Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 6070, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988348

ABSTRACT

During the development of the sympathetic nervous system, signals from tropomyosin-related kinase receptors (Trks) and p75 neurotrophin receptors (p75) compete to regulate survival and connectivity. During this process, nerve growth factor (NGF)- TrkA signaling in axons communicates NGF-mediated trophic responses in signaling endosomes. Whether axonal p75 signaling contributes to neuronal death and how signaling endosomes contribute to p75 signaling has not been established. Using compartmentalized sympathetic neuronal cultures (CSCGs) as a model, we observed that the addition of BDNF to axons increased the transport of p75 and induced death of sympathetic neurons in a dynein-dependent manner. In cell bodies, internalization of p75 required the activity of JNK, a downstream kinase mediating p75 death signaling in neurons. Additionally, the activity of Rab5, the key GTPase regulating early endosomes, was required for p75 death signaling. In axons, JNK and Rab5 were required for retrograde transport and death signaling mediated by axonal BDNF-p75 in CSCGs. JNK was also required for the proper axonal transport of p75-positive endosomes. Thus, our findings provide evidence that the activation of JNK by p75 in cell bodies and axons is required for internalization to a Rab5-positive signaling endosome and the further propagation of p75-dependent neuronal death signals.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/pathology , Receptors, Growth Factor/metabolism , rab5 GTP-Binding Proteins/metabolism , Animals , Apoptosis/drug effects , Axons/metabolism , Cells, Cultured , Endosomes/metabolism , Female , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Neurons/cytology , Neurons/metabolism , Primary Cell Culture , Rats , Receptor, trkA/metabolism , Superior Cervical Ganglion/cytology
2.
Br J Anaesth ; 120(4): 745-760, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29576115

ABSTRACT

BACKGROUND: Exposure of the developing brain to propofol results in cognitive deficits. Recent data suggest that inhibition of neuronal apoptosis does not prevent cognitive defects, suggesting mechanisms other than neuronal apoptosis play a role in anaesthetic neurotoxicity. Proper neuronal growth during development is dependent upon growth cone morphology and axonal transport. Propofol modulates actin dynamics in developing neurones, causes RhoA-dependent depolymerisation of actin, and reduces dendritic spines and synapses. We hypothesised that RhoA inhibition prevents synaptic loss and subsequent cognitive deficits. The present study tested whether RhoA inhibition with the botulinum toxin C3 (TAT-C3) prevents propofol-induced synapse and neurite loss, and preserves cognitive function. METHODS: RhoA activation, growth cone morphology, and axonal transport were measured in neonatal rat neurones (5-7 days in vitro) exposed to propofol. Synapse counts (electron microscopy), dendritic arborisation (Golgi-Cox), and network connectivity were measured in mice (age 28 days) previously exposed to propofol at postnatal day 5-7. Memory was assessed in adult mice (age 3 months) previously exposed to propofol at postnatal day 5-7. RESULTS: Propofol increased RhoA activation, collapsed growth cones, and impaired retrograde axonal transport of quantum dot-labelled brain-derived neurotrophic factor, all of which were prevented with TAT-C3. Adult mice previously treated with propofol had decreased numbers of total hippocampal synapses and presynaptic vesicles, reduced hippocampal dendritic arborisation, and infrapyramidal mossy fibres. These mice also exhibited decreased hippocampal-dependent contextual fear memory recall. All anatomical and behavioural changes were prevented with TAT-C3 pre-treatment. CONCLUSION: Inhibition of RhoA prevents propofol-mediated hippocampal neurotoxicity and associated cognitive deficits.


Subject(s)
Axonal Transport/drug effects , Behavior, Animal/drug effects , Growth Cones/drug effects , Propofol , Synapses/drug effects , rhoA GTP-Binding Protein/antagonists & inhibitors , Animals , Botulinum Toxins , Brain/drug effects , Disease Models, Animal , Hypnotics and Sedatives , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurotoxicity Syndromes , Rats , Rats, Sprague-Dawley , rhoA GTP-Binding Protein/genetics
3.
Neurobiol Dis ; 77: 173-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25753471

ABSTRACT

Down Syndrome (DS), trisomy 21, is characterized by synaptic abnormalities and cognitive deficits throughout the lifespan and with development of Alzheimer's disease (AD) neuropathology and progressive cognitive decline in adults. Synaptic abnormalities are also present in the Ts65Dn mouse model of DS, but which synapses are affected and the mechanisms underlying synaptic dysfunction are unknown. Here we show marked increases in the levels and activation status of TrkB and associated signaling proteins in cortical synapses in Ts65Dn mice. Proteomic analysis at the single synapse level of resolution using array tomography (AT) uncovered increased colocalization of activated TrkB with signaling endosome related proteins, and demonstrated increased TrkB signaling. The extent of increases in TrkB signaling differed in each of the cortical layers examined and with respect to the type of synapse, with the most marked increases seen in inhibitory synapses. These findings are evidence of markedly abnormal TrkB-mediated signaling in synapses. They raise the possibility that dysregulated TrkB signaling contributes to synaptic dysfunction and cognitive deficits in DS.


Subject(s)
Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Down Syndrome/pathology , Receptor, trkB/metabolism , Signal Transduction/physiology , Synaptosomes/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Transgenic , RNA, Messenger/metabolism , Signal Transduction/genetics , Synaptosomes/drug effects
4.
Sci Transl Med ; 1(7): 7ra17, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-20368182

ABSTRACT

Down syndrome (trisomy 21) is the most common cause of mental retardation in children and leads to marked deficits in contextual learning and memory. In rodents, these tasks require the hippocampus and are mediated by several inputs, particularly those originating in the locus coeruleus. These afferents mainly use norepinephrine as a transmitter. To explore the basis for contextual learning defects in Down syndrome, we examined the Ts65Dn mouse model. These mice, which have three copies of a fragment of mouse chromosome 16, exhibited significant deficits in contextual learning together with dysfunction and degeneration of locus coeruleus neurons. However, the postsynaptic targets of innervation remained responsive to noradrenergic receptor agonists. Indeed, despite advanced locus coeruleus degeneration, we were able to reverse contextual learning failure by using a prodrug for norepinephrine called l-threo-3,4-dihydroxyphenylserine, or xamoterol, a beta(1)-adrenergic receptor partial agonist. Moreover, an increased gene dosage of App, in the context of Down syndrome, was necessary for locus coeruleus degeneration. Our findings raise the possibility that restoring norepinephrine-mediated neurotransmission could reverse cognitive dysfunction in Down syndrome.


Subject(s)
Disease Models, Animal , Down Syndrome/physiopathology , Memory , Norepinephrine/physiology , Animals , Down Syndrome/psychology , Learning Disabilities , Mice
5.
Gene Ther ; 13(11): 917-25, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16482204

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is caused by an inherited deficiency of alpha-L-iduronidase (IDUA). The result is a progressive, lysosomal storage disease with central nervous system (CNS) as well as systemic involvement. To target gene therapy to the CNS, recombinant adeno-associated virus (AAV) vectors carrying IDUA sequence were administered to MPS I mice via injection into cerebrospinal fluid. In contrast to intravenous administration, this intrathecal administration was effective in generating widespread IDUA activity in the brain, with the cerebellum and olfactory bulbs having highest activities. In general, IDUA levels correlated with vector dose, although this correlation was obscured in cerebellum by particularly high variability. High doses of vector (4 x 10(10) particles) provided IDUA levels approaching or exceeding normal levels in the brain. Histopathology indicated that the number of cells with storage vacuoles was reduced extensively or was eliminated entirely. Elimination of storage material in Purkinje cells was particularly dramatic. A lower vector dose (2 x 10(9) particles) reduced both the number of storage cells and the extent of storage per cell, but the effect was not complete. Some perivascular cells with storage persisted, and this cell type appeared to be more resistant to treatment than neurons or glial cells. We conclude that intrathecal administration of AAV-IDUA delivers vector to brain cells, and that this route of administration is both minimally invasive and effective.


Subject(s)
Brain/metabolism , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Iduronidase/genetics , Mucopolysaccharidosis I/therapy , Animals , Brain/ultrastructure , Fluorescent Antibody Technique, Indirect , Gene Expression , Iduronidase/analysis , Iduronidase/metabolism , Injections, Spinal , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology
6.
Mol Genet Metab ; 86(1-2): 141-9, 2005.
Article in English | MEDLINE | ID: mdl-16006167

ABSTRACT

Central nervous system disease can have devastating consequences in the severe or Hurler form of mucopolysaccharisosis I (MPS I). Intravenously administered recombinant human alpha-L-iduronidase (rhIDU) is not expected to reach and treat the brain disease due to the blood-brain barrier. To determine whether administration of rhIDU into the cerebrospinal fluid could successfully treat the brain, we studied intraventricular administration of rhIDU in rats. RhIDU was stereotactically administered directly to the lateral ventricle of the intact rat brain and the brain tissues assessed by enzyme assays, immunofluorescence and confocal microscopy 30 min, 24 h, or 7 days later. Quantitation of activity revealed that rhIDU was widely distributed throughout the brain following injection into the lateral ventricle, with activities increased by a factor of 3.3 higher than control in most samples 30 min-24 h after injection and highest levels on the side of injection. The enzyme crossed the ependymal lining of the ventricle and entered neurons into lysosomal-like vesicles. The enzyme was able to diffuse through brain tissue as demonstrated by a decreasing signal gradient from 0.2 to 4.8 mm from the ventricle surface. The largest amount of rhIDU, as detected by immunostaining, was observed 24 h after injection and decreased approximately 50% during the first 7 days. Although the immunostaining decreased with time, specific vesicular staining was still detectable 28 days after injection. The data suggest that rhIDU given into the ventricle can diffuse, penetrate at least several millimeters of brain tissue and be taken up into neurons and glial cells.


Subject(s)
Iduronidase/pharmacokinetics , Animals , Diffusion , Fluorescent Antibody Technique, Indirect , Humans , Iduronidase/administration & dosage , Injections, Intraventricular , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics
7.
Neuron ; 32(5): 801-14, 2001 Dec 06.
Article in English | MEDLINE | ID: mdl-11738027

ABSTRACT

The target-derived neurotrophic factor "nerve growth factor" (NGF) signals through TrkA to promote the survival, differentiation, and maintenance of neurons. How the NGF signal in axon terminals is conveyed to the cell body is unknown. The "signaling endosome hypothesis" envisions that NGF-TrkA complexes are internalized at the axon terminal and retrogradely transported to the cell body. Following NGF treatment, we found that clathrin-coated vesicles contained NGF bound to TrkA together with activated signaling proteins of the Ras-MAP kinase pathway. Evidence that these vesicles could signal was their ability in vitro to activate Elk, a downstream target of Erk1/2. Our results point to the existence of a population of signaling endosomes derived from clathrin-coated membranes in NGF-treated cells.


Subject(s)
Clathrin-Coated Vesicles/physiology , Endosomes/physiology , MAP Kinase Signaling System/physiology , Nerve Growth Factor/physiology , ras Proteins/physiology , Animals , Cells, Cultured , Mice , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/metabolism , Neurons/physiology , PC12 Cells , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptor, trkA/metabolism
8.
Annu Rev Neurosci ; 24: 1217-81, 2001.
Article in English | MEDLINE | ID: mdl-11520933

ABSTRACT

Nerve growth factor (NGF) was discovered 50 years ago as a molecule that promoted the survival and differentiation of sensory and sympathetic neurons. Its roles in neural development have been characterized extensively, but recent findings point to an unexpected diversity of NGF actions and indicate that developmental effects are only one aspect of the biology of NGF. This article considers expanded roles for NGF that are associated with the dynamically regulated production of NGF and its receptors that begins in development, extends throughout adult life and aging, and involves a surprising variety of neurons, glia, and nonneural cells. Particular attention is given to a growing body of evidence that suggests that among other roles, endogenous NGF signaling subserves neuroprotective and repair functions. The analysis points to many interesting unanswered questions and to the potential for continuing research on NGF to substantially enhance our understanding of the mechanisms and treatment of neurological disorders.


Subject(s)
Nerve Growth Factor/physiology , Nerve Regeneration/physiology , Nervous System Physiological Phenomena , Neurons/physiology , Animals , Humans , Nerve Degeneration/physiopathology , Nerve Growth Factor/genetics , Neuroglia/physiology , Neurons/cytology , Signal Transduction/physiology
9.
Proc Natl Acad Sci U S A ; 98(18): 10439-44, 2001 Aug 28.
Article in English | MEDLINE | ID: mdl-11504920

ABSTRACT

Age-related degeneration of basal forebrain cholinergic neurons (BFCNs) contributes to cognitive decline in Alzheimer's disease and Down's syndrome. With aging, the partial trisomy 16 (Ts65Dn) mouse model of Down's syndrome exhibited reductions in BFCN size and number and regressive changes in the hippocampal terminal fields of these neurons with respect to diploid controls. The changes were associated with significantly impaired retrograde transport of nerve growth factor (NGF) from the hippocampus to the basal forebrain. Intracerebroventricular NGF infusion reversed well established abnormalities in BFCN size and number and restored the deficit in cholinergic innervation. The findings are evidence that even BFCNs chronically deprived of endogenous NGF respond to an intervention that compensates for defective retrograde transport. We suggest that age-related cholinergic neurodegeneration may be a treatable disorder of failed retrograde NGF signaling.


Subject(s)
Down Syndrome/metabolism , Nerve Growth Factor/metabolism , Aging/metabolism , Aging/pathology , Animals , Biological Transport, Active , Cell Count , Cells , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Cholinergic Fibers/pathology , Disease Models, Animal , Down Syndrome/drug therapy , Down Syndrome/pathology , Hippocampus/metabolism , Humans , Infusions, Parenteral , Mice , Mice, Mutant Strains , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Nerve Growth Factor/administration & dosage , Phenotype , Prosencephalon/drug effects , Prosencephalon/metabolism , Prosencephalon/pathology , Trisomy
10.
J Neurosci ; 21(15): 5406-16, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466412

ABSTRACT

We investigated a role for endogenous Rap1, a small monomeric GTP-binding protein of the Ras family, in nerve growth factor (NGF) signaling in PC12 cells. Although both epidermal growth factor (EGF) and NGF caused transient activation of Ras, only NGF induced the activation of Rap1. Moreover, Rap1 activation was sustained for hours, an effect that matched the sustained activation of the mitogen-activated protein kinase (MAPK) pathway. To investigate the molecular basis for Rap1 activation, we examined complexes containing C3G, a guanine nucleotide exchange factor for Rap1, and CrkL, an adapter protein known to influence Rap1 signaling. NGF induced the formation of a long-lived complex containing C3G/CrkL/Shp2/Gab2/TrkA. Linking the complex to Rap1 activation, we coprecipitated activated TrkA and activated MAPK with activated Rap1 in NGF-treated cells. Confocal microscopy and subcellular fractionation showed that activated Rap1 and the other proteins of the signaling complex were present in endosomes. Pretreatment of PC12 cells with brefeldin A (BFA), which disrupts the Golgi and endosomal compartments, had little effect on Ras activation but strongly inhibited NGF-induced Rap1 activation and continuing MAPK activation. We propose that endosomes are a site from which NGF induces the prolonged activation of Rap1 and MAPK.


Subject(s)
Adaptor Proteins, Signal Transducing , Endosomes/metabolism , Nerve Growth Factor/pharmacology , Receptor, trkA , Signal Transduction/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Brefeldin A/pharmacology , Carrier Proteins/metabolism , Cell Compartmentation/drug effects , Cell Fractionation , Electrophoresis, Polyacrylamide Gel , Endosomes/drug effects , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Guanine Nucleotide-Releasing Factor 2/metabolism , Helminth Proteins/metabolism , Immunoblotting , MAP Kinase Signaling System/drug effects , Macromolecular Substances , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , PC12 Cells , Phosphoproteins/metabolism , Rats , Signal Transduction/drug effects
12.
J Biol Chem ; 276(14): 11294-301, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11150291

ABSTRACT

The TrkA receptor is activated primarily by nerve growth factor (NGF), but it can also be activated by high concentrations of neurotrophin 3 (NT-3). The pan-neurotrophin receptor p75(NTR) strongly inhibits activation of TrkA by NT-3 but not by NGF. To examine the role of p75(NTR) in regulating the specificity of TrkA signaling, we expressed both receptors in Xenopus oocytes. Application of NGF or NT-3 to oocytes expressing TrkA alone resulted in efflux of (45)Ca(2+) by a phospholipase C-gamma-dependent pathway. Coexpression of p75(NTR) with TrkA inhibited (45)Ca(2+) efflux in response to NT-3 but not NGF. The inhibitory effect on NT-3 activation of TrkA increased with increasing expression of p75(NTR). Coexpression of a truncated p75(NTR) receptor lacking all but the first 9 amino acids of the cytoplasmic domain inhibited NT-3 stimulation of (45)Ca(2+) efflux, whereas coexpression of an epidermal growth factor receptor/p75(NTR) chimera (extracellular domain of epidermal growth factor receptor with transmembrane and cytoplasmic domains of p75(NTR)) did not inhibit NT-3 signaling through TrkA. These studies demonstrated that the extracellular domain of p75(NTR) was necessary to inhibit NT-3 signaling through TrkA. Remarkably, p75(NTR) binding to NT-3 was not required to prevent signaling through TrkA, since occupying p75(NTR) with brain-derived neurotrophic factor or anti-p75 antibody (REX) did not rescue the ability of NT-3 to activate (45)Ca(2+) efflux. These data suggested a physical association between TrkA and p75(NTR). Documenting this physical interaction, we showed that p75(NTR) and TrkA could be coimmunoprecipitated from Xenopus oocytes. Our results suggest that the interaction of these two receptors on the cell surface mediated the inhibition of NT-3-activated signaling through TrkA.


Subject(s)
Neurotrophin 3/metabolism , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Humans , Neurotrophin 3/chemistry , Receptor, Nerve Growth Factor , Receptor, trkA/chemistry , Receptors, Nerve Growth Factor/chemistry , Recombinant Proteins/metabolism , Signal Transduction , Xenopus laevis
13.
J Neurosci ; 20(19): 7325-33, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11007890

ABSTRACT

Neurotrophin (NT) signals may be moved from axon terminals to neuron cell bodies via signaling endosomes-organelles in which NTs continue to be bound to their activated receptors. Suggesting that clathrin-coated membranes serve as one source of signaling endosomes, in earlier studies we showed that nerve growth factor (NGF) treatment increased clathrin at the plasma membrane and resulted in colocalization of clathrin with TrkA, the receptor tyrosine kinase for NGF. Strikingly, however, we also noted that most clathrin puncta at the surface of NGF-treated cells did not colocalize with TrkA, raising the possibility that NGF induces a general increase in clathrin-coated membrane formation. To explore this possibility further, we examined the distribution of clathrin in NGF- and BDNF-treated cells. NGF signaling in PC12 cells robustly redistributed the adaptor protein AP2 and the clathrin heavy chain (CHC) to surface membranes. Using confocal and epifluorescence microscopy, as well as biochemical assays, we showed the redistribution of clathrin to be attributable to the activation of TrkA. Significantly, NGF signaled through TrkA to induce an increase in clathrin-mediated membrane trafficking, as revealed in the increased endocytosis of transferrin. In that BDNF treatment increased AP2 and clathrin at the surface membranes of hippocampal neurons, these findings may represent a physiologically significant response to NTs. We conclude that NT signaling increases clathrin-coated membrane formation and clathrin-mediated membrane trafficking and speculate that this effect contributes to their trophic actions via the increased internalization of receptors and other proteins that are present in clathrin-coated membranes.


Subject(s)
Cell Membrane/metabolism , Clathrin/metabolism , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction/physiology , Adaptor Protein Complex 2 , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cell Membrane/ultrastructure , Cells, Cultured , Clathrin Heavy Chains , Endocytosis/drug effects , Hippocampus/cytology , Hippocampus/metabolism , Humans , Membrane Proteins/metabolism , Mice , Nerve Growth Factor/pharmacology , Neurons/cytology , Neurons/metabolism , PC12 Cells , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley
14.
J Exp Med ; 191(10): 1799-806, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10811872

ABSTRACT

Multiple sclerosis is a demyelinating disorder of the central nervous system (CNS), in which an immune attack directed against myelin constituents causes myelin destruction and death of oligodendrocytes, the myelin-producing cells. Here, the efficacy of nerve growth factor (NGF), a growth factor for neurons and oligodendrocytes, in promoting myelin repair was evaluated using the demyelinating model of experimental allergic encephalomyelitis (EAE) in the common marmoset. Surprisingly, we found that NGF delayed the onset of clinical EAE and, pathologically, prevented the full development of EAE lesions. We demonstrate by immunocytochemistry that NGF exerts its antiinflammatory effect by downregulating the production of interferon gamma by T cells infiltrating the CNS, and upregulating the production of interleukin 10 by glial cells in both inflammatory lesions of EAE and normal-appearing CNS white matter. Thus, NGF, currently under investigation in human clinical trials as a neuronal trophic factor, may be an attractive candidate for therapy of autoimmune demyelinating disorders.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Nerve Growth Factor/pharmacology , Animals , Callithrix , Central Nervous System/drug effects , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunohistochemistry , Recombinant Proteins/pharmacology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology
15.
J Biol Chem ; 274(51): 36707-14, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593976

ABSTRACT

Nerve growth factor (NGF) binding to its receptors TrkA and p75(NTR) enhances the survival, differentiation, and maintenance of neurons. Recent studies have suggested that NGF receptor activation may occur in caveolae or caveolae-like membranes (CLM). This is an intriguing possibility because caveolae have been shown to contain many of the signaling intermediates in the TrkA signaling cascade. To examine the membrane localization of TrkA and p75(NTR), we isolated caveolae from 3T3-TrkA-p75 cells and CLM from PC12 cells. Immunoblot analysis showed that TrkA and p75(NTR) were enriched about 13- and 25-fold, respectively, in caveolae and CLM. Binding and cross-linking studies demonstrated that the NGF binding to both TrkA and p75(NTR) was considerably enriched in CLM and that about 90% of high affinity binding to TrkA was present in CLM. When PC12 cells were treated with NGF, virtually all activated (i.e. tyrosine phosphorylated) TrkA was found in the CLM. Remarkably, in NGF-treated cells, it was only in CLM that activated TrkA was coimmunoprecipitated with phosphorylated Shc and PLCgamma. These results document a signaling role for TrkA in CLM and suggest that both TrkA and p75(NTR) signaling are initiated from these membranes.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Nerve Growth Factor/metabolism , Receptor, Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Animals , PC12 Cells , Phosphorylation , Rats , Signal Transduction
16.
Neurobiol Dis ; 6(5): 321-34, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10527801

ABSTRACT

Batten disease, a degenerative neurological disorder with juvenile onset, is the most common form of the neuronal ceroid lipofuscinoses. Mutations in the CLN3 gene cause Batten disease. To facilitate studies of Batten disease pathogenesis and treatment, a murine model was created by targeted disruption of the Cln3 gene. Mice homozygous for the disrupted Cln3 allele had a neuronal storage disorder resembling that seen in Batten disease patients: there was widespread and progressive intracellular accumulation of autofluorescent material that by EM displayed a multilamellar rectilinear/fingerprint appearance. Inclusions contained subunit c of mitochondrial ATP synthase. Mutant animals also showed neuropathological abnormalities with loss of certain cortical interneurons and hypertrophy of many interneuron populations in the hippocampus. Finally, as is true in Batten disease patients, there was increased activity in the brain of the lysosomal protease Cln2/TPP-1. Our findings are evidence that the Cln3-deficient mouse provides a valuable model for studying Batten disease.


Subject(s)
Hippocampus/pathology , Membrane Glycoproteins , Molecular Chaperones , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/pathology , Proteins/genetics , Animals , Disease Models, Animal , Female , Genotype , Hippocampus/metabolism , Hippocampus/ultrastructure , Humans , Hypertrophy , Interneurons/pathology , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Knockout , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/physiopathology , Neurons/metabolism , Neurons/ultrastructure , Proteins/physiology , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction , Tripeptidyl-Peptidase 1
17.
Exp Neurol ; 159(1): 297-308, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10486198

ABSTRACT

Much more is known about nerve growth factor (NGF) signaling than that initiated by brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), or NT-4. We sought to study early BDNF, NT-3, and NT-4 signaling events. Using TrkB-expressing cells, we found that BDNF and NT-4 individually induced tyrosine phosphorylation of TrkB in a dose-dependent fashion. At maximally effective concentrations, BDNF or NT-4 induced robust TrkB tyrosine phosphorylation at 5 min; this progressively declined at 15, 30, and 60 min. Using immunoprecipitation, PI3-kinase and tyrosine phosphorylated PLC-gamma1 and SHC were shown to be associated with tyrosine phosphorylated TrkB in response to both BDNF and NT-4. BDNF and NT-4 induced similar intensities of phosphorylation of TrkB and signaling intermediates at equivalent doses. NT-3 treatment of TrkC-expressing cells induced very similar patterns for induction of TrkC tyrosine phosphorylation and recruitment of signaling intermediates. BDNF, NT-3, and NT-4 caused rapid tyrosine phosphorylation of ERK and SNT. These data suggest that the earliest signaling events for BDNF, NT-3, and NT-4 are very similar to those for NGF.


Subject(s)
Mitogen-Activated Protein Kinases , Nerve Growth Factors/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , 3T3 Cells/chemistry , 3T3 Cells/physiology , Adaptor Proteins, Signal Transducing , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Gene Expression/physiology , Humans , Isoenzymes/metabolism , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinase 3 , Neuroprotective Agents/pharmacology , Neurotrophin 3 , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma , Phosphoproteins/metabolism , Phosphorylation , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Ciliary Neurotrophic Factor , Receptor, trkC , Receptors, Nerve Growth Factor/metabolism , Transfection , Type C Phospholipases/metabolism , Tyrosine/metabolism , src Homology Domains/physiology
18.
Cell ; 96(5): 677-87, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10089883

ABSTRACT

Epidermal growth factor (EGF) binding to its receptor causes rapid phosphorylation of the clathrin heavy chain at tyrosine 1477, which lies in a domain controlling clathrin assembly. EGF-mediated clathrin phosphorylation is followed by clathrin redistribution to the cell periphery and is the product of downstream activation of SRC kinase by EGF receptor (EGFR) signaling. In cells lacking SRC kinase, or cells treated with a specific SRC family kinase inhibitor, EGF stimulation of clathrin phosphorylation and redistribution does not occur, and EGF endocytosis is delayed. These observations demonstrate a role for SRC kinase in modification and recruitment of clathrin during ligand-induced EGFR endocytosis and thereby define a novel effector mechanism for regulation of endocytosis by receptor signaling.


Subject(s)
Clathrin/metabolism , Endocytosis/drug effects , Epidermal Growth Factor/metabolism , ErbB Receptors/physiology , Protein Processing, Post-Translational/drug effects , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Biological Transport , Cattle , Humans , Ligands , Mice , Molecular Sequence Data , Phosphorylation/drug effects , Proto-Oncogene Proteins pp60(c-src)/metabolism , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
19.
J Neurosci ; 19(7): 2556-67, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10087069

ABSTRACT

The neuronal ceroid lipofuscinoses (NCL) are progressive neurodegenerative disorders with onset from infancy to adulthood that are manifested by blindness, seizures, and dementia. In NCL, lysosomes accumulate autofluorescent proteolipid in the brain and other tissues. The mnd/mnd mutant mouse was first characterized as exhibiting adult-onset upper and lower motor neuron degeneration, but closer examination revealed early, widespread pathology similar to that seen in NCL. We used the autofluorescent properties of accumulated storage material to map which CNS neuronal populations in the mnd/mnd mouse show NCL-like pathological changes. Pronounced, early accumulation of autofluorescent lipopigment was found in subpopulations of GABAergic neurons, including interneurons in the cortex and hippocampus. Staining for phenotypic markers normally present in these neurons revealed progressive loss of staining in the cortex and hippocampus of mnd/mnd mice, with pronounced hypertrophy of remaining detectable interneurons. In contrast, even in aged mutant mice, many hippocampal interneurons retained staining for glutamic acid decarboxylase. Treatment with insulin-like growth factor-1 partially restored interneuronal number and reduced hypertrophy in some subregions. These results provide the first evidence for the involvement of interneurons in a mouse model of NCL. Moreover, our findings suggest that at least some populations of these neurons persist in a growth factor-responsive state.


Subject(s)
Insulin-Like Growth Factor I/therapeutic use , Interneurons/physiology , Neuronal Ceroid-Lipofuscinoses/drug therapy , Aging/pathology , Animals , Atrophy , Cerebral Cortex/pathology , Cerebral Ventricles , Fluorescence , Hypertrophy/drug therapy , Hypertrophy/pathology , Infusions, Parenteral , Mice , Mice, Inbred C57BL , Nerve Degeneration , Neuronal Ceroid-Lipofuscinoses/pathology , Parvalbumins/analysis , Phenotype
20.
Biochemistry ; 37(48): 16846-52, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9836577

ABSTRACT

Neurotrophin-3 (NT-3) is a cystine knot growth factor that promotes the survival, proliferation, and differentiation of developing neurons and is a potential therapeutic for neurodegenerative diseases. To clarify the structural basis of receptor specificity and the role of neurotrophin dimerization in receptor activation, the structure of the NT-3 homodimer was determined using X-ray crystallography. The orthorhombic crystals diffract to 2.4 A, with dimer symmetry occurring about a crystallographic 2-fold axis. The overall structure of NT-3 resembles that of the other neurotrophins, NGF and BDNF; each protomer forms a twisted four-stranded beta sheet, with three intertwined disulfide bonds. There are notable differences, however, between NT-3 and NGF in the surface loops and in three functionally important regions, shown in previous mutagenesis studies to be critical for binding. One such difference implies that NT-3's binding affinity and specificity depend on a novel hydrogen bond between Gln 83, a residue important for binding specificity with TrkC, and Arg 103, a residue crucial for binding affinity with TrkC. NT-3's extensive dimer interface buries much of the otherwise solvent-accessible hydrophobic surface area and suggests that the dimeric state is stabilized through the formation of this hydrophobic core. A comparison of the dimer interface between the NT-3 homodimer and the BDNF/NT-3 heterodimer reveals similar patterns of hydrogen bonds and nonpolar contacts, which reinforces the notion that the evolutionarily conserved neurotrophin interface resulted from the need for receptor dimerization in signal initiation.


Subject(s)
Nerve Growth Factors/chemistry , Amino Acid Sequence , Computer Simulation , Crystallography, X-Ray , Dimerization , Humans , Models, Molecular , Molecular Sequence Data , Nerve Growth Factors/metabolism , Neurotrophin 3 , Protein Conformation , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/chemistry , Receptors, Nerve Growth Factor/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...