10.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 08/25/2008] [Accepted: 10/10/2008] [Indexed: 11/25/2022]
Abstract
Neurons are continuously added to the hippocampal dentate gyrus throughout life. These neurons must develop dendritic arbors and spines by which they form synapses for making functional connections with existing neurons. The molecular mechanisms that regulate dendritic development and synaptic formation of postnatal-born granular neurons in the dentate gyrus are largely unknown. Hippocampal dentate gyrus (HDG) has been shown to express high level of brain-derived neurotrophic factor (BDNF). Here we reported that when BDNF is conditionally knockout in the postnatal-born granular neurons of the HDG, the mutant neurons exhibit aberrant morphological development with less dendritic branches, shorter dendritic length, and lower density of dendritic spines, while their primary dendrites are not obviously affected. Even though, these BDNF-deficient granular neurons develop immature dendritic spines to initiate synaptic contacts with afferent axons, they fail to develop or maintain mature spine structures. Thus, these postnatal-born neurons have fewer numbers of synapses, particularly mature synaptic spines. These results suggest that BDNF plays an important role during dendritic development, synaptic formation and synaptic maturation in postnatal-born granular neurons of the HDG in vivo.
Collapse
Affiliation(s)
- Xiang Gao
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | | | | |
Collapse
|
282
|
Napoli I, Mercaldo V, Boyl PP, Eleuteri B, Zalfa F, De Rubeis S, Di Marino D, Mohr E, Massimi M, Falconi M, Witke W, Costa-Mattioli M, Sonenberg N, Achsel T, Bagni C. The fragile X syndrome protein represses activity-dependent translation through CYFIP1, a new 4E-BP. Cell 2008; 134:1042-54. [PMID: 18805096 DOI: 10.1016/j.cell.2008.07.031] [Citation(s) in RCA: 477] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 04/25/2008] [Accepted: 07/15/2008] [Indexed: 11/29/2022]
Abstract
Strong evidence indicates that regulated mRNA translation in neuronal dendrites underlies synaptic plasticity and brain development. The fragile X mental retardation protein (FMRP) is involved in this process; here, we show that it acts by inhibiting translation initiation. A binding partner of FMRP, CYFIP1/Sra1, directly binds the translation initiation factor eIF4E through a domain that is structurally related to those present in 4E-BP translational inhibitors. Brain cytoplasmic RNA 1 (BC1), another FMRP binding partner, increases the affinity of FMRP for the CYFIP1-eIF4E complex in the brain. Levels of proteins encoded by known FMRP target mRNAs are increased upon reduction of CYFIP1 in neurons. Translational repression is regulated in an activity-dependent manner because BDNF or DHPG stimulation of neurons causes CYFIP1 to dissociate from eIF4E at synapses, thereby resulting in protein synthesis. Thus, the translational repression activity of FMRP in the brain is mediated, at least in part, by CYFIP1.
Collapse
Affiliation(s)
- Ilaria Napoli
- Department of Biology, University Tor Vergata, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
283
|
Johnson AW, Chen X, Crombag HS, Zhang C, Smith DR, Shokat KM, Gallagher M, Holland PC, Ginty DD. The brain-derived neurotrophic factor receptor TrkB is critical for the acquisition but not expression of conditioned incentive value. Eur J Neurosci 2008; 28:997-1002. [PMID: 18671735 PMCID: PMC2825165 DOI: 10.1111/j.1460-9568.2008.06383.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Stimuli paired with reward acquire incentive properties that are important for many aspects of motivated behavior, such as feeding and drug-seeking. Here we used a novel chemical-genetic strategy to determine the role of the brain-derived neurotrophic factor (BDNF) receptor TrkB, known to be critical to many aspects of neural development and plasticity, during acquisition and expression of positive incentive value by a cue paired with food. We assessed that cue's learned incentive value in a conditioned reinforcement task, in which its ability to reinforce instrumental responding later, in the absence of food itself, was examined. In TrkB (F616A) knock-in mice, TrkB kinase activity was suppressed by administering the TrkB inhibitor 1NMPP1 during the period of initial cue incentive learning only (i.e. Pavlovian training), during nose-poke conditioned reinforcement testing only, during both phases, or during neither phase. All mice acquired cue-food associations as indexed by approach responses. However, TrkB (F616A) mice that received 1NMPP1 during initial cue incentive learning failed to show conditioned reinforcement of nose-poking, regardless of their treatment in testing, whereas administration of 1NMMP1 only during the testing phase had no effect. The effects of 1NMPP1 administration were due to inhibition of TrkB(F616A), because the performance of wild-type mice was unaffected by administration of the compound during either phase. These data indicate that BDNF or NT4 signaling through TrkB receptors is required for the acquisition of positive incentive value, but is not needed for the expression of previously acquired incentive value in the reinforcement of instrumental behavior.
Collapse
Affiliation(s)
- Alexander W Johnson
- Department of Psychological and Brain Sciences, Neurogenetics and Behavior Center, Johns Hopkins University, Baltimore, MD 21218, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
284
|
Abstract
It is widely accepted that changes in gene expression contribute to enduring modifications of synaptic strength and are required for long-term memory. This is an exciting, wide-open area of research at this moment, one of those areas where it is clear that important work is underway but where the surface has just been scratched in terms of our understanding. Much attention has been given to the mechanisms of gene transcription; however, the regulation of transcription is only one route of manipulating gene expression. Regulation of mRNA translation is another route, and is the ultimate step in the control of gene expression, enabling cells to regulate protein production without altering mRNA synthesis or transport. One of the main advantages of this mechanism over transcriptional control in the nucleus lies in the fact that it endows local sites with independent decision-making authority, a consideration that is of particular relevance in neurons with complex synapto-dendritic architecture. There are a growing number of groups that are taking on the challenge of identifying the mechanisms responsible for regulating the process of mRNA translation during synaptic plasticity and memory formation. In this chapter we will discuss what has been discovered with regard to the localization and regulation of mRNA translation during specific types of neuronal activity in the mammalian central nervous system. The data are most complete for cap-dependent translation; therefore, particular attention will be paid to the machinery that initiates cap-dependent translation and its regulation during synaptic plasticity as well as the behavioral phenotypes consequent to its dysregulation.
Collapse
Affiliation(s)
- Jessica L Banko
- Department of Molecular Medicine, University of South Florida - Health, 12901 Bruce B. Downs Boulevard, MDC 61, Tampa, FL, USA
| | | |
Collapse
|
285
|
Sánchez-Carbente MDR, Desgroseillers L. Understanding the importance of mRNA transport in memory. PROGRESS IN BRAIN RESEARCH 2008; 169:41-58. [PMID: 18394467 DOI: 10.1016/s0079-6123(07)00003-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
RNA localization is an important mechanism to sort proteins to specific subcellular domains. In neurons, several mRNAs are localized in dendrites and their presence allows autonomous control of local translation in response to stimulation of specific synapses. Active constitutive and activity-induced mechanisms of mRNA transport have been described that represent critical steps in the establishment and maintenance of synaptic plasticity. In recent years, the molecular composition of different transporting units has been reported and the identification of proteins and mRNAs in these RNA granules contributes to our understanding of the key steps that regulate mRNA transport and translation. Although RNA granules are heterogeneous, several proteins are common to different RNA granule populations, suggesting that they play important roles in the formation of the granules and/or their regulation during transport and translation. About 1-4% of the neuron transcriptome is found in RNA granules and the characterization of bound mRNAs reveal that they encode proteins of the cytoskeleton, the translation machinery, vesicle trafficking, and/or proteins involved in synaptic plasticity. Non-coding RNAs and microRNAs are also found in dendrites and likely regulate RNA translation. These mechanisms of mRNA transport and local translation are critical for synaptic plasticity mediated by activity or experience and memory.
Collapse
|
286
|
Abstract
MicroRNAs (miRNAs), a class of small, non-protein-coding transcripts about 21 nucleotides long, have recently entered center stage in the study of posttranscriptional gene regulation. They are now thought to be involved in the control of about one third of all protein-coding genes and play a role in the majority of cellular processes that have been studied. We focus on the role of the miRNA pathway in brain development, function, and disease by highlighting recent observations with respect to miRNA-mediated gene regulation in neuronal differentiation, synaptic plasticity, and the circadian clock. We also discuss the implications of these findings with respect to the involvement of miRNAs in the etiopathology of brain disorders and pinpoint the emerging therapeutic potential of miRNAs for the treatment of human diseases.
Collapse
|
287
|
Tapia-Arancibia L, Aliaga E, Silhol M, Arancibia S. New insights into brain BDNF function in normal aging and Alzheimer disease. ACTA ACUST UNITED AC 2008; 59:201-20. [PMID: 18708092 DOI: 10.1016/j.brainresrev.2008.07.007] [Citation(s) in RCA: 429] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/30/2008] [Accepted: 07/30/2008] [Indexed: 01/13/2023]
Abstract
The decline observed during aging involves multiple factors that influence several systems. It is the case for learning and memory processes which are severely reduced with aging. It is admitted that these cognitive effects result from impaired neuronal plasticity, which is altered in normal aging but mainly in Alzheimer disease. Neurotrophins and their receptors, notably BDNF, are expressed in brain areas exhibiting a high degree of plasticity (i.e. the hippocampus, cerebral cortex) and are considered as genuine molecular mediators of functional and morphological synaptic plasticity. Modification of BDNF and/or the expression of its receptors (TrkB.FL, TrkB.T1 and TrkB.T2) have been described during normal aging and Alzheimer disease. Interestingly, recent findings show that some physiologic or pathologic age-associated changes in the central nervous system could be offset by administration of exogenous BDNF and/or by stimulating its receptor expression. These molecules may thus represent a physiological reserve which could determine physiological or pathological aging. These data suggest that boosting the expression or activity of these endogenous protective systems may be a promising therapeutic alternative to enhance healthy aging.
Collapse
|
288
|
Yudin D, Hanz S, Yoo S, Iavnilovitch E, Willis D, Gradus T, Vuppalanchi D, Segal-Ruder Y, Ben-Yaakov K, Hieda M, Yoneda Y, Twiss JL, Fainzilber M. Localized regulation of axonal RanGTPase controls retrograde injury signaling in peripheral nerve. Neuron 2008; 59:241-52. [PMID: 18667152 PMCID: PMC2538677 DOI: 10.1016/j.neuron.2008.05.029] [Citation(s) in RCA: 181] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 05/26/2008] [Accepted: 05/27/2008] [Indexed: 12/30/2022]
Abstract
Peripheral sensory neurons respond to axon injury by activating an importin-dependent retrograde signaling mechanism. How is this mechanism regulated? Here, we show that Ran GTPase and its associated effectors RanBP1 and RanGAP regulate the formation of importin signaling complexes in injured axons. A gradient of nuclear RanGTP versus cytoplasmic RanGDP is thought to be fundamental for the organization of eukaryotic cells. Surprisingly, we find RanGTP in sciatic nerve axoplasm, distant from neuronal cell bodies and nuclei, and in association with dynein and importin-alpha. Following injury, localized translation of RanBP1 stimulates RanGTP dissociation from importins and subsequent hydrolysis, thereby allowing binding of newly synthesized importin-beta to importin-alpha and dynein. Perturbation of RanGTP hydrolysis or RanBP1 blockade at axonal injury sites reduces the neuronal conditioning lesion response. Thus, neurons employ localized mechanisms of Ran regulation to control retrograde injury signaling in peripheral nerve.
Collapse
Affiliation(s)
- Dmitry Yudin
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Shlomit Hanz
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Soonmoon Yoo
- Nemours Biomedical Research Institute, A.I. Dupont Hospital, Wilmington, DE, U.S.A
| | - Elena Iavnilovitch
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Dianna Willis
- Nemours Biomedical Research Institute, A.I. Dupont Hospital, Wilmington, DE, U.S.A
| | - Tal Gradus
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Deepika Vuppalanchi
- Nemours Biomedical Research Institute, A.I. Dupont Hospital, Wilmington, DE, U.S.A
- Department of Biological Sciences, University of Delaware, Newark, DE, U.S.A
| | - Yael Segal-Ruder
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Keren Ben-Yaakov
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Miki Hieda
- Dept. of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshihiro Yoneda
- Dept. of Cell Biology and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Jeffery L. Twiss
- Nemours Biomedical Research Institute, A.I. Dupont Hospital, Wilmington, DE, U.S.A
- Department of Biological Sciences, University of Delaware, Newark, DE, U.S.A
| | - Mike Fainzilber
- Dept. of Biological Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
289
|
An JJ, Gharami K, Liao GY, Woo NH, Lau AG, Vanevski F, Torre ER, Jones KR, Feng Y, Lu B, Xu B. Distinct role of long 3' UTR BDNF mRNA in spine morphology and synaptic plasticity in hippocampal neurons. Cell 2008; 134:175-87. [PMID: 18614020 PMCID: PMC2527207 DOI: 10.1016/j.cell.2008.05.045] [Citation(s) in RCA: 522] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 03/26/2008] [Accepted: 05/16/2008] [Indexed: 01/01/2023]
Abstract
The brain produces two brain-derived neurotrophic factor (BDNF) transcripts, with either short or long 3' untranslated regions (3' UTRs). The physiological significance of the two forms of mRNAs encoding the same protein is unknown. Here, we show that the short and long 3' UTR BDNF mRNAs are involved in different cellular functions. The short 3' UTR mRNAs are restricted to somata, whereas the long 3' UTR mRNAs are also localized in dendrites. In a mouse mutant where the long 3' UTR is truncated, dendritic targeting of BDNF mRNAs is impaired. There is little BDNF in hippocampal dendrites despite normal levels of total BDNF protein. This mutant exhibits deficits in pruning and enlargement of dendritic spines, as well as selective impairment in long-term potentiation in dendrites, but not somata, of hippocampal neurons. These results provide insights into local and dendritic actions of BDNF and reveal a mechanism for differential regulation of subcellular functions of proteins.
Collapse
Affiliation(s)
- Juan Ji An
- Department of Pharmacology, Georgetown University, Washington, DC 20057, USA
| | - Kusumika Gharami
- Department of Pharmacology, Georgetown University, Washington, DC 20057, USA
| | - Guey-Ying Liao
- Department of Pharmacology, Georgetown University, Washington, DC 20057, USA
| | - Newton H. Woo
- Section on Neural Development and Plasticity, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Anthony G. Lau
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| | - Filip Vanevski
- Department of Pharmacology, Georgetown University, Washington, DC 20057, USA
| | - Enrique R. Torre
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
| | - Kevin R. Jones
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Yue Feng
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| | - Bai Lu
- Section on Neural Development and Plasticity, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Baoji Xu
- Department of Pharmacology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
290
|
A drug-controllable tag for visualizing newly synthesized proteins in cells and whole animals. Proc Natl Acad Sci U S A 2008; 105:7744-9. [PMID: 18511556 DOI: 10.1073/pnas.0803060105] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Research on basic cellular processes involving local production or delivery of proteins, such as activity-dependent synaptic modification in neurons, would benefit greatly from a robust, nontoxic method to visualize selectively newly synthesized copies of proteins of interest within cells, tissues, or animals. We report a technique for covalent labeling of newly synthesized proteins of interest based on drug-dependent preservation of epitope tags. Epitope tags are removed from proteins of interest immediately after translation by the activity of a sequence-specific protease until the time a protease inhibitor is added, after which newly synthesized protein copies retain their tags. This method, which we call TimeSTAMP for time-specific tagging for the age measurement of proteins, allows sensitive and nonperturbative visualization and quantification of newly synthesized proteins of interest with exceptionally tight temporal control. We demonstrate applications of TimeSTAMP in retrospectively identifying growing synapses in cultured neurons and in visualizing the distribution of recently synthesized proteins in intact fly brains.
Collapse
|
291
|
|
292
|
Weil D. [GW bodies and stress granules, two cytoplasmic structures for mRNA degradation and storage in mammalian cells]. ACTA ACUST UNITED AC 2008; 201:349-58. [PMID: 18533095 DOI: 10.1051/jbio:2007905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
What does mRNA become at the issue of translation in eukaryotic cells? It can be directly degraded or stored for further use. In some cases, the underlying molecular mechanisms have been studied in detail by biochemical approaches, as examplified by the most recently discovered regulation pathway, RNA interference. However, the cellular context of these regulations has often been ignored, as if these reactions took place diffusely throughout the cytoplasm. Two new structures involved therein have now been described: GW bodies (or P-bodies) and stress granules. The first studies suggested that they were specifically devoted to mRNA degradation and mRNA storage, respectively. This framework is changing rapidly with obvious functional overlapping between both structures.
Collapse
Affiliation(s)
- Dominique Weil
- CNRS FRE2937, Institut André Lwoff, 7 rue Guy Moquet, 94801 Villejuif cedex.
| |
Collapse
|
293
|
Chen WQ, Diao WF, Viidik A, Skalicky M, Höger H, Lubec G. Modulation of the hippocampal protein machinery in voluntary and treadmill exercising rats. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2008; 1784:555-62. [DOI: 10.1016/j.bbapap.2008.01.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Revised: 12/05/2007] [Accepted: 01/03/2008] [Indexed: 11/17/2022]
|
294
|
Hernandez PJ, Abel T. The role of protein synthesis in memory consolidation: progress amid decades of debate. Neurobiol Learn Mem 2008; 89:293-311. [PMID: 18053752 PMCID: PMC2745628 DOI: 10.1016/j.nlm.2007.09.010] [Citation(s) in RCA: 177] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Accepted: 09/30/2007] [Indexed: 12/30/2022]
Abstract
A major component of consolidation theory holds that protein synthesis is required to produce the synaptic modification needed for long-term memory storage. Protein synthesis inhibitors have played a pivotal role in the development of this theory. However, these commonly used drugs have unintended effects that have prompted some to reevaluate the role of protein synthesis in memory consolidation. Here we review the role of protein synthesis in memory formation as proposed by consolidation theory calling special attention to the controversy involving the non-specific effects of a group of protein synthesis inhibitors commonly used to study memory formation in vivo. We argue that molecular and genetic approaches that were subsequently applied to the problem of memory formation confirm the results of less selective pharmacological studies. Thus, to a certain extent, the debate over the role of protein synthesis in memory based on interpretational difficulties inherent to the use of protein synthesis inhibitors may be somewhat moot. We conclude by presenting avenues of research we believe will best provide answers to both long-standing and more recent questions facing field of learning and memory.
Collapse
Affiliation(s)
- Pepe J Hernandez
- Department of Biology, University of Pennsylvania, 433 S. University Avenue, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
295
|
Cox LJ, Hengst U, Gurskaya N, Lukyanov KA, Jaffrey SR. Intra-axonal translation and retrograde trafficking of CREB promotes neuronal survival. Nat Cell Biol 2008; 10:149-59. [PMID: 18193038 PMCID: PMC3153364 DOI: 10.1038/ncb1677] [Citation(s) in RCA: 233] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Accepted: 12/05/2007] [Indexed: 12/17/2022]
Abstract
During development of the nervous system, axons and growth cones contain mRNAs such as beta-actin, cofilin and RhoA, which are locally translated in response to guidance cues. Intra-axonal translation of these mRNAs results in local morphological responses; however, other functions of intra-axonal mRNA translation remain unknown. Here, we show that axons of developing mammalian neurons contain mRNA encoding the cAMP-responsive element (CRE)-binding protein (CREB). CREB is translated within axons in response to nerve growth factor (NGF) and is retrogradely trafficked to the cell body. In neurons that are selectively deficient in axonal CREB transcripts, increases in nuclear pCREB, CRE-mediated transcription and neuronal survival elicited by axonal application of NGF are abolished, indicating a signalling function for axonally synthesized CREB. These studies identify a signalling role for axonally derived CREB, and indicate that signal-dependent synthesis and retrograde trafficking of transcription factors enables specific transcriptional responses to signalling events at distal axons.
Collapse
Affiliation(s)
- Llewellyn J. Cox
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10021, USA
| | - Ulrich Hengst
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10021, USA
| | - Nadya. Gurskaya
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow 117991, Russia
| | - Konstantin A. Lukyanov
- Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, Moscow 117991, Russia
| | - Samie R. Jaffrey
- Department of Pharmacology, Weill Medical College, Cornell University, New York, NY 10021, USA
| |
Collapse
|
296
|
Homeostatic regulation of AMPA receptor expression at single hippocampal synapses. Proc Natl Acad Sci U S A 2008; 105:775-80. [PMID: 18174334 DOI: 10.1073/pnas.0706447105] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Homeostatic synaptic response is an important measure in confining neuronal activity within a narrow physiological range. Whether or not homeostatic plasticity demonstrates synapse specificity, a key feature characteristic of Hebbian-type plasticity, is largely unknown. Here, we report that in cultured hippocampal neurons, alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid subtype glutamate receptor (AMPAR) accumulation is increased selectively in chronically inhibited single synapses, whereas the neighboring normal synapses remain unaffected. This synapse-specific homeostatic regulation depends on the disparity of synaptic activity and is mediated by GluR2-lacking AMPARs and PI3-kinase signaling. These results demonstrate the existence of synaptic specificity and the crucial role of AMPAR-gated calcium in homeostatic plasticity in central neurons.
Collapse
|
297
|
Synapse-specific stabilization of plasticity processes: The synaptic tagging and capture hypothesis revisited 10 years later. Neurosci Biobehav Rev 2008; 32:831-51. [DOI: 10.1016/j.neubiorev.2008.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 12/28/2007] [Accepted: 01/07/2008] [Indexed: 11/22/2022]
|
298
|
Leung KM, Holt CE. Live visualization of protein synthesis in axonal growth cones by microinjection of photoconvertible Kaede into Xenopus embryos. Nat Protoc 2008; 3:1318-27. [PMID: 18714300 PMCID: PMC3687492 DOI: 10.1038/nprot.2008.113] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Photoconvertible fluorescent proteins, such as Kaede, can be switched irreversibly from their native color to a new one. This property can be exploited to visualize de novo mRNA translation, because newly synthesized proteins can be distinguished from preexisting ones by their color. In this protocol, Kaede cDNA linked to the 3' untranslated region (UTR) of beta-actin is delivered into cells fated to become the retina by injection into Xenopus blastomeres. Brief exposure (6-10 s) to UV light (350-410 nm) of Kaede-positive retinal axons/growth cones efficiently converts Kaede from its native green fluorescence to red. The reappearance of the green signal reports the synthesis of new Kaede protein. This approach can be used to investigate the spatiotemporal control of translation of specific mRNAs in response to external stimuli and to test the efficiency of full-length versus mutant UTRs. The 3-d protocol can be adapted for broad use with other photoactivatable fluorescent proteins.
Collapse
Affiliation(s)
- Kin-Mei Leung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | | |
Collapse
|
299
|
Abstract
Dendritic spines are the primary recipients of excitatory input in the central nervous system. They provide biochemical compartments that locally control the signaling mechanisms at individual synapses. Hippocampal spines show structural plasticity as the basis for the physiological changes in synaptic efficacy that underlie learning and memory. Spine structure is regulated by molecular mechanisms that are fine-tuned and adjusted according to developmental age, level and direction of synaptic activity, specific brain region, and exact behavioral or experimental conditions. Reciprocal changes between the structure and function of spines impact both local and global integration of signals within dendrites. Advances in imaging and computing technologies may provide the resources needed to reconstruct entire neural circuits. Key to this endeavor is having sufficient resolution to determine the extrinsic factors (such as perisynaptic astroglia) and the intrinsic factors (such as core subcellular organelles) that are required to build and maintain synapses.
Collapse
Affiliation(s)
- Jennifer N. Bourne
- Center for Learning and Memory, Department of Neurobiology, University of Texas, Austin, TX 78712-0805, ;
| | - Kristen M. Harris
- Center for Learning and Memory, Department of Neurobiology, University of Texas, Austin, TX 78712-0805, ;
| |
Collapse
|
300
|
Dahm R, Zeitelhofer M, Götze B, Kiebler MA, Macchi P. Visualizing mRNA localization and local protein translation in neurons. Methods Cell Biol 2008; 85:293-327. [PMID: 18155468 DOI: 10.1016/s0091-679x(08)85013-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Fluorescent proteins (FPs) have been successfully used to study the localization and interactions of proteins in living cells. They have also been instrumental in analyzing the proteins involved in the localization of RNAs in different cell types, including neurons. With the development of methods that also tag RNAs via fluorescent proteins, researchers now have a powerful tool to covisualize RNAs and associated proteins in living neurons. Here, we review the current status of the use of FPs in the study of transport and localization of ribonucleoprotein particles (RNPs) in neurons and provide key protocols used to introduce transgenes into cultured neurons, including calcium-phosphate-based transfection and nucleofection. These methods allow the fast and efficient expression of fluorescently tagged fusion proteins in neurons at different stages of differentiation and form the basis for fluorescent protein-based live cell imaging in neuronal cultures. Additional protocols are given that allow the simultaneous visualization of RNP proteins and cargo RNAs in living neurons and aspects of the visualization of fluorescently tagged proteins in neurons, such as colocalization studies, are discussed. Finally, we review approaches to visualize the local synthesis of proteins in distal dendrites and axons.
Collapse
Affiliation(s)
- Ralf Dahm
- Center for Brain Research, Division of Neuronal Cell Biology, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | | | | | | | | |
Collapse
|