201
|
Abstract
In addition to its role as a survival factor, nerve growth factor (NGF) has been implicated in initiating apoptosis in restricted cell types both during development and after terminal cell differentiation. NGF binds to the TrkA tyrosine kinase and the p75 neurotrophin receptor, a member of the tumor necrosis factor cytokine family. To understand the mechanisms underlying survival versus death decisions, the TrkA receptor was introduced into oligodendrocyte cell cultures that undergo apoptosis in a p75-dependent manner. Here we report that activation of the TrkA NGF receptor in oligodendrocytes negates cell death by the p75 receptor. TrkA-mediated rescue from apoptosis correlated with mitogen-activated protein kinase activation. Concurrently, activation of TrkA in oligodendrocytes resulted in suppression of c-jun kinase activity initiated by p75, whereas induction of NFkappaB activity by p75 was unaffected. These results indicate that TrkA-mediated rescue involves not only activation of survival signals but also simultaneous suppression of a death signal by p75. The selective interplay between tyrosine kinase and cytokine receptors provides a novel mechanism that achieves alternative cellular responses by merging signals from different ligand-receptor systems.
Collapse
|
202
|
Kahle PJ, Shooter EM, Johnson RM, Verity AN. Phosphatidylcholine-specific phospholipase inhibitor D609 differentially affects MAP kinases and immediate-early genes in PC12 cells. Cell Signal 1998; 10:321-30. [PMID: 9692675 DOI: 10.1016/s0898-6568(98)00010-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The effects of tricyclodecan-9-yl xanthogenate (D609), an inhibitor of phosphatidylcholine-specific phospholipases, on PC12 cells were investigated. D609 repressed nerve growth factor (NGF)-mediated induction of c-fos mRNA with an IC50 approximately 50 microg/ml. Interestingly, maximal c-fos-suppressing doses of D609 did not affect activity of extracellular signal-regulated kinases. Surprisingly, D609 enhanced the extracellular acidification rate of PC12 cells, even in the absence of NGF. D609 alone induced c-jun mRNA with the same potency as it repressed the NGF-induced expression of c-fos. Like NGF, D609 alone induced c-jun even in the presence of dominant-negative Ras. Immediate-early induction of c-jun mRNA by NGF and D609 was abrogated by pretreatment with the kinase inhibitor olomoucine. Jun kinase, which is inhibited by olomoucine, was found to be activated by D609. Thus, D609 might induce c-jun in PC12 cells as a consequence of Jun kinase activation through a Ras-independent pathway. Under the same conditions, D609 repressed NGF-mediated induction of c-fos mRNA.
Collapse
Affiliation(s)
- P J Kahle
- Department of Neurobiology, Stanford University School of Medicine, CA 94305-5401, USA
| | | | | | | |
Collapse
|
203
|
van Kesteren RE, Fainzilber M, Hauser G, van Minnen J, Vreugdenhil E, Smit AB, Ibáñez CF, Geraerts WP, Bulloch AG. Early evolutionary origin of the neurotrophin receptor family. EMBO J 1998; 17:2534-42. [PMID: 9564036 PMCID: PMC1170595 DOI: 10.1093/emboj/17.9.2534] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurotrophins and their Trk receptors play a crucial role in the development and maintenance of the vertebrate nervous system, but to date no component of this signalling system has been found in invertebrates. We describe a molluscan Trk receptor, designated Ltrk, from the snail Lymnaea stagnalis. The full-length sequence of Ltrk reveals most of the characteristics typical of Trk receptors, including highly conserved transmembrane and intracellular tyrosine kinase domains, and a typical extracellular domain of leucine-rich motifs flanked by cysteine clusters. In addition, Ltrk has a unique N-terminal extension and lacks immunoglobulin-like domains. Ltrk is expressed during development in a stage-specific manner, and also in the adult, where its expression is confined to the central nervous system and its associated endocrine tissues. Ltrk has the highest sequence identity with the TrkC mammalian receptor and, when exogenously expressed in fibroblasts or COS cells, binds human NT-3, but not NGF or BDNF, with an affinity of 2.5 nM. These findings support an early evolutionary origin of the Trk family as neuronal receptor tyrosine kinases and suggest that Trk signalling mechanisms may be highly conserved between vertebrates and invertebrates.
Collapse
Affiliation(s)
- R E van Kesteren
- Graduate School of Neurosciences Amsterdam, Research Institute Neurosciences, Vrije Universiteit, De Boelelaan 1087, 1081 HV Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Loeb JA, Susanto ET, Fischbach GD. The neuregulin precursor proARIA is processed to ARIA after expression on the cell surface by a protein kinase C-enhanced mechanism. Mol Cell Neurosci 1998; 11:77-91. [PMID: 9608535 DOI: 10.1006/mcne.1998.0676] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have investigated how the transmembrane precursor proARIA is processed to ARIA (acetylcholine receptor-inducing activity). Pulse-chase labeling in transfected Chinese hamster ovary (CHO) cells showed that proARIA was cleaved to release ARIA into the medium. Cell surface biotin-labeling experiments demonstrated that proARIA was first expressed on the cell surface before being rapidly cleaved to release biotin-labeled ARIA into the medium. While not essential for proteolytic cleavage of proARIA, serum or phorbol-12-myristate-13-acetate (PMA), which activates protein kinase C (PKC), was needed for the efficient release of the processed ARIA. Proteolytic cleavage was blocked by brefeldin A, suggesting that processing occurred distal to Golgi compartments, and by NH4Cl, suggesting a need for intracellular acidic compartments. Serum and PMA also stimulated ARIA release from cultured sensory neurons, suggesting that a similar regulated release mechanism occurs in neurons and may be important in determining where ARIA is released in the developing nervous system.
Collapse
Affiliation(s)
- J A Loeb
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
205
|
Lesser SS, Sherwood NT, Lo DC. Neurotrophins differentially regulate voltage-gated ion channels. Mol Cell Neurosci 1998; 10:173-83. [PMID: 9532579 DOI: 10.1006/mcne.1997.0656] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neurotrophic factors profoundly affect neuronal differentiation, but whether they influence neuronal phenotype in instructive ways remains unclear: do different neurotrophic factors always trigger identical programs of differentiation or can each impose distinct functional properties even when acting upon the same population of target neurons? We addressed this issue by examining the regulatory effects of the four neurotrophins on the molecular components of electrical excitability, voltage-gated ion channels, within a single cellular context. Using patch clamp methods, we studied neurotrophin regulation of voltage-gated sodium, calcium, and potassium currents in SK-N-SH neuroblastoma cells. We found that each neurotrophin induced a unique pattern of expression of ionic currents despite similar activation of initial signal transduction events. Thus, each neurotrophin imposed a different excitable phenotype even when acting upon the same target cells.
Collapse
Affiliation(s)
- S S Lesser
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
206
|
Nakamura T, Muraoka S, Sanokawa R, Mori N. N-Shc and Sck, two neuronally expressed Shc adapter homologs. Their differential regional expression in the brain and roles in neurotrophin and Src signaling. J Biol Chem 1998; 273:6960-7. [PMID: 9507002 DOI: 10.1074/jbc.273.12.6960] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Shc adapter protein is ubiquitously expressed and has been implicated in phosphotyrosine signalings following a variety of extracellular stimulation, e.g. growth factor stimulation, Ca2+ elevation, and G-protein-coupled receptor stimulation. In neuronal cells such as PC12, Shc was demonstrated to be involved in vitro in Ras-dependent mitogen-activated protein kinase activation following nerve growth factor stimulation and Ca2+ entry. However, Shc mRNA was hardly detectable in the brain, and therefore, Shc is unlikely to participate in phosphotyrosine signaling in the central nervous system. Two recently isolated Shc homologs, N-Shc and Sck, have been shown to be expressed in the brain and are expected to function as neuronal adapters instead of Shc. In this study, the neuronal distribution and function of these novel Shc members were investigated. In human and rat central nervous systems, the expression profiles of N-Shc and Sck mRNAs considerably overlapped, although some distinct localization between them was observed: in the adult rat brain, the level of N-Shc mRNA was the highest in the thalamus, whereas that of Sck mRNA was the highest in the hippocampus. In the peripheral nervous system, transcripts of Shc and Sck, but not of N-Shc, were detected. Immunoprecipitation experiments demonstrated functional differences between N-Shc and Sck: (i) N-Shc was a higher affinity adapter molecule than Sck in nerve growth factor and brain-derived neurotrophic factor signaling; and (ii) N-Shc, but not Sck, was efficiently phosphorylated by activated Src tyrosine kinase, whereas Sck, but not N-Shc, formed a complex with pp135, a protein highly phosphorylated by v-Src. These results suggest that neurally expressed N-Shc and Sck may have distinct roles in neuronal signaling in the brain.
Collapse
MESH Headings
- 3T3 Cells
- Adaptor Proteins, Signal Transducing
- Adaptor Proteins, Vesicular Transport
- Amino Acid Sequence
- Animals
- COS Cells
- Cell Line
- Cloning, Molecular
- DNA, Complementary
- Epidermal Growth Factor/metabolism
- Humans
- Mice
- Molecular Sequence Data
- Protein Binding
- Proteins/genetics
- Proteins/metabolism
- Proto-Oncogene Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor, Ciliary Neurotrophic Factor
- Receptor, trkA
- Receptors, Nerve Growth Factor/metabolism
- Sequence Homology, Amino Acid
- Shc Signaling Adaptor Proteins
- Signal Transduction
- Src Homology 2 Domain-Containing, Transforming Protein 1
- Src Homology 2 Domain-Containing, Transforming Protein 2
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- T Nakamura
- Biomedical Research and Development Department, Sumitomo Electric Industries, Sakae-ku, Yokohama 244, Japan
| | | | | | | |
Collapse
|
207
|
Holtmaat AJ, Oestreicher AB, Gispen WH, Verhaagen J. Manipulation of gene expression in the mammalian nervous system: application in the study of neurite outgrowth and neuroregeneration-related proteins. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 1998; 26:43-71. [PMID: 9600624 DOI: 10.1016/s0165-0173(97)00044-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A fundamental issue in neurobiology entails the study of the formation of neuronal connections and their potential to regenerate following injury. In recent years, an expanding number of gene families has been identified involved in different aspects of neurite outgrowth and regeneration. These include neurotrophic factors, cell-adhesion molecules, growth-associated proteins, cytoskeletal proteins and chemorepulsive proteins. Genetic manipulation technology (transgenic mice, knockout mice, viral vectors and antisense oligonucleotides) has been instrumental in defining the function of these neurite outgrowth-related proteins. The aim of this paper is to provide an overview of the above-mentioned four approaches to manipulate gene expression in vivo and to discuss the progress that has been made using this technology in helping to understand the molecular mechanisms that regulate neurite outgrowth. We will show that work with transgenic mice and knockout mice has contributed significantly to the dissection of the function of several proteins with a key role in neurite outgrowth and neuronal survival. Recently developed viral vectors for gene transfer in postmitotic neurons have opened up new avenues to analyze the function of a protein following local expression in naive adult rodents. The initial results with viral vector-based gene transfer provide a conceptual framework for further studies on genetic therapy of neuroregeneration and neurodegenerative diseases.
Collapse
Affiliation(s)
- A J Holtmaat
- Graduate School of Neurosciences Amsterdam, Netherlands Institute for Brain Research
| | | | | | | |
Collapse
|
208
|
Vecino E, Caminos E, Ugarte M, Martín-Zanca D, Osborne NN. Immunohistochemical distribution of neurotrophins and their receptors in the rat retina and the effects of ischemia and reperfusion. GENERAL PHARMACOLOGY 1998; 30:305-14. [PMID: 9510078 DOI: 10.1016/s0306-3623(97)00361-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. Neurotrophins are molecules that regulate the survival, development and maintenance of specific functions in different populations of nerve cells. 2. In the present work, we studied the localization, at the cellular level, of the different neurotrophins and their receptors within the rat retina in control and after ischemia-reperfusion of the retina. We found variations in the localization of some of these molecules depending on the reperfusion time of the retina after the ischemic lesion. 3. Thus it is suggested that the changes in the distribution and concentration of neurotrophins and their receptors caused by ischemia are protective reactions related to neuronal damage and synaptic reorganization.
Collapse
Affiliation(s)
- E Vecino
- Departamento de Biología Celular y Ciencias Morfológicas, Facultad de Medicina, Universidad del País Vasco, Vizcaya, Spain
| | | | | | | | | |
Collapse
|
209
|
Tanabe K, Kiryu-Seo S, Nakamura T, Mori N, Tsujino H, Ochi T, Kiyama H. Alternative expression of Shc family members in nerve-injured motoneurons. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1998; 53:291-6. [PMID: 9473699 DOI: 10.1016/s0169-328x(97)00264-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Expression of Shc family protein (Shc/ShcA, SCK/ShcB and N-Shc/ShcC) and Grb2 mRNAs in the hypoglossal motoneurons after axotomy was examined by in situ hybridization. In normal hypoglossal motor neurons, N-Shc mRNA was expressed predominantly, whereas the Shc mRNA level is very low. Rat hypoglossal nerve injury reversed the expressions of these two molecules in hypoglossal motoneurons. Shc mRNA expression was up-regulated markedly whereas N-Shc was down-regulated after nerve injury. Expression levels of SCK, another Shc family member, and Grb2 were unaffected by nerve injury. These results suggest that, whereas the N-Shc-mediated pathway dominates under normal conditions, an alternative Shc-mediated pathway is utilized in the event of nerve injury. By changing the expression of the Shc family members, the signaling pathway can be altered and various responses induced for nerve regeneration.
Collapse
Affiliation(s)
- K Tanabe
- Department of Anatomy, Asahikawa Medical College, Nishikagura 4-5-3-11, Asahikawa 078, Japan
| | | | | | | | | | | | | |
Collapse
|
210
|
Abstract
Retrograde signaling from the postsynaptic cell to the presynaptic neuron is essential for the development, maintenance, and activity-dependent modification of synaptic connections. This review covers various forms of retrograde interactions at developing and mature synapses. First, we discuss evidence for early retrograde inductive events during synaptogenesis and how maturation of presynaptic structure and function is affected by signals from the postsynaptic cell. Second, we review the evidence that retrograde interactions are involved in activity-dependent synapse competition and elimination in developing nervous systems and in long-term potentiation and depression at mature synapses. Third, we review evidence for various forms of retrograde signaling via membrane-permeant factors, secreted factors, and membrane-bound factors. Finally, we discuss the evidence and physiological implications of the long-range propagation of retrograde signals to the cell body and other parts of the presynaptic neuron.
Collapse
Affiliation(s)
- R M Fitzsimonds
- Department of Biology, University of California at San Diego, La Jolla, USA
| | | |
Collapse
|
211
|
Tessarollo L, Tsoulfas P, Donovan MJ, Palko ME, Blair-Flynn J, Hempstead BL, Parada LF. Targeted deletion of all isoforms of the trkC gene suggests the use of alternate receptors by its ligand neurotrophin-3 in neuronal development and implicates trkC in normal cardiogenesis. Proc Natl Acad Sci U S A 1997; 94:14776-81. [PMID: 9405689 PMCID: PMC25113 DOI: 10.1073/pnas.94.26.14776] [Citation(s) in RCA: 161] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We have generated null mutant mice that lack expression of all isoforms encoded by the trkC locus. These mice display a behavioral phenotype characterized by a loss of proprioceptive neurons. Neuronal counts of sensory ganglia in the trkC mutant mice reveal less severe losses than those in NT-3 null mutant mice, strongly suggesting that NT-3, in vivo, may signal through receptors other than trkC. Mice lacking either NT-3 or all trkC receptor isoforms die in the early postnatal period. Histological examination of trkC-deficient mice reveals severe cardiac defects such as atrial and ventricular septal defects, and valvular defects including pulmonic stenosis. Formation of these structures during development is dependent on cardiac neural crest function. The similarities in cardiac defects observed in the trkC and NT-3 null mutant mice indicate that the trkC receptor mediates most NT-3 effects on the cardiac neural crest.
Collapse
Affiliation(s)
- L Tessarollo
- Neural Development Group, ABL-Basic Research Program, National Cancer Institute-Frederick Cancer Research and Development Center, Frederick, MD 21702, USA.
| | | | | | | | | | | | | |
Collapse
|
212
|
Katagiri Y, Hirata Y, Milbrandt J, Guroff G. Differential regulation of the transcriptional activity of the orphan nuclear receptor NGFI-B by membrane depolarization and nerve growth factor. J Biol Chem 1997; 272:31278-84. [PMID: 9395454 DOI: 10.1074/jbc.272.50.31278] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The immediate-early gene NGFI-B (also called nur77) encodes an orphan nuclear receptor that activates transcription through a unique response element (NBRE). NGFI-B is rapidly induced and modified via phosphorylation by a variety of stimuli that induce cells to differentiate or to proliferate. We have shown that the in vitro phosphorylation of Ser350 located within the "A-box," a motif necessary for DNA binding by NGFI-B, results in a decrease in the binding of NGFI-B to its response element (Hirata, Y., Kiuchi, K., Chen, H.-C., Milbrandt, J., and Guroff, G. (1993) J. Biol. Chem. 268, 24808-24812). We show here that nerve growth factor (NGF)-induced changes in the in vivo phosphorylation of Ser350 accompany transcriptional deactivation of NGFI-B in PC12 cells, that membrane depolarization and NGF treatment cause differential phosphorylation of NGFI-B, and that the transcriptional activation caused by exogenous expression of NGFI-B or membrane depolarization can be inhibited by NGF treatment. In addition, the mutation of Ser350 to Ala abolished the inhibitory effect of NGF on the transcriptional activation of NGFI-B in PC12 cells. These data could provide new insights into the regulation of transcriptional activity required for some neurons to switch from activity-dependent survival to neurotrophin-dependent survival during development.
Collapse
Affiliation(s)
- Y Katagiri
- Section on Growth Factors, NICHD, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
213
|
Althaus HH, Hempel R, Klöppner S, Engel J, Schmidt-Schultz T, Kruska L, Heumann R. Nerve growth factor signal transduction in mature pig oligodendrocytes. J Neurosci Res 1997; 50:729-42. [PMID: 9418961 DOI: 10.1002/(sici)1097-4547(19971201)50:5<729::aid-jnr10>3.0.co;2-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
It has previously been shown that nerve growth factor (NGF) is of functional significance for mature pig oligodendrocytes (OLs) in culture. The present data give evidence for the expression of TrkA, the so-called high-affinity NGF receptor, and of p75NTR, the so-called low-affinity NGF receptor. TrkA is upregulated during culturing, in contrast to the p75 receptor. Exposure of OLs to NGF induces an autophosphorylation of TrkA via its intrinsic tyrosine kinase. K-252a inhibits the TrkA autophosphorylation, which reduces the OL process formation to control levels. To the tyrosine-phosphorylated sites of TrkA several proteins, such as phospholipase C-gamma1, the adaptor protein SHC, the phosphotyrosine phosphatase SH-PTP2 (SYP) associate via their SH2 phosphotase SH-PTP2 domain. The association of SHC to TrkA is shown by co-immunoprecipitation. Indirect evidence for a possible activation of PLC-gamma1 is given by an NGF-induced increase of oligodendroglial [Ca2+]i. Downstream from TrkA, a mitogen-activated protein kinase cascade, which includes Erk1 and Erk2, is operating. An in-gel myelin basic protein kinase assay revealed that NGF activates predominantly Erk1. Finally, it is shown that NGF stimulates expression of c-fos.
Collapse
Affiliation(s)
- H H Althaus
- Max-Planck-Institute for Experimental Medicine, AG Neural Regeneration, Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
214
|
Yamada M, Ohnishi H, Sano SI, Nakatani A, Ikeuchi T, Hatanaka H. Insulin receptor substrate (IRS)-1 and IRS-2 are tyrosine-phosphorylated and associated with phosphatidylinositol 3-kinase in response to brain-derived neurotrophic factor in cultured cerebral cortical neurons. J Biol Chem 1997; 272:30334-9. [PMID: 9374521 DOI: 10.1074/jbc.272.48.30334] [Citation(s) in RCA: 112] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a member of the neurotrophins, promotes differentiation and survival of various types of neurons in the central nervous system. BDNF binds to and activates the tyrosine kinase receptor, TrkB, initiating intracellular signaling and exerting its effects. Phosphatidylinositol 3-kinase (PI3-K), which has been implicated in promotion of neuronal survival by neurotrophic factors, is a component in the signaling pathway of BDNF. We examined how BDNF activates PI3-K in cultured cerebral cortical neurons. We found that insulin receptor substrate (IRS)-1 and -2 are involved in the BDNF signaling pathway that activates PI3-K. IRS-1 and -2 were tyrosine-phosphorylated and bound to PI3-K in response to BDNF. This BDNF-stimulated signaling via IRS-1 and -2 was inhibited by K-252a, an inhibitor of Trk tyrosine kinase. In addition, signaling via IRS-1 and -2 was markedly sustained as well as the BDNF-induced tyrosine phosphorylation of TrkB. On the other hand, we observed no association of PI3-K with TrkB in response to BDNF. These results indicate that the activation of TrkB by BDNF induces the activation of PI3-K via IRS-1 and -2 rather than by a direct interaction of TrkB with PI3-K in cultured cortical neurons.
Collapse
Affiliation(s)
- M Yamada
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565, Japan.
| | | | | | | | | | | |
Collapse
|
215
|
Rius RA, Edsall LC, Spiegel S. Activation of sphingosine kinase in pheochromocytoma PC12 neuronal cells in response to trophic factors. FEBS Lett 1997; 417:173-6. [PMID: 9395290 DOI: 10.1016/s0014-5793(97)01277-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nerve growth factor (NGF), basic fibroblast growth factor (bFGF), dibutyryl cAMP and forskolin, known differentiating agents for pheochromocytoma PC12 cells, induced sustained activation of sphingosine kinase, the enzyme responsible for the formation of the sphingolipid second messenger, sphingosine-1-phosphate, which mediates the mitogenic effects of certain growth factors. In contrast, epidermal growth factor and insulin-like growth factor-1, which stimulate proliferation of PC12 cells, induced only small and transient increases in sphingosine kinase activity. Of the growth factors examined, NGF was the most potent activator of sphingosine kinase, inducing a 4-fold increase in Vmax. Sphingosine kinase activity induced by NGF, but not FGF, was blocked by the protein kinase inhibitor K252a when added simultaneously, with minimal effect when added after 60 min. Thus, activation of sphingosine kinase may have an important role in neural differentiation.
Collapse
Affiliation(s)
- R A Rius
- Department of Biochemistry and Molecular Biology, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | |
Collapse
|
216
|
Abstract
The neurotrophins are important for their long-term effects on the survival and differentiation of many types of neurons during development. They also appear to protect mature neurons from injury caused by nutrient or oxygen deprivation. More recently, the neurotrophins have been implicated in such short-term processes as synaptic plasticity. A great deal of evidence suggests that intracellular calcium levels play a key role in neuronal survival during normal development, in neuronal injury following nutrient or oxygen deprivation, and in synaptic plasticity as well. Maintaining appropriate intracellular levels of calcium is important for proper biological function and it has been shown that one of the actions of the neurotrophins is to modulate intracellular calcium levels in a number of in vivo and in vitro systems. Some information about the mechanism(s) by which this is accomplished is now available. Understanding the mechanisms of neurotrophin action should provide insights into the processes by which the brain functions and, further, provide therapeutic tools for the treatment of neuronal injury and neurodegenerative diseases.
Collapse
Affiliation(s)
- H Jiang
- Section on Growth Factors, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
217
|
Finkbeiner S, Tavazoie SF, Maloratsky A, Jacobs KM, Harris KM, Greenberg ME. CREB: a major mediator of neuronal neurotrophin responses. Neuron 1997; 19:1031-47. [PMID: 9390517 DOI: 10.1016/s0896-6273(00)80395-5] [Citation(s) in RCA: 733] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neurotrophins regulate neuronal survival, differentiation, and synaptic function. To understand how neurotrophins elicit such diverse responses, we elucidated signaling pathways by which brain-derived neurotrophic factor (BDNF) activates gene expression in cultured neurons and hippocampal slices. We found, unexpectedly, that the transcription factor cyclic AMP response element-binding protein (CREB) is an important regulator of BDNF-induced gene expression. Exposure of neurons to BDNF stimulates CREB phosphorylation and activation via at least two signaling pathways: by a calcium/calmodulin-dependent kinase IV (CaMKIV)-regulated pathway that is activated by the release of intracellular calcium and by a Ras-dependent pathway. These findings reveal a previously unrecognized, CaMK-dependent mechanism by which neurotrophins activate CREB and suggest that CREB plays a central role in mediating neurotrophin responses in neurons.
Collapse
Affiliation(s)
- S Finkbeiner
- Department of Neurology, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
218
|
Acute morphogenic and chemotropic effects of neurotrophins on cultured embryonic Xenopus spinal neurons. J Neurosci 1997. [PMID: 9315906 DOI: 10.1523/jneurosci.17-20-07860.1997] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neurotrophins constitute a family of trophic factors with profound effects on the survival and differentiation of the nervous system. Addition of brain-derived neurotrophic factor (BDNF) or neurotrophin-3 (NT-3), but not nerve growth factor (NGF), increased the survival of embryonic Xenopus spinal neurons in culture, although all three neurotrophins enhanced neurite outgrowth. Here we report that neurotrophins also exert acute actions on the morphology and motility of 1-day-old cultured Xenopus spinal neurons. Bath application of BDNF induced extensive formation of lamellipodia simultaneously at multiple sites along the neurite shaft as well as at the growth cone. The BDNF-induced lamellipodia appeared within minutes, rapidly protruded to their greatest extent in about 10 min, and gradually disappeared thereafter, leaving behind newly formed thin lateral processes. When applied as microscopic concentration gradients, both BDNF and NT-3, but not NGF, induced the growth cone to grow toward the neurotrophin source. Our results suggest that neurotrophic factors, when delivered to responsive neurons, may serve as morphogenic and chemotropic agents during neuronal development.
Collapse
|
219
|
Absence of p75NTR causes increased basal forebrain cholinergic neuron size, choline acetyltransferase activity, and target innervation. J Neurosci 1997. [PMID: 9315882 DOI: 10.1523/jneurosci.17-20-07594.1997] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Emerging evidence suggests that the p75 neurotrophin receptor (p75NTR) mediates cell death; however, it is not known whether p75NTR negatively regulates other neuronal phenotypes. We found that mice null for p75NTR displayed highly significant increases in the size of basal forebrain cholinergic neurons, including those that are TrkA-positive. Cholinergic hippocampal target innervation also was increased significantly. Activity of the cholinergic neurotransmitter synthetic enzyme choline acetyltransferase (ChAT) was increased in both the medial septum and hippocampus. Upregulation of these cholinergic features was not associated with increased basal forebrain or hippocampal target NGF levels. In contrast, striatal cholinergic neurons, which do not express p75NTR, showed no difference in neuronal number, size, or ChAT activity between wild-type and p75NTR null mutant mice. These findings indicate that p75NTR negatively regulates cholinergic neuronal phenotype of the basal forebrain cholinergic neurons, including cell size, target innervation, and neurotransmitter synthesis.
Collapse
|
220
|
Involvement of sphingosine 1-phosphate in nerve growth factor-mediated neuronal survival and differentiation. J Neurosci 1997. [PMID: 9278531 DOI: 10.1523/jneurosci.17-18-06952.1997] [Citation(s) in RCA: 212] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sphingolipid metabolites, such as ceramide and sphingosine-1-phosphate (SPP), are emerging as a new class of second messengers involved in cellular proliferation, differentiation, and apoptosis. Nerve growth factor (NGF), a neurotrophic factor for pheochromocytoma PC12 cells, induced a biphasic increase in the activity of sphingosine kinase, the enzyme that catalyzes the formation of SPP. This activation was blocked by K252a, an inhibitor of tyrosine kinase A (trkA). A rapid 1.7-fold increase was followed by a marked prolonged increase reaching a maximum of fourfold to fivefold stimulation with a concomitant increase in SPP levels and a corresponding decrease in endogenous sphingosine levels. Levels of ceramide, the precursor of sphingosine, were only slightly decreased by NGF in serum-containing medium. However, NGF decreased the elevation of ceramide induced by serum withdrawal. Treatment of PC12 cells with SPP did not induce neurite outgrowth or neurofilament expression, yet it enhanced neurofilament expression elicited by suboptimal doses of NGF. Moreover, SPP also protected PC12 cells from apoptosis induced by serum withdrawal. To further substantiate a role for SPP in the cytoprotective actions of NGF, we found that N, N-dimethylsphingosine, a competitive inhibitor of sphingosine kinase, also induced apoptosis and interfered with the survival effect of NGF. These effects were counteracted by exogenous SPP. Moreover, other structurally related compounds, such as dihydrosphingosine 1-phosphate and lysophosphatidic acid, had no significant protective effects. Our results suggest that activation of sphingosine kinase and subsequent formation of SPP may play an important role in the differentiation and survival effects induced by NGF.
Collapse
|
221
|
Transgenic mice expressing the intracellular domain of the p75 neurotrophin receptor undergo neuronal apoptosis. J Neurosci 1997. [PMID: 9278534 DOI: 10.1523/jneurosci.17-18-06988.1997] [Citation(s) in RCA: 137] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have asked whether p75(NTR) may play a role in neuronal apoptosis by producing transgenic mice that express the p75(NTR) intracellular domain within peripheral and central neurons. These animals showed profound reductions in numbers of sympathetic and peripheral sensory neurons as well as cell loss in the neocortex, where there is normally little or no p75(NTR) expression. Developmental loss of facial motor neurons was not observed, but induced expression of the p75(NTR) intracellular domain within adult animals led to increased motor neuron death after axotomy. Biochemical analyses suggest that these effects were not attributable to a p75(NTR)-dependent reduction in trk activation but instead indicate that the p75(NTR) intracellular domain may act as a constitutive activator of signaling cascades that regulate apoptosis in both peripheral and central neurons.
Collapse
|
222
|
Yoon SO, Soltoff SP, Chao MV. A dominant role of the juxtamembrane region of the TrkA nerve growth factor receptor during neuronal cell differentiation. J Biol Chem 1997; 272:23231-8. [PMID: 9287331 DOI: 10.1074/jbc.272.37.23231] [Citation(s) in RCA: 26] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
All receptor tyrosine kinases share a common intracellular signaling machinery, including ras activation, whereas cellular responses vary from mitogenesis to cell differentiation. To investigate the structural basis for receptor tyrosine kinase action for nerve growth factor, the juxtamembrane region of TrkA was transferred to a corresponding region of the epidermal growth factor (EGF) receptor. The resulting chimeric receptor contains an additional Shc site, Tyr490, in the juxtamembrane region. In transfected PC12 cell lines, neuronal differentiation was observed with EGF treatment, as evidenced by increased neurite extension. The action of the chimeric receptor was correlated with prolonged activation of MAP kinases and a 3-4-fold increase in phosphatidylinositol 3-kinase activity. The effect of the juxtamembrane chimera was dependent upon the Shc site at Tyr490, because expression of a chimeric receptor containing a Y490F mutation resulted in a complete loss of neuritogenesis by EGF treatment. These findings indicate that the juxtamembrane region of the TrkA receptor serves as a key functional domain that can confer a dominant effect upon neuronal differentiation.
Collapse
Affiliation(s)
- S O Yoon
- Beth Israel Deaconess Medical Center, Division of Signal Transduction, Department of Medicine, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
223
|
Grimes ML, Beattie E, Mobley WC. A signaling organelle containing the nerve growth factor-activated receptor tyrosine kinase, TrkA. Proc Natl Acad Sci U S A 1997; 94:9909-14. [PMID: 9275225 PMCID: PMC23291 DOI: 10.1073/pnas.94.18.9909] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/1997] [Accepted: 07/03/1997] [Indexed: 02/05/2023] Open
Abstract
The topology of signal transduction is particularly important for neurons. Neurotrophic factors such as nerve growth factor (NGF) interact with receptors at distal axons and a signal is transduced by retrograde transport to the cell body to ensure survival of the neuron. We have discovered an organelle that may account for the retrograde transport of the neurotrophin signal. This organelle is derived from endocytosis of the receptor tyrosine kinase for NGF, TrkA. In vitro reactions containing semi-intact PC12 cells and ATP were used to enhance recovery of a novel organelle: small vesicles containing internalized NGF bound to activated TrkA. These vesicles were distinct from clathrin coated vesicles, uncoated primary endocytic vesicles, and synaptic vesicles, and resembled transport vesicles in their sedimentation velocity. They contained 10% of the total bound NGF and almost one-third of the total tyrosine phosphorylated TrkA. These small vesicles are compelling candidates for the organelles through which the neurotrophin signal is conveyed down the axon.
Collapse
Affiliation(s)
- M L Grimes
- Department of Biochemistry, Massey University, Palmerston North, New Zealand.
| | | | | |
Collapse
|
224
|
Nilsson G, Forsberg-Nilsson K, Xiang Z, Hallböök F, Nilsson K, Metcalfe DD. Human mast cells express functional TrkA and are a source of nerve growth factor. Eur J Immunol 1997; 27:2295-301. [PMID: 9341772 DOI: 10.1002/eji.1830270925] [Citation(s) in RCA: 167] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Mast cells are the principal effector cells in IgE-dependent hypersensitivity reactions. Despite reports that rodent mast cells proliferate in the presence of nerve growth factor (NGF), human mast cells reportedly do not respond to this factor. To determine if human mast cells express the NGF receptors, TrkA tyrosine receptor and the low affinity NGF receptor (LNGFR), we first analyzed the mRNA expression by RT-PCR of TrkA and LNGFR in a human mast cell line (HMC-1) and in human mast cells cultured in the presence of stem cell factor. Both HMC-1 and cultured human mast cells were found to express TrkA but not LNGFR. TrkA protein was demonstrated by Western blot analysis of HMC-1 lysates. Using flow cytometric analysis and mast cell tryptase as a mast cell marker, both HMC-1 cells and cultured human mast cells were shown to coexpress tryptase and TrkA. Treatment of mast cells with NGF resulted in phosphorylation of TrkA on tyrosine residues as detected by immunoblotting with an antiphosphotyrosine antibody. Furthermore, NGF induced the immediate early gene c-fos in HMC-1 cells. HMC-1 cells and cultured human mast cells were also found to express NGF mRNA, and conditioned medium from HMC-1 cells stimulated neurite outgrowth from chicken embryonic sensory ganglia in culture. This effect was blocked by anti-NGF. Thus, mast cells express functional TrkA and synthesize NGF, suggesting a mechanism by which NGF may act as an autocrine factor for human mast cells, and by which mast cells and nerves may interact.
Collapse
Affiliation(s)
- G Nilsson
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, USA.
| | | | | | | | | | | |
Collapse
|
225
|
Abstract
The members of the neurotrophin family play key biological roles in the development of the nervous system. Based on studies initially in cell lines (e.g., the rat pheochromocytoma PC12 cells), neurotrophins have been found to be important mediators of proliferation, differentiation, and survival in the normal brain, but their role in brain tumors remains unclear. Since neurotrophins and neurotrophin receptors are frequently detected in biopsy samples of central nervous system medulloblastomas, efforts have been undertaken in several laboratories to elucidate the potential effects of neurotrophins on the growth and differentiation of these tumors. Results from these studies may have both basic and clinical implications because medulloblastomas resemble embryonic neuroectodermal stem cells and/or their immature neuronal and glial progeny. This review focuses on recent developments in our understanding of the role of neurotrophins in medulloblastomas, especially the ability of nerve growth factor to induce apoptosis in vitro in medulloblastomas.
Collapse
Affiliation(s)
- T T Chou
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia 19104-4283, USA
| | | | | |
Collapse
|
226
|
Jiang H, Movsesyan V, Fink, Jr. DW, Fasler M, Whalin M, Katagiri Y, Monshipouri M, Dickens G, Lelkes PI, Guroff G, Lazarovici P. Expression of human p140trk receptors in p140trk-deficient, PC12/endothelial cells results in nerve growth factor-induced signal transduction and DNA synthesis. J Cell Biochem 1997. [DOI: 10.1002/(sici)1097-4644(19970801)66:2<229::aid-jcb10>3.0.co;2-c] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
227
|
Yao L, Zhang D, Bernd P. Differential regulation of substance P by all members of the nerve growth factor family of neurotrophins in avian dorsal root ganglia throughout development. Neuroscience 1997; 79:1197-206. [PMID: 9219978 DOI: 10.1016/s0306-4522(96)00698-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
This study examined the effects of nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin-4/5 on substance P levels in dorsal root ganglia of the quail shortly after ganglia formation (stage 26, embryonic day 4.5), during the middle of development (stage 33, embryonic day 7.5) and during late development (stage 44, embryonic day 14). It has already been shown that nerve growth factor increases levels of substance P during the middle and late stages of development, and that messenger RNA for the neurotrophin receptors, trkA, trkB and trkC is present at all of these stages. Dorsal root ganglia were isolated, rinsed with defined medium to dilute endogenous neurotrophins and exposed to one of the neurotrophins for either 4 or 20 h. Substance P levels were quantitated using enzyme immunoassay. None of the neurotrophins had any effect on substance P levels in dorsal root ganglia obtained at stage 26 after either a 4 or 20 h exposure time. Nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin-4/5 all significantly increased levels of substance P after either a 4 h or 20 h incubation in ganglia obtained at stages 33 and 44. The effects of nerve growth factor and neurotrophin-3 were specific: increases in substance P were completely blocked by simultaneous exposure to antibodies against either nerve growth factor or neurotrophin-3. The absence of any effect of neurotrophins on substance P expression during early development was unexpected, since dorsal root ganglia exhibit substantial levels of substance P and receptors for the neurotrophins are present and are apparently functional. It was also surprising that brain-derived neurotrophic factor, neurotrophin-3 and neurotrophin-4/5 induced increases in substance P levels during the middle and late stages of development, since substance P was thought to be exclusively localized to small TrkA neurons in dorsal root ganglia. However, immunocytochemical examination of dorsal root ganglia at stages 33 and 44 revealed substance P-like immunoreactivity in larger neurons as well as in small neurons. The results of this study have shown that different cellular responses to neurotrophins, such as effects on survival and/or peptide expression, may be acquired with differing temporal patterns not strictly related to expression of their receptors. Further, the regulation of neuropeptide synthesis in dorsal root ganglia is not due to any one neurotrophic factor. and the factors that regulate expression during early development are still unknown.
Collapse
Affiliation(s)
- L Yao
- Department of Anatomy and Cell Biology, SUNY Health Science Center at Brooklyn, NY 11203, USA
| | | | | |
Collapse
|
228
|
Abstract
We have developed an in vitro system for studying the interaction of chick dorsal root ganglion neuronal growth cones with a localized source of nerve growth factor (NGF) covalently conjugated to polystyrene beads. Growth cones rapidly turned and migrated under NGF-coated beads in a process that involved the initial formation of persistent contact with a bead, followed by directed flow of cytoplasm toward the point of contact. A role for the local activation of the high-affinity NGF receptor trkA was suggested by a strong inhibition of the turning response by (1) the addition of an antibody against the extracellular portion of trkA, (2) the elevation of the background concentration of NGF to saturate trkA, or (3) the presence of a concentration of the drug K252a that inhibits trkA activation. NGF binding to the pan-neurotrophin receptor p75 is also involved but is not required for turning. These data show a new role for both the trkA and the p75 receptors: the mediation of local events in the guidance of nerve growth cones.
Collapse
|
229
|
Ivkovic S, Polonskaia O, Fariñas I, Ehrlich ME. Brain-derived neurotrophic factor regulates maturation of the DARPP-32 phenotype in striatal medium spiny neurons: studies in vivo and in vitro. Neuroscience 1997; 79:509-16. [PMID: 9200733 DOI: 10.1016/s0306-4522(96)00684-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The medium spiny neuron is the predominant striatal neuronal subtype. The striatum, a participant in motor and cognitive functions, is a site of pathophysiology in prevalent neuropsychiatric diseases and is the target of many currently utilized pharmacologic agents. DARPP-32, a dopamine and cyclic AMP-regulated phosphoprotein, is a widely-used marker of mature striatal medium-sized neurons, but the molecules regulating DARPP-32 transcription have not been identified. We show that a null mutation in the mouse brain-derived neurotrophic factor gene leads to decreased DARPP-32 immunoreactivity in striatal medium spiny neurons at birth and postnatal day 10. Striatal DARPP-32 messenger RNA and protein are decreased relative to wild-type littermate controls. In densely plated (1 x 10(6) cells/cm2) primary cultures derived from the ganglionic eminences, addition of brain-derived neurotrophic factor (100 ng/ml) to defined media results in a greater than 3-fold increase in the number of DARPP-32-immunopositive cells after 12 h and greater than 4-fold (P<0.005) after 24 h. The increase in DARPP-32-immunopositivity is abolished by the addition of 2 microg/ml actinomycin D without a significant effect on cell viability. These data suggest that brain-derived neurotrophic factor directly or indirectly regulates DARPP-32 transcription in medium spiny neurons. This is the first demonstration of transcriptional regulation of DARPP-32, and the first evidence of a forebrain abnormality in a newborn neurotrophin "knockout" mouse.
Collapse
Affiliation(s)
- S Ivkovic
- Department of Psychiatry, New York University Medical Center, New York 10016, U.S.A
| | | | | | | |
Collapse
|
230
|
Abstract
The ongoing dissection of the roles of p75NTR and TrkA, -B and -C in neurotrophin signaling has generated a number of apparent paradoxes. Limiting consideration to the role of p75NTR in cell death, a theory is proposed that is based on the following postulates: (1) that p75NTR displays a pro-apoptotic intrinsic (ligand-independent, Trk-independent) receptor effect (IRE), which is inhibited by ligand binding; (2) that p75NTR and TrkA exhibit mutual repression of signaling; and (3) that p75NTR and TrkA are required for the efficient generation of high-affinity NGF binding sites.
Collapse
|
231
|
Deshmukh M, Johnson EM. Programmed cell death in neurons: focus on the pathway of nerve growth factor deprivation-induced death of sympathetic neurons. Mol Pharmacol 1997; 51:897-906. [PMID: 9187255 DOI: 10.1124/mol.51.6.897] [Citation(s) in RCA: 180] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Extensive programmed cell death (PCD) occurs in the developing nervous system. Neuronal death occurs, at least in part, because neurons are produced in excess during development and compete with each other for the limited amounts of the survival-promoting trophic factors secreted by target tissues. Neuronal death is apoptotic and utilizes components that are conserved in other PCD pathways. In this review, we discuss the mechanism of trophic factor-dependent neuronal cell death by focusing on the pathway of nerve growth factor (NGF) deprivation-induced sympathetic neuronal death. We describe the biochemical and genetic events that occur in NGF-deprived sympathetic neurons undergoing PCD. Participation of the Bcl-2 family of proteins and the interleukin-1beta-converting enzyme family of proteases (caspases) in this and other models of neuronal death is also examined. The order and importance of these components during NGF deprivation-induced sympathetic neuronal death are discussed.
Collapse
Affiliation(s)
- M Deshmukh
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
232
|
Abstract
Activation specific tyrosine kinase receptors by neurotrophins accounts for the longest known biological actions of the neurotrophins, in particular the promotion of neuronal survival. However, recent studies have revealed that nerve growth factor, the neurotrophin regarded as best understood, also activates a signalling pathway by binding to the neurotrophin receptor p75(NTR). This receptor belongs to the tumor necrosis factor receptor family and lacks intrinsic catalytic activity. The p75(NTR) receptor binds all neurotrophins with nanomolar affinity; however, nerve growth factor seems to be uniquely able to activate it, causing the death of trkA-negative neurons during normal development. Thus, nerve growth factor prevents programmed cell death through its receptor TrkA, but promotes it by signalling through p75(NTR).
Collapse
Affiliation(s)
- G Dechant
- Max-Planck Institute for Psychiatry, Department of Neurobiochemistry Am Klopferspitz 18A, 82152 Martinsried, Germany.
| | | |
Collapse
|
233
|
Cunningham ME, Stephens RM, Kaplan DR, Greene LA. Autophosphorylation of activation loop tyrosines regulates signaling by the TRK nerve growth factor receptor. J Biol Chem 1997; 272:10957-67. [PMID: 9099755 DOI: 10.1074/jbc.272.16.10957] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Many receptor tyrosine kinases possess an "activation loop" containing three similarly placed tyrosine autophosphorylation sites. To examine their roles in the TRK NGF receptor, these residues (Tyr-670, Tyr-674, and Tyr-675) were mutated singly and in all combinations to phenylalanine and stably expressed in Trk-deficient PC12nnr5 cells. All mutant receptors showed significantly diminished nerve growth factor (NGF)-stimulated autophosphorylation, indicating impaired catalytic activity. NGF-induced neurite outgrowth exhibited dose-responsive behavior when transfectants were compared by relative receptor expression and exhibited a functional hierarchy: wild type > Y670F >/= Y674F >> Y675F >/= YY670/674FF = YY670/675FF >> YY674/675FF > YYY670/674/675FFF. NGF-induced tyrosine phosphorylation of Shc, ERKs, and SNT and immediate early gene inductions generally paralleled neurogenic potential. However, activation of phosphatidylinositol 3'-kinase and tyrosine phosphorylation of phospholipase Cgamma-1 was essentially abolished. The latter effect appears due to selective inability of the mutated TRKs to autophosphorylate the tyrosine residue (Tyr-785) required for binding phospholipase Cgamma-1 and indicates that the "activation loop" tyrosines participate in NGF-dependent changes in receptor conformation. Our findings stress the importance that expression levels play in assessing the consequences of receptor mutations and that all three activation loop tyrosines have roles regulating both overall and specific NGF-mediated signaling through TRK.
Collapse
Affiliation(s)
- M E Cunningham
- Department of Pathology and Center of Neurobiology and Behavior, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | |
Collapse
|
234
|
Longo FM, Manthorpe M, Xie YM, Varon S. Synthetic NGF peptide derivatives prevent neuronal death via a p75 receptor-dependent mechanism. J Neurosci Res 1997; 48:1-17. [PMID: 9086177 DOI: 10.1002/(sici)1097-4547(19970401)48:1<1::aid-jnr1>3.0.co;2-k] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Cyclized peptides corresponding to beta-loop regions of NGF were purified by HPLC and assayed for neurotrophic activity using DRG neurons. Peptides with the highest activity corresponded to loop region 29-35, a domain likely to interact with the p75 receptor. Unexpectedly, activity was confined to late-eluting HPLC fractions containing peptide multimers and primarily promoted neuronal survival without neurite outgrowth. Directed synthesis of dimer and monomer cyclized peptides demonstrated that dimers acted as partial NGF agonists in that they had both survival-promoting and NGF-inhibiting activity while monomer and linear peptides were inactive. Dimer activity was not affected by the Trk inhibitor K252a but was blocked by p75 receptor antibody and absent using p75 null mutant neurons. These studies suggest that region 29-35 peptide derivatives inhibit neuronal death via a structure- and p75-dependent mechanism.
Collapse
Affiliation(s)
- F M Longo
- Department of Neurology, UCSF/VAMC, San Francisco, California 94121, USA
| | | | | | | |
Collapse
|
235
|
Abstract
The neurotrophins signal cell survival, differentiation, growth cessation, and apoptosis through two cell surface receptors, the Trks and p75NTR (p75 neurotrophin receptor). Recent advances indicate that the particular events that are mediated by neurotrophins are dependent upon the cell type and the expression pattern of each neurotrophin receptor. For example, TrkA activation induces cell death of neural tumor cells, and survival and differentiation of neurons. Likewise, p75NTR, when activated in the absence of a strong Trk signal, induces apoptosis of neurons, while in the presence of Trk it enhances responses to neurotrophin. These differing responses point to a complex interplay between neurotrophin-stimulated survival, differentiation, and apoptosis pathways.
Collapse
Affiliation(s)
- D R Kaplan
- Brain Tumor Research Centre, Montreal Neurological Institute, 3801 University Street, Montreal, PQ, Canada, H3A 2B4.
| | | |
Collapse
|
236
|
Chen DF, Schneider GE, Martinou JC, Tonegawa S. Bcl-2 promotes regeneration of severed axons in mammalian CNS. Nature 1997; 385:434-9. [PMID: 9009190 DOI: 10.1038/385434a0] [Citation(s) in RCA: 341] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Most neurons of the mammalian central nervous system (CNS) lose the ability to regenerate severed axons in vivo after a certain point in development. At least part of this loss in regenerative potential is intrinsic to neurons. Although embryonic retinal ganglion cells (RGCs) can grow axons into tectum of any age, most RGCs from older animals fail to extend axons into CNS tissue derived from donors of any age, including the embryonic tectum. Here we report that the proto-oncogene bcl-2 plays a key role in this developmental change by promoting the growth and regeneration of retinal axons. This effect does not seem to be an indirect consequence of its well-known anti-apoptotic activity. Another anti-apoptotic drug, ZVAD, supported neuronal survival but did not promote axon regeneration in culture. This finding could lead to new strategies for the treatment of injuries to the CNS.
Collapse
Affiliation(s)
- D F Chen
- Howard Hughes Medical Institute, Center for Learning and Memory, Center for Cancer Research, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | | | | | |
Collapse
|
237
|
Wu C, Butz S, Ying Y, Anderson RG. Tyrosine kinase receptors concentrated in caveolae-like domains from neuronal plasma membrane. J Biol Chem 1997; 272:3554-9. [PMID: 9013605 DOI: 10.1074/jbc.272.6.3554] [Citation(s) in RCA: 198] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Recent evidence suggests that tyrosine kinases are highly organized in caveolae of tissue culture cells. We now report the isolation of a membrane domain from neuronal plasma membranes that has the biochemical characteristics of caveolae. A low density membrane (LDM) fraction with the same density as caveolae was highly enriched in tyrosine kinases such as insulin receptors, neurotrophin receptors, Eph family receptors, and Fyn. Grb2, Ras, heterotrimeric GTP-binding proteins, and Erk2 were also concentrated in the LDM. Incubation of the LDM fraction at 37 degrees C stimulated the phosphorylation on tyrosine of multiple, resident proteins, whereas the bulk membrane fraction was devoid of tyrosine kinase activity. The LDM, which makes up approximately 5-10% of the plasma membrane protein, appears to be organized for signal transduction.
Collapse
Affiliation(s)
- C Wu
- Department of Cell Biology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75235-9039, USA
| | | | | | | |
Collapse
|
238
|
Holm NR, Christophersen P, Olesen SP, Gammeltoft S. Activation of calcium-dependent potassium channels in mouse [correction of rat] brain neurons by neurotrophin-3 and nerve growth factor. Proc Natl Acad Sci U S A 1997; 94:1002-6. [PMID: 9023372 PMCID: PMC19629 DOI: 10.1073/pnas.94.3.1002] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The neurotrophins are signaling factors that are essential for survival and differentiation of distinct neuronal populations during the development and regeneration of the nervous system. The long-term effects of neurotrophins have been studied in detail, but little is known about their acute effects on neuronal activity. Here we use permeabilized whole-cell patch clamp to demonstrate that neurotrophin-3 (NT-3) and nerve growth factor activate calcium-dependent, paxilline-sensitive potassium channels (BK channels) in cortical neurons. Application of NT-3 or nerve growth factor produced a rapid and gradual rise in BK current that was sustained for 30-50 min; brain-derived neurotrophic factor, ciliary neurotrophic factor, and insulin-like growth factor-1 had no significant effect. The response to NT-3 was blocked by inhibitors of protein kinases, phospholipase C, and serine/threonine protein phosphatase 1 and 2a. Omission of Ca2+ from the extracellular medium prevented the NT-3 effect. Our results indicate that NT-3 stimulates BK channel activity in cortical neurons through a signaling pathway that involves Trk tyrosine kinase, phospholipase C, and protein dephosphorylation and is calcium-dependent. Activation of BK channels may be a major mechanism by which neurotrophins acutely regulate neuronal activity.
Collapse
|
239
|
Van der Zee CE, Ross GM, Riopelle RJ, Hagg T. Survival of cholinergic forebrain neurons in developing p75NGFR-deficient mice. Science 1996; 274:1729-32. [PMID: 8939868 DOI: 10.1126/science.274.5293.1729] [Citation(s) in RCA: 166] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The functions of the low-affinity p75 nerve growth factor receptor (p75(NGFR)) in the central nervous system were explored in vivo. In normal mice, approximately 25 percent of the cholinergic basal forebrain neurons did not express TrkA and died between postnatal day 6 and 15. This loss did not occur in p75(NGFR)-deficient mice or in normal mice systemically injected with a p75(NGFR)-inhibiting peptide. Control, but not p75(NGFR)-deficient, mice also had fewer cholinergic striatal interneurons. Apparently, p75(NGFR) mediates apoptosis of these developing neurons in the absence of TrkA, and modulation of p75(NGFR) can promote neuronal survival. Cholinergic basal forebrain neurons are involved in learning and memory.
Collapse
Affiliation(s)
- C E Van der Zee
- Department of Anatomy and Neurobiology, Tupper Building, Dalhousie University, Halifax, Nova Scotia B3H 4H7, Canada.
| | | | | | | |
Collapse
|
240
|
Kang H, Schuman EM. A requirement for local protein synthesis in neurotrophin-induced hippocampal synaptic plasticity. Science 1996; 273:1402-6. [PMID: 8703078 DOI: 10.1126/science.273.5280.1402] [Citation(s) in RCA: 725] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Two neurotrophic factors, brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), are able to produce a long-lasting enhancement of synaptic transmission in the hippocampus. Unlike other forms of plasticity, neurotrophin-induced plasticity exhibited an immediate requirement for protein synthesis. Plasticity in rat hippocampal slices in which the synaptic neuropil was isolated from the principal cell bodies also required early protein synthesis. Thus, the neurotrophins may stimulate the synthesis of proteins in either axonal or dendritic compartments, allowing synapses to exert local control over the complement of proteins expressed at individual synaptic sites.
Collapse
Affiliation(s)
- H Kang
- Division of Biology 216-76, California Institute of Technology, Pasadena, CA 91125, USA
| | | |
Collapse
|
241
|
Rasouly D, Shavit D, Zuniga R, Elejalde RB, Unsworth BR, Yayon A, Lazarovici P, Lelkes PI. Staurosporine induces neurite outgrowth in neuronal hybrids (PC12EN) lacking NGF receptors. J Cell Biochem 1996; 62:356-71. [PMID: 8872607 DOI: 10.1002/(sici)1097-4644(199609)62:3<356::aid-jcb6>3.0.co;2-q] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A novel neuronal model (PC12EN cells), obtained by somatic hybridization of rat adrenal medullary pheochromocytoma (PC12) and bovine adrenal medullary endothelial (BAME) cells, was developed. PC12EN cells maintained numerous neuronal characteristics: they expressed neuronal glycolipid conjugates, synthesized and secreted catecholamines, and responded to differentiative agents with neurite outgrowth. PC12EN lacked receptors for EGF and both the p75 and trk NGF receptors, while FGF receptor expression was maintained. Staurosporine (5-50 nM), but not other members of the K252a family of protein kinase inhibitors, rapidly induced neurite outgrowth in PC12EN, as also found in the parental PC12 cells, but not in BAME cells. Similarly, both acidic and basic FGF (1-100 ng/ml) were neurotropic in PC12EN. In contrast to the mechanism by which FGF promoted neurite outgrowth in PC12EN, the neurotropic effect of staurosporine did not involve activation of established signalling pathways, such as tyrosine phosphorylation of erk (ras pathway) or SNT (a specific target of neuronal differentiation). In addition, staurosporine induced the tyrosine phosphorylation of the focal adhesion kinase p125FAK. However, since the latter effect was also observed with other protein kinase inhibitors of the K252a family, which induced PC12EN cells flattening but no neurite extension, we propose that FAK tyrosine phosphorylation may be related to ubiquitous changes in cell shape. We anticipate that PC12EN neuronal hybrids will become useful models in neuroscience research for evaluating unique cellular signalling mechanisms of novel neurotropic compounds.
Collapse
Affiliation(s)
- D Rasouly
- Department of Pharmacology and Experimental Therapeutics, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
242
|
Borasio GD, Markus A, Heumann R, Ghezzi C, Sampietro A, Wittinghofer A, Silani V. Ras p21 protein promotes survival and differentiation of human embryonic neural crest-derived cells. Neuroscience 1996; 73:1121-7. [PMID: 8809829 DOI: 10.1016/0306-4522(96)00084-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We have previously shown that the oncogene product p21 Ras is essential for the survival and neurite outgrowth-promoting activity of nerve growth factor on cultured chick embryonic sensory, but not sympathetic neurons. In order to extend our observations to the human system and to non-neuronal cells, we introduced the oncogenic form of p21 Ras into the cytoplasm of three different types of cultured human embryonic neural crest derivatives (8th-11th gestational week): dorsal root ganglion neurons, sympathetic neurons, and adrenal chromaffin cells. These cells are dependent on nerve growth factor for survival and/or fibre outgrowth in vitro. In dorsal root ganglion neurons, p21 Ras promoted survival and fibre outgrowth which was quantitatively and qualitatively comparable to the nerve growth factor effect (84% vs. 95%, control 18%). Sympathetic neurons showed a similar effect, albeit with a higher background survival (91% vs. 93%, control 58%). On chromaffin cells, which respond to nerve growth factor with pronounced fibre outgrowth in culture, the effect of p21 Ras was again comparable to that of nerve growth factor (35% vs. 30%, control 5%). The survival and fibre outgrowth-promoting effects of p21 Ras on human embryonic dorsal root ganglion neurons, sympathetic neurons and chromaffin cells suggest an involvement of p21 Ras in the intracellular signal transduction of nerve growth factor in human neural crest-derived cell populations.
Collapse
Affiliation(s)
- G D Borasio
- Neurologische Klinik, Ludwig-Maximilians-Universität, Klinikum Grosshadern, München, Germany
| | | | | | | | | | | | | |
Collapse
|
243
|
Nerve growth factor (NGF)-mediated protection of neural crest cells from antimitotic agent-induced apoptosis: the role of the low-affinity NGF receptor. J Neurosci 1996. [PMID: 8656283 DOI: 10.1523/jneurosci.16-12-03895.1996] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Prevention by nerve growth factor (NGF) of apoptotic death in neural cells has been variously ascribed to binding of NGF to its low-affinity (p75) or high-affinity (trkA) receptor or to a cooperative interaction between the two. In a series of studies using, in turn, neuroblastoma cell lines that express only p75, mutant NGF species that bind selectively to either p75 or trkA, and a polyclonal antibody that binds to the NGF-binding domain of p75, we demonstrate that NGF binding to p75 is both necessary and sufficient for the abrogation of apoptosis in neuroblastoma cells treated with antimitotic agents.
Collapse
|
244
|
Synaptic modulation by neurotrophic factors: differential and synergistic effects of brain-derived neurotrophic factor and ciliary neurotrophic factor. J Neurosci 1996. [PMID: 8627363 DOI: 10.1523/jneurosci.16-10-03256.1996] [Citation(s) in RCA: 156] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Extracellular application of brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF) to developing neuromuscular junctions in Xenopus nerve-muscle cultures resulted in an increase in the frequency of spontaneous synaptic currents (SSCs) and in the amplitude of nerve-evoked synaptic currents. Analyses of the amplitude and time course of the SSCs suggest that these effects are attributable to elevation of presynaptic transmitter release. The actions of these two factors on the transmitter secretion process, however, are distinctly different. Fura-2 Ca2+ imaging showed that an increase in presynaptic cytosolic Ca2+ ([Ca2+]i) accompanied the synaptic potentiation by BDNF, whereas no change in [Ca2+]i was observed during synaptic potentiation by CNTF. Removing external Ca2+ also abolished the potentiating effect of BDNF but did not influence the CNTF effect. Moreover, the two factors exerted different effects on the short-term synaptic plasticity. Paired-pulse facilitation normally found at these synapses was reduced by BDNF but unaffected by CNTF; CNTF, but not BDNF, reduced the extent of synaptic depression during high-frequency tetanic stimulation. Finally, the potentiation effect of BDNF and CNTF on spontaneous transmitter release was additive when both factors were applied together to the synapse at saturating concentrations (100 ng/ml) and was highly synergistic when low doses (1 and 10 ng/ml) of both factors were used. These results suggest that because of their differential effects on the secretory machinery, BDNF and CNTF may act cooperatively in modulating the development and functioning of synapses.
Collapse
|
245
|
Abstract
A diversity of neurotrophic factors are required for the differentiation and survival of neurons and for maintaining their phenotype. By virtue of the rapid time scale of signal transduction in the cytosol, many of these factors also acutely regulate neuronal functions as diverse as synaptic transmission and nerve growth. These fast actions greatly expand the regulatory role of neurotrophic factors, particularly in the synaptic plasticity of developing nervous systems.
Collapse
Affiliation(s)
- B Berninger
- Department of Neurochemistry, Max Planck Institute for Psychiatry, Am Klopferspitz 18a, D-82152 Martinsried, Germany.
| | | |
Collapse
|