101
|
Flavell SW, Greenberg ME. Signaling mechanisms linking neuronal activity to gene expression and plasticity of the nervous system. Annu Rev Neurosci 2008; 31:563-90. [PMID: 18558867 PMCID: PMC2728073 DOI: 10.1146/annurev.neuro.31.060407.125631] [Citation(s) in RCA: 662] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Sensory experience and the resulting synaptic activity within the brain are critical for the proper development of neural circuits. Experience-driven synaptic activity causes membrane depolarization and calcium influx into select neurons within a neural circuit, which in turn trigger a wide variety of cellular changes that alter the synaptic connectivity within the neural circuit. One way in which calcium influx leads to the remodeling of synapses made by neurons is through the activation of new gene transcription. Recent studies have identified many of the signaling pathways that link neuronal activity to transcription, revealing both the transcription factors that mediate this process and the neuronal activity-regulated genes. These studies indicate that neuronal activity regulates a complex program of gene expression involved in many aspects of neuronal development, including dendritic branching, synapse maturation, and synapse elimination. Genetic mutations in several key regulators of activity-dependent transcription give rise to neurological disorders in humans, suggesting that future studies of this gene expression program will likely provide insight into the mechanisms by which the disruption of proper synapse development can give rise to a variety of neurological disorders.
Collapse
Affiliation(s)
- Steven W. Flavell
- F.M. Kirby Neurobiology Center, Children’s Hospital Boston, and Departments of Neurology and Neurobiology, Harvard Medical School, Boston, Massachusetts 02115; ,
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts 02115
| | - Michael E. Greenberg
- F.M. Kirby Neurobiology Center, Children’s Hospital Boston, and Departments of Neurology and Neurobiology, Harvard Medical School, Boston, Massachusetts 02115; ,
| |
Collapse
|
102
|
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: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
103
|
Antidepressant actions of the exercise-regulated gene VGF. Nat Med 2007; 13:1476-82. [PMID: 18059283 DOI: 10.1038/nm1669] [Citation(s) in RCA: 205] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2007] [Accepted: 09/20/2007] [Indexed: 01/30/2023]
Abstract
Exercise has many health benefits, including antidepressant actions in depressed human subjects, but the mechanisms underlying these effects have not been elucidated. We used a custom microarray to identify a previously undescribed profile of exercise-regulated genes in the mouse hippocampus, a brain region implicated in mood and antidepressant response. Pathway analysis of the regulated genes shows that exercise upregulates a neurotrophic factor signaling cascade that has been implicated in the actions of antidepressants. One of the most highly regulated target genes of exercise and of the growth factor pathway is the gene encoding the VGF nerve growth factor, a peptide precursor previously shown to influence synaptic plasticity and metabolism. We show that administration of a synthetic VGF-derived peptide produces a robust antidepressant response in mice and, conversely, that mutation of VGF in mice produces the opposite effects. The results suggest a new role for VGF and identify VGF signaling as a potential therapeutic target for antidepressant drug development.
Collapse
|
104
|
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
|
105
|
Han Y, Chen X, Shi F, Li S, Huang J, Xie M, Hu L, Hoidal JR, Xu P. CPG15, A New Factor Upregulated after Ischemic Brain Injury, Contributes to Neuronal Network Re-Establishment after Glutamate-Induced Injury. J Neurotrauma 2007; 24:722-31. [PMID: 17439354 DOI: 10.1089/neu.2006.0174] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Candidate plasticity-related gene 15 (cpg15) encodes a protein that regulates dendritic and axonal arbor growth and synaptic maturation. In the present study, we investigated the potential role of CPG15 in regulating the neuronal network re-establishment after ischemic brain injury. In the mouse model with transient global ischemia (TGI), CPG15 transcripts and proteins were determined using RT-PCR and Western blot analyses. Cell proliferation was observed using 5'-bromo-2'-deoxyuridine-5'-monophosphate (BrdU) labeling. Double immunostaining and depletion of soluble CPG15 proteins were performed to examine the cellular distribution of CPG15 and the role of soluble CPG15 in the neurite outgrowth during the neuronal network re-establishment in primarily cultured hippocampal cells after glutamate-induced injury. We demonstrated that CPG15 expression in the hippocampus was upregulated at 1-2 weeks after TGI. In the dentate gyrus, the number of CPG15 and BrdU positive cells increased concurrently after the injury. During the neuronal network re-establishment after the glutamate-induced injury of primarily cultured hippocampal cells, CPG15 was mainly located at the ends and turn-off regions of the growth cones and in the vesicles. Depletion of soluble CPG15 proteins secreted from the hippocampal cells in the culture media significantly reduced the neurite outgrowth and neuron-neuron connection. The results indicate that CPG15 may function as a new factor required in re-establishment of neuronal network after the injury. Our findings will be important in developing a new strategy to enhance endogenous neurogenesis after an ischemic brain injury.
Collapse
Affiliation(s)
- Yu Han
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Rickhag M, Teilum M, Wieloch T. Rapid and long-term induction of effector immediate early genes (BDNF, Neuritin and Arc) in peri-infarct cortex and dentate gyrus after ischemic injury in rat brain. Brain Res 2007; 1151:203-10. [PMID: 17397810 DOI: 10.1016/j.brainres.2007.03.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 03/01/2007] [Accepted: 03/02/2007] [Indexed: 01/30/2023]
Abstract
The genomic response following brain ischemia is very complex and involves activation of both protective and detrimental signaling pathways. Immediate early genes (IEGs) represent the first wave of gene expression following ischemia and are induced in extensive regions of the ischemic brain including cerebral cortex and hippocampus. Brain-derived neurotrophic factor (BDNF), Neuritin and Activity-regulated cytoskeleton-associated protein (Arc) belong to a subgroup of immediate early genes implicated in synaptic plasticity known as effector immediate early genes. Here, we investigated the spatial and temporal activation pattern for these genes during the first 24 h of reperfusion following 2-h occlusion of the middle cerebral artery. Neuritin showed a persistent activation in frontal-cingulate cortex while Arc displayed a biphasic response. Also, in dentate gyrus, activation was observed at 0-6 h of reperfusion for Neuritin and 0-12 h of reperfusion for Arc while BDNF was induced 0-9 h of reperfusion. Our study demonstrates a rapid and long-term activation of effector immediate early genes in distinct brain areas following ischemic injury in rat. Effector gene activation may be part of long-term synaptic responses of ischemic brain tissue.
Collapse
Affiliation(s)
- Mattias Rickhag
- Laboratory for Experimental Brain Research, Wallenberg Neuroscience Center, University of Lund, BMC A13, 22184 Lund, Sweden.
| | | | | |
Collapse
|
107
|
Parrish JZ, Emoto K, Kim MD, Jan YN. Mechanisms that regulate establishment, maintenance, and remodeling of dendritic fields. Annu Rev Neurosci 2007; 30:399-423. [PMID: 17378766 DOI: 10.1146/annurev.neuro.29.051605.112907] [Citation(s) in RCA: 209] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although dendrite arborization patterns are hallmarks of neuronal type and critical determinants of neuronal function, how dendritic arbors take shape is still largely unknown. Transcription factors play important roles in specifying neuronal types and have a profound influence on dendritic arbor size and complexity. The space that a dendritic arbor occupies is determined largely by a combination of growth-promoting signals that regulate arbor size, chemotropic cues that steer dendrites into the appropriate space, and neurite-neurite contacts that ensure proper representation of the dendritic field and appropriate synaptic contacts. Dendritic arbors are largely maintained over the neuron's lifetime, but in some cases, dendritic arbors are refined, in large part as a result of neuronal activity. In this review, we summarize our current understanding of the cellular and molecular mechanisms that regulate dendritic field formation and influence the shaping of dendritic arbors.
Collapse
Affiliation(s)
- Jay Z Parrish
- Howard Hughes Medical Institute, Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California 94143, USA
| | | | | | | |
Collapse
|
108
|
Latefi NS, Colman DR. The CNS synapse revisited: gaps, adhesive welds, and borders. Neurochem Res 2006; 32:303-10. [PMID: 17080313 DOI: 10.1007/s11064-006-9181-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2006] [Accepted: 09/22/2006] [Indexed: 12/16/2022]
Abstract
Although processes leading up to the point of synapse formation are fairly well understood, the precise sequence of events in which the membranes of two separate cells "lock in" to form a mature synaptic junctional complex is poorly understood. A careful study of the molecules operating at the synapse indicates that their roles are more multifarious than once imagined. In this review we posit that the synapse is a functional organelle with poorly defined boundaries and a complex biochemistry. The role of adhesion molecules, including the integration of their signaling and adhesive properties in the context of synaptic activity is discussed.
Collapse
Affiliation(s)
- Nazlie S Latefi
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University Street, BT-105, H3A2B4, Montreal, QC, Canada.
| | | |
Collapse
|
109
|
Maier IC, Schwab ME. Sprouting, regeneration and circuit formation in the injured spinal cord: factors and activity. Philos Trans R Soc Lond B Biol Sci 2006; 361:1611-34. [PMID: 16939978 PMCID: PMC1664674 DOI: 10.1098/rstb.2006.1890] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Central nervous system (CNS) injuries are particularly traumatic, owing to the limited capabilities of the mammalian CNS for repair. Nevertheless, functional recovery is observed in patients and experimental animals, but the degree of recovery is variable. We review the crucial characteristics of mammalian spinal cord function, tract development, injury and the current experimental therapeutic approaches for repair. Regenerative or compensatory growth of neurites and the formation of new, functional circuits require spontaneous and experimental reactivation of developmental mechanisms, suppression of the growth-inhibitory properties of the adult CNS tissue and specific targeted activation of new connections by rehabilitative training.
Collapse
Affiliation(s)
- Irin C Maier
- Brain Research Institute, University and ETH Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| | | |
Collapse
|
110
|
Tetzlaff JE, Huppenbauer CB, Tanzer L, Alexander TD, Jones KJ. Motoneuron injury and repair: New perspectives on gonadal steroids as neurotherapeutics. J Mol Neurosci 2006; 28:53-64. [PMID: 16632875 DOI: 10.1385/jmn:28:1:53] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Revised: 11/30/1999] [Accepted: 06/28/2005] [Indexed: 12/26/2022]
Abstract
In this review, we will summarize recent work from our laboratory on the role of gonadal steroids as neuroprotective agents in motoneuron viability following cell stress. Three motoneuron models will be discussed: developing axotomized hamster facial motoneurons (FMNs); adult axotomized mouse FMNs; and immortalized, cultured mouse spinal motoneurons subjected to heat shock. New work on two relevant motoneuron proteins, the survival of motor neuron protein, and neuritin or candidate plasticity-related gene 15, indicates differential steroid regulation of these two proteins after axotomy. The concept of gonadal steroids as cellular stress correction factors and the implications of this for acute neurological injury situations will be presented as well.
Collapse
Affiliation(s)
- Julie E Tetzlaff
- Department of Cell Biology, Neurobiology, and Anatomy, Loyola University of Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|
111
|
Wibrand K, Messaoudi E, Håvik B, Steenslid V, Løvlie R, Steen VM, Bramham CR. Identification of genes co-upregulated with Arc during BDNF-induced long-term potentiation in adult rat dentate gyrus in vivo. Eur J Neurosci 2006; 23:1501-11. [PMID: 16553613 DOI: 10.1111/j.1460-9568.2006.04687.x] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a critical regulator of transcription-dependent adaptive neuronal responses, such as long-term potentiation (LTP). Brief infusion of BDNF into the dentate gyrus of adult anesthetized rats triggers stable LTP at medial perforant path-granule synapses that is transcription-dependent and requires induction of the immediate early gene Arc. Rather than acting alone, Arc is likely to be part of a larger BDNF-induced transcriptional program. Here, we used cDNA microarray expression profiling to search for genes co-upregulated with Arc 3 h after BDNF-LTP induction. Of nine cDNAs encoding for known genes and up-regulated more than four-fold, we selected five genes, Narp, neuritin, ADP-ribosylation factor-like protein-4 (ARL4L), TGF-beta-induced immediate early gene-1 (TIEG1) and CARP, for further validation. Real-time PCR confirmed robust up-regulation of these genes in an independent set of BDNF-LTP experiments, whereas infusion of the control protein cytochrome C had no effect. In situ hybridization histochemistry further revealed up-regulation of all five genes in somata of post-synaptic granule cells following both BDNF-LTP and high-frequency stimulation-induced LTP. While Arc synthesis is critical for local actin polymerization and stable LTP formation, several of the co-upregulated genes have known functions in excitatory synaptogenesis, axon guidance and glutamate receptor clustering. These results provide novel insight into gene expression responses underlying BDNF-induced synaptic consolidation in the adult brain in vivo.
Collapse
Affiliation(s)
- Karin Wibrand
- Department of Biomedicine and Bergen Mental Health Research Center, Section for Physiology, University of Bergen, Jonas Liens vei 91, N-5009 Bergen, Norway
| | | | | | | | | | | | | |
Collapse
|
112
|
Le Jan S, Le Meur N, Cazes A, Philippe J, Le Cunff M, Léger J, Corvol P, Germain S. Characterization of the expression of the hypoxia-induced genes neuritin, TXNIP and IGFBP3 in cancer. FEBS Lett 2006; 580:3395-400. [PMID: 16723126 DOI: 10.1016/j.febslet.2006.05.011] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Revised: 04/27/2006] [Accepted: 05/03/2006] [Indexed: 12/18/2022]
Abstract
By triggering an adaptive response to hypoxia which is a common feature of tumor microenvironments, endothelial cells contribute to the onset of angiogenic responses involved in tumor growth. Therefore, identifying hypoxic markers represent a challenge for a better understanding of tumor angiogenesis and for the optimization of anti-angiogenic therapeutic strategy. Using representational difference analysis combined with microarray, we here report the identification of 133 hypoxia-induced transcripts in human microendothelial cells (HMEC-1). By Northern blot, we confirm hypoxia-induced expression of insulin-like growth factor binding protein 3 (igfbp3), thioredoxin-interacting protein (txnip), neuritin (nrn1). Finally, by performing in situ hybridization on several types of human tumors, we provide evidence for nrn1 and txnip as hypoxic perinecrotic markers and for igfbp3 as a tumor endothelial marker. We propose these hypoxia-induced genes could represent relevant prognostic tools and targets for therapeutic intervention in cancers.
Collapse
|
113
|
Harwell C, Burbach B, Svoboda K, Nedivi E. Regulation of cpg15 expression during single whisker experience in the barrel cortex of adult mice. ACTA ACUST UNITED AC 2005; 65:85-96. [PMID: 16010668 PMCID: PMC3062911 DOI: 10.1002/neu.20176] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Regulation of gene transcription by neuronal activity is thought to be key to the translation of sensory experience into long-term changes in synaptic structure and function. Here we show that cpg15, a gene encoding an extracellular signaling molecule that promotes dendritic and axonal growth and synaptic maturation, is regulated in the somatosensory cortex by sensory experience capable of inducing cortical plasticity. Using in situ hybridization, we monitored cpg15 expression in 4-week-old mouse barrel cortex after trimming all whiskers except D1. We found that cpg15 expression is depressed in the deprived barrels and enhanced in the barrel column corresponding to the spared D1 whisker. Changes in cpg15 mRNA levels first appear in layer IV, peak 12 h after deprivation, and then decline rapidly. In layers II/III, changes in cpg15 expression appear later, peak at 24 h, and persist for days. Induction of cpg15 expression is significantly diminished in adolescent as well as adult CREB knockout mice. cpg15's spatio-temporal expression pattern and its regulation by CREB are consistent with a role in experience-dependent plasticity of cortical circuits. Our results suggest that local structural and/or synaptic changes may be a mechanism by which the adult cortex can adapt to peripheral manipulations.
Collapse
Affiliation(s)
- Corey Harwell
- The Picower Center for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 50 Ames Street, E18-670, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
114
|
Poletti A, Negri-Cesi P, Martini L. Reflections on the diseases linked to mutations of the androgen receptor. Endocrine 2005; 28:243-62. [PMID: 16388114 DOI: 10.1385/endo:28:3:243] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Accepted: 07/13/2005] [Indexed: 11/11/2022]
Abstract
This review summarizes the most recent information on two pathologies linked to mutations of the androgen receptor, namely, the complete androgen insensitivity syndrome (CAIS) and the spinal and bulbar muscular atrophy (SBMA or Kennedy's disease). Data on the clinical manifestations of the two diseases are presented, together with the most relevant findings on their physiopathology and genetics.
Collapse
Affiliation(s)
- Angelo Poletti
- Department of Endocrinology and Centre of Excellence on Neurodegenerative Diseases, University of Milano, Italy
| | | | | |
Collapse
|
115
|
Raggo C, Ruhl R, McAllister S, Koon H, Dezube BJ, Früh K, Moses AV. Novel cellular genes essential for transformation of endothelial cells by Kaposi's sarcoma-associated herpesvirus. Cancer Res 2005; 65:5084-95. [PMID: 15958552 DOI: 10.1158/0008-5472.can-04-2822] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is involved in the development of lymphoproliferative diseases and Kaposi's sarcoma. The oncogenicity of this virus is reflected in vitro by its ability to transform B cells and endothelial cells. Infection of dermal microvascular endothelial cells (DMVEC) transforms the cells from a cobblestone-like monolayer to foci-forming spindle cells. This transformation is accompanied by dramatic changes in the cellular transcriptome. Known oncogenes, such as c-Kit, are among the KSHV-induced host genes. We previously showed that c-Kit is an essential cellular component of the KSHV-mediated transformation of DMVEC. Here, we test the hypothesis that the transformation process can be used to discover novel oncogenes. When expression of a panel of KSHV-induced cellular transcripts was inhibited with antisense oligomers, we observed inhibition of DMVEC proliferation and foci formation using antisense molecules to RDC1 and Neuritin. We further showed that transformation of KSHV-infected DMVEC was inhibited by small interfering RNA directed at RDC1 or Neuritin. Ectopic expression of Neuritin in NIH 3T3 cells resulted in changes in cell morphology and anchorage-independent growth, whereas RDC1 ectopic expression significantly increased cell proliferation. In addition, both RDC1- and Neuritin-expressing cells formed tumors in nude mice. RDC1 is an orphan G protein-coupled receptor, whereas Neuritin is a growth-promoting protein known to mediate neurite outgrowth. Neither gene has been previously implicated in tumorigenesis. Our data suggest that KSHV-mediated transformation involves exploitation of the hitherto unrealized oncogenic properties of RDC1 and Neuritin.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Cell Transformation, Viral/genetics
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- Endothelial Cells/cytology
- Endothelial Cells/virology
- GPI-Linked Proteins
- Gene Expression Profiling
- Herpesvirus 8, Human/genetics
- Herpesvirus 8, Human/physiology
- Humans
- LIM Domain Proteins
- Metalloproteins/biosynthesis
- Metalloproteins/genetics
- Mice
- Mice, Nude
- NIH 3T3 Cells
- Neuropeptides/biosynthesis
- Neuropeptides/genetics
- Oligonucleotide Array Sequence Analysis
- Oligonucleotides, Antisense/genetics
- Oncogenes/physiology
- Osteopontin
- Proto-Oncogene Proteins
- RNA, Small Interfering/genetics
- Receptors, CXCR
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, G-Protein-Coupled/biosynthesis
- Receptors, G-Protein-Coupled/genetics
- Sarcoma, Kaposi/genetics
- Sarcoma, Kaposi/virology
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/genetics
Collapse
Affiliation(s)
- Camilo Raggo
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | | | | | | | | | | |
Collapse
|
116
|
Jüttner R, Moré MI, Das D, Babich A, Meier J, Henning M, Erdmann B, Mu Ller EC, Otto A, Grantyn R, Rathjen FG. Impaired synapse function during postnatal development in the absence of CALEB, an EGF-like protein processed by neuronal activity. Neuron 2005; 46:233-45. [PMID: 15848802 DOI: 10.1016/j.neuron.2005.02.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Revised: 01/14/2005] [Accepted: 02/27/2005] [Indexed: 10/25/2022]
Abstract
In an attempt to characterize the molecular components by which electric activity influences the development of synapses, we searched for cell surface proteins modulated by calcium influx and glutamate receptor activity. Here, we report that neuronal depolarization facilitates the conversion of CALEB, which results in a truncated transmembrane form with an exposed EGF domain. To characterize the role of CALEB in synapse development, synaptic features were investigated in slices of the colliculus superior from CALEB-deficient mice. In the absence of CALEB, the number of synapses and their morphological characteristics remained unchanged. However, in CALEB-deficient mice, synapses displayed higher paired-pulse ratios, less depression during prolonged repetitive activation, a lower rate of spontaneous postsynaptic currents, and a lower release probability at early but not mature postnatal stages. Our findings indicate that CALEB provides a molecular basis for maintaining normal release probability at early developmental stages.
Collapse
Affiliation(s)
- René Jüttner
- Max-Delbrück-Centrum, Robert-Rössle-Strasse 10, 13092 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Javaherian A, Cline HT. Coordinated motor neuron axon growth and neuromuscular synaptogenesis are promoted by CPG15 in vivo. Neuron 2005; 45:505-12. [PMID: 15721237 DOI: 10.1016/j.neuron.2004.12.051] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Revised: 09/03/2004] [Accepted: 12/07/2004] [Indexed: 12/15/2022]
Abstract
We have used in vivo time-lapse two-photon imaging of single motor neuron axons labeled with GFP combined with labeling of presynaptic vesicle clusters and postsynaptic acetylcholine receptors in Xenopus laevis tadpoles to determine the dynamic rearrangement of individual axon branches and synaptogenesis during motor axon arbor development. Control GFP-labeled axons are highly dynamic during the period when axon arbors are elaborating. Axon branches emerge from sites of synaptic vesicle clusters. These data indicate that motor neuron axon elaboration and synaptogenesis are concurrent and iterative. We tested the role of Candidate Plasticity Gene 15 (CPG15, also known as Neuritin), an activity-regulated gene that is expressed in the developing motor neurons in this process. CPG15 expression enhances the development of motor neuron axon arbors by promoting neuromuscular synaptogenesis and by increasing the addition of new axon branches.
Collapse
Affiliation(s)
- Ashkan Javaherian
- Graduate Program in Genetics, Stony Brook University, Stony Brook, New York 11790, USA
| | | |
Collapse
|
118
|
Putz U, Harwell C, Nedivi E. Soluble CPG15 expressed during early development rescues cortical progenitors from apoptosis. Nat Neurosci 2005; 8:322-31. [PMID: 15711540 PMCID: PMC3075944 DOI: 10.1038/nn1407] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Accepted: 01/21/2005] [Indexed: 12/31/2022]
Abstract
The balance between proliferation and apoptosis is critical for proper development of the nervous system. Yet, little is known about molecules that regulate apoptosis of proliferative neurons. Here we identify a soluble, secreted form of CPG15 expressed in embryonic rat brain regions undergoing rapid proliferation and apoptosis, and show that it protects cultured cortical neurons from apoptosis by preventing activation of caspase 3. Using a lentivirus-delivered small hairpin RNA, we demonstrate that endogenous CPG15 is essential for the survival of undifferentiated cortical progenitors in vitro and in vivo. We further show that CPG15 overexpression in vivo expands the progenitor pool by preventing apoptosis, resulting in an enlarged, indented cortical plate and cellular heterotopias within the ventricular zone, similar to the phenotypes of mutant mice with supernumerary forebrain progenitors. CPG15 expressed during mammalian forebrain morphogenesis may help balance neuronal number by countering apoptosis in specific neuroblasts subpopulations, thus influencing final brain size and shape.
Collapse
Affiliation(s)
- Ulrich Putz
- The Picower Center for Learning and Memory, Departments of Brain and Cognitive Sciences and Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
119
|
Marron TU, Guerini V, Rusmini P, Sau D, Brevini TAL, Martini L, Poletti A. Androgen-induced neurite outgrowth is mediated by neuritin in motor neurones. J Neurochem 2005; 92:10-20. [PMID: 15606892 DOI: 10.1111/j.1471-4159.2004.02836.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In the brain, the spinal cord motor neurones express the highest levels of the androgen receptor (AR). Experimental data have suggested that neurite outgrowth in these neurones may be regulated by testosterone or its derivative 5alpha-dihydrotestosterone (DHT), formed by the 5alpha-reductase type 2 enzyme. In this study we have produced and characterized a model of immortalized motor neuronal cells expressing the mouse AR (mAR) [neuroblastoma-spinal cord (NSC) 34/mAR] and analysed the role of androgens in motor neurones. Androgens either activated or repressed several genes; one has been identified as the mouse neuritin, a protein responsible for neurite elongation. Real-time PCR analysis has shown that the neuritin gene is expressed in the basal condition in immortalized motor neurones and is selectively up-regulated by androgens in NSC34/mAR cells; the DHT effect is counteracted by the anti-androgen Casodex. Moreover, DHT induced neurite outgrowth in NSC34/mAR, while testosterone was less effective and its action was counteracted by the 5alpha-reductase type 2 enzyme inhibitor finasteride. Finally, the androgenic effect on neurite outgrowth was abolished by silencing neuritin with siRNA. Therefore, the trophic effects of androgens in motor neurones may be explained by the androgenic regulation of neuritin, a protein linked to neurone development, elongation and regeneration.
Collapse
Affiliation(s)
- T U Marron
- Institute of Endocrinology, Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
120
|
Taha SA, Stryker MP. Molecular substrates of plasticity in the developing visual cortex. PROGRESS IN BRAIN RESEARCH 2005; 147:103-14. [PMID: 15581700 DOI: 10.1016/s0079-6123(04)47008-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ocular dominance plasticity may be the paradigmatic in vivo model of activity-dependent plasticity. More than four decades of intense research has delineated the network-level rules that govern synaptic change in this model. The recent characterization of a murine model for ocular dominance plasticity has facilitated rapid progress on a new front, extending our understanding of the molecular mechanisms underlying ocular dominance plasticity. In this review, we highlight recent advances in this research effort, focusing in particular on signaling pathways mediating shifts in ocular dominance, and mechanisms underlying the timing of the critical period.
Collapse
Affiliation(s)
- Sharif A Taha
- Department of Neurology, University of California at San Francisco, San Francisco, CA 94143, USA
| | | |
Collapse
|
121
|
Abstract
Currently, behavioral development is thought to result from the interplay among genetic inheritance, congenital characteristics, cultural contexts, and parental practices as they directly impact the individual. Evolutionary ecology points to another contributor, epigenetic inheritance, the transmission to offspring of parental phenotypic responses to environmental challenges-even when the young do not experience the challenges themselves. Genetic inheritance is not altered, gene expression is. Organismic pathways for such transmission exist. Maternal stress during the latter half of a daughter's gestation may affect not only the daughter's but also grand-offspring's physical growth. The author argues that temperamental variation may be influenced in the same way. Implications for theory and research design are presented along with testable predictions.
Collapse
Affiliation(s)
- Lawrence V Harper
- Department of Human and Community Development, University of California, Davis, CA 95616, USA.
| |
Collapse
|
122
|
Parker BS, Argani P, Cook BP, Liangfeng H, Chartrand SD, Zhang M, Saha S, Bardelli A, Jiang Y, St Martin TB, Nacht M, Teicher BA, Klinger KW, Sukumar S, Madden SL. Alterations in vascular gene expression in invasive breast carcinoma. Cancer Res 2004; 64:7857-66. [PMID: 15520192 DOI: 10.1158/0008-5472.can-04-1976] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The molecular signature that defines tumor microvasculature will likely provide clues as to how vascular-dependent tumor proliferation is regulated. Using purified endothelial cells, we generated a database of gene expression changes accompanying vascular proliferation in invasive breast cancer. In contrast to normal mammary vasculature, invasive breast cancer vasculature expresses extracellular matrix and surface proteins characteristic of proliferating and migrating endothelial cells. We define and validate the up-regulated expression of VE-cadherin and osteonectin in breast tumor vasculature. In contrast to other tumor types, invasive breast cancer vasculature induced a high expression level of specific transcription factors, including SNAIL1 and HEYL, that may drive gene expression changes necessary for breast tumor neovascularization. We demonstrate the expression of HEYL in tumor endothelial cells and additionally establish the ability of HEYL to both induce proliferation and attenuate programmed cell death of primary endothelial cells in vitro. We also establish that an additional intracellular protein and previously defined metastasis-associated gene, PRL3, appears to be expressed predominately in the vasculature of invasive breast cancers and is able to enhance the migration of endothelial cells in vitro. Together, our results provide unique insights into vascular regulation in breast tumors and suggest specific roles for genes in driving tumor angiogenesis.
Collapse
Affiliation(s)
- Belinda S Parker
- Breast Cancer Program, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Di Giovanni S, Faden AI, Yakovlev A, Duke-Cohan JS, Finn T, Thouin M, Knoblach S, De Biase A, Bregman BS, Hoffman EP. Neuronal plasticity after spinal cord injury: identification of a gene cluster driving neurite outgrowth. FASEB J 2004; 19:153-4. [PMID: 15522907 DOI: 10.1096/fj.04-2694fje] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Functional recovery after spinal cord injury (SCI) may result in part from axon outgrowth and related plasticity through coordinated changes at the molecular level. We employed microarray analysis to identify a subset of genes the expression patterns of which were temporally coregulated and correlated to functional recovery after SCI. Steady-state mRNA levels of this synchronously regulated gene cluster were depressed in both ventral and dorsal horn neurons within 24 h after injury, followed by strong re-induction during the following 2 wk, which paralleled functional recovery. The identified cluster includes neuritin, attractin, microtubule-associated protein 1a, and myelin oligodendrocyte protein genes. Transcriptional and protein regulation of this novel gene cluster was also evaluated in spinal cord tissue and in single neurons and was shown to play a role in axonal plasticity. Finally, in vitro transfection experiments in primary dorsal root ganglion cells showed that cluster members act synergistically to drive neurite outgrowth.
Collapse
Affiliation(s)
- Simone Di Giovanni
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Hua JY, Smith SJ. Neural activity and the dynamics of central nervous system development. Nat Neurosci 2004; 7:327-32. [PMID: 15048120 DOI: 10.1038/nn1218] [Citation(s) in RCA: 331] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2003] [Accepted: 01/21/2004] [Indexed: 11/09/2022]
Abstract
Recent imaging studies show that the formation of neural connections in the central nervous system is a highly dynamic process. The iterative formation and elimination of synapses and neuronal branches result in the formation of a much larger number of trial connections than is maintained in the mature brain. Neural activity modulates development through biasing this process of formation and elimination, promoting the formation and stabilization of appropriate synaptic connections on the basis of functional activity patterns.
Collapse
Affiliation(s)
- Jackie Yuanyuan Hua
- Department of Molecular and Cellular Physiology, Beckman Center B141, Stanford University, Stanford, California 94305, USA
| | | |
Collapse
|
125
|
Abstract
Although development of the nervous system is inherently a process of dynamic change, until recently it has generally been investigated by inference from static images. However, advances in live optical imaging are now allowing direct observation of growth, synapse formation, and even incipient function in the developing nervous system, at length scales from molecules to cortical regions, and over timescales from milliseconds to months. In this review, we provide technical background and present examples of how these new methods, including confocal and two-photon microscopy, GFP-based markers, and functional indicators, are being applied to provide fresh insight into long-standing questions of neural development.
Collapse
Affiliation(s)
- Cristopher M Niell
- Neurosciences Program and Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA.
| | | |
Collapse
|
126
|
Abstract
The development of dendrites is a crucial step in the formation of cortical circuitry. The morphogen brain-derived neurotrophic factor (BDNF) may mediate the effects of activity on dendritic morphology since its expression and release are thought to be activity-dependent. Using two-photon microscopy, the autocrine and paracrine effects of BDNF on dendritic morphology were assessed. Overexpression of BDNF profoundly altered the form and stability of basal dendritic arbors via an autocrine mechanism. Paracrine BDNF also altered dendritic branching, though in a highly local fashion. BDNF is capable of acting as an intercellular morphogen, and could hypothetically shape dendritic arbors to best fit the developing structure and function of the pre-synaptic circuit.
Collapse
|
127
|
Abstract
Dendrite development is an important and unsolved problem in neuroscience. The nervous system is composed of a vast number of neurons with strikingly different morphology. Neurons are highly polarized cells with distinct subcellular compartments, including one or multiple dendritic processes arising from the cell body, and a single, extended axon. Communications between neurons involve synapses formed between axons of the presynaptic neurons and dendrites of the postsynaptic neurons. Extensive studies over the past decade have identified many molecules underlying axonal outgrowth and pathfinding. In contrast, the control of dendrite development is still much less well understood. However, recent progress has begun to shed light on the molecular mechanisms that orchestrate dendrite growth, arborization, and guidance.
Collapse
Affiliation(s)
- Yuh-Nung Jan
- Howard Hughes Medical Institute, Department of Physiology, University of California at San Francisco, San Francisco, CA 94143, USA.
| | | |
Collapse
|
128
|
Fujino T, Lee WCA, Nedivi E. Regulation of cpg15 by signaling pathways that mediate synaptic plasticity. Mol Cell Neurosci 2003; 24:538-54. [PMID: 14664806 PMCID: PMC3065975 DOI: 10.1016/s1044-7431(03)00230-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Transcriptional activation is a key link between neuronal activity and long-term synaptic plasticity. Little is known about genes responding to this activation whose products directly effect functional and structural changes at the synapse. cpg15 is an activity-regulated gene encoding a membrane-bound ligand that regulates dendritic and axonal arbor growth and synaptic maturation. We report that cpg15 is an immediate-early gene induced by Ca(2+) influx through NMDA receptors and L-type voltage-sensitive calcium channels. Activity-dependent cpg15 expression requires convergent activation of the CaM kinase and MAP kinase pathways. Although activation of PKA is not required for activity-dependent expression, cpg15 is induced by cAMP in active neurons. CREB binds the cpg15 promoter in vivo and partially regulates its activity-dependent expression. cpg15 is an effector gene that is a target for signal transduction pathways that mediate synaptic plasticity and thus may take part in an activity-regulated transcriptional program that directs long-term changes in synaptic connections.
Collapse
Affiliation(s)
- Tadahiro Fujino
- The Picower Center for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
129
|
Brain-derived neurotrophic factor mediates activity-dependent dendritic growth in nonpyramidal neocortical interneurons in developing organotypic cultures. J Neurosci 2003. [PMID: 12843269 DOI: 10.1523/jneurosci.23-13-05662.2003] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) promotes postnatal maturation of GABAergic inhibition in the cerebral and cerebellar cortices, and its expression and release are enhanced by neuronal activity, suggesting that it acts in a feedback manner to maintain a balance between excitation and inhibition during development. BDNF promotes differentiation of cerebellar, hippocampal, and neostriatal inhibitory neurons, but its effects on the dendritic development of neocortical inhibitory interneurons remain unknown. Here, we show that BDNF mediates depolarization-induced dendritic growth and branching in neocortical interneurons. To visualize inhibitory interneurons, we biolistically transfected organotypic cortical slice cultures from neonatal mice with green fluorescent protein (GFP) driven by the glutamic acid decarboxylase (GAD)67 promoter. Nearly all GAD67-GFP-expressing neurons were nonpyramidal, many contained GABA, and some expressed markers of neurochemically defined GABAergic subtypes, indicating that GAD67-GFP-expressing neurons were GABAergic. We traced dendritic trees from confocal images of the same GAD67-GFP-expressing neurons before and after a 5 d growth period, and quantified the change in total dendritic length (TDL) and total dendritic branch points (TDBPs) for each neuron. GAD67-GFP-expressing neurons growing in control medium exhibited a 20% increase in TDL, but in 200 ng/ml BDNF or 10 mm KCl, this increase nearly doubled and was accompanied by a significant increase in TDBPs. Blocking action potentials with TTX did not prevent the BDNF-induced growth, but antibodies against BDNF blocked the growth-promoting effect of KCl. We conclude that BDNF, released by neocortical pyramidal neurons in response to depolarization, enhances dendritic growth and branching in nearby inhibitory interneurons.
Collapse
|
130
|
Xu D, Hopf C, Reddy R, Cho RW, Guo L, Lanahan A, Petralia RS, Wenthold RJ, O'Brien RJ, Worley P. Narp and NP1 form heterocomplexes that function in developmental and activity-dependent synaptic plasticity. Neuron 2003; 39:513-28. [PMID: 12895424 DOI: 10.1016/s0896-6273(03)00463-x] [Citation(s) in RCA: 202] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Narp is a neuronal immediate early gene that plays a role in excitatory synaptogenesis. Here, we report that native Narp in brain is part of a pentraxin complex that includes NP1. These proteins are covalently linked by disulfide bonds into highly organized complexes, and their relative ratio in the complex is dynamically dependent upon the neuron's activity history and developmental stage. Complex formation is dependent on their distinct N-terminal coiled-coil domains, while their closely homologous C-terminal pentraxin domains mediate association with AMPA-type glutamate receptors. Narp is substantially more effective in assays of cell surface cluster formation, coclustering of AMPA receptors, and excitatory synaptogenesis, yet their combined expression results in supraadditive effects. These studies support a model in which Narp can regulate the latent synaptogenic activity of NP1 by forming mixed pentraxin assemblies. This mechanism appears to contribute to both activity-independent and activity-dependent excitatory synaptogenesis.
Collapse
Affiliation(s)
- Desheng Xu
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Jin X, Hu H, Mathers PH, Agmon A. Brain-derived neurotrophic factor mediates activity-dependent dendritic growth in nonpyramidal neocortical interneurons in developing organotypic cultures. J Neurosci 2003; 23:5662-73. [PMID: 12843269 PMCID: PMC6741232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) promotes postnatal maturation of GABAergic inhibition in the cerebral and cerebellar cortices, and its expression and release are enhanced by neuronal activity, suggesting that it acts in a feedback manner to maintain a balance between excitation and inhibition during development. BDNF promotes differentiation of cerebellar, hippocampal, and neostriatal inhibitory neurons, but its effects on the dendritic development of neocortical inhibitory interneurons remain unknown. Here, we show that BDNF mediates depolarization-induced dendritic growth and branching in neocortical interneurons. To visualize inhibitory interneurons, we biolistically transfected organotypic cortical slice cultures from neonatal mice with green fluorescent protein (GFP) driven by the glutamic acid decarboxylase (GAD)67 promoter. Nearly all GAD67-GFP-expressing neurons were nonpyramidal, many contained GABA, and some expressed markers of neurochemically defined GABAergic subtypes, indicating that GAD67-GFP-expressing neurons were GABAergic. We traced dendritic trees from confocal images of the same GAD67-GFP-expressing neurons before and after a 5 d growth period, and quantified the change in total dendritic length (TDL) and total dendritic branch points (TDBPs) for each neuron. GAD67-GFP-expressing neurons growing in control medium exhibited a 20% increase in TDL, but in 200 ng/ml BDNF or 10 mm KCl, this increase nearly doubled and was accompanied by a significant increase in TDBPs. Blocking action potentials with TTX did not prevent the BDNF-induced growth, but antibodies against BDNF blocked the growth-promoting effect of KCl. We conclude that BDNF, released by neocortical pyramidal neurons in response to depolarization, enhances dendritic growth and branching in nearby inhibitory interneurons.
Collapse
Affiliation(s)
- Xiaoming Jin
- Department of Neurobiology and Anatomy, West Virginia University, Morgantown, West Virginia 26506-9128, USA
| | | | | | | |
Collapse
|
132
|
Walkley SU. Neurobiology and cellular pathogenesis of glycolipid storage diseases. Philos Trans R Soc Lond B Biol Sci 2003; 358:893-904. [PMID: 12803923 PMCID: PMC1693176 DOI: 10.1098/rstb.2003.1276] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Disorders of lysosomal metabolism often involve the accumulation of specific types of glycolipid, particularly gangliosides, because of either degradative failure or other currently unknown mechanisms. Although the precise role of gangliosides in cells remains enigmatic, the presence of specific abnormalities secondary to ganglioside accumulation in lysosomal diseases has suggested important biological functions. Chief among these is the growth of new dendrites on particular classes of mature neurons secondary to an increase in GM2 ganglioside. That GM2 has also been shown to be elevated in normal immature neurons coincident with dendritic sprouting provides a compelling argument that this ganglioside plays a role in dendritic initiation. This discovery has led to the search for other regulators of dendritic differentiation that may in some way be linked to the expression and/or function of GM2 ganglioside. Principal candidates that have emerged include tyrosine kinase receptors, small GTPases and calcium/calmodulin-dependent protein kinase II. Understanding the mechanism underlying ectopic dendritogenesis in lysosomal diseases can be expected to generate significant insight into the control of dendritic plasticity in normal brain. The detrimental aspects of ganglioside accumulation in storage diseases as well as the potential link between gangliosides and dendritogenesis also provide a strong rationale for developing pharmacological means to manipulate ganglioside expression in neurons.
Collapse
Affiliation(s)
- Steven U Walkley
- Sidney Weisner Laboratory of Genetic Neurological Disease, Department of Neuroscience, Rose F. Kennedy Center for Research in Mental Retardation and Human Development, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
133
|
Affiliation(s)
- Jeffrey L Goldberg
- Department of Neurobiology, Stanford University School of Medicine, California 94305, USA.
| |
Collapse
|
134
|
Maekawa S, Iino S, Miyata S. Molecular characterization of the detergent-insoluble cholesterol-rich membrane microdomain (raft) of the central nervous system. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1610:261-70. [PMID: 12648779 DOI: 10.1016/s0005-2736(03)00023-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Many fundamental neurological issues such as neuronal polarity, the formation and remodeling of synapses, synaptic transmission, and the pathogenesis of the neuronal cell death are closely related to the membrane dynamics. The elucidation of functional roles of a detergent-insoluble cholesterol-rich domain (raft) could therefore provide good clues to the molecular understanding of these important phenomena, for the participation of the raft in the fundamental cell functions, such as signal transduction and selective transport of lipids and proteins, has been elucidated in nonneural cells. Interestingly, the brain is rich in raft and the brain-derived raft differs in its lipid and protein components from other tissue-derived rafts. Since many excellent reviews are written on the membrane lipid dynamics of this microdomain, signal transduction, and neuronal glycolipids, we review on the characterization of the raft proteins recovered in the detergent-insoluble low-density fraction from rat brain. Special focus is addressed on the biochemical characterization of a neuronal enriched protein, NAP-22, for the lipid organizing activity of this protein has become increasingly clear.
Collapse
Affiliation(s)
- Shohei Maekawa
- Department of Life Science, Graduate School of Science and Technology, Kobe University, Rokkodai 1-1, Kobe 657-8501, Japan.
| | | | | |
Collapse
|
135
|
West AE, Griffith EC, Greenberg ME. Regulation of transcription factors by neuronal activity. Nat Rev Neurosci 2002; 3:921-31. [PMID: 12461549 DOI: 10.1038/nrn987] [Citation(s) in RCA: 464] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Anne E West
- Division of Neuroscience, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
136
|
Datwani A, Iwasato T, Itohara S, Erzurumlu RS. NMDA receptor-dependent pattern transfer from afferents to postsynaptic cells and dendritic differentiation in the barrel cortex. Mol Cell Neurosci 2002; 21:477-92. [PMID: 12498788 PMCID: PMC3564661 DOI: 10.1006/mcne.2002.1195] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
N-Methyl-D-aspartate receptors (NMDARs) are important for synaptic refinement during development. In CxNR1KO mice, cortical excitatory neurons lack NR1, the essential subunit of the NMDAR, and in their primary somatosensory (S1) cortex whisker-specific cellular patterns, "barrels," are absent. Despite this cytoarchitectural defect, thalamocortical axons (TCAs) representing the mystacial vibrissae form topographically organized patterns and undergo critical period plasticity. This region-specific knockout mouse model allows for dissection of the mechanisms underlying patterning of the pre- and postsynaptic neural elements in the S1 cortex. In the absence of functional NMDARs, layer IV cell numbers are unaltered, but these cells fail to segregate into barrels. Furthermore, the dendritic fields of spiny stellate cells do not orient toward TCA terminal patches as in normal mice. Instead, they radiate in all directions covering larger territories, exhibiting profuse branching with increased spine density. Comparison of TCA patches with serotonin transporter (5-HTT) immunohistochemistry or Dil labeling also indicates that in the CxNR1KO cortex TCAs form smaller patches and individual axon terminal branching is not as well developed as in control cortex. Our results suggest that postsynaptic NMDAR activation is critical in communicating periphery-related sensory patterns from TCAs to barrel cells. When postsynaptic NMDAR function is disrupted, layer IV spiny stellate cells remain imperceptive to patterning of their presynaptic inputs and elaborate exuberant dendritic specializations.
Collapse
Affiliation(s)
- Akash Datwani
- Department of Cell Biology and Anatomy, and Neuroscience Center of Excellence, LSUHSC, 1901 Perdido Street, New Orleans, Louisiana 70112
| | - Takuji Iwasato
- PRESTO, Japan Science and Technology Corporation (JST), RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
- Laboratory for Behavioral Genetics, Brain Science Institute (BSI), RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, Brain Science Institute (BSI), RIKEN, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Reha S. Erzurumlu
- Department of Cell Biology and Anatomy, and Neuroscience Center of Excellence, LSUHSC, 1901 Perdido Street, New Orleans, Louisiana 70112
- To whom correspondence should be addressed at Department of Cell Biology and Anatomy, LSUHSC, 1901 Perdido Street, New Orleans, LA 70112. Fax: (504) 458-4392.
| |
Collapse
|
137
|
Horch HW, Katz LC. BDNF release from single cells elicits local dendritic growth in nearby neurons. Nat Neurosci 2002; 5:1177-84. [PMID: 12368805 DOI: 10.1038/nn927] [Citation(s) in RCA: 319] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2002] [Accepted: 09/09/2002] [Indexed: 01/14/2023]
Abstract
In cultured neurons, the exogenous application of neurotrophins (in homogenous concentrations) alters many features of axonal and dendritic arbors. In vivo, however, release of endogenous neurotrophins from neuronal processes creates spatially heterogeneous neurotrophin distributions. To probe the consequences of such endogenous neurotrophin distribution, we produced 'donor neurons' in ferret cortex brain slices that co-expressed brain-derived neurotrophic factor (BDNF) and red fluorescent protein (RFP). Using two-photon microscopy, we analyzed their effects on 'recipient neurons' that expressed green fluorescent protein (GFP) alone. BDNF released from dendrites and cell bodies acted directly on nearby recipient neurons to increase dendritic branching in a distance-dependent manner. Three-dimensional analysis of donor and recipient dendrites indicated that the BDNF source had to be within 4.5 microm to induce dendritic growth in the recipient neuron. Thus, BDNF released from an individual cell alters the structure of nearby dendrites on an exquisitely local scale.
Collapse
Affiliation(s)
- Hadley Wilson Horch
- Howard Hughes Medical Institute, Department of Neurobiology, Duke University Medical Center, Box 3209, Durham, North Carolina 27710, USA.
| | | |
Collapse
|
138
|
Groc L, Petanjek Z, Gustafsson B, Ben-Ari Y, Hanse E, Khazipov R. In vivo blockade of neural activity alters dendritic development of neonatal CA1 pyramidal cells. Eur J Neurosci 2002; 16:1931-8. [PMID: 12453057 DOI: 10.1046/j.1460-9568.2002.02264.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
During development, neural activity has been proposed to promote neuronal growth. During the first postnatal week, the hippocampus is characterized by an oscillating neural network activity and a rapid neuronal growth. In the present study we tested in vivo, by injecting tetanus toxin into the hippocampus of P1 rats, whether this neural activity indeed promotes growth of pyramidal cells. We have previously shown that tetanus toxin injection leads to a strong reduction in the frequency of spontaneous GABA and glutamatergic synaptic currents, and to a complete blockade of the early neural network activity during the first postnatal week. Morphology of neurobiotin-filled CA1 pyramidal cells was analyzed at the end of the first postnatal week (P6-10). In activity-reduced neurons, the total length of basal dendritic tree was three times less than control. The number, but not the length, of basal dendritic branches was affected. The growth impairment was restricted to the basal dendrites. The apical dendrite, the axons, or the soma grew normally during activity deprivation. Thus, the in vivo neural activity in the neonate hippocampus seems to promote neuronal growth by initiating novel branches.
Collapse
Affiliation(s)
- Laurent Groc
- Institute of Physiology and Pharmacology, Department of Physiology, Göteborg University, Göteborg, Sweden.
| | | | | | | | | | | |
Collapse
|
139
|
Díaz E, Ge Y, Yang YH, Loh KC, Serafini TA, Okazaki Y, Hayashizaki Y, Speed TP, Ngai J, Scheiffele P. Molecular analysis of gene expression in the developing pontocerebellar projection system. Neuron 2002; 36:417-34. [PMID: 12408845 DOI: 10.1016/s0896-6273(02)01016-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
As an approach toward understanding the molecular mechanisms of neuronal differentiation, we utilized DNA microarrays to elucidate global patterns of gene expression during pontocerebellar development. Through this analysis, we identified groups of genes specific to neuronal precursor cells, associated with axon outgrowth, and regulated in response to contact with synaptic target cells. In the cerebellum, we identified a phase of granule cell differentiation that is independent of interactions with other cerebellar cell types. Analysis of pontine gene expression revealed that distinct programs of gene expression, correlated with axon outgrowth and synapse formation, can be decoupled and are likely influenced by different cells in the cerebellar target environment. Our approach provides insight into the genetic programs underlying the differentiation of specific cell types in the pontocerebellar projection system.
Collapse
Affiliation(s)
- Elva Díaz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Abstract
The first several months of life are a critical period for neuronal plasticity in the visual cortex during which anatomic and physiological development depends on visual experience. In cats, electrophysiologically assessed neuronal plasticity is minimal until approximately 3 weeks, peaks at 5 weeks, gradually declines to low levels at 20 weeks, and disappears at approximately 1 year of age (Daw, 1994). Rearing in darkness slows the entire time course of this critical period, such that at 5 weeks of age, normal cats are more plastic than dark-reared cats, whereas at 20 weeks, dark-reared cats are more plastic (Mower, 1991; Beaver et al., 2001). Thus, a stringent criterion is that genes that are important for plasticity in visual cortex will show differences in expression between normal rearing and dark rearing that are of opposite direction in young versus older animals. The present study reports the identification by differential display PCR of Munc13-3, a mammalian homolog of the Caenorhabditis elegans "uncoordinated" gene (unc-13), as a candidate gene for critical-period neuronal plasticity, the expression of which is regulated according to this criterion specifically in visual cortex and not in frontal cortex. Other members of the Munc13 family (Munc13-1 and Munc13-2) do not meet this criterion in visual cortex, indicating that Munc13-3 is the only family member that is regulated by age and dark rearing in the same manner as physiological plasticity during the visual cortical critical period.
Collapse
|
141
|
Yang CB, Zheng YT, Li GY, Mower GD. Identification of Munc13-3 as a candidate gene for critical-period neuroplasticity in visual cortex. J Neurosci 2002; 22:8614-8. [PMID: 12351735 PMCID: PMC6757768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
The first several months of life are a critical period for neuronal plasticity in the visual cortex during which anatomic and physiological development depends on visual experience. In cats, electrophysiologically assessed neuronal plasticity is minimal until approximately 3 weeks, peaks at 5 weeks, gradually declines to low levels at 20 weeks, and disappears at approximately 1 year of age (Daw, 1994). Rearing in darkness slows the entire time course of this critical period, such that at 5 weeks of age, normal cats are more plastic than dark-reared cats, whereas at 20 weeks, dark-reared cats are more plastic (Mower, 1991; Beaver et al., 2001). Thus, a stringent criterion is that genes that are important for plasticity in visual cortex will show differences in expression between normal rearing and dark rearing that are of opposite direction in young versus older animals. The present study reports the identification by differential display PCR of Munc13-3, a mammalian homolog of the Caenorhabditis elegans "uncoordinated" gene (unc-13), as a candidate gene for critical-period neuronal plasticity, the expression of which is regulated according to this criterion specifically in visual cortex and not in frontal cortex. Other members of the Munc13 family (Munc13-1 and Munc13-2) do not meet this criterion in visual cortex, indicating that Munc13-3 is the only family member that is regulated by age and dark rearing in the same manner as physiological plasticity during the visual cortical critical period.
Collapse
Affiliation(s)
- Cui Bo Yang
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | |
Collapse
|
142
|
Sweeney NT, Li W, Gao FB. Genetic manipulation of single neurons in vivo reveals specific roles of flamingo in neuronal morphogenesis. Dev Biol 2002; 247:76-88. [PMID: 12074553 DOI: 10.1006/dbio.2002.0702] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To study the roles of intracellular factors in neuronal morphogenesis, we used the mosaic analysis with a repressible cell marker (MARCM) technique to visualize identifiable single multiple dendritic (MD) neurons in living Drosophila larvae. We found that individual neurons in the peripheral nervous system (PNS) developed clear morphological polarity and diverse dendritic branching patterns in larval stages. Each MD neuron in the same dorsal cluster developed a unique dendritic field, suggesting that they have specific physiological functions. Single-neuron analysis revealed that Flamingo did not affect the general dendritic branching patterns in postmitotic neurons. Instead, Flamingo limited the extension of one or more dorsal dendrites without grossly affecting lateral branches. The dendritic overextension phenotype was partially conferred by the precocious initiation of dorsal dendrites in flamingo mutant embryos. In addition, Flamingo is required cell autonomously to promote axonal growth and to prevent premature axonal branching of PNS neurons. Our molecular analysis also indicated that the amino acid sequence near the first EGF motif is important for the proper localization and function of Flamingo. These results demonstrate that Flamingo plays a role in early neuronal differentiation and exerts specific effects on dendrites and axons.
Collapse
Affiliation(s)
- Neal T Sweeney
- Gladstone Institute of Neurological Disease, University of California, San Francisco, 94141-9100, USA
| | | | | |
Collapse
|
143
|
Abstract
OBJECTIVE We have analysed pharmacologically induced perturbation of functional and structural neurogenesis in the prefrontal cortex (PFC) and hippocampus. METHOD Juvenile gerbils received a single dose of methamphetamine (METH, 50 mg/kg, i.p.). In adults the following parameters were quantitatively investigated: prefrontal dopaminergic and GABAergic innervation densities (immunocytochemistry), morphogenesis of pyramidal cells (Golgi), dentate granule cell proliferation (BrdU-labelling), working memory and behavioural inhibition (delayed response, open-field). RESULT A single challenge of METH continuously suppresses granule cell proliferation in adult gerbils and initiates rewiring of neuronal networks in the PFC which run concurrently with the development of severe deficits in PFC-related behaviours. CONCLUSION It appears that a continuous remodelling of neuronal circuits is an inherent property of the brain, the biological significance of which seems to be to ascertain adaptive interaction between brain and environment. Learning more about drug-induced neuronal reorganization might be basic for understanding the genesis of psychotic conditions in the brain. This presentation is based both on own research and on a review of the literature.
Collapse
Affiliation(s)
- R R Dawirs
- University of Bielefeld, Faculty of Biology, Department of Neuroanatomy, Germany
| | | |
Collapse
|
144
|
Extended plasticity of visual cortex in dark-reared animals may result from prolonged expression of cpg15-like genes. J Neurosci 2002. [PMID: 11880509 DOI: 10.1523/jneurosci.22-05-01807.2002] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
cpg15 is an activity-regulated gene that encodes a membrane-bound ligand that coordinately regulates growth of apposing dendritic and axonal arbors and the maturation of their synapses. These properties make it an attractive candidate for participating in plasticity of the mammalian visual system. Here we compare cpg15 expression during normal development of the rat visual system with that seen in response to dark rearing, monocular blockade of retinal action potentials, or monocular deprivation. Our results show that the onset of cpg15 expression in the visual cortex is coincident with eye opening, and it increases until the peak of the critical period at postnatal day 28 (P28). This early expression is independent of both retinal activity and visual experience. After P28, a component of cpg15 expression in the visual cortex, lateral geniculate nucleus (LGN), and superior colliculus (SC) develops a progressively stronger dependence on retinally driven action potentials. Dark rearing does not affect cpg15 mRNA expression in the LGN and SC at any age, but it does significantly affect its expression in the visual cortex from the peak of the critical period and into adulthood. In dark-reared rats, the peak level of cpg15 expression in the visual cortex at P28 is lower than in controls. Rather than showing the normal decline with maturation, these levels are maintained in dark-reared animals. We suggest that the prolonged plasticity in the visual cortex that is seen in dark-reared animals may result from failure to downregulate genes such as cpg15 that could promote structural remodeling and synaptic maturation.
Collapse
|
145
|
Lee WCA, Nedivi E. Extended plasticity of visual cortex in dark-reared animals may result from prolonged expression of cpg15-like genes. J Neurosci 2002; 22:1807-15. [PMID: 11880509 PMCID: PMC3062906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
cpg15 is an activity-regulated gene that encodes a membrane-bound ligand that coordinately regulates growth of apposing dendritic and axonal arbors and the maturation of their synapses. These properties make it an attractive candidate for participating in plasticity of the mammalian visual system. Here we compare cpg15 expression during normal development of the rat visual system with that seen in response to dark rearing, monocular blockade of retinal action potentials, or monocular deprivation. Our results show that the onset of cpg15 expression in the visual cortex is coincident with eye opening, and it increases until the peak of the critical period at postnatal day 28 (P28). This early expression is independent of both retinal activity and visual experience. After P28, a component of cpg15 expression in the visual cortex, lateral geniculate nucleus (LGN), and superior colliculus (SC) develops a progressively stronger dependence on retinally driven action potentials. Dark rearing does not affect cpg15 mRNA expression in the LGN and SC at any age, but it does significantly affect its expression in the visual cortex from the peak of the critical period and into adulthood. In dark-reared rats, the peak level of cpg15 expression in the visual cortex at P28 is lower than in controls. Rather than showing the normal decline with maturation, these levels are maintained in dark-reared animals. We suggest that the prolonged plasticity in the visual cortex that is seen in dark-reared animals may result from failure to downregulate genes such as cpg15 that could promote structural remodeling and synaptic maturation.
Collapse
Affiliation(s)
- Wei-Chung Allen Lee
- Center for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
146
|
Tao X, West AE, Chen WG, Corfas G, Greenberg ME. A calcium-responsive transcription factor, CaRF, that regulates neuronal activity-dependent expression of BDNF. Neuron 2002; 33:383-95. [PMID: 11832226 DOI: 10.1016/s0896-6273(01)00561-x] [Citation(s) in RCA: 266] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transcription of the brain-derived neurotrophic factor (BDNF) gene is regulated in a calcium- and neuron-selective manner; however, the mechanisms that underlie this selectivity are not known. We have characterized a new calcium-response element, CaRE1, that is required for activity-dependent transcription of BDNF exon III and have cloned a transcription factor, CaRF, that activates transcription from BDNF promoter III in a CaRE1-dependent manner. The transcriptional activity of CaRF is regulated in a calcium- and neuron-selective manner, suggesting that CaRF may confer selectivity upon the activity-dependent induction of BDNF exon III expression.
Collapse
Affiliation(s)
- Xu Tao
- Division of Neuroscience, Children's Hospital, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
147
|
Takasu MA, Dalva MB, Zigmond RE, Greenberg ME. Modulation of NMDA receptor-dependent calcium influx and gene expression through EphB receptors. Science 2002; 295:491-5. [PMID: 11799243 DOI: 10.1126/science.1065983] [Citation(s) in RCA: 394] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Protein-protein interactions and calcium entry through the N-methyl-d-aspartate (NMDA)-type glutamate receptor regulate synaptic development and plasticity in the central nervous system. The EphB receptor tyrosine kinases are localized at excitatory synapses where they cluster and associate with NMDA receptors. We identified a mechanism whereby EphBs modulate NMDA receptor function. EphrinB2 activation of EphB in primary cortical neurons potentiates NMDA receptor-dependent influx of calcium. Treatment of cells with ephrinB2 led to NMDA receptor tyrosine phosphorylation through activation of the Src family of tyrosine kinases. These ephrinB2-dependent events result in enhanced NMDA receptor-dependent gene expression. Our findings indicate that ephrinB2 stimulation of EphB modulates the functional consequences of NMDA receptor activation and suggest a mechanism whereby activity-independent and activity-dependent signals converge to regulate the development and remodeling of synaptic connections.
Collapse
Affiliation(s)
- Mari A Takasu
- Division of Neuroscience, Children's Hospital, and the Department of Neurobiology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
148
|
Abstract
A distinct feature of the nervous system is the intricate network of synaptic connections among neurons of diverse phenotypes. Although initial connections are formed largely through molecular mechanisms that depend on intrinsic developmental programs, spontaneous and experience-driven electrical activities in the developing brain exert critical epigenetic influence on synaptic maturation and refinement of neural circuits. Selective findings discussed here illustrate some of our current understanding of the effects of electrical activity on circuit development and highlight areas that await further study.
Collapse
Affiliation(s)
- L I Zhang
- Keck Center of Integrative Neuroscience, University of California, San Francisco, California 94143-0732, USA
| | | |
Collapse
|
149
|
Brenman JE, Gao FB, Jan LY, Jan YN. Sequoia, a tramtrack-related zinc finger protein, functions as a pan-neural regulator for dendrite and axon morphogenesis in Drosophila. Dev Cell 2001; 1:667-77. [PMID: 11709187 DOI: 10.1016/s1534-5807(01)00072-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Morphological complexity of neurons contributes to their functional complexity. How neurons generate different dendritic patterns is not known. We identified the sequoia mutant from a previous screen for dendrite mutants. Here we report that Sequoia is a pan-neural nuclear protein containing two putative zinc fingers homologous to the DNA binding domain of Tramtrack. sequoia mutants affect the cell fate decision of a small subset of neurons but have global effects on axon and dendrite morphologies of most and possibly all neurons. In support of sequoia as a specific regulator of neuronal morphogenesis, microarray experiments indicate that sequoia may regulate downstream genes that are important for executing neurite development rather than altering a variety of molecules that specify cell fates.
Collapse
Affiliation(s)
- J E Brenman
- Howard Hughes Medical Institute, Departments of Physiology and Biochemistry, University of California, San Francisco 94143, USA
| | | | | | | |
Collapse
|
150
|
Keith CH, Wilson MT. Factors controlling axonal and dendritic arbors. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 205:77-147. [PMID: 11336394 DOI: 10.1016/s0074-7696(01)05003-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The sculpting and maintenance of axonal and dendritic arbors is largely under the control of molecules external to the cell. These factors include both substratum-associated and soluble factors that can enhance or inhibit the outgrowth of axons and dendrites. A large number of factors that modulate axonal outgrowth have been identified, and the first stages of the intracellular signaling pathways by which they modify process outgrowth have been characterized. Relatively fewer factors and pathways that affect dendritic outgrowth have been described. The factors that affect axonal arbors form an incompletely overlapping set with those that affect dendritic arbors, allowing selective control of the development and maintenance of these critical aspects of neuronal morphology.
Collapse
Affiliation(s)
- C H Keith
- Department of Cellular Biology. University of Georgia, Athens, 30605, USA
| | | |
Collapse
|